Discovering and exploiting bacterial proteins as...

111
UNIVERSIDADE DE LISBOA FACULDADE DE CIÊNCIAS DEPARTAMENTO DE BIOLOGIA ANIMAL Discovering and exploiting bacterial proteins as anticancer agents Gonçalo Emanuel Fialho Mourata da Silva DISSERTAÇÃO Mestrado em Biologia Humana e Ambiente 2013

Transcript of Discovering and exploiting bacterial proteins as...

Page 1: Discovering and exploiting bacterial proteins as ...repositorio.ul.pt/bitstream/10451/9665/1/ulfc103147_tm_goncalo_sil… · Discovering and exploiting bacterial proteins as anticancer

UNIVERSIDADE DE LISBOA

FACULDADE DE CIÊNCIAS

DEPARTAMENTO DE BIOLOGIA ANIMAL

Discovering and exploiting bacterial

proteins as anticancer agents

Gonçalo Emanuel Fialho Mourata da Silva

DISSERTAÇÃO

Mestrado em Biologia Humana e Ambiente

2013

Page 2: Discovering and exploiting bacterial proteins as ...repositorio.ul.pt/bitstream/10451/9665/1/ulfc103147_tm_goncalo_sil… · Discovering and exploiting bacterial proteins as anticancer
Page 3: Discovering and exploiting bacterial proteins as ...repositorio.ul.pt/bitstream/10451/9665/1/ulfc103147_tm_goncalo_sil… · Discovering and exploiting bacterial proteins as anticancer

UNIVERSIDADE DE LISBOA

FACULDADE DE CIÊNCIAS

DEPARTAMENTO DE BIOLOGIA ANIMAL

Discovering and exploiting bacterial

proteins as anticancer agents

Gonçalo Emanuel Fialho Mourata da Silva

DISSERTAÇÃO

Mestrado em Biologia Humana e Ambiente

Dissertação orientada por Professor Doutor Arsénio Fialho e

Professora Doutora Ana Crespo

2013

Page 4: Discovering and exploiting bacterial proteins as ...repositorio.ul.pt/bitstream/10451/9665/1/ulfc103147_tm_goncalo_sil… · Discovering and exploiting bacterial proteins as anticancer
Page 5: Discovering and exploiting bacterial proteins as ...repositorio.ul.pt/bitstream/10451/9665/1/ulfc103147_tm_goncalo_sil… · Discovering and exploiting bacterial proteins as anticancer

I

"To myself I am only a child playing on the beach, while vast oceans of truth lie

undiscovered before me."

Isaac Newton

"I am not apt to follow blindly the lead of other men."

Charles Darwin

Page 6: Discovering and exploiting bacterial proteins as ...repositorio.ul.pt/bitstream/10451/9665/1/ulfc103147_tm_goncalo_sil… · Discovering and exploiting bacterial proteins as anticancer
Page 7: Discovering and exploiting bacterial proteins as ...repositorio.ul.pt/bitstream/10451/9665/1/ulfc103147_tm_goncalo_sil… · Discovering and exploiting bacterial proteins as anticancer

III

ACKNOWLEDGMENTS

I would like to express my sincere gratitude to my supervisor Professor

Arsénio Fialho (Instituto Superior Técnico), for having granted me this

opportunity to work with him and for all the help and support that he gave me,

every time I needed, throughout my thesis.

I am also grateful to Professor Ana Crespo, at the Departamento de Biologia

Animal (Faculdade de Ciências, Universidade de Lisboa), for having gently

accepted to be my supervisor and for being always available whenever I needed.

I would also like to thank Nuno Bernardes, for all the knowledge transmitted

and for teaching me everything I have learned inside a laboratory this year.

In addition, I am thankful to the people in the Biological Sciences Research

Group (Instituto Superior Técnico), especially to Dalila Mil-Homens and Sofia

Abreu which helped me in many occasions.

I sincerely want to address a special thanks to my closest friends Alexsandro

Costa, João Guerreiro, Duarte Silva, João Serafim, Ana Catarina, Andreia

Ferreira, for always been there, for giving me a kind word, for listening, for

making me laugh, and specially for believing in me and my capabilities. I would

like to extend this acknowledge also with my friends from my master degree

course, especially to Sofia Alves.

I especially want to thank Tânia, for being my fountain of joy and happiness,

and for having the amazing ability of making me smile even in the toughest times.

I would also like to thank my family, especially my parents and my brother, for

all their support, patience and strength they gave me through this entire year. I

could not have finished my master degree without them.

I dedicate the achievement of my master degree to my grandparents José and

Mariana, for being the reason why I came to science and whose memories remain

strictly in my heart and still give me the strength to overcome my fears and

difficulties.

Page 8: Discovering and exploiting bacterial proteins as ...repositorio.ul.pt/bitstream/10451/9665/1/ulfc103147_tm_goncalo_sil… · Discovering and exploiting bacterial proteins as anticancer
Page 9: Discovering and exploiting bacterial proteins as ...repositorio.ul.pt/bitstream/10451/9665/1/ulfc103147_tm_goncalo_sil… · Discovering and exploiting bacterial proteins as anticancer

V

ABSTRACT

Azurin is a low molecular weight protein, produced by Pseudomonas

aeruginosa and possesses several antitumor properties, like causing P-cadherin

levels to decrease in invasive breast cancer cells. In this work, we studied the

effect of lysosome and proteasome inhibitors on P-cadherin level, using a breast

cancer cell line, expressing high P-cadherin level (MCF-7/AZ.Pcad), previously

treated with azurin. Additionally, we evaluated how a cholesterol-depleting agent

(MβCD) affects P-cadherin level. The effects of both inhibitors on P-cadherin

were observed by western blot and confirmed that azurin mediates P-cadherin

degradation through lysosome and proteasome proteolytic pathways. We also

described, for the first time, that MβCD causes P-cadherin level to decrease.

Together, these findings have increased our understanding of how the bacterial

protein azurin is acting as anti-cancer agent.

In this work we have also studied the in vitro cytotoxicity of two other bacterial

proteins (MPT 63 and Ndk) against human breast and lung cancer cells. MPT 63

is an antigen secreted by Mycobacterium tuberculosis that induces immunogenic

responses in animal models and its cytotoxicity against several tumor cell lines

was recently described in a patent. Nucleoside diphosphate kinase (Ndk) is a

ubiquitous enzyme which maintains the nucleotide pools within the cells, and can

be secreted by P. aeruginosa. A human Ndk, termed Nm23-H1, also showed an

anti-metastatic role in different cancer models. In order to test possible antitumor

properties of these proteins, MTT cell viability assays were performed in breast

and lung cancer models (MCF-7/AZ.Mock and A549) using increasing azurin,

MPT 63 and Ndk concentrations, and different exposure times. In addition,

matrigel invasion assay was performed in A549 invasive cells treated with Ndk.

Both azurin, MPT 63 and Ndk evidenced cytotoxicity against both cancer models

in a time and dose dependent manner. Ndk revealed cytotoxic activity and

selectivity against tumor cells similar to azurin. We observed a small decrease in

cell invasion using this protein. In summary, we promoted a screening of new

bacterial proteins that demonstrated antitumor potential, especially Ndk.

Keywords: Azurin, MPT 63, Ndk, Cancer, P-cadherin.

Page 10: Discovering and exploiting bacterial proteins as ...repositorio.ul.pt/bitstream/10451/9665/1/ulfc103147_tm_goncalo_sil… · Discovering and exploiting bacterial proteins as anticancer
Page 11: Discovering and exploiting bacterial proteins as ...repositorio.ul.pt/bitstream/10451/9665/1/ulfc103147_tm_goncalo_sil… · Discovering and exploiting bacterial proteins as anticancer

VII

RESUMO

Novas terapias anti-tumorais emergentes baseiam-se em abordagens pouco

convencionais, como a utilização de microorganismos, nomeadamente bactérias

vivas ou produtos purificados a partir das mesmas, como proteínas. A azurina é

uma proteína de baixo peso molecular, produzida por Pseudomonas aeruginosa e

possui diversas propriedades anti-tumorais, entre as quais a indução de apoptose

em células tumorais pela estabilização da proteína supressora de tumores p53.

Mais recentemente, um novo tipo de acção anti-tumoral foi descoberta, tendo sida

descrita a sua capacidade de diminuir os níveis de P-caderina em células tumorais

invasivas de cancro da mama, sem afectar, no entanto, os níveis de E-caderina. O

mecanismo, pelo qual a azurina causa o decréscimo de P-caderina nas células

tumorais não é ainda totalmente conhecido, mas esta parece actuar a nível pós-

transcripcional dado que não se verificam diferenças na expressão de P-caderina

em células tratadas com azurina. Dados relativos a ensaios com análise a

microarrays revelaram que a transcrição de genes associados ao lisossoma e

processos de transporte mediado por vesículas se encontrava mais activa. No

presente trabalho pretendeu-se esclarecer se a diminuição dos níveis de P-

caderina, mediada pela acção da azurina, se deve à sua degradação pelos sistemas

proteolíticos a nível celular, como o lisossoma e proteossoma. Nesse sentido

utilizaram-se células de uma linha celular de cancro da mama, que expressa níveis

elevados de P-caderina (MCF-7/AZ.Pcad), e que foram previamente tratadas com

azurina antes de serem administrados inibidores de lisossoma (cloreto de amónio)

e de proteossoma (MG-132). Do mesmo modo foram também avaliados os efeitos

de um agente sequestrador de colesterol (MβCD) e inibidor de entrada da azurina

nas células, ao nível da P-caderina nesta linha celular tumoral. Os efeitos de

ambos os inibidores, ao nível da degradação da P-caderina, foram observados por

western blot e confirmaram que a azurina medeia a degradação da P-caderina por

sistemas proteolíticos como o lisossoma e o proteossoma. Descrevemos

igualmente, pela primeira vez, que a MβCD provoca a diminuição dos níveis de

P-caderina sem afectar os níveis de E-caderina. Conjuntamente, estes resultados

permitiram aumentar o nosso conhecimento acerca do modo como a azurina actua

como agente anticancerígeno neste caso específico.

Page 12: Discovering and exploiting bacterial proteins as ...repositorio.ul.pt/bitstream/10451/9665/1/ulfc103147_tm_goncalo_sil… · Discovering and exploiting bacterial proteins as anticancer

VIII

Neste trabalho pretendemos também estudar a citotoxicidade in vitro de duas

outras proteínas bacterianas (MPT 63 e Ndk) em células tumorais humanas de

cancro da mama e de pulmão. A MPT 63 é uma proteína antigénica secretada por

Mycobacterium tuberculosis e capaz de induzir respostas imunogénicas em

diversos modelos animais. Esta proteína apresenta uma estrutura semelhante a

imunoglobulinas, uma característica que é partilhada com a azurina.

Recentemente foi descrita como possuindo elevada actividade citotóxica contra

várias linhas celulares tumorais, assim como um péptido derivado desta proteína

(MB30), tendo esta propriedade de ambas as molécula sido registada numa

patente. A nucleosídeo difosfato cinase (Ndk) é uma enzima ubíqua em diversos

organismos e que tem como função manter as reservas de nucleótidos das células.

Esta proteína pode igualmente ser secretada por várias bactérias como P.

aeruginosa. As Ndks humanas estão agrupadas numa família de proteínas

denominada de Nm23, tendo sidas até hoje descritas dez tipos. A primeira destas

proteínas a ser descrita, denominada Nm23-H1, demonstrou possuir

adicionalmente uma importante acção anti-metastática em diferentes modelos de

cancro. Tendo em conta o vasto leque de acção anti-tumoral da azurina, procurou-

se seleccionar duas proteínas bacterianas (MPT 63 e Ndk) com propriedades

interessantes de serem exploradas, no sentido de testar uma possível actividade

citotóxica das mesmas em células cancerígenas. Para esse efeito foram realizados

ensaios de viabilidade celular (ou ensaios de MTT) em modelos tumorais de

cancro da mama e do pulmão (MCF-7/AZ.Mock e A549), usando concentrações

crescentes de azurina, MPT 63 e Ndk, bem como diferentes tempos de exposição,

com o intuito de entender como estes parâmetros podem afectar o nível de

citotoxicidade destas proteínas. Adicionalmente foi testada a actividade anti-

metastática da Ndk, realizando um ensaio de invasão em matrigel, usando uma

linha celular altamente invasiva de cancro de pulmão, A549. A azurina, assim

como a MPT 63 e a Ndk, evidenciaram citotoxicidade contra ambos os modelos

tumorais testados, de um modo dependente do tempo e concentrações

administradas. A Ndk revelou níveis de actividade citotóxica e selectividade de

acção, relativamente a células tumorais, semelhantes à azurina. Observámos ainda

um pequeno decréscimo da invasão celular das células tumorais de pulmão A549,

Page 13: Discovering and exploiting bacterial proteins as ...repositorio.ul.pt/bitstream/10451/9665/1/ulfc103147_tm_goncalo_sil… · Discovering and exploiting bacterial proteins as anticancer

IX

quando esta proteína foi administrada. Em suma, promovemos um rastreio de

novas proteínas bacterianas que demonstraram potencial anti-tumoral,

especialmente a Ndk. O conhecimento acerca destas propriedades necessita de ser

expandido e aprofundado para que, no futuro, se possa avaliar a sua utilização

como agentes anti-cancerígenos úteis, tal como a azurina.

Palavras-chave: Azurina, MPT 63, Ndk, Cancro, P-caderina.

Page 14: Discovering and exploiting bacterial proteins as ...repositorio.ul.pt/bitstream/10451/9665/1/ulfc103147_tm_goncalo_sil… · Discovering and exploiting bacterial proteins as anticancer
Page 15: Discovering and exploiting bacterial proteins as ...repositorio.ul.pt/bitstream/10451/9665/1/ulfc103147_tm_goncalo_sil… · Discovering and exploiting bacterial proteins as anticancer

XI

TABLE OF CONTENTS

ACKNOWLEDGMENTS .................................................................................... III

ABSTRACT ............................................................................................................ V

RESUMO ............................................................................................................. VII

TABLE OF CONTENTS ...................................................................................... XI

LIST OF ABBREVIATIONS ............................................................................. XIII

INDEX OF FIGURES ......................................................................................... XV

INDEX OF TABLES ......................................................................................... XIX

1. INTRODUCTION ........................................................................................... 1

1.1 Cancer ............................................................................................................ 1

1.2 Microorganisms and their products as anti-cancer agents ...................... 2

1.3 Azurin ............................................................................................................ 4

1.4 Cadherins .................................................................................................... 10

1.5 Azurin and P-cadherin interactions ......................................................... 13

1.6 Inhibitors ..................................................................................................... 14

1.7 MPT 63........................................................................................................ 16

1.8 Nucleoside diphosphate kinase ................................................................ 19

2. OBJECTIVES ................................................................................................ 25

3. MATERIALS AND METHODS................................................................... 27

3.1 Bacterial proteins superexpression .......................................................... 27

3.1.1 Bacterial strains and plasmids ......................................................... 27

3.1.2 Inoculum ......................................................................................... 27

3.1.3 Cell sonication ................................................................................. 28

3.1.4 Azurin purification .......................................................................... 28

3.1.5 MPT 63 and Ndk purification ......................................................... 30

3.2 Cell culture and human cell lines ............................................................. 31

3.3 Inhibitors treatment ................................................................................... 32

3.3.1 Lysosome and proteasome inhibitors .............................................. 32

3.3.2 Azurin internalization inhibitor ....................................................... 32

3.4 Protein lysates ............................................................................................ 33

3.5 Sodium dodecyl sulfate polyacrylamide gel electrophoresis (SDS-

PAGE) 33

Page 16: Discovering and exploiting bacterial proteins as ...repositorio.ul.pt/bitstream/10451/9665/1/ulfc103147_tm_goncalo_sil… · Discovering and exploiting bacterial proteins as anticancer

XII

3.6 Western blotting ......................................................................................... 34

3.7 MTT cell viability assay ............................................................................ 35

3.8 Matrigel invasion assay ............................................................................. 36

3.9 Bioinformatics ............................................................................................ 36

3.9.1 Sequence analysis and secondary structures ................................... 36

3.9.2 Phylogenetic and structural alignment analysis............................... 37

4. RESULTS....................................................................................................... 39

4.1 Proteolytic pathways inhibitors effect on P-cadherin level .................. 40

4.2 Azurin internalization ................................................................................ 42

4.3 Inhibition of azurin’s effect on E and P-cadherins level using MβCD

44

4.4 Bacterial proteins to treat cancer .............................................................. 45

4.4.1 Purified proteins .............................................................................. 45

4.5 MTT cell viability assays .......................................................................... 46

4.5.1 Azurin .............................................................................................. 47

4.5.2 MPT 63 ............................................................................................ 48

4.5.3 Ndk .................................................................................................. 49

4.6 Matrigel invasion assay ............................................................................. 51

4.7 Bioinformatic analysis on human Nm23 and bacterial Ndks ............... 51

5. DISCUSSION ................................................................................................ 65

6. CONCLUSION .............................................................................................. 73

7. REFERENCES ............................................................................................... 77

Page 17: Discovering and exploiting bacterial proteins as ...repositorio.ul.pt/bitstream/10451/9665/1/ulfc103147_tm_goncalo_sil… · Discovering and exploiting bacterial proteins as anticancer

XIII

LIST OF ABBREVIATIONS

BCG - Bacille Calmette-Guérin

BSA - Bovine serum albumin

DMBA - Dimethyl-benz-anthracene

DMSO - Dimethyl sulfoxide

E1 - Ubiquitin-activating enzyme

E2 - Ubiquitin-conjugating enzyme

E3 - Ubiquitin-ligase enzyme

ELISA - Enzyme-linked immunosorbent assay

Eph - Ephrin receptor

FBS - Fetal bovine serum

HBS - Hepes buffered saline

HLA-DR - Human Leucocyte Antigen

HUVEC - Human umbilical vein endothelial cells

INF-γ - Gamma interferon

IPTG - Isopropyl-β-D-Thiogalactopyranoside

KSR - Kinase suppressor of ras

Laz - Lipid-modified azurin

LB - Luria Broth

LPS - Lipopolysaccharide

MEM - Minimum Essential Medium

MMP - Matrix metalloproteases

MTT - 3-(4,5 dimethylthiazol-2-yl-2,5 tetrazolium bromide)

MβCD - Methyl-β-cyclodextrin

NaCl - Sodium chloride

NCBI - National Center for Biotechnology Information

Ndk - Nucleoside diphosphate kinase

NDPs - Nucleoside diphosphates

NH4Cl – Ammonium chloride

NSCLC - Non-small cell lung cancer

NTPs - Nucleoside triphosphates

OD640 nm - Optical density

Page 18: Discovering and exploiting bacterial proteins as ...repositorio.ul.pt/bitstream/10451/9665/1/ulfc103147_tm_goncalo_sil… · Discovering and exploiting bacterial proteins as anticancer

XIV

PBMNCs - Peripheral blood mononuclear cells

PBS - Phosphate buffered saline

PDB - Protein Data Bank

PTD - Protein transduction domain

PVP - Polyvinylpyrrolidone

SCLC - Small cell lung cancer

SDS-PAGE - Sodium dodecyl sulfate polyacrylamide gel electrophoresis

sP-cad - Soluble P-cadherin fragment

STRAP - Serine-threonine kinase receptor-associated protein

SURE - Stop Unwanted Rearrangement Events

Th1 - T helper 1

UPS - Ubiquitin-proteasome system

Page 19: Discovering and exploiting bacterial proteins as ...repositorio.ul.pt/bitstream/10451/9665/1/ulfc103147_tm_goncalo_sil… · Discovering and exploiting bacterial proteins as anticancer

XV

INDEX OF FIGURES

Figure 1 – Current diverse strategies to use bacteria and bacterial products in

cancer treatment (Bernardes et al, 2013) [11]. ........................................................ 3

Figure 2 – The three-dimensional structure of azurin from P. aeruginosa

(Bonander et al, 1997) [18]. .................................................................................... 4

Figure 3 – Azurin as a promiscuous protein, which possess both anticancer,

antiparasite and antiviral activities (Fialho et al, 2007) [37]. ............................... 10

Figure 4 – Schematic representation of the classical cadherin-catenin complex

and the structural components of cadherins (adapted from Albergaria et al, 2011)

[40]. ....................................................................................................................... 11

Figure 5– Structure of M. tuberculosis MPT 63 protein (adapted from Goulding

et al, 2002) [58] ..................................................................................................... 17

Figure 6 – Three-dimensional structures of human Ndk-A. (A) Diagram of Ndk-

A monomer. (B) Diagram of the homo-hexamer (Han et al, 2010) [79]. ............. 22

Figure 7 – (A): Western Blot analysis of the effect of lysossome inhibitor,

ammonium chloride (NH4Cl), on E and P-cadherins of MCF-7/AZ.Pcad cells,

previously treated with azurin at 0, 50 and 100 μM. Azurin and the inhibitor were

administrated at 0 and 32 hours, respectively, in a 48 hours assay. (B): Charts

illustrated represent average percentage values of protein level for E and P-

cadherins which signal was normalized with actin levels. .................................... 41

Figure 8 – (A): Western Blot analysis of the effect of proteasome inhibitor, MG-

132, on E and P-cadherins of MCF-7/AZ.Pcad cells, previously treated with

azurin at 0, 50 and 100 μM. Azurin and the inhibitor were administrated at 0 and

32 hours, respectively, in a 48 hours assay. (B): Charts illustrated represent

average percentage values of protein level for E and P-cadherins which signal was

normalized with actin levels.................................................................................. 42

Figure 9 - Effects of MβCD on azurin’s internalization on MCF-7/AZ.Pcad cells

after 8 hours of exposure to this inhibitor and 24 hours with fresh medium

(visualized by western blot). PBS was used as double negative control. DMSO

was used as negative control. Azurin was tested at two different concentrations

(50 and 100 μM). ................................................................................................... 43

Page 20: Discovering and exploiting bacterial proteins as ...repositorio.ul.pt/bitstream/10451/9665/1/ulfc103147_tm_goncalo_sil… · Discovering and exploiting bacterial proteins as anticancer

XVI

Figure 10 – Western blot showing the effect of azurin’s internalization inhibitor

MβCD on E- and P-cadherin levels in MCF-7/AZ.Pcad cells after 8 hours of

exposure and 24 hours with fresh medium. PBS was used as double negative

control. DMSO was used as negative control. A decrease in P-cadherin level,

between control samples and samples treated with MβCD and azurin, is shown in

the figure. ............................................................................................................... 45

Figure 11 - 15% SDS-PAGE of MPT 63 and Ndk purified from E. coli SURE

strain. Proteins appear as a single band at approximately 15 kDa, according to

their expected molecular weight. ........................................................................... 46

Figure 12 – Cytotoxicity (%) caused by azurin on MCF-7/AZ.Mock during 48 h

(one dose) and 72 h (three doses), and A549 48 h (one and two doses). Both cell

lines were tested with 5 different azurin concentrations (0, 10, 25, 50, and 100

μM). Significant values, p-value<0.05 with the Student t-test, are shown as

asterisks (*). ........................................................................................................... 48

Figure 13 – Cytotoxicity (%) caused by MPT 63 on MCF-7/AZ.Mock during 48

h (one dose) and 72 h (three doses), and A549 during 48 h (two doses). Both cell

lines were tested with 5 different MPT 63 concentrations (0, 10, 25, 50, and 100

μM). ....................................................................................................................... 49

Figure 14 - Cytotoxicity (%) caused by Ndk on MCF-7/AZ.Mock during 48 h

(two doses) and 72 h (three doses), A549 48 h (two doses) and 16HBE14o- during

48 h (two doses). Both cell lines were tested with 5 different Ndk concentrations

(0, 10, 25, 50, and 100 μM). Significant values, p-value<0.05 with the Student t-

test, are shown as asterisks (*). ............................................................................. 50

Figure 15 – Cell invasion (%) in a Matrigel invasion assay performed on A549

highly invasive tumor cells, which were treated with Ndk at 0, 50 and 100 μM,

during 24 hours. ..................................................................................................... 51

Figure 16 – ClustalW multiple alignment of human and bacterial Ndks. The

numbers above the sequences indicate the position of amino acid residues.

Secondary structure of E. coli Ndk is represented according to PDB code 2HUR.

Secondary elements are indicate as: single purple curves (turns), yellow arrows

(β-sheets), red lines (bends), blue curved lines (alpha helices), orange curved lines

(3/10-helices), and black lines (no secondary structure assigned). NCBI accession

Page 21: Discovering and exploiting bacterial proteins as ...repositorio.ul.pt/bitstream/10451/9665/1/ulfc103147_tm_goncalo_sil… · Discovering and exploiting bacterial proteins as anticancer

XVII

number for each protein is given below. Human (Homo sapiens) Nm23 proteins:

Nm23-H1 (CAG46912), Nm23-H2 (NP_001018147), Nm23-H3 (EAW85629),

Nm23-H4 (NP_005000), Nm23-H5 (NP_003542), Nm23-H6 (NP_005784),

Nm23-H7 (Q9Y5B8), Nm23-H7B (NP_932076), Nm23-H8 (AAF20909), Nm23-

H9 (NP_835231), Nm23-H10 (NP_008846). Bacterial Ndks: P. aeruginosa

(EPR01938), M. tuberculosis (EQM19968), M. bovis (AGE68451), E. coli

(ERF95659), M. xanthus (P15266), V. cholerae (WP_001162853). .................... 58

Figure 17 – Sequence alignment and secondary structure comparison between

human Nm23-H1 and P. aeruginosa Ndk. Symbols in Clustal Consensus

sequence indicate standard ClustalW nomenclature: (*) identity, (:) high

conservation and (.) conservation. Secondary structure of Nm23-H1 was

represented according to PDB code 4ENO. Secondary structure of P. aeruginosa

Ndk was predicted using PDB code 3VGU as template. This template was aligned

between its 2 to 141 amino acid residues and share a 79% sequence identity with

P. aeruginosa Ndk. Secondary elements are indicate as: single purple curves

(turns), yellow arrows (β-sheets), red lines (bends), blue curved lines (alpha

helices), orange curved lines (3/10-helices), and black lines (no secondary

structure assigned). NCBI accession number for each protein is given below.

Human (Homo sapiens) Nm23-H1 (CAG46912) and P. aeruginosa Ndk

(EPR01938). .......................................................................................................... 61

Figure 18 – Phylogenetic tree representation of human Nm23 protein family as

well as some bacterial Ndks. Splitstree software was used to construct the tree

based on ClustalW multiple alignment generated. All sequences and its NCBI

accession numbers used were the same as those displayed in Figure 16. Protein

three-dimensional structures represented were retrieved from PDB database:

Nm23-H1 (4ENO), Nm23-H2 (3BBF), Nm23-H3 (1ZS6), Nm23-H4 (1EHW),

Nm23-H10 (2BX6), and Ndks from M. tuberculosis (1K44), E. coli (2HUR) and

M. xanthus (2NCK). Quaternary structures are represented as homohexamers

(Nm23-H1, Nm23-H2, Nm23-H3 and Nm23-H4 and Ndk from M. tuberculosis)

and as homotetramers (Ndk from E. coli and M. xanthus). Tertiary structure is

represented as a monomer (Nm23-H10). Ndk from P. aeruginosa is marked with

an asterisk (*) since it is a predicted three-dimensional model created using Swiss-

Page 22: Discovering and exploiting bacterial proteins as ...repositorio.ul.pt/bitstream/10451/9665/1/ulfc103147_tm_goncalo_sil… · Discovering and exploiting bacterial proteins as anticancer

XVIII

model software tools. This representation was modeled using 2 to 141 amino acid

residues of the Ndk sequence (79% identity) from Halomonas sp. 593 (3VGU) as

template. This template forms a homodimer in its quaternary structure, as well as

the model created. .................................................................................................. 62

Figure 19 – Structural alignment between human Nm23-H1 (4ENO) [blue] and P.

aeruginosa Ndk predicted model (green) was performed using PyMOL software.

Three dimensional structures are displayed at different degrees: 0º (A), 90º (B),

180º (C) and 270º (D). ........................................................................................... 63

Page 23: Discovering and exploiting bacterial proteins as ...repositorio.ul.pt/bitstream/10451/9665/1/ulfc103147_tm_goncalo_sil… · Discovering and exploiting bacterial proteins as anticancer

XIX

INDEX OF TABLES

Table 1 - Phosphate buffer 8x composition .......................................................... 29

Table 2 - START and elution buffer compositions .............................................. 29

Table 3 - ÄKTA elution program for protein sample desalting ........................... 29

Table 4 - PBS composition ................................................................................... 30

Table 5 - Resolving Gel 8% and 15% composition for one gel ........................... 34

Table 6 – Percent identity matrix of human Nm23 proteins and different bacterial

Ndks. ..................................................................................................................... 59

Page 24: Discovering and exploiting bacterial proteins as ...repositorio.ul.pt/bitstream/10451/9665/1/ulfc103147_tm_goncalo_sil… · Discovering and exploiting bacterial proteins as anticancer
Page 25: Discovering and exploiting bacterial proteins as ...repositorio.ul.pt/bitstream/10451/9665/1/ulfc103147_tm_goncalo_sil… · Discovering and exploiting bacterial proteins as anticancer

1

1. INTRODUCTION

1.1 Cancer

Currently cancer represents one of the greatest burdens of our society and will

remain a serious and challenging major public health problem in the future years.

Each year about 12,7 million people are diagnosed with cancer, and

approximately 7,6 million die from it, clearly demonstrating the magnitude of this

disease in human population [1].

During the last decades lung cancer has been the most common cancer in the

world, as well as the most common cancer in men [1], [2]. This type of cancer

alone is accountable for 1,4 million deaths each year (18,2% of overall cancers),

being unveiled 1,6 million new cases annually [1]. Among women, lung cancer is

the fourth most frequent, as well as the second most common cause of death from

cancer [1]. Regarding its histopathologic classification, lung cancer can be divided

in 2 major types: Small cell lung cancer (SCLC) and Non-small cell lung cancer

(NSCLC), which comprises 3 subtypes (adenocarcinoma, squamous cell

carcinoma and large cell carcinoma) and accounts for more than 85% of all lung

cancers [3], [4]. NSCLC is typically chemo-resistant and treated primarily by

surgery at early stages, while SCLC progresses more rapidly and metastasizes

earlier than NSCLC, being usually treated by chemotherapy and radiotherapy [3],

[4].

Breast cancer is the second most common cancer worldwide, with 1,4 million

cases being diagnosed annually, and the first cause of cancer-related death among

women (458,000 deaths/year) [1]. This type of cancer changes the size and/or

shape of the breast and can be classified into 2 histopathological categories: ductal

and lobular carcinomas. Each one of these carcinomas can be designated as in situ

or invasive, according to whether the tumor is confined to the glandular area of

the organ or whether it has invaded the stroma [5]. Ductal carcinoma represents

80% of breast cancer cases and it arises from epithelial lining the mammary ducts,

whereas lobular carcinoma is a less common form of breast cancer, that is

originated in the milk-producing lobules of the breast [5], [6].

Page 26: Discovering and exploiting bacterial proteins as ...repositorio.ul.pt/bitstream/10451/9665/1/ulfc103147_tm_goncalo_sil… · Discovering and exploiting bacterial proteins as anticancer

2

Current cancer treatments rely on surgery, chemo and radiotherapy, or even

hormone therapy, in the case of breast cancer [5]. However these therapies can

reveal serious and systemic side-effects in patient’s health due to its high toxicity

and lack of cancerous tissue specificity [7]. Additionally not every patient

responds efficiently to chemotherapy or other treatments, since cancer cells can

undergo micro-evolution and rapidly render cancer cells resistant to drug therapy

[8], [9]. Another relevant issue is that not always the primary tumor is responsible

for the death of cancer patients, but rather the metastases of cancerous cells to

secondary sites, as brain, bones or lungs for instance [6], [10]. Therefore today we

face new challenges regarding cancer treatment and cancer patients, especially

those which do not respond to conventional therapies, demand for new and more

efficient and selective drugs or therapies to fight this disease.

1.2 Microorganisms and their products as anti-cancer agents

One of the new paths considered in the search for new anticancer therapies

resides on an unconventional approach using microorganisms, namely live

bacteria or their purified products (Figure 1). Although it can be considered, at

first sight, as a revolutionary method, the use of live bacteria and their products

has been investigated and initially proposed a long time ago. In fact, in late

nineteenth century, William Coley, a surgeon in the Memorial Hospital in New

York, used a mixture of extracts of killed bacteria for the first time against

different types of cancer. Surprisingly he observed anti-tumor activity and

complete remission of tumor in some cases, although some patients have

developed systemic infections and died [11], [12]. More recently this line of work

has regained a new insight and several patents, regarding the use of bacteria or its

derived products, have been issued [12].

Since Coley’s experiments until now several live, attenuated or engineered

bacteria, namely Mycobaterium, Clostridium, Salmonella or Listeria, have shown

the ability to act as anticancer agents [13]. The main disadvantage of this method

is clearly the risk of originating undesired infections on patients, caused by live

bacteria themselves, making it a serious risk if applied on humans. Therefore,

Page 27: Discovering and exploiting bacterial proteins as ...repositorio.ul.pt/bitstream/10451/9665/1/ulfc103147_tm_goncalo_sil… · Discovering and exploiting bacterial proteins as anticancer

3

attenuated or genetic engineered bacteria have been explored in order to minimize

that problem.

However not only bacteria may have applications in cancer therapies, since

bacterial purified products such as protein, enzymes, immunotoxins, antibiotics or

other secondary metabolites have been extensively studied concerning this matter

[11]. With these approaches one could overcome the limitation of using living

bacteria, eliminating the risk of infection. Moreover, some of these products have

proven to cause significant and promising results, such as tumor regression

through growth inhibition, cell cycle arrest or even apoptosis induction [12]. The

use of some bacterial products (as proteins for instance) which are able to target

and lead to the death of tumor cells specifically, would probably overcome the

flaws unspecific cancer treatments.

Figure 1– Current diverse strategies to use bacteria and bacterial products in cancer treatment

(Bernardes et al, 2013) [11].

Page 28: Discovering and exploiting bacterial proteins as ...repositorio.ul.pt/bitstream/10451/9665/1/ulfc103147_tm_goncalo_sil… · Discovering and exploiting bacterial proteins as anticancer

4

1.3 Azurin

Azurin is a low molecular weight (128 aa-14 kDa), water-soluble, type I

copper-containing protein that belongs to the cupredoxin family [13], [14], [15].

This periplasmic redox protein is secreted by Pseudomonas aeruginosa and act as

electron donor to nitrite reductase during denitrification in this pathogenic

bacterium [14], [16].

Azurin possesses a characteristic single domain structure, which consists of a

rigid β-sandwich core (an immunoglobulin fold), formed by eight antiparallel

strands (Greek key β-barrel structure) stabilized by a disulfide bridge and its

tridimensional structure is presented in Figure 2 [13], [17]. In addition, it has a

neutral hydrophobic patch surrounding the copper site [17]. Azurin displays

structural similarity with variable domains of various immunoglobulins, thereby

demonstrating its single antibody-like structure [13].

However a new and interesting role regarding azurin was revealed in 2000,

when Zaborina et al. reported azurin cytotoxic and apoptosis-inducing activities

towards murine macrophage cell line J774 [19]. Since J774 is a transformed cell

line, derived from reticulum cell sarcoma, it was relevant to verify if azurin could

Figure 2 – The three-dimensional structure of azurin from P. aeruginosa (Bonander et al, 1997)

[18].

Page 29: Discovering and exploiting bacterial proteins as ...repositorio.ul.pt/bitstream/10451/9665/1/ulfc103147_tm_goncalo_sil… · Discovering and exploiting bacterial proteins as anticancer

5

cause cytotoxic effects in human tumor cell lines. In fact, later it was shown that

azurin can also trigger apoptosis and lead to significant cytotoxicity in different

human tumor cell lines as breast cancer (MCF-7), melanoma (UISO-Mel-2) and

osteosarcoma (U2OS) cells [20], [21], [22]. Moreover, in the referred cell types

the elevation of p53 intracellular levels led to enhanced pro-apoptotic Bax

formation and a rearrangement in its distribution from the cytosol to the

mitochondria, due to azurin treatment, which it is known to occur along with the

release of cytochrome c from the mitochondria to the cytosol [21]. In the case of

MCF-7 cells it was also shown that not only enhanced Bax formation occurs but

also anti-apoptotic Bcl2 levels decrease with the time of azurin treatment [21].

Interestingly azurin exhibits preferentially selectivity against tumor cell lines,

showing much less cytotoxic and apoptotic effects towards normal cell lines [21].

A domain with only 28 amino acids, designated Azu 50-77 or p28, was

identified as the preferential entry domain of azurin and can act as a potential

protein transduction domain (PTD) in cancer cells [23]. Although azurin entry

mode remains unclear, recently some studies regarding p28 showed that this

peptide requires caveolae-mediated endocytosis in order to enter cells, since

microtubule and caveolae-disrupting agents, that inhibit caveosome formation and

transport, were able to inhibit considerably the entry of p28 in different cancer cell

lines [15], [24]. Caveolae are a 50- to 100-nm subset of lipid raft invaginations of

the plasma membrane, which contains caveolin-specific proteins, as caveolin-1,

that act as regulators of signal transduction [24]. Another evidence that supports

the importance of caveolae in p28 internalization is that p28 co-localizes with

caveolin-1 [24], [25]. Moreover cancer cells showed to be more sensitive to the

effects of these inhibitors comparing to normal cells, which suggests that a higher

number of membrane receptors or structures present in cancer cells, such as

caveolae, can help to explain p28 preferential entry in this type of cells [15], [24].

On the other hand several inhibitors of energy-dependent transport mechanisms,

as Na+K

+ ATPase pump, had no inhibitory effect on p28 penetration, suggesting

that non-endocytic pathways may also be involved in the internalization of this

peptide [15], [24].

Page 30: Discovering and exploiting bacterial proteins as ...repositorio.ul.pt/bitstream/10451/9665/1/ulfc103147_tm_goncalo_sil… · Discovering and exploiting bacterial proteins as anticancer

6

After entering cancer cells, azurin can be found in the cytosol and nuclear

fractions [20]. Azurin has shown the ability to bind and to form a complex with

the tumor suppressor protein p53, thereby stabilizing it and favoring its

intracellular level to raise [20]. This protein is a central and major player in a

complex network responsible for regulating processes like cell growth, genomic

stability and cell death [26]. These different processes can be regulated by the

same protein as it is a transcription factor that acts as a sequence-specific

transcription regulator for many pro-apoptotic genes, namely bax and p21, which

encode protein Bax, involved in apoptosis, and protein 21, involved in the

inhibition of cell cycle progression as well as growth arrest [26]. Although p53-

mediated apoptosis is not yet a fully understood phenomenon, it is clear that p53

has a fundamental role on azurin triggering-apoptosis. As a matter of fact

evidence shows that apoptosis rate and Bax levels are almost inexistent in p53

null cell lines or p53 nonfunctional cell lines, thus supporting a mandatory

relationship between p53 positive cells and azurin apoptotic induction [21].

Therefore azurin can cause an increase in p53 intracellular level which, ultimately,

lead to apoptosis induction in cancer cells, via caspase-mediated mitochondrial

pathways [20]. Another interesting fact is that azurin was shown to be localized in

cytosol and in mitochondria but not in the nucleus of p53 null cell lines, thereby

demonstrating that azurin nuclear transport is p53-dependent [20], [21]. Further

studies on this matter revealed that azurin binds preferentially to the N-terminal

and central domain of p53, but very weakly to the C-terminal domain of this

protein [21]. It was suggested that azurin forms a stable complex by binding the

N-terminal domain of p53 in a 4:1 stoichiometry, and that it can also bind the

DNA-binding domain of this tumor suppressor protein [15]. MDM2, a repressor

oncoprotein which inhibits p53 transcriptional activity and favors its degradation,

also binds p53 in the N-terminal region [27]. However it seems that azurin does

not overlap the MDM2 binding site, although it may sterically shield p53 from

interacting with MDM2 or other ubiquitin ligases [27]. Therefore the increase in

p53 in response to azurin results essentially from a reduction in proteasome

degradation of the tumor suppressor protein, and not from p53 enhanced

transcription [15].

Page 31: Discovering and exploiting bacterial proteins as ...repositorio.ul.pt/bitstream/10451/9665/1/ulfc103147_tm_goncalo_sil… · Discovering and exploiting bacterial proteins as anticancer

7

More recently, azurin in vitro anti-tumor effects were extended to liquid

cancers as well. Azurin demonstrated selective entry and cytotoxic effects against

HL60, an acute myeloid leukemia cell line, and K562, a chronic myeloid leukemia

cell line [28]. It also showed significant effect on arresting K562 cells at G2/M

checkpoint, which often leads to induction of apoptosis.

Additionally to its entry specificity in cancer cells, azurin-derived peptide p28

has been shown to be capable of interfering with angiogenesis by inhibiting the

formation of capillary tube formation of human umbilical vein endothelial cells

(HUVEC), in a dose-related manner [25]. Although p28 could alter p53

intracellular levels in these cells, inhibition of angiogenesis was not suggested to

be accomplished by a p53-mediated inhibition of cell cycle. In this study azurin

also revealed the same anti-angiogenic activity but not the same efficacy of

inhibition as p28.

Azurin, as well as p28, seems additionally capable of interfering in oncogenic

transformation, since it was shown to inhibit the development of precancerous

lesions in a mouse mammary gland organ culture model, previously exposed to a

carcinogen Dimethyl-benz-anthracene (DMBA) [16].

Considering azurin’s remarkable p53-mediated apoptosis and cytotoxic effects

against several tumor cell lines as in vitro models, it was of interest to verify

whether or not similar results could be observed within in vivo models. On this

subject azurin has shown to be effective on tumor growth inhibition in nude

(athymic) mice with xenotransplanted UISO-Mel-2 and MCF-7 cells [20], [21]. In

both studies tumor regression was justified by an increase of apoptosis in tumor

cells. In another study, using a Dalton’s lymphoma bearing ascites mice model,

Ramachandran et al. also showed azurin’s ability to induce apoptosis in tumor

cells, therefore leading to tumor regression. In this study Bax and caspase-3 levels

in tumor cells, treated with azurin, were shown to be increased, whereas Bcl-2

levels were diminished [29]. These results clearly support in vitro evidence,

already described, regarding p53-mediated apoptosis by azurin, which alters the

balance between pro and anti-apoptotic protein levels in tumor cells in favor of the

first. Another important factor, verified in these three in vivo models, was the lack

Page 32: Discovering and exploiting bacterial proteins as ...repositorio.ul.pt/bitstream/10451/9665/1/ulfc103147_tm_goncalo_sil… · Discovering and exploiting bacterial proteins as anticancer

8

of azurin’s toxic effects on the animals [20], [21], [29]. In both cases no body

weight changes or any histologic evidence of toxicity were observed.

Since p28 was considered accountable for a significant amount of the overall

tumoricidal activity of azurin, this peptide was tested originally in vivo towards

MCF-7 xenografs in athymic mice, resulting in tumor growth inhibition [15]. A

more recent and broader study evaluated p28 activity against HCT-116 (colon

cancer), UISO-Mel-23 (melanoma) and MDA-MB-231 (breast cancer) xenografs

in athymic mice [30]. In this study tumor cell proliferation decreased in a dose-

related manner in all three xenografs, whereas tumor regression was observed in a

dose-related way in UISO-Mel-23 and MDA-MB-231 xenografs [30]. This

azurin-derived peptide was shown to be non-immunogenic and non-toxic in mice

and non-human primates, which indicates that it can become a promising drug to

be used in a near future [30].

Taking into account all these promising findings about p28, it was proposed its

entry into a phase I clinical trial in order to access its potential role as an anti-

tumor drug in humans. Intravenous p28 was administrated three times a week, for

4 weeks, in 15 adult patients with p53-positive advanced solid tumors (7

melanoma, 4 colon cancer, 1 sarcoma, 1 gastrointestinal stromal cell tumor, 1

prostate cancer and 1 pancreatic cancer) [31]. Five escalating doses of p28 were

used although no significant adverse effects, toxicity, nor any immune response

were observed in any patient. This phase I clinical trial resulted in one patient with

a complete tumor regression, three patients with partial regression, and seven

patients with stable disease [31].

Cupredoxins, like azurin, exhibit topological similarity to a eucaryotic family

of ligands named ephrins, having a common type of Greek key β-barrel [32].

Ephrins are endogenous ligands that bind with Ephrin receptors (Eph), which

constitute the largest family of receptor protein tyrosine kinases [33]. Binding and

heterodimerization between an ephrin and its receptor leads to the trans-

autophosphorylation of the tyrosine kinase domains of the Eph receptor, leading

to several signaling transduction cascades involved in developmental processes

that require organized patterning and movement of cells, as in the remodeling of

blood vessels [32], [33]. However Eph receptors as well as ephrins have shown

Page 33: Discovering and exploiting bacterial proteins as ...repositorio.ul.pt/bitstream/10451/9665/1/ulfc103147_tm_goncalo_sil… · Discovering and exploiting bacterial proteins as anticancer

9

also to be linked to pathological processes, such as tumor progression,

angiogenesis, migration and invasion [32]. Both types of proteins were shown to

be up-regulated in several different tumors, like EphB2 in breast carcinoma or

lung cancer, for instance [32]. Azurin can bind the EphB2 receptor tyrosine kinase

with a higher affinity than its endogenous ligand, ephrinB2, competing for this

receptor and diminishing tyrosine phosphorylation, thereby interfering with cell

signaling and cancer growth [32].

Despite being involved in the denitrification in P.aeruginosa, an obligatory

role of azurin in this process was excluded, thereby the biological role of this

protein still requires clarification [23]. It has been suggested, however, that

azurin’s physiological function could be involved in bacterial virulence,

characteristic of pathogenic bacteria like P.aeruginosa [23]. Experiments

involving this bacterium and cancer cells have demonstrated that azurin secretion

occurs mainly in the presence of cancer cells in the medium, whereas in the

absence of cancer cells very little secretion of this protein was verified [34]. These

findings point out the possible existence of a sensing mechanism in bacteria that

could lead to azurin secretion in the presence of cancer cells, which they could

sense as a threat or competitor to their own growth [35].

Interestingly it was shown that azurin can inhibit not only growth in cancer

cells, but also in different pathogens as virus (AIDS virus HIV-1), parasites

(Plasmodium falciparum) and protozoans (Toxoplasma gondii) [14], [36]. It

appears that azurin’s ability for binding some pathogen surface proteins interferes

in the entry and inhibit the growth of different pathogens and cancer cells [14],

[36]. This promiscuity in binding different proteins, as seen in Figure 3, may be

attributable to its structural similarity with the variable folds of immunoglobulins,

which could represent a “progenitor” immune response used by prokaryotes, as

suggested by some authors [14].

Azurin’s promiscuity and broader anti-tumor action represent two major

aspects of this bacterial protein. So far it has been shown capable of 3 different

modes of anti-tumor action: induction of apoptosis through p53 stabilization;

inhibition of angiogenesis; and binding ephrin receptor kinases. Acting on

different pathways in cancer progression, azurin differentiates itself from any

Page 34: Discovering and exploiting bacterial proteins as ...repositorio.ul.pt/bitstream/10451/9665/1/ulfc103147_tm_goncalo_sil… · Discovering and exploiting bacterial proteins as anticancer

10

other current anti-tumor drug available, which could show to be more effective

against cancer cells.

Figure 3 - Azurin as a promiscuous protein, which possess both anticancer, antiparasite and

antiviral activities (Fialho et al, 2007) [37].

1.4 Cadherins

Classical cadherins constitute a family of transmembrane glycoproteins that

mediate calcium-dependent cell-cell adhesion and present themselves as the major

components of cell-cell adhesive junctions [38], [39]. These particular family

includes four different types of cadherins, designated accordingly to their tissue

distribution: CDH1/E-cadherin (epithelial), CDH2/N-cadherin (neuronal),

CDH3/P-cadherin (placental) and CDH4/R-cadherin (retinal) [40]. E- and P-

cadherin can be divided in three major structural domains: 1) an extracellular

domain, which is responsible for cadherins adhesion properties since it’s where

Ca2+

ions will bind to stabilize cadherins conformation; 2) a single membrane-

spanning segment, accountable for protein anchorage to the cellular membrane; 3)

a highly conserved cytoplasmic domain that bind directly to α, β and γ-catenins

and p120-catenin, forming a complex that acts as a bridge between cadherins and

the actin cytoskeleton [38], [39]. This binding is supported by α-catenin and

Page 35: Discovering and exploiting bacterial proteins as ...repositorio.ul.pt/bitstream/10451/9665/1/ulfc103147_tm_goncalo_sil… · Discovering and exploiting bacterial proteins as anticancer

11

provides the molecular basis for stable cell-cell interactions and its represented in

Figure 4 [40].

The epithelial-calcium dependent cell-cell adhesion is accomplished by the

establishment of hemophilic interactions between proteins, as two cadherin

molecules of adjacent cells to form a homodimer [39]. The cadherin/catenin

complex stability, as well as the signaling pathways controlled by this structure, is

therefore essential for maintaining some cell properties, like cell-cell adhesion and

homeostatic tissue architecture [39]. By regulating these properties the

cadherin/catenin complex has a major role on processes like cell growth,

differentiation, motility and survival [40]. In fact there is wide evidence that

alterations in the adhesion properties between adjacent cells provide them with an

invasive and migratory phenotype. Several data was reported regarding changes in

normal E- and P-cadherin function or expression, which has been associated with

all steps involved in tumor progression [39].

E-cadherin is predominantly expressed in all epithelial tissues, playing a major

role in the formation of epithelia, and being responsible for the maintenance of

cell shape and polarity [40]. E-cadherin gene, CDH1, acts as a tumor suppressor

Figure 4 – Schematic representation of the classical cadherin-catenin complex and the structural

components of cadherins (adapted from Albergaria et al, 2011) [40].

Page 36: Discovering and exploiting bacterial proteins as ...repositorio.ul.pt/bitstream/10451/9665/1/ulfc103147_tm_goncalo_sil… · Discovering and exploiting bacterial proteins as anticancer

12

gene, regulating the invasion and metastasis of tumor cells. In fact loss of

expression or abnormal function of E-cadherin, by mutations or loss of

heterozygosity, can result in increased ability of tumor cells to invade and to

create metastasis in neighbouring tissues, namely in breast cancer [39], [41].

Another relevant factor that inhibits E-cadherin expression is hypermethylation of

the promotor region of CDH1, which has been implicated at the same time with

the induction of migration in breast cancer cell lines [41]. Reduced expression of

E-cadherin was associated with tumor progression in several types of cancer,

including breast and stomach carcinomas [38], [39].

P-cadherin is a 118 kDa cadherin, that is expressed in ectodermal tissues,

namely in the basal layers of stratified epithelia (as skin, uterine cervix, prostate,

lung) and in myoepithelial cells of the breast [38], [40], [42]. This cadherin has

been associated in growth and differentiation processes, as those during

embryogenesis, for instance, and low levels of this protein were detected in

normal tissues [40]. Unlike CDH1, which is undoubtedly recognized as a tumor

suppressor gene, CDH3 role in cancer is contradictory and less characterized [40].

Distinct P-cadherin behavior was verified in different types of cancer, since it

appears to act as an invasion suppressor in colorectal cancer and melanoma

models whereas it resembles an oncogene in other models, as breast cancer, by

inducing tumor cell motility and invasiveness when overexpressed [43]. Increased

P-cadherin expression was also correlated with cell dedifferentiation and

increased cell proliferation [39]. The opposite effects of E- and P-cadherin in

breast cancer (as well as other models) are moreover unexpected since these two

molecules share more than 67% of homology [40].

Before acquiring an invading behavior, tumor cells often suffer a process

termed epithelial-to-mesenchymal transition, on which epithelial cadherins suffers

downregulation whereas mesenchymal cadherins are expressed de novo [44]. This

cadherin switch results in inhibition of cell-cell contacts and elicits active signals

which prompt cell migration and invasion. In breast cancer, many of the highly

aggressive tumors do not show, however, this cadherin switch. By contrast these

tumors show P-cadherin overexpression while maintaining the normal E-cadherin

expression [44]. P-cadherin is preferentially overexpressed in basal like

Page 37: Discovering and exploiting bacterial proteins as ...repositorio.ul.pt/bitstream/10451/9665/1/ulfc103147_tm_goncalo_sil… · Discovering and exploiting bacterial proteins as anticancer

13

carcinomas and it was verified that 20% to 40% of invasive breast carcinomas, as

well as 25% of ductal carcinomas in situ showed overexpression of this cadherin

[42], [43]. Another relevant observation is that P-cadherin expression has been

reported as a marker of poor prognosis and reduced patient survival in high

histological grade tumors, with decreased cell polarity [42]. Expression of this

cadherin can also have an important role in the prognosis of invasive breast

carcinomas that maintains normal E-cadherin expression [39]. An in vitro study

supports these findings as it revealed that P-cadherin overexpression, in wild-type

E-cadherin breast cancer cell lines, as MCF-7/AZ, can induce increased cell

invasion, motility and migration [44]. Moreover it was shown that P-cadherin can

only induce invasion in breast cancer cell lines which already express and

endogenous cadherin like E-cadherin, by disrupting the interaction between E-

cadherin and two catenins (p120 and β-catenin) [40].

Additionally it was found that the presence of P-cadherin, in these E-cadherin

positive cancer cells, can provoke the secretion of pro-invasive factors, as matrix

metalloproteases (MMPs), MMP-1 and MMP-2, which in turn lead to P-cadherin

ectodomain cleavage [44]. The 80 kDa soluble P-cadherin fragment (sP-cad)

formed in that cleavage has pro-invasive activity, being responsible for in vitro

invasion of these cancer cells [42], [44]. This protein was found also in body

fluids, as milk from the lactating breast, although its biological role in that context

is still unknown [42]. Although the mechanism by which P-cadherin

overexpression induces the secretion of metalloproteases is unknown, it is clear

that these proteases are responsible for extracellular matrix degradation, for

instance, which increases cell invasion induction [44].

1.5 Azurin and P-cadherin interactions

Considering azurin’s broader anti-tumor properties and knowing that it exhibits

a high affinity level with P-cadherin, it was of interest to evaluate this protein’s

activity in an invasive breast cancer cell lines (MCF-7/AZ.Pcad) that express

endogenous levels of E-cadherin and higher levels of P-cadherin [45]. After

azurin administration with sub-lethal dosages (50-100 µM) to these cells, it was

verified, by western blotting, a specific decrease in P-cadherin total protein level

Page 38: Discovering and exploiting bacterial proteins as ...repositorio.ul.pt/bitstream/10451/9665/1/ulfc103147_tm_goncalo_sil… · Discovering and exploiting bacterial proteins as anticancer

14

[45]. P-cadherin distribution across the plasma membrane was analyzed through

immunofluorescence, and it was shown that it suffered a decrease, as well, in cells

treated with azurin [45]. On the contrary, evidence showed that E-cadherin level

and its membrane distribution were not affected by azurin’s treatment.

Additionally, it was found that a reduction in P-cadherin’s level (caused by

azurin) was correlated with a less invasive behavior of breast cancer cells in a

Matrigel system, as well as with a lower activity of metalloproteases MMP-2 and

9 in cell conditioned medium. Another extreme relevant finding was that P-

cadherin soluble fragment (sP-cad) level, which has pro-invasive activity, was

found to be decreased when the conditioned medium, on which cells treated with

azurin where cultured, was analyzed. Complementary assays regarding CDH1 and

CDH3 genes showed that P-cadherin decreased levels were not a consequence of

changes in gene expression [45]. Therefore it was suggested that post-

transcriptional regulation processes, mediated by azurin, should be involved in P-

cadherin diminished levels. Microarray analysis on mRNA profiles of MCF-

7/AZ.Pcad cells treated with azurin showed that genes were associated with

membrane organization, vesicle-mediated and endosome transport, and lysosome

were up-regulated (Bernardes et al., 2013, submitted for publication).

This work intends to pursue those evidences and to demonstrate that azurin can

display an important role on mediating P-cadherin degradation through proteolytic

systems.

1.6 Inhibitors

In order to maintain regular homeostasis and meet their nutritional and

energetic demands, cells have devolved proteolytic systems which eliminate

instable and incorrect folded proteins. These systems are able to protect cells from

cytotoxic damage, caused by intracellular accumulation of damaged proteins or

organelles, and to recycle amino acids that result from protein degradation,

replenishing the intracellular reserve of these macromolecules, which are essential

in the absence of nutrients [46]. The two main cellular degradation systems in

eukaryotic cells are the autophagy-lysossome system and the ubiquitin-

Page 39: Discovering and exploiting bacterial proteins as ...repositorio.ul.pt/bitstream/10451/9665/1/ulfc103147_tm_goncalo_sil… · Discovering and exploiting bacterial proteins as anticancer

15

proteasome system (UPS), although the relative contribution of each one of these

pathways may vary greatly between cell types [47], [48].

Lysosomes are single membrane vesicles that contain in their lumen a large

diversity of hydrolytic enzymes, such as proteases, lipases, glycosidases and

nucleotidases [46]. These enzymes reach their optimal enzymatic activity at the

acidic pH verified in the lysosomal lumen, which is maintained by an ATP-

dependent proton pump (V-ATPase) present at the lysosomal membrane [46],

[47]. The lysosomal degradation pathway is preferentially used in proteolysis of

membrane proteins, as receptors or channels, although it can act on cytoplasm

proteins as well [47]. In order to reach the lysosomes, tagged proteins must be

recognized and delivered to this organelle by three different ways:

macroautophagy, microautophagy and chaperone-mediated autophagy [48].

Macroautophagy involves a de novo formed double membrane vesicle or

autophagosome, which is responsible for protein sequestering, and that will fuse

with late endosomes or lysosomes [46]. In microautophagy, it occurs a lysosomal

membrane invagination, resulting in sequestration of regions of the cytosol

directly by the lysosomal membrane into its lumen. Finally, chaperone-mediated

autophagy is a process that requires the recognition of the target protein by a

cytosolic chaperone, and its binding to a lysosomal membrane receptor, which in

turn allows target protein translocation into lysosome’s lumen. There are several

chemical agents which act as lysosome inhibitors, such as ammonium chloride

and chloroquine, two weak bases that accumulate inside the lysosome and

dissipate its low acidic pH, by neutralizing H+

ions [49].

The UPS represents the major pathway accountable for the degradation of

proteins present in the cytosol, nucleus and endoplasmic reticulum [46]. This

proteolytic system is composed by ubiquitin, a small tagging protein that is

covalently linked to proteins, through a three step ATP-consuming reaction, that

involves E1 (ubiquitin-activating enzyme), E2 (ubiquitin-conjugating enzyme)

and E3 (ubiquitin-ligase enzyme), selecting the substrate protein for degradation

[48]. These three enzymes are responsible for selecting misfolded proteins to

degradation, as well as catalyzing the binding reaction between ubiquitin

molecules and target proteins [48]. After ubiquitination target proteins are

Page 40: Discovering and exploiting bacterial proteins as ...repositorio.ul.pt/bitstream/10451/9665/1/ulfc103147_tm_goncalo_sil… · Discovering and exploiting bacterial proteins as anticancer

16

delivered to the proteasome, a multicatalytic enzyme complex composed by the

catalytic 20S core and the 19S regulator [47]. The 20S core complex its divided in

three catalytic subunits, with distinct activities as chymotrypsin-like, trypsin-like

and caspase-like activities [48]. Finally, polyubiquitinated proteins are degraded

to peptides by the proteasome and then free ubiquitin is recycled [48]. Currently

available proteasome inhibitors interfere with chymotryptic-like of the 20S core

complex such as MG132, a reversible but potent peptide aldehyde [48].

Lipid rafts are considered subdomains of cell membrane, and are involved in

processes like cell proliferation, differentiation and apoptosis, which often are

altered during tumor development [50]. These membrane regions are enriched in

cholesterol, or caveolins, for instance. Methyl-β-cyclodextrin (MβCD) is a cyclic

oligosaccharide that binds preferentially to cholesterol from the plasma

membrane, altering lipid composition and causing cholesterol depletion [51], [52].

By disrupting the assembly of lipid rafts and caveolae, this compound can

therefore inhibit caveolae-mediated endocytosis, since caveolin can’t form further

vesicle [53], [54].

Since azurin enter cells through caveolae-mediated endocytosis, we sought to

use MβCD to inhibit azurin entry into breast cancer cells, in order to evaluate its

impact on P-cadherin degradation level.

1.7 MPT 63

MPT 63 is a low molecular weight (16 kDa) protein that comprises 130 amino

acid residues preceded by a secretion signal peptide [55], [56]. Firstly described as

one of the three most abundant extracellular proteins secreted by Mycobacterium

tuberculosis, MPT 63 was later found to be specific of mycobacteria of the M.

tuberculosis complex, as M. africanum, M. bovis and attenuated M. bovis bacille

Calmette-Guérin (BCG) and it is absent in mycobacterial species that do not

belong to this complex, as M. avium [56], [57].

The X-ray crystal structure of MPT 63 (represented in Figure 5) was

determined by Goulding et al as a β-sandwich, consisting of two antiparallel β-

sheets, similar to an immunoglobulin-like fold, with and additional antiparallel β-

sheet [58]. Interestingly this protein exhibits some structural similarity to cell

Page 41: Discovering and exploiting bacterial proteins as ...repositorio.ul.pt/bitstream/10451/9665/1/ulfc103147_tm_goncalo_sil… · Discovering and exploiting bacterial proteins as anticancer

17

surface-binding proteins such as Homo sapiens β-2 adaptin, bovine arrestin,

eukaryotic fibronectin-binding proteins, major histocompability domains or T-cell

receptors, for instance [58]. This similarity has suggested a possible role of MPT

63 in cell-host interactions to facilitate endocytosis as well as phagocytosis during

bacterial internalization [58]. In fact MPT 63, as well as other proteins secreted by

M. tuberculosis, is considered relevant to the survival of the bacterium within its

host, since it has been shown to be a cell envelope-associated protein that may act

as a virulence factor [58]. Moreover, secretion of proteins by intracellular

pathogens, as M. bacterium, has a central role in determining pathways of antigen

presentation and recognition by effector T-cells involved in protective immunity

[58]. Although it shares some structural similarities with proteins of other

organisms, this protein doesn’t exhibit a very strong homology with any of them,

which leaves its physiological role yet to be unveiled [55].

Figure 5– Structure of M. tuberculosis MPT 63 protein (adapted from Goulding et al, 2002) [58]

As mentioned earlier, MPT 63 is one of the three most abundant extracellular

proteins secreted by M. tuberculosis, and the other two most abundant proteins

were already described as antigens (antigens 85A and 85B), which can indicate

that this protein can also act as an antigen [56]. In fact so far several data have

Page 42: Discovering and exploiting bacterial proteins as ...repositorio.ul.pt/bitstream/10451/9665/1/ulfc103147_tm_goncalo_sil… · Discovering and exploiting bacterial proteins as anticancer

18

been pointing in that direction, as a study that has shown that guinea pigs,

previously aerosol infected with virulent M. tuberculosis, were able to induce

humoral immune responses, leading to the production of high-level antibodies

against MPT 63 [55]. Moreover, guineas pigs injected with purified MPT 63

revealed cutaneous delayed-type hypersensitivity responses, which indicate that

this protein can induce immunogenic responses, through different pathways [55].

MPT 63 was also shown to be immunogenic in rabbits, since immunization with

the purified protein led to antibodies production against it [59]. Antibodies against

this protein were also identified in serum from tuberculosis patients by enzyme-

linked immunosorbent assay (ELISA) [60]. In a different study, a DNA vaccine

encoding both the MPT 63 and ESAT-6 antigens, has demonstrated to be capable

of inducing a robust protective response in mice, namely through generation of

gamma interferon (INF-γ)-secreting CD4+ T cells [61]. Since MPT 63 has shown,

at some level, structural similarities with major histocompability domains, as well

as T-cell receptors, it is important to mention a study where these two factors were

tested in humans. In this study it was shown that MPT 63 induced a moderate T

helper 1 (Th1) cell reactivity in peripheral blood mononuclear cells (PBMNCs),

obtained from M. bovis BCG vaccinated healthy subjects [62]. Th1 cells reactivity

were evaluated through proliferation and IFN-γ secretion screening tests.

Furthermore the presentation of MPT 63 to Th1 cells were also analyzed, as

PBMNCs from MPT 63-responding donors were typed for Human Leucocyte

Antigen (HLA-DR) molecules revealing heterogenecity between them [62] .

Therefore it seems that MPT 63 exhibits important requirements for a potential

use as a vaccine against tuberculosis, since it could be able to induce a positive

response throughout heterogeneous groups of donors.

Although little is known about the mechanisms behind this protein’s

immunogenic properties it was suggested that these properties could be explained,

at least in part, by the first 30 amino acid sequence in its N-terminal region, which

has shown a high density of T-cell epitopes recognized in immunized guinea pigs

[56].

As mentioned before MPT 63 is not present in mycobacteria which do not

belong to the Mycobacterium complex, and this specificity is also supported by

Page 43: Discovering and exploiting bacterial proteins as ...repositorio.ul.pt/bitstream/10451/9665/1/ulfc103147_tm_goncalo_sil… · Discovering and exploiting bacterial proteins as anticancer

19

the fact that MPT 63 lacks epitopes that cross-react serologically with M. avium

antigens [55]. Taken altogether, this results regarding the immunogenic properties

of MPT 63, led several authors to propose this protein as a target for vaccine

design and diagnostic tools development in tuberculosis [57], [60].

MPT 63 and azurin share some important features, as they both are low

molecular weight proteins secreted by bacteria. More interestingly, however, is

the fact that both their structures are a β-sandwich, which demonstrates evident

structural similarities with immunoglobulin-like folds. Another interesting

observation is the promiscuity evidenced by these two proteins, which grant them

unique properties, allowing them t be capable of binding to different proteins.

Additionally to MPT 63’s ability to bind T-cell receptors, it also shares some

structural similarity with bovine arrestin, a protein that inhibits receptor activity,

by binding to the cytoplasmic surface, occluding the interaction with G-proteins

[58]. Taking account that azurin can also bind to surface cell receptors, as Eph

receptors in cancer cells, thereby interfering with cancer growth, it is intriguing to

verify if these proteins can provoke similar anti-tumor effects.

Recently a 30 amino acid peptide derived from MPT 63 and termed MB30, has

shown significant cytotoxic activity against several bladder (HTB-9, UM-UC-3),

colon (COLO 205, HCT 116), and cervical (SiHa, CaSki) cancer cell lines [63].

Moreover, MPT 63 and MB30 also showed cytotoxicity in brain (U87), liver

(HepG2) and breast (MDA-MB-231) cancer cell lines, although the MB30 peptide

has demonstrated an even higher cytotoxicity [63].

Since MPT 63, as well as its derived peptide MB30, has shown cytotoxic

effects against different cancer cell lines, our purpose in this work was to unveil a

possible role of MPT 63 as an anticancer agent. Therefore we sought to verify if

MPT 63 could cause possible cytotoxic effects on a breast (MCF-7/AZ.Mock) and

lung (A549) cancer cell lines.

1.8 Nucleoside diphosphate kinase

Nucleoside diphosphate kinase (Ndk) is an enzyme which catalyzes the

reversible transfer of the 5’-terminal phosphate from nucleoside triphosphates

(NTPs) to nucleoside diphosphates (NDPs), thus playing a key role as a

Page 44: Discovering and exploiting bacterial proteins as ...repositorio.ul.pt/bitstream/10451/9665/1/ulfc103147_tm_goncalo_sil… · Discovering and exploiting bacterial proteins as anticancer

20

housekeeping enzyme, as it maintains the nucleotide pools for the synthesis of

nucleic acids [64]. Ndk is a small protein, with approximately 150 amino acids,

which form an homohexamer in eukaryotes, archae and gram-positive bacteria,

whereas in gram-negative bacteria it forms homotetramers [64], [65]. So far it has

been described in humans and others eukaryotes as Droshophila melanogaster, as

well as in several bacteria such as P. aeruginosa, Escherichia coli, Myxococcus

xanthus, M. tuberculosis or Vibrio cholerae for instance [66], [67]. Interestingly

there were reported two forms of this protein in P. aeruginosa: a 16 kDa

cytoplasmic form (during the early stages of cellular growth), and a 12 kDa

membrane-associated form (at the onset of the stationary phase) which is

originated from a cleavage in the 16 kDa form, by a periplasmic protease termed

elastase [64], [68]. Although the importance of having two forms of this protein is

not clear, it is known that the membrane-associated form, constitutes a complex

with pyruvate kinase, which predominantly synthesizes GTP [69]. In mammalian

cells Ndk can be localized in the cytosol, as membrane-associated or as an

ectoenzyme in the cell surface exposed to the outside medium [70].

Despite the fact that Ndk exhibits a different structure in humans and some

prokaryotes, it was revealed that Ndk form P. aeruginosa shows 40-45% identity

with eukaryotic Ndks, as well as 50-60% identity with other bacterial Ndks,

evidencing that it is a highly conserved enzyme among different species [66] .

Extracellular secretion of Ndk was reported in M. bovis BCG, M. tuberculosis,

P. aeruginosa, Trichenella spiralis and V. cholerae [67]. The physiological

relevance for the secretory nature of Ndk in these bacteria is not clear, although it

has been mentioned to be an important enzyme in host-pathogen interactions [67].

This suggestion is supported from the fact that Ndk from M. tuberculosis was

reported to interfere and block phagosome maturation in murine macrophages

[71]. Moreover, Ndks secreted by M. tuberculosis and V. cholerae were shown to

display cytotoxic effects on mouse macrophage cell lines, which can indicate that

secretable Ndk plays a major role in the modulation of virulence in these

pathogenic species [67], [72].

Additionally to its kinase function, Ndk has been associated in further

biological functions in eukaryotic cells, namely in humans, where it plays a role in

Page 45: Discovering and exploiting bacterial proteins as ...repositorio.ul.pt/bitstream/10451/9665/1/ulfc103147_tm_goncalo_sil… · Discovering and exploiting bacterial proteins as anticancer

21

normal embryonic development, cell differentiation, tumor progression and cell

migration [73], [74]. In humans, 10 genes have been already identified as part of

the nm23 (non metastatic) gene family, which represents the human Ndk gene

family [75]. Nm23-H1 was the first metastatic gene in humans to be identified in

1988 by Steeg et al. , since the accumulation of correspondent transcripts were

shown higher in tumor cells of low metastatic potential in murine melanoma cell

lines of varying metastatic potentials [76]. The two more expressed nm23 genes in

humans are nm23-H1 and nm23-H2, localized in chromosome 17q21, and encode

the Ndk-A and Ndk-B, respectively [10], [75]. The three-dimensional structure of

human Ndk-A subunit is shown in Figure 6.

Although it is known mainly by its role in tumor progression, Ndk-B has been

also described as a transcription factor for the c-Myc oncogene, and it was

proposed to act as a repair protein in humans, on the basis of sequence homology

with certain glycosylases [73].

Since Steeg et al. initial observations it has been verified, by several authors, a

correlation between reduced Ndk-A expression and high tumor metastatic

potential in different carcinomas: liver, melanoma, colon, breast, ovarian, gastric

and hepatocellular [75]. Transfection experiments with nm23-H1 and nm23-H2

support these findings, since the overexpression of these genes in breast cancer

MDA cell lines resulted in decreased metastatic potential of these cells [77]. The

overexpression of this protein in tumor cells was found to reduce tumor cell

motility and invasion, promote cellular differentiation and inhibits anchorage-

independent growth, as well as adhesion to fibronectin, laminin and vascular

endothelial cells [10]. However, although Ndk-A has shown anti-metastatic

effects on the majority of carcinomas, there are some exceptions. It was reported

that high levels of this protein were detected in aggressive carcinomas derived

from thyroid, pancreatic, squamous cell lung carcinomas, neuroblastoma and

acute myelogenous leukemia [75]. Moreover high tissue levels of Ndk-A were

found in patients with breast carcinoma [78].

Page 46: Discovering and exploiting bacterial proteins as ...repositorio.ul.pt/bitstream/10451/9665/1/ulfc103147_tm_goncalo_sil… · Discovering and exploiting bacterial proteins as anticancer

22

Figure 6 – Three-dimensional structures of human Ndk-A. (A) Diagram of Ndk-A monomer. (B)

Diagram of the homo-hexamer (Han et al, 2010) [79].

The mechanisms by which human Ndk achieves anti-metastatic effects in

several tumor cell lines is unknown, and contradictory effects of this protein in

other carcinomas constitutes a challenge that requires further investigation on this

subject.

Mutations in nm23 genes are rare in cancer, therefore it has been proposed that

this genes can become deregulated through expression changes at the protein

level, instead through mutation [78].

Recently in vitro studies revealed that extracellular Ndk-B was secreted by

breast cancer, colon and pancreas cell lines, while Ndk-A was secreted by human

leukemia cell lines into the extracellular environment [78]. The same phenomenon

was verified in MDA-MB-435 and MDA-MB-231 metastatic human breast

carcinoma cell lines, which secreted Ndk into their surroundings, while the non-

metastatic breast cancer cell line MCF-12 didn’t exhibit the same effect [78].

Extracellular Ndk-A was also found in blood and its level was shown to be

correlated with poor prognosis in acute myelogenous leukemia, malignant

lymphoma and neuroblastoma patients [74].

Ndk, as well as azurin and MPT 63, are low molecular weight bacterial

proteins, and secreted by some bacteria. Recent evidence has attributed an anti-

metastatic role for human Ndk in several tumor cell lines, which presents this

protein with an interesting property to be explored in cancer research. Contrasting

Page 47: Discovering and exploiting bacterial proteins as ...repositorio.ul.pt/bitstream/10451/9665/1/ulfc103147_tm_goncalo_sil… · Discovering and exploiting bacterial proteins as anticancer

23

with the human Ndk, bacterial Ndk effects on tumor cells weren’t studied until

now, as far as we know. Therefore, and since bacterial and human Ndk share great

homology we intended to evaluate possible anti-tumor and anti-metastatic effects

caused by a bacterial Ndk in a human breast cancer cell line (MCF-7/AZ.Mock)

and a human lung carcinoma cell line (A549), using a normal lung cell line as

control (16HBE14o-). Moreover it was of interest to verify if a bacterial Ndk

(from P.aeruginosa) could cause cytotoxic effects, as demonstrated in murine

macrophage cell lines by other bacterial Ndks, and if those effects could be higher

in tumor cell lines, as shown by azurin.

Page 48: Discovering and exploiting bacterial proteins as ...repositorio.ul.pt/bitstream/10451/9665/1/ulfc103147_tm_goncalo_sil… · Discovering and exploiting bacterial proteins as anticancer
Page 49: Discovering and exploiting bacterial proteins as ...repositorio.ul.pt/bitstream/10451/9665/1/ulfc103147_tm_goncalo_sil… · Discovering and exploiting bacterial proteins as anticancer

25

2. OBJECTIVES

The present work intends to help clarify how does azurin causes a decrease in

P-cadherin level on a breast cancer cell line, with a normal E-cadherin expression

but a high P-cadherin expression level (MCF-7/AZ.Pcad cells). Taking into

account previous experiments, we seek to unveil a strongly suggested link

between azurin and P-cadherin decrease through proteolytic pathways. Our

approach resides in blocking the proteasome and the lysosome systems, using two

inhibitors, MG-132 and ammonium chloride respectively, and analyze by western

blot how that affects P-cadherin level when cells are previously treated with

azurin. Additionally, we want to verify what effect can MβCD originate on E- and

P-cadherin levels, by inhibiting azurin internalization on these cells.

The second main objective in this work was to use two different bacterial

proteins, termed MPT 63 (from Mycobacterium tuberculosis) and Nucleoside

diphosphate kinase (from Pseudomonas aeruginosa), in order to describe and

elucidate their possible anti-tumor and cytotoxic effects on cancer cell lines from

breast and lung carcinomas (MCF-7/AZ.Mock and A549, respectively). Azurin

(from Pseudomonas aeruginosa) was used as positive control in order to compare

their relative effects, since its cytotoxicity toward tumor cells was already

described. We also used a normal cell line, 16HBE14o-, to evaluate if Ndk’s

cytotoxicity is selective towards only tumor cells. Cell viability assay was

performed using the 3 different cell lines and the three bacterial proteins at 5

different concentrations (0, 10, 25, 50 and 100 µM). Also different exposure time

(48, 72 h) and number of protein doses (1 to 3) applied were tested to see how

these parameters affect cell viability.

Additionally, since a human Ndk, termed Nm23-H1, displays anti-metastatic

behavior, we intended to test if Ndk also possesses anti-metastatic potential

against a highly invasive tumor cell line, A549, using a matrigel invasion system.

Page 50: Discovering and exploiting bacterial proteins as ...repositorio.ul.pt/bitstream/10451/9665/1/ulfc103147_tm_goncalo_sil… · Discovering and exploiting bacterial proteins as anticancer
Page 51: Discovering and exploiting bacterial proteins as ...repositorio.ul.pt/bitstream/10451/9665/1/ulfc103147_tm_goncalo_sil… · Discovering and exploiting bacterial proteins as anticancer

27

3. MATERIALS AND METHODS

3.1 Bacterial proteins superexpression

3.1.1 Bacterial strains and plasmids

The azurin-encoding gene azu, derived from P. aeruginosa PAO 1 strain, was

amplified and cloned into pWH844 plamid. The same pWH844 plasmid was also

used for cloning the mpt63 and ndk genes, derived from M. bacterium and P.

aeruginosa, respectively. Azurin, MPT 63 and Ndk were expressed in competent

Escherichia coli SURE (Stop Unwanted Rearrangement Events) strain containing

the pWH844 plasmid. E. coli SURE is a protease-deficient strain, which was

chosen to improve the yield of expressed proteins. Each one of the genes that

encode these proteins is regulated by an IPTG (Isopropyl-β-D-

Thiogalactopyranoside) inducible lac promoter. Upstream region of these genes

also contain a nucleotide sequence encoding a six histidine tag, thus allowing

further protein purification through a nickel affinity column.

3.1.2 Inoculum

E. coli SURE cells were pre-inoculated in Luria Broth (LB) medium [10 g/L of

tryptone; 10 g/L of sodium chloride (NaCl); 5 g/L of yeast extract] with ampicillin

(150 µg/mL) and maintained at 37º C, with continuous shaking at 250 rpm

(revolution per minute) overnight. On the next day Super Broth medium (32 g/L

of tryptone; 20 g/L of yeast extract; 5 g/L of NaCl) was inoculated with a volume

of the pre-inoculum equivalent to an OD640 nm (optical density) of 0.1 and

incubated at 37º C with continuous shaking. The antibiotic added to the culture

medium was ampicillin with the same concentration used for the pre-inoculum

(150 µg/mL). When the inoculum OD640 nm value was between 0.6 and 0.8, IPTG

(0.2 mM) was added to the medium in order to induce gene superexpression.

Induced inoculums were incubated for 4-5 hours at 37º C.

After the incubation period the inoculums were centrifuged at 8000 rpm for 10

minutes (Beckman J2-MC centrifuge) with a constant temperature of 4º C. The

retrieved pellet was ressuspended in 15 mL of START buffer (10 mM of

Page 52: Discovering and exploiting bacterial proteins as ...repositorio.ul.pt/bitstream/10451/9665/1/ulfc103147_tm_goncalo_sil… · Discovering and exploiting bacterial proteins as anticancer

28

imidazole, 0.2 mM of sodium phosphate, 0.5 M of NaCl, pH 7.4) for each 350 mL

of the initial culture medium and stored at -80ºC until its use.

3.1.3 Cell sonication

The cells were sonicated on ice with a Branson 250 sonifier, applying 9 cycles

of 15 pulses each at 50% duty cycle and output control 10. Between each cycle,

cells were left to rest during 5 minutes. The sonicated cells were centrifuged at

17600 g (B. Braun Sigma-Aldrich 2K15) and 4º C, for 5 minutes, after which time

the resulting supernatant was collected and centrifuged for 1 hour at the same

conditions.

3.1.4 Azurin purification

Azurin purification was accomplished using a HisTrapTM

FF 5 mL purification

column (GE Healthcare Life Sciences), previously equilibrated with 25 mL of

START buffer. Cell lysates were then loaded into the column and washed with 25

mL of START buffer before sequential elution with 25 mL of elution buffers. The

elution buffers contain a constant phosphate buffer and an increasing imidazole

concentration (20, 40, 60, 100, 200, 300 and 500 mM) (Table 1, Table 2). The

purified protein was obtained by collecting the 100 and 200 mM imidazole

fractions.

In order to remove excess salt and imidazole from our protein sample, the

collected fractions were desalted through size-exclusion chromatography using

the ÄKTA Prime liquid chromatography system (Amersham Biosciences).

Samples were injected through a HiPrepTM

26/10 Desalting column (GE

Healthcare Life Sciences) with Sephadex G-25, according to the elution program

shown in Table 3. Phosphate buffered saline (PBS) was used as the elution buffer

(Table 4). The eluted fractions were collected according to the chromatogram

generated by PrimeView software, which measures the desalted solution OD280 nm,

thus allowing protein detection.

Page 53: Discovering and exploiting bacterial proteins as ...repositorio.ul.pt/bitstream/10451/9665/1/ulfc103147_tm_goncalo_sil… · Discovering and exploiting bacterial proteins as anticancer

29

Table 1 - Phosphate buffer 8x composition

Phosphate buffer 8x

Na2HPO4.2H2O (M) 0.08

NaH2PO4.H2O (M) 0.08

NaCl (M) 4

pH 7.4

Table 2 - START and elution buffer compositions

START buffer Elution buffers

Phosphate buffer 8x (mL) 12 3

Imidazole (mM) 10 20; 40; 60; 100; 200; 300; 400; 500

Water until final volume of (mL) 100 24

pH 7.4 7.4

Table 3 - ÄKTA elution program for protein sample desalting

Step Volume (mL) Flow rate (mL/min) Inject valve

Tube wash 50 40 waste

Column wash 5 5 load

Protein injection 15 5 inject

Protein elution 100 5 load

Column wash 30 5 load

After the desalting step the collected fractions were concentrated in a 3 kDa

cut-off column with a cellulose membrane (Ultracel-3K Amicon Ultra centrifugal

filter from Millipore) through successive centrifuging at 5000 rpm and 4º C

(Eppendorf Centrifuge 5804R) until concentrate the sample to an approximated

volume of 2 mL.

The concentrated sample was then applied onto a Detoxi-GelTM

Endotoxin

Removing column (Thermo Scientific) with a gel matrix composed by crosslinked

6% beaded agarose, that has polymixin B immobilized onto it. This antibiotic can

bind bacterial lipopolysaccharide (LPS) therefore assuring that our protein sample

was free of endotoxin contamination. Before applying the sample, the detoxi-gel

column was previously equilibrated with 5 mL of each of these reagents:

Page 54: Discovering and exploiting bacterial proteins as ...repositorio.ul.pt/bitstream/10451/9665/1/ulfc103147_tm_goncalo_sil… · Discovering and exploiting bacterial proteins as anticancer

30

distillated water, 1% sodium deoxycholate, distillated water and PBS. Protein

sample was then applied and left to rest at 4º C, for 1 hour, after which time it was

collected by gravity flow by eluting it with PBS until reaching a volume of 30

mL. After each detoxing step the columns were regenerated with 5 mL of destilled

water, 1% sodium deoxycholate, distilled water and 20% ethanol. The collected

sample was concentrated until reaching a volume of 2 mL, following the same

specifications as the first concentration step. This final volume obtained was

centrifuged in a 100 kDa cut-off column (Ultracel-100K Amicon Ultra centrifugal

filter from Millipore) in order to guaranty that no contamination with bacterial

cells took place.

Azurin final concentration was calculated after OD280nm reading and using the

Lambert-Beer equation, where azurin’s extinction coefficient ε (280 nm) value

applied was 9.1 x 103

M-1

.cm-1

[80]. Spot test was performed in LB plates at 37º

C overnight to exclude any possible contamination. Azurin was kept at 4º C until

further use.

Table 4 - PBS composition

PBS Concentration (mM)

NaCl 137

KCl 2.7

Na2HPO4.2H2O 4.3

KH2PO4 1.47

3.1.5 MPT 63 and Ndk purification

MPT 63 purification was performed following exactly the same procedures as

in azurin purification, with the exception that the MPT 63 sample was collected at

60 and 100 mM imidazole fractions.

On its turn Ndk samples were collected at 200 and 300 mM imidazole

fractions. Additionally, after being collected from the detoxing column, Ndk

sample was readily passed through a 0.2 µm PuradiscTM

filter (Whatman), which

removes any bacterial cells present from our sample. After this, protein was

concentrated with the 3 kDa cut-off column but did not pass through the 100 kDa

cut-off column.

Page 55: Discovering and exploiting bacterial proteins as ...repositorio.ul.pt/bitstream/10451/9665/1/ulfc103147_tm_goncalo_sil… · Discovering and exploiting bacterial proteins as anticancer

31

The quantification of MPT 63 and Ndk was done according to the Bradford

method using a bovine serum albumin (BSA) standard solution kit (Thermo

Scientific). Ten microliters of BSA standard solutions and 10 µl of protein sample

diluted in PBS (1:10, 1:50 and 1:100) were loaded, in duplicated, into a 96-well

microplate and 200 µl of diluted Bio-Rad Protein assay dye concentrate (1:5) were

added to each well. The microplate was incubated in the dark, at room

temperature for 15 minutes, and OD was measured at 595 nm using

SPECTROstar Nano (BMG LABTECH) microplate reader. PBS was used as

blank. To convert the obtained mass concentration in molar concentration it were

considered MPT 63 and Ndk molecular weights: 16 kDa and 15 kDa respectively.

3.2 Cell culture and human cell lines

In this study, two human breast cancer cell lines were used: MCF-7/AZ.Mock

and MCF-7/AZ.Pcad [(transduced with empty vector and CHD3/P-cadherin

cDNA, respectively, and kindly provided by Doctor Joana Paredes (IPATIMUP)].

It were also used a human lung cancer cell line A549 and a human bronchial

epithelial cell line 16HBE14o- [kindly provided by Doctor Dieter Gruenert

(University of California, San Francisco, USA].

MCF-7/Az.Mock and MCF-7/Az.Pcad cell lines were routinely maintained in

Dulbecco’s Modified Eagle Medium (DMEM, Invitrogen) and A549 cells were

maintained in Ham’s F-12 medium (Gibco). Both cell media were supplemented

with 10% of heat-inactivated fetal bovine serum (FBS, Lonza), 100 IU/ml of

penicillin (Invitrogen) and 100 mg/ml of streptomycin (Invitrogen). In each

passage, culture medium was firstly removed and cells were washed twice with

PBS and trypsinized before being resuspended in fresh medium and transferred to

a new flask with a 1:3 dilution (1 mL of cell suspension to 2 mL of fresh

medium). Cell scrapers were used to remove A549 cells from the bottom of the

flask, instead of using trypsine.

Human bronchial cell line 16HBE14o- was routinely maintained in

fibronectin/vitrogen coated flasks in Minimum Essential Medium with Earle’s salt

(MEM, Gibco) supplemented with 10% FBS, 0.292 g/L of L-Glutamine (Sigma-

Aldrich) and 100 U/mL of Penicillin/Streptomycin (Gibco). On each passage cells

Page 56: Discovering and exploiting bacterial proteins as ...repositorio.ul.pt/bitstream/10451/9665/1/ulfc103147_tm_goncalo_sil… · Discovering and exploiting bacterial proteins as anticancer

32

were washed with Hepes buffered saline (HBS, Sigma-Aldrich) and detached

using PET solution (Sigma-Aldrich), which contained HBS, 10%

polyvinylpyrrolidone solution (PVP), 0.2% EGTA in HBS and 0.25% trypsin in

0.02% EDTA.

Both cell lines here mentioned were maintained in a humidified incubator at

37ºC, with a 5% carbon dioxide atmosphere (Binder CO2 incubator C150).

3.3 Inhibitors treatment

3.3.1 Lysosome and proteasome inhibitors

In order to test each inhibitor, all cells were previously treated with azurin at

different cell concentrations (0, 50 and 100 µM).

Breast cancer cells were seeded in a 6-well plate with 5x105 cells (MFC-

7/AZ.Pcad) for each well. The following day, all cells were treated with azurin or

PBS (control) and fresh medium was added. After 32 hours cell were treated with

20 µl of the lysosome inhibitor, ammonium chloride (NH4Cl 0.75 M), or 20 µl of

distilled water as control. In the case of the proteasome inhibitor MG-132 (Sigma-

Aldrich), 2.5 µl of this compound or 2.5 µl of ethanol absolute (control) were

added. Both inhibitors were applied during 16 hours, after which time the cells

were lysed.

3.3.2 Azurin internalization inhibitor

In order to test the effects of MβCD on azurin internalization, breast cancer

cells were seeded with the same number of cells used for the experiments with

lysosome and proteasome inhibitors. The following day fresh medium was added

and cells were treated with PBS (control) or azurin (50 and 100 µM) and

simultaneously with 20 µl of MβCD (Sigma-Aldrich) or 20 µl of Dimethyl

sulfoxide (DMSO), used as control. After 8 hours culture medium was exchanged

and, at the end of 24 hours, protein lysates were prepared.

Page 57: Discovering and exploiting bacterial proteins as ...repositorio.ul.pt/bitstream/10451/9665/1/ulfc103147_tm_goncalo_sil… · Discovering and exploiting bacterial proteins as anticancer

33

3.4 Protein lysates

Culture media were firstly removed from the 6-well plates and washed twice

with 3 mL of PBS. Cell lysis was accomplished by adding 100 µl of catenin lysis

buffer (1% Triton X-100, 1% Nonidet P-40 in deionized PBS) and a 1:7 dilution

of proteases inhibitor cocktail (Roche Diagnostics) to each well. Plates were kept

at 4º C for 10 minutes, after which time cells were scratched from the wells,

ressuspended and transferred to microtubes. These tubes were vortexed 3 times,

for 10 seconds, and centrifuged at 14000 rpm with a constant temperature of 4º C

for 10 minutes (B.Braun Sigma-Aldrich 2K15). If the cell lysates weren’t used

immediately they were stored at -80ºC.

Protein quantification was done according to the Bradford method as explained

earlier for MPT 63 and Ndk quantification. The only difference was that protein

samples were diluted in PBS with a 1:4 dilution for quantification. Protein

samples were prepared to have 20 µg of total protein dissolved in sample buffer

(Laemmli with 5% (v/v) 2-beta-mercaptoethanol and 5% (v/v) bromophenol blue)

and were stored at -20º C if not used immediately.

3.5 Sodium dodecyl sulfate polyacrylamide gel electrophoresis (SDS-

PAGE)

Polyacrylamide gels were assembled with a gel caster (Amersham Biosciences)

and polymerized between a glass and a porcelain plate separated by two spacers.

Approximately 3 mL of resolving gel were casted and simultaneously overlaid

with isopropanol to create a smooth surface above the gel. After gel

polymerization, the isopropanol was removed and the stacking gel was casted to

the top of the plates, with the comb (10 wells) already inserted. The resolving gel

and stacking gels were left to polymerize for approximately 20 minutes.

In order to separate azurin, MPT 63 and Ndk it was used a resolving gel with a

15% acrylamide concentration, whereas to separate E- and P- cadherins it was

used a resolving gel with a lower acrylamide concentration (8%). Resolving and

stacking gel composition is shown in Table 5.

Page 58: Discovering and exploiting bacterial proteins as ...repositorio.ul.pt/bitstream/10451/9665/1/ulfc103147_tm_goncalo_sil… · Discovering and exploiting bacterial proteins as anticancer

34

Table 5 - Resolving Gel 8% and 15% composition for one gel

Resolving Gel 8% Resolving Gel 15% Stacking Gel 5%

Distilled water 2.3 mL 850 µl 1.35 mL

Acrylamide-bisacrylamide 30% 1.35 mL 1.9 mL 335 µl

Tris Base 1.25 mL (1.5 M) 950 µl (1.5 M) 250 µl (1M)

10% SDS 50 µl 34 µl 20 µl

APS 50 µl 34 µl 20 µl

TEMED 3 µl 1.5 µl 2 µl

Before electrophoresis, the comb was removed from the top of the gel and the

gel cassette was placed in the electrophoresis apparatus (Amersham Biosciences)

and electrophoresis buffer was added. Protein samples were boiled in dry bath at

100º C, for 5 minutes, and 20 µl of each sample were loaded to the gel wells, as

well as 3 µl of PageRuler Prestained Protein Ladder (Fermentas).

The electrophoresis voltage was set to 150 V (azurin, MPT 63 and Ndk), and

120 V (E- and P-cadherin).

After electrophoresis the gels containing purified MPT 63 and Ndk proteins

were stained with Coomassie Brilliant Blue staining solution for 2 hours and

destained for half an hour with acetic acid destaining solution until the protein

bands were clearly visible against a translucid background. The destaining

solution was removed and the gel was washed with water and finally dried.

3.6 Western blotting

For western blotting purposes protein samples were separated by SDS-PAGE

following the protocol described earlier. After electrophoresis proteins were

transferred into nitrocellulose membranes using Trans-Blot Turbo RTA Transfer

Kit (BioRad). Protein transference was accomplished by using Trans-Blot Turbo

Transfer System (BioRad) at 2.5 V for 7 minutes. Membranes were blocked with

5% (w/v) non-fat dry milk in PBS-Tween-20 (0.5% v/v) for 1 hour and incubated

with primary antibodies overnight at 4° C. On the next day membranes were

washed three times with PBS-Tween 20 for 5 minutes and incubated for 1 hour, at

room temperature, with secondary antibodies conjugated with horseradish

peroxidase enzyme. After washing the membranes for 5 minutes with PBS-Tween

Page 59: Discovering and exploiting bacterial proteins as ...repositorio.ul.pt/bitstream/10451/9665/1/ulfc103147_tm_goncalo_sil… · Discovering and exploiting bacterial proteins as anticancer

35

20, proteins were detected by adding ECL reagent (Pierce) as substrate and

chemiluminescence was capture by Fusion Solo system (Vilber Fourmat). The

band intensity obtained was measured with ImageJ software, and the results

represent the ration between the signal intensities in azurin treated and non-treated

samples. Moreover, cadherin levels were normalized by the respective actin level

in each sample.

Anti-E-cadherin (HECD 1, Sigma-Aldrich), anti-azurin (AB0048-200 SicGen)

and anti-actin (sc-1616, Santa Cruz Biotechnology) primary antibodies were

diluted 1:1000 in 5% non-fat milk, whereas anti-P-cadherin (clone 56, BD

Transduction Laboratories) antibody was diluted 1:500.

Anti-mouse (sc-2005, Santa Cruz Biotechnology) secondary antibody was used

against cadherins, whereas anti-goat (sc-2354, Santa Cruz Biotechnology)

secondary antibody was used for actin and azurin.

3.7 MTT cell viability assay

MTT [3-(4,5 dimethylthiazol-2-yl-2,5 tetrazolium bromide) ; Sigma-Aldrich]

assay was used to determine cell viability in breast (MCF-7/AZ.Mock) and lung

(A549) cancer cells, as well as a normal lung cell line (16HBE14o-) after

exposing them to azurin, MPT 63 and Ndk. This assay is based on the reduction

of MTT, by the mitochondrial dehydrogenase of intact cells, to a purple formazan

product which can be quantified. Breast cancer cells (MCF-7/AZ.Mock) were

suspended in a 96-well plate at a density of 1 x 104 cells per well, whereas lung

cancer cells (A549) were seeded at a density of 5 x 103 cells per well. A normal

lung cell line, 16HBE14o-, was used as control and seeded at a density of 1 x 104

cells per well. After 24 hours, medium was exchanged and azurin, MPT 63 and

Ndk were administrated. Both proteins were administrated at 10, 25, 50 and 100

uM, and PBS was used as control. After another 24 hours medium was removed

and a new protein dosage was applied. Forty eight hours after the first protein

administration, 20 µl of MTT solution (5 mg/ml, Sigma-Aldrich) were added to

each well and incubated for 4 hours. The supernatant was carefully removed and

150 ul of MTT solvent (3.4 µl of HCl, 10 µl of Nonidet-P40, 10 ml of

isopropanol) was added to each well. The 96-well plate was shacked in an orbital

Page 60: Discovering and exploiting bacterial proteins as ...repositorio.ul.pt/bitstream/10451/9665/1/ulfc103147_tm_goncalo_sil… · Discovering and exploiting bacterial proteins as anticancer

36

shaker for 15 minutes at 300 rpm, and SPECTROStar Nano microplate reader was

used to measure the absorbance at 590 nm (with a 620 nm reference filter). Each

condition tested was performed in triplicated.

3.8 Matrigel invasion assay

Matrigel invasion assay was performed using BD Biocoat MatrigelTM

Invasion Chambers with an 8 µm pore size PET membrane with a thin

MatrigelTM layer (BD Biosciences), following manufacturer’s instructions.

Chambers were pre-incubated with serum-free medium during 2 hours at 37ºC. In

the chamber’s upper compartment 2.5 x 104 A549 cells were added with complete

medium and Ndk (at 0, 50 and 100 µM), whereas in the lower compartment only

complete medium was added. After 24 hours at 37ºC, non-invasive cells were

cleared and chambers were washed four times with PBS. Cells were fixed in cold

methanol during 10 minutes at 4ºC. Invasive cells attached to the lower surface

were stained with DAPI and counted under the microscope (Zeiss).

3.9 Bioinformatics

3.9.1 Sequence analysis and secondary structures

Primary protein sequences from bacterial Ndks and all human Nm23 family

members were retrieved from the National Center for Biotechnology Information

(NCBI; http://www.ncbi.nlm.nih.gov/pubmed/). Multiple sequence alignment

between human and bacterial Ndks was performed by ClustalW algorithm default

settings (http://www.ebi.ac.uk/Tools/msa/clustalw2/) and edited on Bioedit

software (http://www.mbio.ncsu.edu/). Secondary structures of E. coli Ndk and

human Nm23-H1 were represented according to structural information provided

by their solved structures on Protein Data Bank (PDB, http://www.rcsb.org/).

Swiss-Model default settings were used (http://swissmodel.expasy.org/) in order

to predict P. aeruginosa Ndk secondary structure and a three-dimensional model,

based on the closest template found in the database.

Page 61: Discovering and exploiting bacterial proteins as ...repositorio.ul.pt/bitstream/10451/9665/1/ulfc103147_tm_goncalo_sil… · Discovering and exploiting bacterial proteins as anticancer

37

3.9.2 Phylogenetic and structural alignment analysis

Multiple sequence alignment, generated by ClustalW algoritm, was used to

construct the phylogenetic tree representation using Splitstree software

(http://www.splitstree.org/) on default settings. All currently protein three-

dimensional structures available on PDB database were retrieved and added to the

phylogenetic tree, as well as the Ndk P. aeruginosa model built. Structural

alignment was performed between Nm23-H1 and Ndk P. aeruginosa model using

PyMOL software (http://www.pymol.org/) default settings.

Page 62: Discovering and exploiting bacterial proteins as ...repositorio.ul.pt/bitstream/10451/9665/1/ulfc103147_tm_goncalo_sil… · Discovering and exploiting bacterial proteins as anticancer
Page 63: Discovering and exploiting bacterial proteins as ...repositorio.ul.pt/bitstream/10451/9665/1/ulfc103147_tm_goncalo_sil… · Discovering and exploiting bacterial proteins as anticancer

39

4. RESULTS

Recently, several experiments revealed that azurin can cause a decrease in P-

cadherin levels in MCF-7/AZ.Pcad breast cancer cells, which display invasive

behavior and express high levels of this protein [45]. Interestingly, azurin did not

exhibit a similar behavior on E-cadherin levels, since they remained unchanged.

These results were also corroborated with immunofluorescence analysis regarding

the membrane localization of both cadherins. Since it was found that this decrease

in protein level was not a consequence of changes in gene expression, it was

suggested that post-transcriptional processes should be involved in this process

[45]. Further observations, regarding microarray analysis on mRNA profiles of

MCF-7/AZ.Pcad cells treated with azurin, showed that several genes were up-

regulated in comparison with untreated cells (Bernardes et al., 2013, submitted for

publication). Interestingly, some of these genes were associated with membrane

organization, vesicle-mediated and endosome transport, or even with lysosome.

This work intends to pursue those evidences and to demonstrate that azurin can

display an important role on mediating P-cadherin degradation through proteolytic

systems.

Tacking account that azurin can enter cells preferentially through lipid rafts

and since tumor cells exhibit a higher number of lipid rafts than normal cells, it

has been suggested that P-cadherin can be mainly localized on these regions.

Azurin internalization on tumor cells through lipid rafts involves the formation of

endocytic vesicles, which thereby make us believe that P-cadherin can also suffer

endocytosis along with this internalization process. These vesicles that sequester

P-cadherin can cause its removal from the cell membrane and further degradation

through proteolytic systems, as mentioned before.

In order to support this theory we sought to use MβCD, which has been known

to remove cholesterol from lipid rafts, thereby inhibiting azurin internalization. By

using this inhibitor, we intended to verify what impact occurs on P-cadherin level

in MCF-7/AZ.Pcad cells treated with azurin.

Page 64: Discovering and exploiting bacterial proteins as ...repositorio.ul.pt/bitstream/10451/9665/1/ulfc103147_tm_goncalo_sil… · Discovering and exploiting bacterial proteins as anticancer

40

4.1 Proteolytic pathways inhibitors effect on P-cadherin level

The mechanism by which azurin cause a decrease on P-cadherin level in breast

cancer cells is not fully understood, although it was excluded an effect on gene

expression. Post-transcriptional processes should probably be relevant in this case

and, in fact, a microarray analysis on MCF-7/AZ.Pcad cells previously treated

with azurin revealed that several genes up-regulated involved in vesicle-mediated,

endosome transport and lysosome. One possible mechanism by which azurin

mediates P-cadherin decrease could then be related to its degradation by

proteolytic systems. We intended to support this theory with our results.

Therefore we selected two major proteolytic systems present in eukaryotic cells

and tested a lysosome inhibitor (ammonium chloride) and a proteasome inhibitor

(MG-132) to inhibit them. We tested the effect of both inhibitors on an invasive

breast cancer cell line, MCF-7/AZ.Pcad. We performed a 48 hours assay, were

azurin was in contact with tumor cells during the first 32 hours, after which time

medium was removed and each inhibitor was added in the last 16 hours. Cell

samples were analyzed by western blot and protein bands were quantified by

ImageJ software. Cadherin levels were normalized by their respective actin levels

and the average results are shown in Figure 7 and Figure 8.

Observing the results obtained from western blot we can see that MCF-

7/AZ.Pcad cells, on which the two inhibitors were not administrated, showed a

decrease tendency in P-cadherin level when azurin’s concentration increases. On

the other hand, in the presence of the inhibitors, P-cadherin level increases at the

same time as azurin concentration administrated increases.

Regarding to E-cadherin level, it suffered a slight increase when cells where

treated with each inhibitor. In samples untreated with MG-132, on the other hand,

we see a decrease in E-cadherin level when azurin concentration increases. In

samples untreated with ammonium chloride we observed that E-cadherin level

remained constant. Although actin level between samples were not constant, the

charts presented in Figure 7 and Figure 8 include average cadherin level

normalized with their respective actin level, and exhibit the same tendency as the

one shown by western blot results.

Page 65: Discovering and exploiting bacterial proteins as ...repositorio.ul.pt/bitstream/10451/9665/1/ulfc103147_tm_goncalo_sil… · Discovering and exploiting bacterial proteins as anticancer

41

The results obtained clearly show that both the lysossome and the proteosome

are involved in P-cadherin degradation, since when they are inhibited, P-cadherin

does not continue to decrease as azurin concentration increases, but rather the

oposite, it starts to accumulates inside the cells. Thereby it seems that both these

proteolytic systems have a synergetic action on P-cadherin degradation, when

tumor cells are treated with azurin.

Figure 7 – (A): Western Blot analysis of the effect of lysossome inhibitor, ammonium chloride

(NH4Cl), on E and P-cadherins of MCF-7/AZ.Pcad cells, previously treated with azurin at 0, 50

and 100 μM. Azurin and the inhibitor were administrated at 0 and 32 hours, respectively, in a 48

hours assay. (B): Charts illustrated represent average percentage values of protein level for E and

P-cadherins which signal was normalized with actin levels.

Page 66: Discovering and exploiting bacterial proteins as ...repositorio.ul.pt/bitstream/10451/9665/1/ulfc103147_tm_goncalo_sil… · Discovering and exploiting bacterial proteins as anticancer

42

Figure 8 – (A): Western Blot analysis of the effect of proteasome inhibitor, MG-132, on E and P-

cadherins of MCF-7/AZ.Pcad cells, previously treated with azurin at 0, 50 and 100 μM. Azurin

and the inhibitor were administrated at 0 and 32 hours, respectively, in a 48 hours assay. (B):

Charts illustrated represent average percentage values of protein level for E and P-cadherins which

signal was normalized with actin levels.

4.2 Azurin internalization

Azurin’s ability for entering cancer cell lines, such as those from breast cancer,

has been known for some years now. Previously, it had been demonstrated, by

confocal imaging, that an Alexa-labeled p28-azurin peptide was less efficient

entering MCF-7 cells, upon MβCD treatment [81]. This compound causes

cholesterol depletion, thereby disrupting structures as lipid rafts which are rich in

cholesterol [52]. In this work we tested the effect of MβCD on azurin’s

Page 67: Discovering and exploiting bacterial proteins as ...repositorio.ul.pt/bitstream/10451/9665/1/ulfc103147_tm_goncalo_sil… · Discovering and exploiting bacterial proteins as anticancer

43

internalization in the human breast cancer cell line MCF-7/AZ.Pcad. Cells were

incubated with azurin and MβCD for 8 hours, and left another 24 hours with fresh

medium. Western blot was performed in order to evaluate the effects of this

inhibitor and two different azurin concentrations were tested (50 and 100 μM).

Cells treated with DMSO were used as negative control since MβCD was

dissolved in this reagent. PBS was used as double negative control.

The results of this experiment are shown in Figure 9, where it can be seen that

anti-azurin antibody was only detected in samples which were previously treated

with azurin at 50 and 100 μM. Anti-azurin antibody signal was stronger in

samples treated with azurin at 100 μM than the ones treated with a 50 μM

concentration. This observation was verifiable in both cells treated with DMSO

and MβCD.

Analyzing azurin corresponding bands it is possible to identify a decrease in

their intensity when comparing samples treated with DMSO and MβCD. This

decrease was verified in both two azurin concentration used, although it was

clearer between DMSO and MβCD samples treated with azurin at 50 μM. Actin

level was constant between all samples thus showing that azurin and MβCD

treatments did not affect this protein.

β-actin

Azurin

Figure 9 - Effects of MβCD on azurin’s internalization on MCF-7/AZ.Pcad cells after 8 hours of

exposure to this inhibitor and 24 hours with fresh medium (visualized by western blot). PBS was

used as double negative control. DMSO was used as negative control. Azurin was tested at two

different concentrations (50 and 100 μM).

Page 68: Discovering and exploiting bacterial proteins as ...repositorio.ul.pt/bitstream/10451/9665/1/ulfc103147_tm_goncalo_sil… · Discovering and exploiting bacterial proteins as anticancer

44

4.3 Inhibition of azurin’s effect on E and P-cadherins level using MβCD

Recently it has been described that azurin can cause P-cadherin levels to

decrease in MCF-7/AZ-Pcad cell line, without affecting its E-cadherin levels [82].

Additionally, as we mentioned earlier, microarray analysis on the same cell line

revealed that cells treated with azurin show several up-regulated genes involved

with membrane organization and vesicular transport (Bernardes et al., 2013,

submitted for publication). Thereby, it was of interest within this work to evaluate

the effect of MβCD on E- and P-cadherin levels through a possible decrease in

azurin’s internalization on MCF-7/AZ.Pcad cells.

In Figure 10 we can observe a decrease in P-cadherin level in samples treated

simultaneously with MβCD and azurin at 50 and 100 μM, comparing to control. It

seems that even though MβCD causes a decrease in azurin internalization (as

Figure 9 shows), a decrease in P-cadherin level still continues to occur, which can

mean that MβCD contributes to this effect. Regarding to the samples treated with

PBS and DMSO there were no significant differences on P-cadherin level, which

indicates that DMSO does not affect P-cadherin level. E-cadherin level seems to

remain constant in all samples, apparently not being affected with azurin and

MβCD treatments.

These observations represent an important breakthrough in studying the

mechanisms behind azurin and P-cadherin interactions. It appears that, when

MβCD is administrated alone there are no visible effects on P-cadherin level.

However, when azurin is added simultaneously with this inhibitor, it occurs a

decrease in P-cadherin level when both two different protein concentrations are

tested. From previous studies it was known that azurin does not affect E-cadherin

level, which was also seen here when MβCD was tested. Additionally,

microarrays analysis mentioned earlier, showed that MCF-7/AZ.Pcad cells

previously treated with azurin, display several up-regulated genes, including some

involving membrane organization or vesicle-mediated transport. Considering

these evidence altogether we believe that azurin and MβCD have a synergetic

effect on P-cadherin degradation. This inhibitor helps to deplete cholesterol from

lipid rafts at the cell membrane, where azurin enters preferentially, which could

mean that diminishing azurin internalization could cause a smaller decrease on P-

Page 69: Discovering and exploiting bacterial proteins as ...repositorio.ul.pt/bitstream/10451/9665/1/ulfc103147_tm_goncalo_sil… · Discovering and exploiting bacterial proteins as anticancer

45

cadherin level. However we saw the opposite, which can probably mean that P-

cadherin is highly present in lipid rafts regions. Thereby a depletion of these sites

by MβCD and the formation of transporting vesicles in the cell membrane, caused

by azurin as microarray analysis suggests, can mean that P-cadherin may suffer

endocytosis and vesicles containing this protein should be conducted to

proteolytic systems, as the lysosome. Since E-cadherin was not affect by azurin or

MβCD, we suggest that this protein may be highly expressed in other sites on the

cell membrane but poorly expressed in lipid rafts.

β-actin

β-actin

E-cadherin

P-cadherin

Figure 10 – Western blot showing the effect of azurin’s internalization inhibitor MβCD on E- and

P-cadherin levels in MCF-7/AZ.Pcad cells after 8 hours of exposure and 24 hours with fresh

medium. PBS was used as double negative control. DMSO was used as negative control. A

decrease in P-cadherin level, between control samples and samples treated with MβCD and azurin,

is shown in the figure.

4.4 Bacterial proteins to treat cancer

4.4.1 Purified proteins

Before applying MPT 63 and Ndk proteins in MTT (cell viability) assays it

was mandatory to establish if these proteins were successfully purified using the

purification methods already described. As we can see in Figure 11, an SDS-

PAGE was performed in order to confirm this, and the single bands visualized on

Page 70: Discovering and exploiting bacterial proteins as ...repositorio.ul.pt/bitstream/10451/9665/1/ulfc103147_tm_goncalo_sil… · Discovering and exploiting bacterial proteins as anticancer

46

the gel clearly show that both proteins were purified from E. coli SURE with

success.

~15 kDa

MPT63 Ndk

Figure 11 - 15% SDS-PAGE of MPT 63 and Ndk purified from E. coli SURE strain. Proteins

appear as a single band at approximately 15 kDa, according to their expected molecular weight.

4.5 MTT cell viability assays

Azurin has already been described as a protein capable of producing effects on

several tumor cell lines, including some derived from humans, namely through

p53 stabilization and apoptosis induction. In this work we tested azurin, as well as

MPT 63 and Ndk, other two bacterial proteins with different origins and

properties.

Since MPT 63 shares some structural similarities with azurin and some

experiments supported anti-tumor activity of this protein, we sought to verify the

same properties [63]. Human Ndk-A isoform (or Nm23-H1) has been described as

an anti-metastatic protein, and it was of interest to verify if bacterial Ndk from P.

aeruginosa could exhibit similar properties, as well as cytotoxic activity, since

human and bacterial Ndks share a high homology level, as we will show after

bioinformatic analysis (section 4.7).

Both cytotoxic effects of these proteins were tested by MTT cell viability

assays, that were performed in two tumor cell lines from breast (MCF-

7/AZ.Mock) and lung (A549) and one normal lung epithelial cell line

(16HBE14o-).

Page 71: Discovering and exploiting bacterial proteins as ...repositorio.ul.pt/bitstream/10451/9665/1/ulfc103147_tm_goncalo_sil… · Discovering and exploiting bacterial proteins as anticancer

47

4.5.1 Azurin

In order to verify if azurin display its cytotoxic properties against tumor cell

lines in a dose dependent-manner, MTT assay was performed using increasing

number of doses and exposure time. Azurin was administrated on MCF-

7/AZ.Mock for 48 (one dose) and 72 hours (three doses). A549 tumor cells were

incubated during 48 hours, where it was tested one and two protein doses. Five

different azurin concentrations (0, 10, 25, 50 and 100 µM) were tested and

cytotoxicity percentages are presented for each condition tested.

Regarding MCF-7/AZ.Mock cells our results shows that azurin caused a

general increase in cell cytotoxicity across increasing concentrations of this

protein, in both 48 and 72 hours assay, as it can be seen in

Figure 12. Moreover, we see that this increase in cytotoxicity occurs not only as

azurin concentration increases, but also when we increase the exposure time and

number of doses applied. About 26% of cell population was killed using three

doses of this protein during 72 hours, in MCF-7/AZ.Mock cells.

A549 cells exhibited a similar cytotoxic response across the different azurin

concentrations tested, as the one observed in MCF-7/AZ.Mock cell line, when

azurin was used. Contrarily to what we have seen on MCF-7/AZ.Mock cells,

adding a second dose of azurin to A549 cells does not appear to cause a

significant increase in cytotoxicity, which was only verified when the highest

protein concentration was used.

Overall we can verify that azurin caused a slightly higher cytotoxicity on MCF-

7/AZ.Mock than on A549 tumor cell line. Statistical analysis using T-student test

on these results revealed a significant value when A549 cells were treated with

azurin at the highest concentration, as shown in

Figure 12. Considering a 0.05 significance level tested, we may assume that

such decrease in cell viability was provoked by azurin administration and its

cytotoxic activity.

Page 72: Discovering and exploiting bacterial proteins as ...repositorio.ul.pt/bitstream/10451/9665/1/ulfc103147_tm_goncalo_sil… · Discovering and exploiting bacterial proteins as anticancer

48

Figure 12 – Cytotoxicity (%) caused by azurin on MCF-7/AZ.Mock during 48 h (one dose) and 72

h (three doses), and A549 48 h (one and two doses). Both cell lines were tested with 5 different

azurin concentrations (0, 10, 25, 50, and 100 μM). Significant values, p-value<0.05 with the

Student t-test, are shown as asterisks (*).

4.5.2 MPT 63

In the same way as azurin, MPT 63 was tested in two different tumor models.

This protein was administrated in MCF-7/AZ.Mock cells during 48 (one dose)

and 72 hours (three doses), and in A549 for 48 hours (two doses). When it was

applied on MCF-7/AZ.Mock breast cancer cells, MPT 63 caused an increase in

cell citotoxicity across all different concentrations tested, as it can be seen in

Figure 13. It can also be observable that MPT 63 cytotoxicity increases, in this

cell line, when a higher number of protein doses and exposure time are applied.

Regarding A549 cells, MPT 63 caused only a maximal 10% decrease on cell

viability, which is about half of the cytotoxic level seen for azurin, within the

same conditions tested.

Tacking an overview of these results we observed that, once again, A549 cell

line appears to be not as sensitive, as MCF-7/AZ.Mock cells, to cytotoxic effects

caused by MPT 63. It also seems that this protein does not exhibit a cytotoxic

activity as higher as azurin demonstrates.

*

Page 73: Discovering and exploiting bacterial proteins as ...repositorio.ul.pt/bitstream/10451/9665/1/ulfc103147_tm_goncalo_sil… · Discovering and exploiting bacterial proteins as anticancer

49

Figure 13 – Cytotoxicity (%) caused by MPT 63 on MCF-7/AZ.Mock during 48 h (one dose) and

72 h (three doses), and A549 during 48 h (two doses). Both cell lines were tested with 5 different

MPT 63 concentrations (0, 10, 25, 50, and 100 μM).

4.5.3 Ndk

Ndk was also administrated as double dose, for 48 hours, against breast and

lung cancer cell lines (MCF-7/AZ.Mock and A549), in order to verify if it could

require more doses to provoke cytotoxic effects in those cells. Additionally, a lung

epithelial cell line (16HBE14o-) was used to analyze if Ndk could exhibit

selective cytotoxic effects against cancer cells or if it was effective against normal

cell lines as well. A 72 hours assay (three doses) was also performed with MCF-

7/AZ.Mock tumor cell line.

Analyzing the results showed on Figure 14 we can see that, when two doses of

Ndk are administrated for 48 or 72 hours, with two or three doses respectively,

cell cytotoxicity increases in MCF-7/AZ.Mock and A549 cells, across each

increasing protein concentrations tested. The higher cytotoxicity values were

found when the 100 µM concentration was used. In this case, A549 cells suffered

a 27% decrease in cell survival, which was the same value verified in MCF-

7/AZ.Mock cells after three doses were applied during 72 hours. Therefore, it

seems that also Ndk cytotoxic effect on both tumor cell lines follows a dose and

time dependent tendency. More interestingly, Ndk appears to be slightly more

Page 74: Discovering and exploiting bacterial proteins as ...repositorio.ul.pt/bitstream/10451/9665/1/ulfc103147_tm_goncalo_sil… · Discovering and exploiting bacterial proteins as anticancer

50

effective on A549 lung cancer cell line, which was not verified for azurin or MPT

63.

Another interesting fact retrieved from Figure 14 is that Ndk has shown a

similar cytotoxic effect as potent as azurin, in MCF-7/AZ.Mock cells. A similar

observation was made in A549 tumor cell line as well, although for a two dose

treatment only, as already described.

Concerning to 16HBE14o- cells, it can be seen, in Figure 14, that Ndk

produces cytotoxic effect on this normal cell line, although not as extended as in

the breast and lung cancer cell lines tested. Moreover, in all Ndk concentrations

tested, the lower cytotoxicity values are seen in 16HBE14o- cell line. This can

possibly indicate that Ndk displays selective cytotoxicity towards tumor cell lines,

in the same way as azurin does.

Statistical analysis using T-student test revealed significant values, with a

significance level inferior to 0.05, in A549 cells treated with Ndk. These results

strongly support the fact that Ndk’s cytotoxic effect is responsible for a decrease

in cell viability in this tumor cell line. When we used the same T-student function

on data retrieved from MTT assay using 16HBE14o- cells, we also found that

samples treated with Ndk showed significant values, thereby there is strong

evidence that Ndk clearly has cytotoxic effect on this epithelial lung cell line.

Figure 14 - Cytotoxicity (%) caused by Ndk on MCF-7/AZ.Mock during 48 h (two doses) and 72

h (three doses), A549 48 h (two doses) and 16HBE14o- during 48 h (two doses). Both cell lines

were tested with 5 different Ndk concentrations (0, 10, 25, 50, and 100 μM). Significant values, p-

value<0.05 with the Student t-test, are shown as asterisks (*).

* *

*

* *

Page 75: Discovering and exploiting bacterial proteins as ...repositorio.ul.pt/bitstream/10451/9665/1/ulfc103147_tm_goncalo_sil… · Discovering and exploiting bacterial proteins as anticancer

51

4.6 Matrigel invasion assay

Since Ndk exhibited significant cytotoxicity towards tumor cell lines, we

intended to test if it could also display anti-metastatic activity, like Nm23-H1,

against A549 which is a highly invasive tumor cell line. Ndk was administrated,

as single dose, to A549 cells in a matrigel invasion system, during 24 hours.

However, only a 6-8% decrease in cell invasion was observed in Figure 15, which

is not significant. Moreover, no differences were seen between the two different

protein concentrations tested.

Figure 15 – Cell invasion (%) in a Matrigel invasion assay performed on A549 highly invasive

tumor cells, which were treated with Ndk at 0, 50 and 100 μM, during 24 hours.

4.7 Bioinformatic analysis on human Nm23 and bacterial Ndks

As mentioned earlier, a human Ndk (Nm23-H1) has been described as an

anti-metastatic protein is several human tumor models, even though exhibiting

opposite results in a minority of cases. In this work we intended to use a bacterial

Ndk, from P. aeruginosa, in order to evaluate if other Ndks manage to inflict the

same cytotoxic and/or anti-metastatic effects on human cancer cells. Since our

MTT cell viability assays also revealed promising results about the cytotoxicity of

this protein, we thought to explore how similar human Nm23 proteins and

bacterial Ndks can be, especially Nm23-H1 (which has anti-metastatic properties)

and P. aeruginosa Ndk, on which we hope to have a similar effect. With that

Page 76: Discovering and exploiting bacterial proteins as ...repositorio.ul.pt/bitstream/10451/9665/1/ulfc103147_tm_goncalo_sil… · Discovering and exploiting bacterial proteins as anticancer

52

thought in mind we used several bio-informatic tools, thereby taking a different,

but quite relevant approach on this subject.

A multiple sequence alignment was performed using ClustalW algorithm to

align primary protein sequences from all human Nm23 family members, and some

bacterial Ndks, namely from P. aeruginosa. This alignment is shown in Figure 16,

and as it can be seen, there is a clear similarity between the sizes of each bacterial

Ndk sequence. On the other hand, regarding human Nm23 sequences we face a

great disparity, since some of them (as Nm23-H1) have approximately the same

size of bacterial Ndks but others (like Nm23-H8) are larger. The most significant

and interesting fact that is raised from the multiple alignment is that between all

different Ndks it appears to be several blocks composed by segments of

approximately 40/50 amino acid residues exhibiting a high level of similarity

between the sequences. Since these conserved regions within the protein

sequences, could be attributable to important protein secondary structures, the

secondary structure of E. coli Ndk is represented in Figure 16. If we look at this

structure it is observable that typical secondary elements, as β-sheets or α-helices

for instance, clearly overlap with the blocks of amino acids aligned. This

observation could possibly demonstrate the importance of these conserved

structures, throughout evolution, for the activity of this protein. Moreover, this

may help to explain the fact that P. aeruginosa Ndk seems to exhibit similar

cytotoxic effects on tumor cells, as human Nm23-H1.

Analyzing the percent identity matrix, in Table 6, generated by the multiple

alignment results, we can observe that all bacterial Ndks sequences share higher

identity values (44-65%) between themselves. Relatively to the identity shared

between bacterial Ndks and human Ndks, the highest value was reached with M.

tuberculosis and M. bovis sequences against Nm23-H3 protein (51%), although

not far from Nm23-H1 (45%). Regarding exclusively to Nm23-H1, and the P.

aeruginosa Ndk used in our MTT assays, there is an approximately 43% identity

between these two protein primary sequences. In the case of this specie, the

highest identity value was also found against Nm23-H3 (45%).

Through the evaluation of the results in Table 6, it is possible to verify that

Nm23-H10 is by far the Ndk that reveals the lowest identity level across all Ndks

Page 77: Discovering and exploiting bacterial proteins as ...repositorio.ul.pt/bitstream/10451/9665/1/ulfc103147_tm_goncalo_sil… · Discovering and exploiting bacterial proteins as anticancer

53

analyzed, including between the other members of human the Nm23 protein

family. Regarding human Nm23 proteins, the ones that could be more related to

each other are Nm23-H1, -H2, -H3 and -H4, since they share between 53-88%

identity level, where the highest value is seen between Nm23-H1 and Nm23-H2.

Taking into account the results obtained with multiple sequence alignment, the

E. coli Ndk secondary structure revealed and the overlapping between secondary

elements and conserved amino acid sequences, we sought to compare Nm23-H1

and P. aeruginosa Ndk regarding their secondary structure. In Figure 17 it can be

seen the result of the alignment between these two proteins as well as a

representation of their respective secondary structures. Both primary sequences

are extremely similar in size. The secondary structure of Nm23-H1 was

represented according to a solved three-dimensional structure presented in PDB

database, whereas P. aeruginosa Ndk represented was based on a protein template

(79% identity) and modeling software (Swiss-model). Through comparison

between the two secondary structures we can see that Nm23-H1 structure exhibits

more detailed information on different elements present in that protein, such as the

localization of turns and bends. However the main observation was that we found

an almost perfect overlapping between the location of β-sheets and α-helices on

both proteins. More importantly, it is evident the higher number of identical

amino acid residues between the two sequences, especially within β-sheets and α-

helices. The same observation is verified for conserved and high conserved

residues, which can indicate the importance of those amino acid residues for

maintaining the correct structure and function of these proteins throughout the

different species.

The multiple sequences involving several Ndks were also used to build a

phylogenetic tree using Splistree software. The resultant representation is

exhibited in Figure 18, and additionally to the phylogenetic tree, all the three-

dimensional structures of Nm23 and bacterial Ndks are displayed. As it can be

visualized in this figure, the closest related human Nm23 seems to be Nm23-H1, -

H2, -H3 and -H4. Nm23-H1 and Nm23-H2 are the two closest members of the

Nm23 protein family. Nm23-H4, a mitochondrial Ndk, shows to have the closest

phylogenetic relationship between a human Nm23 protein and a bacterial Ndk (M.

Page 78: Discovering and exploiting bacterial proteins as ...repositorio.ul.pt/bitstream/10451/9665/1/ulfc103147_tm_goncalo_sil… · Discovering and exploiting bacterial proteins as anticancer

54

tuberculosis and M. bovis). They both diverge from an initial common branch,

which is very close to the one respective to M. tuberculosis and M. bovis Ndks.

These findings were also verified when analyzing the percent identity matrix.

These Ndks seem to be originated from a different branch than the one from all

the other bacterial Ndks analyzed.

A second higher Nm23 branch diverges in a second group of Nm23 proteins,

where Nm23-H8 and Nm23-H9 are closer to each other, as well as Nm23-H7 and

Nm23-H5 between themselves. As mentioned earlier, Nm23-H10 is by far the

most divergent Nm23 protein relatively to the other members of this human Ndk

protein family.

The protein three-dimensional structures represented on Figure 18 indicate all

solved structures available regarding the proteins presented in the phylogenetic

tree. Human Nm23 (-H1, -H2,-H3 and -H4) quaternary structures are defined as

homohexamers, clearly contrasting with the majority of bacterial Ndks present,

which are known to form homotetramers. Nm23-H1 and Nm23-H2 quaternary

structures also reveal to be extremely resemblant. Interestingly, not only M.

tuberculosis Ndk branch is closest to human Nm23 proteins, but it also shares the

same quaternary structure as homohexamers, contrarily to all the others bacterial

Ndks. Concerning P. aeruginosa Ndk, since no solved structure is yet available, a

model was built using Swiss-model software. Surprisingly, the model predicted as

well as the template used (Halomonas sp. 593 Ndk) are displayed as homodimers.

Nm23-H10 protein is represented as a monomer, thereby it only reveals a tertiary

structure, contrarily to both other human and bacterial Ndks and supports the idea

that this protein is evolutionarily distant from these proteins.

Page 79: Discovering and exploiting bacterial proteins as ...repositorio.ul.pt/bitstream/10451/9665/1/ulfc103147_tm_goncalo_sil… · Discovering and exploiting bacterial proteins as anticancer

55

....|....| ....|....| ....|....| ....|....| ....|....| ....|....| ....|....| ....|....| ....|....|

10 20 30 40 50 60 70 80 90

Nm23-H8 MASKKREVQL QTVINNQSLW DEMLQNKGLT VIDVYQAWCG PCRAMQPLFR KLKNELNEDE ILHFAVAEAD NIVTLQPFRD KCEPVFLFSV

Nm23-H9 ---------- ---------- --MLSSKGLT VVDVYQGWCG PCKPVVSLFQ KMRIEVGLD- LLHFALAEAD RLDVLEKYRG KCEPTFLFYA

Nm23-H1 ---------- ---------- ---------- ---------- ---------- ---------- ---------- ---------- ----------

Nm23-H2 ---------- ---------- ---------- ---------- ---------- ---------- ---------- ---------- ----------

Nm23-H3 ---------- ---------- ---------- ---------- ---------- ---------- ---------- ---------- ----------

Nm23-H4 ---------- ---------- ---------- ---------- ---------- ---------- ---------- ---------- ----------

Ndk(M.tuberculosis) ---------- ---------- ---------- ---------- ---------- ---------- ---------- ---------- ----------

Ndk(M.bovis) ---------- ---------- ---------- ---------- ---------- ---------- ---------- ---------- ----------

Ndk(E.coli)* ---------- ---------- ---------- ---------- ---------- ---------- ---------- ---------- ----------

Ndk(V.cholerae) ---------- ---------- ---------- ---------- ---------- ---------- ---------- ---------- ----------

Ndk(P.aeruginosa) ---------- ---------- ---------- ---------- ---------- ---------- ---------- ---------- ----------

Ndk(M.xanthus) ---------- ---------- ---------- ---------- ---------- ---------- ---------- ---------- ----------

Nm23-H7 ---------- ---------- ---------- ---------- ---------- ---------- --MNHSERFV FIAEWYDPNA SLLRRYELLF

Nm23-H7B ---------- ---------- ---------- ---------- ---------- ---------- ---------- ---------- ----------

Nm23-H5 ---------- ---------- ---------- ---------- ---------- ---------- ---------- ---------- ----------

Nm23-H6 ---------- ---------- ---------- ---------- ---------- ---------- ---------- ---------- ----------

Nm23-H10 ---------- ---------- ---MGCFFSK RRKADKESRP ENEEERPKQY SWDQREKVDP KDYMFSGLKD ETVGRLPGTV AGQQFLIQDC

....|....| ....|....| ....|....| ....|....| ....|....| ....|....| ....|....| ....|....| ....|....|

100 110 120 130 140 150 160 170 180

Nm23-H8 NGKIIEKIQG ANAPLVNKKV INLIDEERKI AAGEMARPQY PEIPLVDSDS EVSEESPCES VQELYSIAII KPDAVISK-- ---KVLEIKR

Nm23-H9 ---------- -------IKD EALSDEDECV SHGKNNG--- ------EDED MVSSERTCT- ------LAII KPDAVAHG-- ---KTDEIIM

Nm23-H1 ---------- ---------- ---------- ---------- ---------- MAN------- --CERTFIAI KPDGVQRG-- ---LVGEIIK

Nm23-H2 ---------- ---------- ---------- ---------- ---------- MAN------- --LERTFIAI KPDGVQRG-- ---LVGEIIK

Nm23-H3 ---------- ---------- ---------- ---------- --MICLVLTI FANLFPAACT GAHERTFLAV KPDGVQRR-- ---LVGEIVR

Nm23-H4 ---------- ---------- ------MGGL FWRSALRGLR CGPRAPGPSL LVRHGSGGPS WTRERTLVAV KPDGVQRR-- ---LVGDVIQ

Ndk(M.tuberculosis) ---------- ---------- ---------- ---------- ---------M LGT------- -VTERTLVLI KPDGIERQ-- ---LIGEIIS

Ndk(M.bovis) ---------- ---------- ---------- ---------- ---------M LGT------- -VTERTLVLI KPDGIERQ-- ---LIGEIIS

Ndk(E.coli)* ---------- ---------- ---------- ---------- ---------- MAI------- ---ERTFSII KPNAVAKN-- ---VIGNIFA

Ndk(V.cholerae) ---------- ---------- ---------- ---------- ---------- MAL------- ---ERTFSII KPDAVKRN-- ---LIGEIYH

Ndk(P.aeruginosa) ---------- ---------- ---------- ---------- ---------- MAL------- ---QRTLSII KPDAVSKN-- ---VIGEILT

Ndk(M.xanthus) ---------- ---------- ---------- ---------- ---------- MAI------- ---ERTLSII KPDGLEKG-- ---VIGKIIS

Nm23-H7 YPGDGSVEMH DVKNHRTFLK RTKYDNLHLE DLFIGNKVNV FSRQLVLIDY GDQYTARQLG SRKEKTLALI KPDAISK--- ----AGEIIE

Nm23-H7B --------MH DVKNHRTFLK RTKYDNLHLE DLFIGNKVNV FSRQLVLIDY GDQYTARQLG SRKEKTLALI KPDAISK--- ----AGEIIE

Nm23-H5 ---------- ---------- ---------- ---------- ---------- -MEISMPPPQ IYVEKTLAII KPDIVDK--- ----EEEIQD

Nm23-H6 ---------- ---------- ---------- ---------- ----MTQNLG SEMASILRSP QALQLTLALI KPDAVAHPL- ---ILEAVHQ

Nm23-H10 ENCNIYIFDH SATVTIDDCT NCIIFLGPVK GSVFFRNCRD CKCTLACQQF RVRDCRKLEV FLCCATQPII ESSSNIKFGC FQWYYPELAF

* Ndk (E.coli)

Secondary structure

Page 80: Discovering and exploiting bacterial proteins as ...repositorio.ul.pt/bitstream/10451/9665/1/ulfc103147_tm_goncalo_sil… · Discovering and exploiting bacterial proteins as anticancer

56

....|....| ....|....| ....|....| ....|....| ....|....| ....|....| ....|....| ....|....| ....|....|

190 200 210 220 230 240 250 260 270

Nm23-H8 KITKAGFIIE A--------- ------EHKT VLTEEQVVNF YSRIADQCDF EEFVSFMTSG LSYILVVSQG SKHNPPSEET EPQTDTEPNE

Nm23-H9 KIQEAGFEIL T--------- ------NEER TMTEAEVRLF YQHKAGEEAF EKLVHHMCSG PSHLLILT-- ---------- ----------

Nm23-H1 RFEQKGFRLV G--------- ------LKFM QASEDLLKEH YVDLKDRPFF AGLVKYMHSG PVVAMVWEG- ---------- ----------

Nm23-H2 RFEQKGFRLV A--------- ------MKFL RASEEHLKQH YIDLKDRPFF PGLVKYMNSG PVVAMVWEG- ---------- ----------

Nm23-H3 RFERKGFKLV A--------- ------LKLV QASEELLREH YAELRERPFY GRLVKYMASG PVVAMVWQG- ---------- ----------

Nm23-H4 RFERRGFTLV G--------- ------MKML QAPESVLAEH YQDLRRKPFY PALIRYMSSG PVVAMVWEG- ---------- ----------

Ndk(M.tuberculosis) RIERKGLTIA A--------- ------LQLR TVSAELASQH YAEHEGKPFF GSLLEFITSG PVVAAIVEG- ---------- ----------

Ndk(M.bovis) RIERKGLTIA A--------- ------LQLR TVSAELASQH YAEHEGKPFF GSLLEFITSG PVVAAIVEG- ---------- ----------

Ndk(E.coli)* RFEAAGFKIV G--------- ------TKML HLTVEQARGF YAEHDGKPFF DGLVEFMTSG PIVVSVLEG- ---------- ----------

Ndk(V.cholerae) RIEKAGLQII A--------- ------AKMV RLSEEQASGF YAEHEGKPFF EPLKEFMTSG PIMVQVLEG- ---------- ----------

Ndk(P.aeruginosa) RFEKAGLRVV A--------- ------AKMV QLSEREAGGF YAEHKERPFF KDLVSFMTSG PVVVQVLEG- ---------- ----------

Ndk(M.xanthus) RFEEKGLKPV A--------- ------IRLQ HLSQAQAEGF YAVHKARPFF KDLVQFMISG PVVLMVLEG- ---------- ----------

Nm23-H7 IINKAGFTIT K--------- ------LKMM MLSRKEALDF HVDHQSRPFF NELIQFITTG PIIAMEILR- ---------- ----------

Nm23-H7B IINKAGFTIT K--------- ------LKMM MLSRKEALDF HVDHQSRPFF NELIQFITTG PIIAMEILR- ---------- ----------

Nm23-H5 IILRSGFTIV Q--------- ------RRKL RLSPEQCSNF YVEKYGKMFF PNLTAYMSSG PLVAMILAR- ---------- ----------

Nm23-H6 QILSNKFLIV R--------- ------MREL LWRKEDCQRF YREHEGRFFY QRLVEFMASG PIRAYILAH- ---------- ----------

Nm23-H10 QFKDAGLSIF NNTWSNIHDF TPVSGELNWS LLPEDAVVQD YVPIPTTEEL KAVRVSTEAN RSIVPISRG- ---------- ----------

....|....| ....|....| ....|....| ....|....| ....|....| ....|....| ....|....| ....|....| ....|....|

280 290 300 310 320 330 340 350 360

Nm23-H8 RSEDQPE--- -VEAQVTPGM MKNKQDSLQE YLERQHLAQL CDIEEDAANV AKFMDAFFPD FKKMKSMKLE KTLALLRPNL FHERKDDVLR

Nm23-H9 RTEG------ -FEDVVTT-- ---------- ---------- ---------- ---------- ---------- ---------- ----------

Nm23-H1 ---------- --LNVVKT-- ---------- ---------- ---------- ---------- ---------- ---------- ----------

Nm23-H2 ---------- --LNVVKT-- ---------- ---------- ---------- ---------- ---------- ---------- ----------

Nm23-H3 ---------- --LDVVRT-- ---------- ---------- ---------- ---------- ---------- ---------- ----------

Nm23-H4 ---------- --YNVVRA-- ---------- ---------- ---------- ---------- ---------- ---------- ----------

Ndk(M.tuberculosis) ---------- --TRAIAA-- ---------- ---------- ---------- ---------- ---------- ---------- ----------

Ndk(M.bovis) ---------- --TRAIAA-- ---------- ---------- ---------- ---------- ---------- ---------- ----------

Ndk(E.coli) ---------- --ENAVQR-- ---------- ---------- ---------- ---------- ---------- ---------- ----------

Ndk(V.cholerae) ---------- --ENAIAR-- ---------- ---------- ---------- ---------- ---------- ---------- ----------

Ndk(P.aeruginosa) ---------- --EDAIAK-- ---------- ---------- ---------- ---------- ---------- ---------- ----------

Ndk(M.xanthus) ---------- --ENAVLA-- ---------- ---------- ---------- ---------- ---------- ---------- ----------

Nm23-H7 ---------- --DDAICE-- ---------- ---------- ---------- ---------- ---------- ---------- ----------

Nm23-H7B ---------- --DDAICE-- ---------- ---------- ---------- ---------- ---------- ---------- ----------

Nm23-H5 ---------- --HKAISY-- ---------- ---------- ---------- ---------- ---------- ---------- ----------

Nm23-H6 ---------- --KDAIQL-- ---------- ---------- ---------- ---------- ---------- ---------- ----------

Nm23-H10 --QRQKSSDE S--------- ---------- ---------- ---------- ---------- ---------- ---------- ----------

* Ndk (E.coli)

Secondary structure

* Ndk (E.coli)

Secondary structure

Page 81: Discovering and exploiting bacterial proteins as ...repositorio.ul.pt/bitstream/10451/9665/1/ulfc103147_tm_goncalo_sil… · Discovering and exploiting bacterial proteins as anticancer

57

....|....| ....|....| ....|....| ....|....| ....|....| ....|....| ....|....| ....|....| ....|....|

370 380 390 400 410 420 430 440 450

Nm23-H8 IIKDEDFKIL EQRQVVLSEK EAQALCKEYE NEDYFNKLIE NMTSGPSLAL VLLRDNGLQY WKQLLGPRTV EEAIEYFPES LCAQFAMDSL

Nm23-H9 ---------- ---------- ---------- ---------- ---------- ---------- WRTVMGPRDP NVARREQPES LRAQYGTE-M

Nm23-H1 ---------- ---------- ---------- ---------- ---------- ---------- GRVMLGETN- -PADSK-PGT IRGDFCIQ-V

Nm23-H2 ---------- ---------- ---------- ---------- ---------- ---------- GRVMLGETN- -PADSK-PGT IRGDFCIQ-V

Nm23-H3 ---------- ---------- ---------- ---------- ---------- ---------- SRALIGATN- -PADAP-PGT IRGDFCIE-V

Nm23-H4 ---------- ---------- ---------- ---------- ---------- ---------- SRAMIGHTD- -SAEAA-PGT IRGDFSVH-I

Ndk(M.tuberculosis) ---------- ---------- ---------- ---------- ---------- ---------- VRQLAGGTD- -PVQAAAPGT IRGDFALE-T

Ndk(M.bovis) ---------- ---------- ---------- ---------- ---------- ---------- VRQLAGGTD- -PVQAAAPGT IRGDFALE-T

Ndk(E.coli)* ---------- ---------- ---------- ---------- ---------- ---------- HRDLLGATN- -PANAL-AGT LRADYADS-L

Ndk(V.cholerae) ---------- ---------- ---------- ---------- ---------- ---------- YRELMGKTN- -PEEAA-CGT LRADYALS-M

Ndk(P.aeruginosa) ---------- ---------- ---------- ---------- ---------- ---------- NRELMGATD- -PKKAD-AGT IRADFAVS-I

Ndk(M.xanthus) ---------- ---------- ---------- ---------- ---------- ---------- NRDIMGATN- -PAQAA-EGT IRKDFATS-I

Nm23-H7 ---------- ---------- ---------- ---------- ---------- ---------- WKRLLGPANS GVARTDASES IRALFGTD-G

Nm23-H7B ---------- ---------- ---------- ---------- ---------- ---------- WKRLLGPANS GVARTDASES IRALFGTD-G

Nm23-H5 ---------- ---------- ---------- ---------- ---------- ---------- WLELLGPNNS LVAKETHPDS LRAIYGTD-D

Nm23-H6 ---------- ---------- ---------- ---------- ---------- ---------- WRTLMGPTRV FRARHVAPDS IRGSFGLT-D

Nm23-H10 ---------- ---------- ---------- ---------- ---------- --------CL VVLFAGDYTI ANARKLIDEM VGKGFFLVQT

....|....| ....|....| ....|....| ....|....| ....|....| ....|....| ....|....| ....|....| ....|....|

460 470 480 490 500 510 520 530 540

Nm23-H8 PVNQLYGSDS LETAEREIQH FFPLQ----- ---------- STLGLIKPHA TSEQREQILK IVKEAGFDLT QVKKMFLTPE QIEKIYPKVT

Nm23-H9 PFNAVHGSRD REDADRELAL LFP------- ---------- -SLKFSDKDT EAPQGE---- ---------- ---------- ----------

Nm23-H1 GRNIIHGSDS VESAEKEIGL WFH------- ---------- --PEELVDYT SCAQNWIYE- ---------- ---------- ----------

Nm23-H2 GRNIIHGSDS VKSAEKEISL WFK------- ---------- --PEELVDYK SCAHDWVYE- ---------- ---------- ----------

Nm23-H3 GKNLIHGSDS VESARREIAL WFR------- ---------- --ADELLCWE DSAGHWLYE- ---------- ---------- ----------

Nm23-H4 SRNVIHASDS VEGAQREIQL WFQ------- ---------- --SSELVSWA DGGQHSSIHP A--------- ---------- ----------

Ndk(M.tuberculosis) QFNLVHGSDS AESAQREIAL WFP------- ---------- --GA------ ---------- ---------- ---------- ----------

Ndk(M.bovis) QFNLVHGSDS AESAQREIAL WFP------- ---------- --GA------ ---------- ---------- ---------- ----------

Ndk(E.coli)* TENGTHGSDS VESAAREIAY FFG------- ---------- --EGEVCPRT R--------- ---------- ---------- ----------

Ndk(V.cholerae) RYNSVHGSDS PASAAREIEF FFP------- ---------- --ESEICPRP ---------- ---------- ---------- ----------

Ndk(P.aeruginosa) DENAVHGSDS EASAAREIAY FFA------- ---------- --ATEVCERI R--------- ---------- ---------- ----------

Ndk(M.xanthus) DKNTVHGSDS LENAKIEIAY FFR------- ---------- --ETEIHSYP YQK------- ---------- ---------- ----------

Nm23-H7 IRNAAHGPDS FASAAREMEL FFPSSGGCGP ANTAKFTNCT CCIVKPHAVS EGLLGKILMA IRDAGFEISA MQMFNMDRVN VEEFYEVYKG

Nm23-H7B IRNAAHGPDS FASAAREMEL FFPSSGGCGP ANTAKFTNCT CCIVKPHAVS EGLLGKILMA IRDAGFEISA MQMFNMDRVN VEEFYEVYKG

Nm23-H5 LRNALHGSND FAAAEREIRF MFP------- ---------- EVIVEPIPIG QAAKDYLNLH IMPTLLEG-- ---------- ----------

Nm23-H6 TRNTTHGSDS VVSASREIAA FFP------- ---------- -DFSEQRWYE EEEP------ ---------- ---------- ----------

Nm23-H10 KEVSMKAEDA QRVFREKAPD FLPLLN---- ---------- --KGPVIALE FNGDGAVEVC QLIVN----- ---------- ----------

* Ndk (E.coli)

Secondary structure

* Ndk (E.coli)

Secondary structure

Page 82: Discovering and exploiting bacterial proteins as ...repositorio.ul.pt/bitstream/10451/9665/1/ulfc103147_tm_goncalo_sil… · Discovering and exploiting bacterial proteins as anticancer

58

....|....| ....|....| ....|....| ....|....| ....|....| ....|....| ....|....| ....|....| ....|..

550 560 570 580 590 600 610 620

Nm23-H8 GKDFYKDLLE MLSVGPSMVM ILTKWNAVAE WRRLMGPTDP EEAKLLSPDS IRAQFGISKL KNIVHGASNA YEAKEVVNRL FEDPEEN

Nm23-H9 ---------- ---------- ---------- ---------- --------SS TQPRLKITDL D--------- ---------- -------

Nm23-H1 ---------- ---------- ---------- ---------- ---------- ---------- ---------- ---------- -------

Nm23-H2 ---------- ---------- ---------- ---------- ---------- ---------- ---------- ---------- -------

Nm23-H3 ---------- ---------- ---------- ---------- ---------- ---------- ---------- ---------- -------

Nm23-H4 ---------- ---------- ---------- ---------- ---------- ---------- ---------- ---------- -------

Ndk(M.tuberculosis) ---------- ---------- ---------- ---------- ---------- ---------- ---------- ---------- -------

Ndk(M.bovis) ---------- ---------- ---------- ---------- ---------- ---------- ---------- ---------- -------

Ndk(E.coli) ---------- ---------- ---------- ---------- ---------- ---------- ---------- ---------- -------

Ndk(V.cholerae) ---------- ---------- ---------- ---------- ---------- ---------- ---------- ---------- -------

Ndk(P.aeruginosa) ---------- ---------- ---------- ---------- ---------- ---------- ---------- ---------- -------

Ndk(M.xanthus) ---------- ---------- ---------- ---------- ---------- ---------- ---------- ---------- -------

Nm23-H7 VVTEYHDMVT EMYSGPCVAM EIQQNNATKT FREFCGPADP EIARHLRPGT LRAIFGKTKI QNAVHCTDLP EDGLLEVQYF FKILDN-

Nm23-H7B VVTEYHDMVT EMYSGPCVAM EIQQNNATKT FREFCGPADP EIARHLRPGT LRAIFGKTKI QNAVHCTDLP EDGLLEVQYF FKILDN-

Nm23-H5 ---------- ---------- ---------- LTELCK---- -----QKPAD PLIWLADWLL KNNPNKPKLC HHPIVEEPY- -------

Nm23-H6 ---------- ---------- ---------- ---------- -----QLRCG PVCYSPEGGV HYVAGTGGLG PA-------- -------

Nm23-H10 ---------- ---------- ---------- ---------- -----EIFNG TKMFVSESKE TASGDVDSFY NFADIQMGI- -------

Figure 16 – ClustalW multiple alignment of human and bacterial Ndks. The numbers above the sequences indicate the position of amino acid residues. Secondary

structure of E. coli Ndk is represented according to PDB code 2HUR. Secondary elements are indicate as: single purple curves (turns), yellow arrows (β-sheets), red

lines (bends), blue curved lines (alpha helices), orange curved lines (3/10-helices), and black lines (no secondary structure assigned). NCBI accession number for each

protein is given below. Human (Homo sapiens) Nm23 proteins: Nm23-H1 (CAG46912), Nm23-H2 (NP_001018147), Nm23-H3 (EAW85629), Nm23-H4

(NP_005000), Nm23-H5 (NP_003542), Nm23-H6 (NP_005784), Nm23-H7 (Q9Y5B8), Nm23-H7B (NP_932076), Nm23-H8 (AAF20909), Nm23-H9 (NP_835231),

Nm23-H10 (NP_008846). Bacterial Ndks: P. aeruginosa (EPR01938), M. tuberculosis (EQM19968), M. bovis (AGE68451), E. coli (ERF95659), M. xanthus

(P15266), V. cholerae (WP_001162853).

Page 83: Discovering and exploiting bacterial proteins as ...repositorio.ul.pt/bitstream/10451/9665/1/ulfc103147_tm_goncalo_sil… · Discovering and exploiting bacterial proteins as anticancer

59

Table 6 – Percent identity matrix of human Nm23 proteins and different bacterial Ndks.

Nm23-H8 Nm23-H9 Nm23-H7 Nm23-H7B Nm23-H5 Nm23-H6 Nm23-H1 Nm23-H2 Nm23-H3

Nm23-H8 100.00 41.83 21.05 23.08 26.42 20.73 24.34 24.34 21.89

Nm23-H9 41.83 100.00 22.58 25.93 32.32 28.99 27.78 25.69 24.68

Nm23-H7 21.05 22.58 100.00 100.00 33.49 27.23 30.67 28.00 23.95

Nm23-H7B 23.08 25.93 100.00 100.00 33.49 27.23 30.67 28.00 23.95

Nm23-H5 26.42 32.32 33.49 33.49 100.00 33.15 29.53 32.21 27.85

Nm23-H6 20.73 28.99 27.23 27.23 33.15 100.00 28.57 31.29 30.86

Nm23-H1 24.34 27.78 30.67 30.67 29.53 28.57 100.00 88.16 67.11

Nm23-H2 24.34 25.69 28.00 28.00 32.21 31.29 88.16 100.00 65.13

Nm23-H3 21.89 24.68 23.95 23.95 27.85 30.86 67.11 65.13 100.00

Nm23-H4 19.25 21.21 20.54 20.54 26.25 27.16 55.26 53.95 52.66

Ndk

M.tuberculosis 25.71 30.60 37.68 37.68 34.56 36.43 45.26 45.26 51.08

Ndk

M.bovis 25.71 30.60 37.68 37.68 34.56 36.43 45.26 45.26 51.08

Ndk

E.coli 32.87 32.85 34.04 34.04 35.71 39.57 42.66 42.66 44.76

Ndk

V.cholerae 33.10 32.85 37.86 37.86 38.85 36.69 39.44 41.55 41.55

Ndk

P.aeruginosa 31.47 32.12 40.43 40.43 32.86 35.97 42.66 41.96 45.45

Ndk

M.xanthus 26.90 30.66 30.77 30.77 30.28 32.37 45.14 45.83 46.53

Nm23-H10 11.59 8.95 9.19 10.12 7.96 9.33 10.53 9.21 8.28

Page 84: Discovering and exploiting bacterial proteins as ...repositorio.ul.pt/bitstream/10451/9665/1/ulfc103147_tm_goncalo_sil… · Discovering and exploiting bacterial proteins as anticancer

60

Table 6 – Percent identity matrix of human Nm23 proteins and different bacterial Ndks. (continued)

Nm23-H4 Ndk

M.tuberculosis

Ndk

M.bovis

Ndk

E.coli

Ndk

V.cholerae

Ndk

P.aeruginosa

Ndk

M.xanthus Nm23-H10

Nm23-H8 19.25 25.71 25.71 32.87 33.10 31.47 26.90 11.59

Nm23-H9 21.21 30.60 30.60 32.85 32.85 32.12 30.66 8.95

Nm23-H7 20.54 37.68 37.68 34.04 37.86 40.43 30.77 9.19

Nm23-H7B 20.54 37.68 37.68 34.04 37.86 40.43 30.77 10.12

Nm23-H5 26.25 34.56 34.56 35.71 38.85 32.86 30.28 7.96

Nm23-H6 27.16 36.43 36.43 39.57 36.69 35.97 32.37 9.33

Nm23-H1 55.26 45.26 45.26 42.66 39.44 42.66 45.14 10.53

Nm23-H2 53.95 45.26 45.26 42.66 41.55 41.96 45.83 9.21

Nm23-H3 52.66 51.08 51.08 44.76 41.55 45.45 46.53 8.28

Nm23-H4 100.00 45.32 45.32 37.06 34.51 37.76 37.93 9.09

Ndk

M.tuberculosis 45.32 100.00 100.00 44.12 53.68 48.53 50.00 13.57

Ndk

M.bovis 45.32 100.00 100.00 44.12 53.68 48.53 50.00 13.57

Ndk

E.coli 37.06 44.12 44.12 100.00 64.79 62.24 56.64 13.29

Ndk

V.cholerae 34.51 53.68 53.68 64.79 100.00 65.49 52.11 14.08

Ndk

P.aeruginosa 37.76 48.53 48.53 62.24 65.49 100.00 60.84 13.99

Ndk

M.xanthus 37.93 50.00 50.00 56.64 52.11 60.84 100.00 12.41

Nm23-H10 9.09 13.57 13.57 13.29 14.08 13.99 12.41 100.00

Page 85: Discovering and exploiting bacterial proteins as ...repositorio.ul.pt/bitstream/10451/9665/1/ulfc103147_tm_goncalo_sil… · Discovering and exploiting bacterial proteins as anticancer

61

....|....| ....|....| ....|....| ....|....| ....|....| ....|....| ....|....| ....|....| ....|....|

10 20 30 40 50 60 70 80 90

Nm23-H1 MANCERTFIA IKPDGVQRGL VGEIIKRFEQ KGFRLVGLKF MQASEDLLKE HYVDLKDRPF FAGLVKYMHS GPVVAMVWEG LNVVKTGRVM

Clustal Consensus ** :**: ****.*.:.: :***:.***: *:*:*. *: :* ** .*.: *:*** * .**.:* * ****. * ** :.: ..* :

Ndk (P.aeruginosa) MA-LQRTLSI IKPDAVSKNV IGEILTRFEK AGLRVVAAKM VQLSEREAGG FYAEHKERPF FKDLVSFMTS GPVVVQVLEG EDAIAKNREL

....|....| ....|....| ....|....| ....|....| ....|....| ....|....| ..

100 110 120 130 140 150

Nm23-H1 LGETNPADSK PGTIRGDFCI QVGRNIIHGS DSVESAEKEI GLWFHPEELV DYTSCAQNWI YE

Clustal Consensus :* *:* .:. .****.**.: .:..* :*** ** ** :** . :* . *: :

Ndk (P.aeruginosa) MGATDPKKAD AGTIRADFAV SIDENAVHGS DSEASAAREI AYFFAATEVC ERIR------ --

Figure 17 – Sequence alignment and secondary structure comparison between human Nm23-H1 and P. aeruginosa Ndk. Symbols in Clustal Consensus sequence

indicate standard ClustalW nomenclature: (*) identity, (:) high conservation and (.) conservation. Secondary structure of Nm23-H1 was represented according to PDB

code 4ENO. Secondary structure of P. aeruginosa Ndk was predicted using PDB code 3VGU as template. This template was aligned between its 2 to 141 amino acid

residues and share a 79% sequence identity with P. aeruginosa Ndk. Secondary elements are indicate as: single purple curves (turns), yellow arrows (β-sheets), red

lines (bends), blue curved lines (alpha helices), orange curved lines (3/10-helices), and black lines (no secondary structure assigned). NCBI accession number for each

protein is given below. Human (Homo sapiens) Nm23-H1 (CAG46912) and P. aeruginosa Ndk (EPR01938).

Prediction of Ndk

(P.aeruginosa)

Secondary structure

Nm23-H1

Secondary structure

Nm23-H1

Secondary structure

Prediction of Ndk

(P.aeruginosa)

Secondary structure

Page 86: Discovering and exploiting bacterial proteins as ...repositorio.ul.pt/bitstream/10451/9665/1/ulfc103147_tm_goncalo_sil… · Discovering and exploiting bacterial proteins as anticancer

62

Figure 18 – Phylogenetic tree representation of human Nm23 protein family as well as some

bacterial Ndks. Splitstree software was used to construct the tree based on ClustalW multiple

alignment generated. All sequences and its NCBI accession numbers used were the same as those

displayed in Figure 16. Protein three-dimensional structures represented were retrieved from PDB

database: Nm23-H1 (4ENO), Nm23-H2 (3BBF), Nm23-H3 (1ZS6), Nm23-H4 (1EHW), Nm23-

H10 (2BX6), and Ndks from M. tuberculosis (1K44), E. coli (2HUR) and M. xanthus (2NCK).

Quaternary structures are represented as homohexamers (Nm23-H1, Nm23-H2, Nm23-H3 and

Nm23-H4 and Ndk from M. tuberculosis) and as homotetramers (Ndk from E. coli and M.

xanthus). Tertiary structure is represented as a monomer (Nm23-H10). Ndk from P. aeruginosa is

marked with an asterisk (*) since it is a predicted three-dimensional model created using Swiss-

model software tools. This representation was modeled using 2 to 141 amino acid residues of the

Ndk sequence (79% identity) from Halomonas sp. 593 (3VGU) as template. This template forms a

homodimer in its quaternary structure, as well as the model created.

*

Page 87: Discovering and exploiting bacterial proteins as ...repositorio.ul.pt/bitstream/10451/9665/1/ulfc103147_tm_goncalo_sil… · Discovering and exploiting bacterial proteins as anticancer

63

Structural alignment between human Nm23-H1 and the predicted model,

created for P. aeruginosa Ndk, was performed using PyMOL software tools and is

presented in Figure 19. This kind of analysis clearly supports other data retrieved

from bioinformatics approaches mentioned above, since a remarkable structural

superimposition between the two proteins was successfully accomplished.

Moreover, this superimposition is even more patent between important secondary

elements as β-sheets and α-helices, which indicate that the three-dimension model

created for P. aeruginosa Ndk is extremely resemblant to human Nm23-H1.

Figure 19 – Structural alignment between human Nm23-H1 (4ENO) [blue] and P. aeruginosa

Ndk predicted model (green) was performed using PyMOL software. Three dimensional structures

are displayed at different degrees: 0º (A), 90º (B), 180º (C) and 270º (D).

Page 88: Discovering and exploiting bacterial proteins as ...repositorio.ul.pt/bitstream/10451/9665/1/ulfc103147_tm_goncalo_sil… · Discovering and exploiting bacterial proteins as anticancer
Page 89: Discovering and exploiting bacterial proteins as ...repositorio.ul.pt/bitstream/10451/9665/1/ulfc103147_tm_goncalo_sil… · Discovering and exploiting bacterial proteins as anticancer

65

5. DISCUSSION

Recently, azurin was shown to be capable of cause a decrease in P-cadherin

levels, without interfering with E-cadherin levels, in three different breast cancer

cell lines, including MCF-7/AZ.Pcad [45]. In the same study, an evaluation on the

CDH3/P-cadherin gene expression revealed that no evident decrease took place.

Further experiments based on microarray analysis revealed that when MCF-

7/AZ.Pcad cells were treated with azurin, several genes showed to be up-

regulated. Some of these genes are involved in membrane organization, vesicular

transport and lysosome. Considering these results altogether it can mean that other

mechanisms must be responsible for a decrease in P-cadherin levels when azurin

is administrated. Since post-transcriptional mechanisms should be behind these

observations, we intended to verify if proteolytic systems are involved in P-

cadherin decreased levels, as microarray analysis also pointed out. Following that

line of thought we tested a proteasome and a lysosome inhibitor (MG-132 and

ammonium chloride, respectively) on MCF-7/AZ.Pcad cells, in order to study

how those inhibitory effects could affect P-cadherin levels, after the cells have

been treated with azurin.

Regarding to the samples treated with azurin, but which did not receive any

inhibitor, we showed that P-cadherin level diminishes with increasing azurin

concentrations, as Bernardes et al already described [82]. Also these authors

reported that E-cadherin levels are not affected when azurin is administrated and

this was corroborated looking at the samples untreated with ammonium chloride.

Nevertheless, we found a decrease in E-cadherin level in samples untreated with

MG-132. Since MG-132 was dissolved in ethanol, we administrated absolute

ethanol on control (untreated) samples, whereas water was used in the case of

samples untreated with ammonium chloride. Some authors found that ethanol can

increase E-cadherin expression on B16-BL6 melanoma cells, which is a contrary

observation to what our results show [83]. On the other hand, it was described that

ethanol causes a decrease in vascular endothelial (VE)-cadherin levels by

disrupting junctional VE-cadherin and inducing endocytosis of this protein in

human umbilical vein endothelial cells (HUVEC) [84]. Even though VE-cadherin

is a different cadherin, and that HUVEC are not tumor cells, it does not exclude

Page 90: Discovering and exploiting bacterial proteins as ...repositorio.ul.pt/bitstream/10451/9665/1/ulfc103147_tm_goncalo_sil… · Discovering and exploiting bacterial proteins as anticancer

66

completely the possibility that ethanol can cause a similar decrease in E-cadherin

level in MCF-7/AZ.Pcad cells, since no similar study was ever done in this case.

When we treated MCF-7/AZ.Pcad cells with each inhibitor, we saw that E and

P-cadherin levels increased, especially when azurin at 100 μM, comparing to

control samples. MDM2 is a protein involved in negative regulation of p53

pathway by inhibiting p53 transactivation and promoting p53 degradation via a

ubiquitin-mediated proteolytic process [85]. However it has been also reported

that this MDM2 can mediate E-cadherin degradation by promoting its

ubiquitination and that MG-132 was able to block E-cadherin degradation by this

protein, in breast cancer cells [85]. It seems that those findings clearly support the

fact that we observed an increase on E-cadherin levels when cells were treated

with MG-132. Even though similar findings have not been yet described regarding

P-cadherin, it is likely that the same can happen since both cadherins are very

similar between themselves. The higher cadherin levels were seen at the highest

azurin concentration tested. This could probably be explained by the fact that

azurin can disrupt p53-MDM2 association and, therefore, contribute for a higher

free intercellular MDM2 concentration, which in turn causes E- or P-cadherin

endocytosis and its cytoplasmic accumulation, since proteolytic systems are being

inhibited.

Regarding to ammonium chloride, this inhibitor seemed to exhibit a similar

effect as MG-132 on both cadherin levels. In fact, E-cadherin degradation was

already found to be blocked by ammonium chloride in MCF-7 cells, since

lysosome has been described to be involved in E-cadherin degradation by several

authors [85], [86]. Unfortunately no studies were made about the proteolytic

systems involved in P-cadherin degradation, although once again it is probable to

be similar to E-cadherin. Our results showed thereby that two major proteolytic

systems in eukaryotic cells, as proteasome and lysosome, appear both to be

involved in E- and P-cadherin degradation.

MβCD depletes cholesterol from the cell membranes and blocks azurin

internalization, especially in lipid rafts, where this molecule is extensively present

and where azurin seems to enter preferentially [25], [81]. Therefore, we thought

to study how MβCD affects azurin internalization on MCF-7/AZ.Pcad cells and,

Page 91: Discovering and exploiting bacterial proteins as ...repositorio.ul.pt/bitstream/10451/9665/1/ulfc103147_tm_goncalo_sil… · Discovering and exploiting bacterial proteins as anticancer

67

more importantly, what kind of effect does it provokes on E and P-cadherin levels.

Our results showed that as we were expecting, when MβCD is applied on MCF-

7/AZ.Pcad it occurs a decrease in azurin internalization (in both concentrations

used). These observations are supported by the mechanism of action of this

inhibitor, and the theory that argues that the formation of caveolae/caveossome

complex in the cell membrane can be an important route of entry for azurin,

especially in tumor cells where these membrane receptors are overexpressed [25],

[81]. The most remarkable observation was, nevertheless, that when MβCD and

azurin were applied simultaneously in these cells, a decrease on P-cadherin level

was verified. On the contrary, E-cadherin level remained constant when both these

molecules were applied together. Since only P-cadherin level decreases in MCF-

7/AZ.Pcad cells when both MβCD and azurin are applied, and as both these

molecules act preferentially on lipid rafts sites, we suggest that P-cadherin should

be highly expressed in lipid rafts, within tumor cells. These would also explain

why some genes, involved in membrane organization and vesicular transport were

seen to be up-regulated when breast cancer cells were treated with azurin. It

appears that azurin can enter tumor cells through lipid rafts and interact directly

with P-cadherin or just cause the formation of vesicles containing this protein and

its endocytosis. Moreover, as our treatment with lysosome and proteasome

inhibitors showed, it is likely that azurin can also be involved in these proteolytic

pathways, and somehow directing vesicles, containing P-cadherin, for endosome,

lysosome or proteasome, thereby causing P-cadherin degradation. As reported by

Bernardes et al, E-cadherin level is not affected by azurin, and here we found that

also MβCD does not interfere with this protein level [45]. In light of these facts,

this can signify that E-cadherins should be present mainly in other sites in the cell

membrane other than lipid rafts, contrarily to P-cadherin.

In this work we used cell viability assay (MTT) to evaluate the impact of

azurin’s cytotoxicity in human breast (MCF-7/AZ.Mock) and lung (A549) cancer

cell lines when tested regarding different exposure time and protein doses. In the

case of both cell lines this protein induced a clear dose and time dependent

reduction of cell viability. These results are supported by other similar MTT

assays, performed with azurin, towards other tumor cell lines from breast cancer

Page 92: Discovering and exploiting bacterial proteins as ...repositorio.ul.pt/bitstream/10451/9665/1/ulfc103147_tm_goncalo_sil… · Discovering and exploiting bacterial proteins as anticancer

68

and melanoma, on which a decrease in cell viability was observed at 24, 48 or 72

hours [20], [24], [87]. Recently, a cell viability assay showed a maximal 20%

cytotoxicity when azurin at 100 µM was used on MCF-7/AZ.Mock cells after 48

hours (one dose), which was the same value verified when we treated those cells

during the same conditions [45]. Moreover, this was the same cytotoxicity level

observed on A549 after 48 hours, but after two doses have been applied, which

can mean that this lung cancer cell line is slightly more resistant to azurin’s

cytotoxic effects. Regarding the three-dose treatment on MCF-7/AZ.Mock,

during 72h, 74% of cell viability was reached at the highest protein concentration,

which was exactly the same value observed on an identical MTT assay where

UISO-Mel 2 cells were used [24]. Therefore our results confirmed azurin

cytotoxicity towards different tumor cell lines.

MPT 63 shares an immunoglobulin-like fold as azurin, and recently this protein

and a derived peptide (MB30) were described to display high cytotoxicity in

several tumor cell lines [63]. Taking into account these findings and considering

the fact that this protein’s activity toward cancer cells is far less studied than

azurin, we thought to evaluate its effects on MCF-7/AZ.Mock and A549 cell

lines. Ours results showed that MPT 63 displayed cytotoxicity towards both tumor

cell lines tested, even though, once again, A549 cells seemed to be not as much

sensitive to cytotoxicity as MCF-7/AZ.Mock, since a 10% decrease in cell

viability was the highest value verified in the latter cell line. On the other hand,

MPT 63 produced cytotoxic effects comparable to azurin between 48 and 72

hours (with one and three doses respectively), in MCF-7/AZ.Mock cells.

Therefore, we can affirm that this protein displays anti-tumor activity against

different cancer cell lines, and that its effect can be intensified by increasing

exposure time and protein doses applied. These results are supported by a study

where MPT 63 and a derived peptide, MB30, were able to cause high cytotoxic

levels in a wide variety of different tumor cell lines, including breast cancer cells

[63]. In the same study it were seen slight differences between cell viability

results across different tumor cells, and the same also clearly happened in our

case. Moreover, it was also observed a time dependent effect regarding this

protein anti-tumor activity of this protein. Concerning to MB30 peptide, it showed

Page 93: Discovering and exploiting bacterial proteins as ...repositorio.ul.pt/bitstream/10451/9665/1/ulfc103147_tm_goncalo_sil… · Discovering and exploiting bacterial proteins as anticancer

69

to be more effective comparing to MPT 63 against all tumor cell lines tested,

thereby it could be relevant to explore this peptide in future experiments, since it

can harbor the protein sequence accountable for this protein anti-tumorigenic

properties, in the same way that p28 has higher anti-tumor activity than azurin

itself [63].

Nucleotide diphosphate kinases are extremely common in all organisms since

they have important functional activities in cells as maintaining the nucleotide

pools for the synthesis of nucleic acids. In humans, however, these proteins

constitute a Nm23 protein family which includes 10 proteins so far, and have

shown to be involved in other biological functions. Nm23-H1 has been known for

its anti-tumor and anti-metastatic properties, for some years now, and a higher

expression was associated with the inhibition of cell invasion in several tumor

models [75]. In this work we intended to verify the same cytotoxic and anti-

metastatic properties shown by Nm23-H1, using P. aeruginosa Ndk.

The results retrieved from cell viability assays indicate that also Ndk caused a

decrease in cell viability of MCF-7/AZ.Mock and A549 cells, treated with this

protein during 48 hours and when two doses were applied during this period of

time. Unlike the other two proteins tested, this one revealed cell viability values

very similar between the two different tumor cell lines within the same assay.

Moreover, in this case when the highest concentration was used, a 27% cell

mortality was seen in A549 cells (48 hours, 2 doses applied), which was the same

value verified for MCF-7/AZ.Mock in a 72 hours assay (with 3 doses applied).

This can probably means that A549 cells are more sensitive to the cytotoxic

effects of this protein than MCF-7/AZ.Mock cells. Regarding to the cell viability

assay using Ndk, a normal epithelial lung cell line 16HBE14o- was also tested to

evaluate if Ndk’s cytotoxicity activity was selective towards cancer cells or not.

After two doses have been applied, during 48 hours, it was visible a much lower

decrease in cell viability, which never reached below 90%. Considering the results

obtained, we can say that Ndk appears to exhibit a selective cytotoxic activity

against distinct tumor cells in a dose and time dependent response, which starts to

be visualized after a two dose treatment, during 48 hours. It seems that Ndk has

Page 94: Discovering and exploiting bacterial proteins as ...repositorio.ul.pt/bitstream/10451/9665/1/ulfc103147_tm_goncalo_sil… · Discovering and exploiting bacterial proteins as anticancer

70

higher cytotoxic activity than MPT 63 against both cell lines tested, and displays

much similar results comparing to azurin than MPT 63 does.

In some tumor cells, as MCF-7, it was already described that Nm23-H1 and its

binding partner STRAP (serine-threonine kinase receptor-associated protein)

interact with p53 at the DNA binding domain and increases p53 transcription and

at the same time, activate this protein by removing the negative regulator MDM2

from the p53-MDM2 complex [88], [89]. In MTT assay we only tested p53-

positive cells, thereby it cannot be excluded that Ndk could also have increased

p53 expression or even stabilize it, as azurin does, in the same manner that Nm23-

H1.

Another extremely relevant point would be to know if MPT 63 and Ndk can

enter tumor cells or not, which could help us understand what mechanism

underlies their cytotoxic effects. Actually, we tried to answer this question by

treating tumor cells with these proteins and then using an anti-his-probe antibody,

against the histidine-tag present in both purified proteins, to visualize them by

western blot. Unfortunately, our results were inconclusive since this antibody

seems to be unsuitable for this purpose. On future experiments specific anti-MPT

63 and anti-Ndk antibodies should be designed in order to evaluate the

internalization of these two proteins in human tumor cell lines.

Since human Nm23-H1 was described as an anti-metastatic and that P.

aeruginosa Ndk exhibited relevant cytotoxic effects, it was of interest to use

bioinformatics tools to compare the two proteins at a sequence and structure level.

Taking account all the results obtained, we can see that human Nm23 proteins

seem to be arranged in the phylogenetic tree (Figure 18) as two distinct clusters: a

Group I, which includes Nm23-H1, -H2, -H3 and –H4; and a Group II, which

includes Nm23-H5,-H6,-H7,-H7B,-H8 and –H9. Nm23-H10, on the other hand,

appears to have a different ancestor. This phylogenetic analysis on human Nm23

proteins is corroborated by results obtained by Desvignes et al., on which the

same clusters were identified when analyzing different Nm23 proteins from

chordate organisms, including the human proteins [90]. In the same study it was

found that Nm23-H4 revealed a strongly supported divergence from other Nm23

proteins of the Group I, which was also verified in our results. Since this protein is

Page 95: Discovering and exploiting bacterial proteins as ...repositorio.ul.pt/bitstream/10451/9665/1/ulfc103147_tm_goncalo_sil… · Discovering and exploiting bacterial proteins as anticancer

71

present predominantly in mitochondria, it is very plausible that this may represent

the human Nm23 ancestor and its closest evolutionary proximity to M.

tuberculosis Ndk clearly supports this theory [91]. Moreover, regarding

quaternary structures of Group I cluster, they are displayed as homohexamers, as

well as M. tuberculosis Ndk. Surprisingly, the main bacterial Ndk cluster seen in

the phylogenetic tree includes Ndks as homotetramers, as the one from M.

xanthus, which seems to be closer to the other mitochondrial Nm23 (-H6). This

can possibly indicate that the Group II cluster of human Nm23 proteins could

have diverged from the main bacterial Ndk cluster and that Nm23-H6 could have

been the first protein to appear within this group. Nm23-H10 is the most divergent

human Nm23 protein as the results show. One of the reasons of its divergence

could be the fact that it only possesses a partial Ndk domain in chordate

organisms, and none at all in some cases [90]. In humans the lack of Ndk activity

of this protein is also known [90].

When comparing secondary structures of Nm23-H1 and the model created for

P. aeruginosa Ndk we see that all α-helices and β-sheets present in Nm23-H1

were also proposed to be present in the bacterial Ndk, in the same relative

positions. Even though the model was proposed as a homodimeric protein, it was

built according to its template, thereby no relevant conclusions can be made

concerning to its quaternary structure. Even so, the structural alignment performed

(Figure 19) showed that both protein structural units represented overlap each

other almost perfectly. These results clearly indicate that the model created for P.

aeruginosa Ndk is very robust and that Ndks have maintained higher sequence

and function conservation throughout evolution, as it has been analyzed by some

authors [92]. More importantly this may support the idea that Ndk and Nm23-H1

resemblance at a sequence and structural level, could permit an association of Ndk

to p53, or other important proteins that bind to Nm23-H1.

Although we used P. aeruginosa Ndk in MTT assays, and this protein shares a

very relevant identity percentage with Nm23-H1, M. tuberculosis Ndk revealed a

slightly higher sequence identity. Another important fact is that the three-

dimensional model built for P. aeruginosa Ndk shows a homodimeric protein,

where M. tuberculosis Ndk is a homohexameric protein and therefore have a

Page 96: Discovering and exploiting bacterial proteins as ...repositorio.ul.pt/bitstream/10451/9665/1/ulfc103147_tm_goncalo_sil… · Discovering and exploiting bacterial proteins as anticancer

72

similar quaternary structure as human Nm23-H1. This can suggest that this

bacterial Ndk may be suitable to achieve higher cytotoxic and anti-metastatic

effects against tumor cells, when compared to P. aeruginosa Ndk.

We attempt to verify the same anti-metastatic potential reported for Nm23-H1

in different tumor cells, using P. aeruginosa Ndk to complement our study upon

this protein. As mentioned before, Nm23-H1 has been successively associated

with a decrease in tumor cells motility and migration, thereby we performed a

matrigel invasion assay on A549, a highly invasive cell line, and administrated

Ndk in hope to verify similar results [93], [94]. Our preliminary results only

showed a minimal decrease in cell invasion when using Ndk, comparing to

control (between 6 and 8%), and no differences between the two concentrations

applied were visible. However, additionally to the fact that this assay requires to

be repeated to acquire statistical relevance, it is important to refer that this was a

24 hours assay, on which only one protein dose was applied. This fact could be

extremely important since we only observed cytotoxic effects on A549 cells with

Ndk, after 48 hours and a two dose treatment. Therefore, perhaps it is necessary to

increase the number of protein doses and the exposure time to Ndk in future

experiments.

Additionally to its nucleotide diphosphate kinase activity, Nm23-H1 was also

reported to exhibit a protein histidine kinase activity that mediates its anti-motility

function, probably via phosphorylation of the kinase suppressor of ras (KSR) and

signaling mechanisms associated to this pathway [95]. Experiments involving

site-directed mutagenesis and protein activity analysis, regarding Nm23-H1

protein, revealed a requirement of K12 and Y52 residues for histidine kinase

activity and H118 for the metastasis suppressor activity of this protein [96]. Both

these amino acid residues are identical between human Nm23-H1 and P.

aeruginosa Ndk sequences, which can mean that this histidine kinase activity may

also be present on Ndk, and thus it may display anti-metastatic activity. Another

relevant question here may be the importance of quaternary structure in Nm23-H1

anti-metastatic function, since Ndk is not a hexameric protein, which indicate that

more experiments must be done regarding its putative anti-metastatic effect.

Page 97: Discovering and exploiting bacterial proteins as ...repositorio.ul.pt/bitstream/10451/9665/1/ulfc103147_tm_goncalo_sil… · Discovering and exploiting bacterial proteins as anticancer

73

6. CONCLUSION

Cancer remains one of the major human diseases, representing the leading

cause of death in the world and a terrible burden, which is specially accentuated in

developed countries. Newer and alternative anti-cancer therapies are under

development and are inevitable to reach more successful results against cancer

than the conventional radio- and chemotherapies. The use of microorganisms and

its derived products, especially proteins, to treat this disease are being broadly

explored and can create new windows of hope for these patients.

In this work we used azurin, a well described P. aeruginosa protein with anti-

tumor properties, to understand if it could also mediate a decrease in P-cadherin

levels by inducing its degradation within an invasive tumor cell line which

express high P-cadherin levels, like MCF-7/AZ.Pcad. From our results, it is clear

that the two major proteolytic systems in eukaryotic cells, lysosome and

proteasome, are involved in P-cadherin degradation, even though it was not

possible to declare what is the exact contribution of each system for that matter.

We also found that, when MβCD and azurin were administrated simultaneously to

this tumor cell line, a decrease in P-cadherin level was evident, contrarily to E-

cadherin level, which remained unaltered. Thus we suggest that, by depleting

cholesterol and interfering with lipid rafts on cell membrane, MβCD must be

causing P-cadherin to be displaced from these sites where it is supposedly mainly

present. Since azurin internalization is not completely blocked by this compound,

it is likely that this protein enters tumor cells preferentially through lipid rafts, and

mediates P-cadherin endocytosis and latter proteolysis by systems like the

lysosome. Further investigation is required in order to clarify how exactly does

azurin mediate P-cadherin degradation and what pathways can be involved.

Here we also intended to explore a possible anti-tumor role for new bacterial

proteins as MPT 63 (from M. tuberculosis) and Ndk (from P. aeruginosa) using

two tumor cell lines, derived from two of the deadliest cancers, lung and breast

cancer. Both these proteins share some important features as azurin, since they are

low molecular weight and secreted by pathogenic bacteria, probably as a survival

mechanism against other organisms. MPT 63 also shares an immunoglobulin-like

folding with azurin, which may be relevant for the cytotoxic effect verified on

Page 98: Discovering and exploiting bacterial proteins as ...repositorio.ul.pt/bitstream/10451/9665/1/ulfc103147_tm_goncalo_sil… · Discovering and exploiting bacterial proteins as anticancer

74

tumor cells as our results showed. Both proteins evidenced cytotoxicity against

cancer cells in a time and dose dependent manner.

Nevertheless, Ndk exhibited a far extensive cytotoxicity than MPT 63 against

both MCF-7/AZ.Mock and A549 cells. In fact, this protein revealed a similar

effect on cell viability as azurin. Another interesting observation was that it

appears to display selective cytotoxicity against tumor cell lines and a lower effect

on normal cells, a feature that also azurin presents. This could be a promising

feature since we seek anti-tumor agents that should act mainly on tumor cells and

limit side effects on normal cells.

Although we have shown that MPT 63 and Ndk display cytotoxicity against

cancer cells in vitro, the mechanisms by which that occurs remain a mystery. It is

likely that they increase apoptotic rate by activating p53, Bax and other pro-

apoptotic proteins in some way, as it was already described for azurin. For this

motive it will be crucial to study the effect of these bacterial proteins on several

major pro-apoptotic proteins, which could provide other important clues and tell

us more about their anti-tumor activities, since it cannot be excluded that they

share similar mechanisms as azurin.

In this work, we also tested anti-his-probe antibody towards these purified

proteins in order to evaluate their internalization in tumor cells, but the results

obtained clearly showed that this is not a suitable antibody to test this. It should be

designed specific antibodies against each one of these proteins to assure the real

impact of their internalization in tumor cell lines. This finding will help us to

understand if these proteins require full cell internalization or not to exert their

cytotoxic activity. If internalization reveals to be a truly requirement for these

protein anti-tumor activities we must develop strategies to increase protein

delivery as the use of liposomes, for instance.

Since human Ndk homologue protein Nm23-H1 is known by its anti-metastatic

activity in several tumor models, we sought to test Ndk in A549 invasion assay.

We only observed a 10% approximated decrease in cell invasion using this

protein, which still is a lower value even though we only have made a single

experiment. It is then important to repeat this experiment using increased

exposure time and higher number of protein doses to test Ndk’s anti-metastatic

Page 99: Discovering and exploiting bacterial proteins as ...repositorio.ul.pt/bitstream/10451/9665/1/ulfc103147_tm_goncalo_sil… · Discovering and exploiting bacterial proteins as anticancer

75

effect, which could extent the relevance of the anti-tumor effects of this bacterial

protein toward human cancers.

Our study on multiple sequence alignment between Ndks with different origins

showed that M. tuberculosis Ndk has a higher identity level to Nm23-H1 than P.

aeruginosa Ndk. Moreover, this was so far the unique bacterial Ndk to display a

hexameric three-dimensional strucuture, which is the same kind of structure seen

in human Nm23-H1. Therefore, we think that it should be important to consider

this protein in future similar experiments since three-dimensional structure may be

relevant for the anti-metastatic activity of Ndk.

In summary, our results showed that some bacterial proteins with anti-tumor

properties as azurin, Ndk and MPT 63, can induce important cytotoxic effects on

tumor cells but, in order to represent a solid alternative to conventional therapies

in a near future, some issues still need to be solved. The scientific knowledge

gathered until now and the tools at our disposal in the present could be the key to

enhance the anti-tumor properties of these proteins in order to make them more

efficient. For instance, the use of genetically modified bacteria derived from

human microbiome, to express these bacterial proteins can represent one way of

delivering these proteins with efficiency and to increase bioavailability in the site

of the tumors. Moreover, this could limit side effects on normal cells and a more

targeted anti-tumor response. The possibility of these proteins to act on different

pathways, like controlling apoptosis and cell invasion processes make them a

unique weapon to be explored in the future years.

Page 100: Discovering and exploiting bacterial proteins as ...repositorio.ul.pt/bitstream/10451/9665/1/ulfc103147_tm_goncalo_sil… · Discovering and exploiting bacterial proteins as anticancer
Page 101: Discovering and exploiting bacterial proteins as ...repositorio.ul.pt/bitstream/10451/9665/1/ulfc103147_tm_goncalo_sil… · Discovering and exploiting bacterial proteins as anticancer

77

7. REFERENCES

[1] J. Ferlay, H.-R. Shin, F. Bray, D. Forman, C. Mathers, and D. M. Parkin,

“Estimates of worldwide burden of cancer in 2008: GLOBOCAN 2008.,”

International journal of cancer. Journal international du cancer, vol. 127,

pp. 2893–917, Dec. 2010.

[2] R. Meuwissen and A. Berns, “Mouse models for human lung cancer.,”

Genes & development, vol. 19, pp. 643–64, Mar. 2005.

[3] J. Yokota and T. Kohno, “Molecular footprints of human lung cancer

progression.,” Cancer science, vol. 95, pp. 197–204, Mar. 2004.

[4] A. Fucic, M. Gamulin, Z. Ferencic, D. S. Rokotov, J. Katic, A. Bartonova,

I. B. Lovasic, and D. F. Merlo, “Lung cancer and environmental chemical

exposure: a review of our current state of knowledge with reference to the

role of hormones and hormone receptors as an increased risk factor for

developing lung cancer in man.,” Toxicologic pathology, vol. 38, pp. 849–

55, Oct. 2010.

[5] B.-R. Yi, K. J. Choi, S. U. Kim, and K.-C. Choi, “Therapeutic potential of

stem cells expressing suicide genes that selectively target human breast

cancer cells: evidence that they exert tumoricidal effects via tumor tropism

(review).,” International journal of oncology, vol. 41, pp. 798–804, Sep.

2012.

[6] H. Lahlou and W. J. Muller, “Β1-Integrins Signaling and Mammary Tumor

Progression in Transgenic Mouse Models: Implications for Human Breast

Cancer.,” Breast cancer research : BCR, vol. 13, p. 229, Jan. 2011.

[7] K.-L. Dao and R. N. Hanson, “Targeting the estrogen receptor using

steroid-therapeutic drug conjugates (hybrids).,” Bioconjugate chemistry,

vol. 23, pp. 2139–58, Nov. 2012.

[8] S. Douglass, S. Ali, A. P. Meeson, D. Browell, and J. a Kirby, “The role of

FOXP3 in the development and metastatic spread of breast cancer.,”

Cancer metastasis reviews, vol. 31, pp. 843–54, Dec. 2012.

[9] K. Yanagisawa, B. J. Xu, D. P. Carbone, and R. M. Caprioli, “Molecular

fingerprinting in human lung cancer.,” Clinical lung cancer, vol. 5, pp.

113–8, Sep. 2003.

[10] A. W. Wong, Q. X. Paulson, J. Hong, R. E. Stubbins, K. Poh, E. Schrader,

and N. P. Nunez, “Alcohol promotes breast cancer cell invasion by

regulating the Nm23-ITGA5 pathway.,” Journal of experimental & clinical

cancer research : CR, vol. 30, pp. 1–11, Jan. 2011.

Page 102: Discovering and exploiting bacterial proteins as ...repositorio.ul.pt/bitstream/10451/9665/1/ulfc103147_tm_goncalo_sil… · Discovering and exploiting bacterial proteins as anticancer

78

[11] N. Bernardes, A. M. Chakrabarty, and A. M. Fialho, “Engineering of

bacterial strains and their products for cancer therapy.,” Applied

microbiology and biotechnology, vol. 97, pp. 5189–5199, May 2013.

[12] A. M. Fialho, N. Bernardes, and A. M. Chakrabarty, “Recent Patents on

Live Bacteria and their Products as Potential Anticancer Agents,” Recent

Patents on Anti-Cancer Drug Discovery, vol. 7, pp. 31–55, Jan. 2012.

[13] N. Bernardes, R. Seruca, A. M. Chakrabarty, and A. M. Fialho, “Microbial-

based therapy of cancer: current progress and future prospects.,”

Bioengineered bugs, vol. 1, pp. 178–90, 2010.

[14] A. Naguleswaran, A. M. Fialho, A. Chaudhari, C. S. Hong, A. M.

Chakrabarty, and W. J. Sullivan, “Azurin-like protein blocks invasion of

Toxoplasma gondii through potential interactions with parasite surface

antigen SAG1.,” Antimicrobial agents and chemotherapy, vol. 52, pp. 402–

8, Feb. 2008.

[15] T. Yamada, R. R. Mehta, F. Lekmine, K. Christov, M. L. King, D.

Majumdar, A. Shilkaitis, A. Green, L. Bratescu, C. W. Beattie, and T. K.

Das Gupta, “A peptide fragment of azurin induces a p53-mediated cell

cycle arrest in human breast cancer cells.,” Molecular cancer therapeutics,

vol. 8, pp. 2947–58, Oct. 2009.

[16] R. R. Mehta, M. Hawthorne, X. Peng, A. Shilkaitis, R. G. Mehta, C. W.

Beattie, and T. K. Das Gupta, “A 28-amino-acid peptide fragment of the

cupredoxin azurin prevents carcinogen-induced mouse mammary lesions.,”

Cancer prevention research (Philadelphia, Pa.), vol. 3, pp. 1351–60, Oct.

2010.

[17] A. M. Fialho, P. Salunkhe, S. Manna, S. Mahali, and A. M. Chakrabarty,

“Glioblastoma multiforme: novel therapeutic approaches.,” ISRN

neurology, vol. 2012, pp. 1–10, Jan. 2012.

[18] N. Bonander, T. Vänngård, L. C. Tsai, V. Langer, H. Nar, and L. Sjölin,

“The metal site of Pseudomonas aeruginosa azurin, revealed by a crystal

structure determination of the Co(II) derivative and Co-EPR

spectroscopy.,” Proteins, vol. 27, pp. 385–94, Mar. 1997.

[19] O. Zaborina, N. Dhiman, M. L. Chen, J. Kostal, I. A. Holder, and A. M.

Chakrabarty, “Secreted products of a nonmucoid Pseudomonas aeruginosa

strain induce two modes of macrophage killing : external-ATP- dependent ,

P2Z-receptor-mediated necrosis apoptosis,” Microbiology, vol. 146, pp.

2521–2530, 2000.

[20] T. Yamada, M. Goto, V. Punj, O. Zaborina, M. L. Chen, K. Kimbara, D.

Majumdar, E. Cunningham, T. K. Das Gupta, and A. M. Chakrabarty,

“Bacterial redox protein azurin, tumor suppressor protein p53, and

Page 103: Discovering and exploiting bacterial proteins as ...repositorio.ul.pt/bitstream/10451/9665/1/ulfc103147_tm_goncalo_sil… · Discovering and exploiting bacterial proteins as anticancer

79

regression of cancer.,” Proceedings of the National Academy of Sciences of

the United States of America, vol. 99, pp. 14098–103, Oct. 2002.

[21] V. Punj, S. Bhattacharyya, D. Saint-Dic, C. Vasu, E. a Cunningham, J.

Graves, T. Yamada, A. I. Constantinou, K. Christov, B. White, G. Li, D.

Majumdar, A. M. Chakrabarty, and T. K. Das Gupta, “Bacterial cupredoxin

azurin as an inducer of apoptosis and regression in human breast cancer.,”

Oncogene, vol. 23, pp. 2367–78, Mar. 2004.

[22] D. Yang, X. Miao, Z. Ye, and J. Feng, “Bacterial redox protein azurin

induce apoptosis in human osteosarcoma U2OS cells,” Pharmacological

Research, vol. 52, pp. 413–421, 2005.

[23] T. Yamada, A. M. Fialho, V. Punj, L. Bratescu, T. K. Das Gupta, and A.

M. Chakrabarty, “Internalization of bacterial redox protein azurin in

mammalian cells: entry domain and specificity.,” Cellular microbiology,

vol. 7, pp. 1418–31, Oct. 2005.

[24] B. N. Taylor, R. R. Mehta, T. Yamada, F. Lekmine, K. Christov, A. M.

Chakrabarty, A. Green, L. Bratescu, A. Shilkaitis, C. W. Beattie, and T. K.

Das Gupta, “Noncationic peptides obtained from azurin preferentially enter

cancer cells.,” Cancer research, vol. 69, pp. 537–46, Jan. 2009.

[25] R. R. Mehta, T. Yamada, B. N. Taylor, K. Christov, M. L. King, D.

Majumdar, F. Lekmine, C. Tiruppathi, A. Shilkaitis, L. Bratescu, A. Green,

C. W. Beattie, and T. K. Das Gupta, “A cell penetrating peptide derived

from azurin inhibits angiogenesis and tumor growth by inhibiting

phosphorylation of VEGFR-2, FAK and Akt.,” Angiogenesis, vol. 14, pp.

355–69, Sep. 2011.

[26] T. Yamada, Y. Hiraoka, M. Ikehata, K. Kimbara, B. S. Avner, T. K. Das

Gupta, and A. M. Chakrabarty, “Apoptosis or growth arrest: Modulation of

tumor suppressor p53’s specificity by bacterial redox protein azurin.,”

Proceedings of the National Academy of Sciences of the United States of

America, vol. 101, pp. 4770–5, Apr. 2004.

[27] E. Gabellieri, M. Bucciantini, M. Stefani, and P. Cioni, “Does azurin bind

to the transactivation domain of p53? A Trp phosphorescence study.,”

Biophysical chemistry, vol. 159, pp. 287–93, Dec. 2011.

[28] J. M. Kwan, A. M. Fialho, M. Kundu, J. Thomas, C. S. Hong, T. K. Das

Gupta, and A. M. Chakrabarty, “Bacterial proteins as potential drugs in the

treatment of leukemia.,” Leukemia research, vol. 33, pp. 1392–9, Oct.

2009.

[29] S. Ramachandran and M. Mandal, “Induction of apoptosis of azurin

synthesized from P. aeruginosa MTCC 2453 against Dalton’s lymphoma

Page 104: Discovering and exploiting bacterial proteins as ...repositorio.ul.pt/bitstream/10451/9665/1/ulfc103147_tm_goncalo_sil… · Discovering and exploiting bacterial proteins as anticancer

80

ascites model.,” Biomedicine & pharmacotherapy, vol. 65, pp. 461–6, Oct.

2011.

[30] L. Jia, G. S. Gorman, L. U. Coward, P. E. Noker, D. McCormick, T. L.

Horn, J. B. Harder, M. Muzzio, B. Prabhakar, B. Ganesh, T. K. Das Gupta,

and C. W. Beattie, “Preclinical pharmacokinetics, metabolism, and toxicity

of azurin-p28 (NSC745104) a peptide inhibitor of p53 ubiquitination.,”

Cancer chemotherapy and pharmacology, vol. 68, pp. 513–24, Aug. 2011.

[31] M. a Warso, J. M. Richards, D. Mehta, K. Christov, C. Schaeffer, L. Rae

Bressler, T. Yamada, D. Majumdar, S. a Kennedy, C. W. Beattie, and T. K.

Das Gupta, “A first-in-class, first-in-human, phase I trial of p28, a non-

HDM2-mediated peptide inhibitor of p53 ubiquitination in patients with

advanced solid tumours.,” British journal of cancer, vol. 108, pp. 1061–70,

Mar. 2013.

[32] A. Chaudhari, M. Mahfouz, A. M. Fialho, T. Yamada, A. T. Granja, Y.

Zhu, W. Hashimoto, B. Schlarb-Ridley, W. Cho, T. K. Das Gupta, and A.

M. Chakrabarty, “Cupredoxin-cancer interrelationship: azurin binding with

EphB2, interference in EphB2 tyrosine phosphorylation, and inhibition of

cancer growth.,” Biochemistry, vol. 46, pp. 1799–810, Feb. 2007.

[33] C. Hafner, G. Schmitz, S. Meyer, F. Bataille, P. Hau, T. Langmann, W.

Dietmaier, M. Landthaler, and T. Vogt, “Differential gene expression of

Eph receptors and ephrins in benign human tissues and cancers.,” Clinical

chemistry, vol. 50, pp. 490–9, Mar. 2004.

[34] M. Mahfouz, W. Hashimoto, T. K. Das Gupta, and A. M. Chakrabarty,

“Bacterial proteins and CpG-rich extrachromosomal DNA in potential

cancer therapy.,” Plasmid, vol. 57, pp. 4–17, Jan. 2007.

[35] A. M. Fialho and A. M. Chakrabarty, “Recent patents on bacterial proteins

as potential anticancer agents.,” Recent patents on anti-cancer drug

discovery, vol. 2, pp. 224–34, Nov. 2007.

[36] A. Chaudhari, A. M. Fialho, D. Ratner, P. Gupta, C. S. Hong, S. Kahali, T.

Yamada, K. Haldar, S. Murphy, W. Cho, V. S. Chauhan, T. K. Das Gupta,

and A. M. Chakrabarty, “Azurin, Plasmodium falciparum Malaria and

HIV/AIDS,” Cell Cycle, vol. 5, pp. 1642–1648, 2006.

[37] A. M. Fialho, F. J. Stevens, T. K. Das Gupta, and A. M. Chakrabarty,

“Beyond host-pathogen interactions: microbial defense strategy in the host

environment.,” Current opinion in biotechnology, vol. 18, pp. 279–86, Jun.

2007.

[38] J. Paredes, C. Stove, V. Stove, F. Milanezi, V. Van Marck, L. Derycke, M.

Mareel, M. Bracke, and F. Schmitt, “P-cadherin is up-regulated by the

Page 105: Discovering and exploiting bacterial proteins as ...repositorio.ul.pt/bitstream/10451/9665/1/ulfc103147_tm_goncalo_sil… · Discovering and exploiting bacterial proteins as anticancer

81

antiestrogen ICI 182,780 and promotes invasion of human breast cancer

cells.,” Cancer research, vol. 64, pp. 8309–17, Nov. 2004.

[39] J. Paredes, J. Figueiredo, A. Albergaria, P. Oliveira, J. Carvalho, A. S.

Ribeiro, J. Caldeira, A. M. Costa, J. Simões-Correia, M. J. Oliveira, H.

Pinheiro, S. S. Pinho, R. Mateus, C. a Reis, M. Leite, M. S. Fernandes, F.

Schmitt, F. Carneiro, C. Figueiredo, C. Oliveira, and R. Seruca, “Epithelial

E- and P-cadherins: role and clinical significance in cancer.,” Biochimica et

biophysica acta, vol. 1826, pp. 297–311, Dec. 2012.

[40] A. Albergaria, A. Ribeiro, A. Vieira, and B. Sousa, “P-cadherin role in

normal breast development and cancer,” The international journal of

developmental biology, vol. 55, pp. 811–822, 2011.

[41] R. van Horssen, A. Hollestelle, J. a P. Rens, A. M. M. Eggermont, M.

Schutte, and T. L. M. Ten Hagen, “E-cadherin promotor methylation and

mutation are inversely related to motility capacity of breast cancer cells.,”

Breast cancer research and treatment, vol. 136, pp. 365–77, Nov. 2012.

[42] J. Paredes, A. L. Correia, A. S. Ribeiro, A. Albergaria, F. Milanezi, and F.

C. Schmitt, “The effect of psychosocial factors on breast cancer outcome: a

systematic review.,” Breast cancer research : BCR, vol. 9, pp. 1–12, Jan.

2007.

[43] A. Albergaria, C. Resende, A. R. Nobre, A. S. Ribeiro, B. Sousa, J. C.

Machado, R. Seruca, J. Paredes, and F. Schmitt, “CCAAT/enhancer

binding protein β (C/EBPβ) isoforms as transcriptional regulators of the

pro-invasive CDH3/P-cadherin gene in human breast cancer cells.,” PloS

one, vol. 8, pp. 1–9, Jan. 2013.

[44] A. S. Ribeiro, A. Albergaria, B. Sousa, A. L. Correia, M. Bracke, R.

Seruca, F. C. Schmitt, and J. Paredes, “Extracellular cleavage and shedding

of P-cadherin: a mechanism underlying the invasive behaviour of breast

cancer cells.,” Oncogene, vol. 29, pp. 1–11, Jan. 2009.

[45] N. Bernardes, A. S. Ribeiro, S. Abreu, B. Mota, R. G. Matos, C. M.

Arraiano, R. Seruca, J. Paredes, and A. M. Fialho, “The bacterial protein

azurin impairs invasion and FAK/Src signaling in P-cadherin-

overexpressing breast cancer cell models.,” PloS one, vol. 8, pp. 1-8, Jan.

2013.

[46] E. Wong and A. M. Cuervo, “Integration of clearance mechanisms: the

proteasome and autophagy.,” Cold Spring Harbor perspectives in biology,

vol. 2, pp. 1–19, Dec. 2010.

[47] M. J. Clague and S. Urbé, “Ubiquitin: same molecule, different degradation

pathways.,” Cell, vol. 143, pp. 682–5, Nov. 2010.

Page 106: Discovering and exploiting bacterial proteins as ...repositorio.ul.pt/bitstream/10451/9665/1/ulfc103147_tm_goncalo_sil… · Discovering and exploiting bacterial proteins as anticancer

82

[48] X. Chen and X.-M. Yin, “Coordination of autophagy and the proteasome in

resolving endoplasmic reticulum stress.,” Veterinary pathology, vol. 48, pp.

245–53, Jan. 2011.

[49] A. Ciechanover, “Intracellular protein degradation: from a vague idea,

through the lysosome and the ubiquitin-proteasome system, and onto

human diseases and drug targeting (Nobel lecture).,” Angewandte Chemie

(International ed. in English), vol. 44, pp. 5944–67, Sep. 2005.

[50] B. Park, Y.-M. Lee, J.-S. Kim, Y. Her, J. H. Kang, S.-H. Oh, and H.-M.

Kim, “Neutral sphingomyelinase 2 modulates cytotoxic effects of

protopanaxadiol on different human cancer cells.,” BMC complementary

and alternative medicine, vol. 13, p. 194, Jan. 2013.

[51] R. Cubí, L. a Matas, M. Pou, J. Aguilera, and C. Gil, “Differential

sensitivity to detergents of actin cytoskeleton from nerve endings.,”

Biochimica et biophysica acta, vol. 1828, pp. 2385–2393, Jun. 2013.

[52] V. S. Silva, L. Oliveira, and P. P. Gonçalves, “Alteration of aluminium

inhibition of synaptosomal (Na(+)/K(+))ATPase by colestipol

administration.,” Journal of inorganic biochemistry, vol. 128, pp. 208-214,

Nov. 2013.

[53] D. Jensen, C. B. Godfrey, C. Niklas, M. Canals, M. Kocan, D. P. Poole, J.

E. Murphy, F. Alemi, G. S. Cottrell, C. Korbmacher, N. a Lambert, N. W.

Bunnett, and C. U. Corvera, “The Bile Acid Receptor TGR5 Does Not

Interact with β-arrestins or Traffic to Endosomes but Transmits Sustained

Signals from Plasma Membrane Rafts.,” The Journal of biological

chemistry, vol. 288, pp. 22942-22960, Jul. 2013.

[54] Z.-J. Huang, S.-T. Kang, J.-H. Leu, and L.-L. Chen, “Endocytic pathway is

indicated for white spot syndrome virus (WSSV) entry in shrimp.,” Fish &

shellfish immunology, vol. 35, pp. 707-715, Jun. 2013.

[55] C. Manca, K. Lyashchenko, H. G. Wiker, D. Usai, R. Colangeli, and M. L.

Gennaro, “Molecular cloning, purification, and serological characterization

of MPT63, a novel antigen secreted by Mycobacterium tuberculosis.,”

Infection and immunity, vol. 65, pp. 16–23, Jan. 1997.

[56] B. Y. Lee and M. a Horwitz, “T-cell epitope mapping of the three most

abundant extracellular proteins of Mycobacterium tuberculosis in outbred

guinea pigs.,” Infection and immunity, vol. 67, pp. 2665–70, May 1999.

[57] T. Redchuk, N. Korotkevich, O. Gorbatiuk, P. Gilchuk, A. Kaberniuk, O.

Oliynyk, D. Kolibo, and S. Komisarenko, “Expression of Mycobacterium

tuberculosis proteins MPT63 and MPT83 as a fusion: purification,

refolding and immunological characterization,” Journal of Applied

Biomedicine, vol. 10, pp. 169–176, Jan. 2012.

Page 107: Discovering and exploiting bacterial proteins as ...repositorio.ul.pt/bitstream/10451/9665/1/ulfc103147_tm_goncalo_sil… · Discovering and exploiting bacterial proteins as anticancer

83

[58] C. W. Goulding, A. Parseghian, M. R. Sawaya, D. Cascio, M. I. Apostol,

M. L. Gennaro, and D. Eisenberg, “Crystal structure of a major secreted

protein of Mycobacterium tuberculosis — MPT63 at 1 . 5-Å resolution,”

Protein Science, vol. 11, pp. 2887–2893, Dec. 2002.

[59] S. Nagai, H. G. Wiker, M. Harboe, and M. Kinomoto, “Isolation and partial

characterization of major protein antigens in the culture fluid of

Mycobacterium tuberculosis.,” Infection and immunity, vol. 59, pp. 372–

82, Jan. 1991.

[60] B. L. Wang, Y. Xu, Z. M. Li, Y. M. Xu, X. H. Weng, and H. H. Wang,

“Antibody response to four secretory proteins from Mycobacterium

tuberculosis and their complex antigen in TB patients.,” The international

journal of tuberculosis and lung disease : the official journal of the

International Union against Tuberculosis and Lung Disease, vol. 9, pp.

1327–34, Dec. 2005.

[61] H. M. C. Shane, R. Brookes, S. C. Gilbert, A. V. S. Hill, and I. N. I. Mmun,

“Enhanced Immunogenicity of CD4 ϩ T-Cell Responses and Protective

Efficacy of a DNA-Modified Vaccinia Virus Ankara Prime-Boost

Vaccination Regimen for Murine Tuberculosis,” Infection and immunity,

vol. 69, pp. 681–686, Feb. 2001.

[62] A. S. Mustafa, “Th1 cell reactivity and HLA-DR binding prediction for

promiscuous recognition of MPT63 (Rv1926c), a major secreted protein of

Mycobacterium tuberculosis.,” Scandinavian journal of immunology, vol.

69, pp. 213–22, Mar. 2009.

[63] A. Suri, D. Kanojia, P. Salunkhe, S. Mahali, S. K. Manna, A. Surolia, and

A. Chakrabarty, “Anticancer agent.” WIPO Patent Application

WO/2012/042540, Apr. 5, 2012.

[64] G. W. Sundin, S. Shankar, and a M. Chakrabarty, “Mutational analysis of

nucleoside diphosphate kinase from Pseudomonas aeruginosa:

characterization of critical amino acid residues involved in

exopolysaccharide alginate synthesis.,” Journal of bacteriology, vol. 178,

pp. 7120–8, Dec. 1996.

[65] H. Tokunaga, M. Ishibashi, F. Arisaka, S. Arai, R. Kuroki, T. Arakawa, and

M. Tokunaga, “Residue 134 determines the dimer-tetramer assembly of

nucleoside diphosphate kinase from moderately halophilic bacteria.,” FEBS

letters, vol. 582, pp. 1049–54, Apr. 2008.

[66] a M. Chakrabarty, “Nucleoside diphosphate kinase: role in bacterial

growth, virulence, cell signalling and polysaccharide synthesis.,” Molecular

microbiology, vol. 28, pp. 875–82, Jun. 1998.

Page 108: Discovering and exploiting bacterial proteins as ...repositorio.ul.pt/bitstream/10451/9665/1/ulfc103147_tm_goncalo_sil… · Discovering and exploiting bacterial proteins as anticancer

84

[67] S. Tiwari, K. V. R. Kishan, T. Chakrabarti, and P. K. Chakraborti, “Amino

acid residues involved in autophosphorylation and phosphotransfer

activities are distinct in nucleoside diphosphate kinase from

Mycobacterium tuberculosis.,” The Journal of biological chemistry, vol.

279, pp. 43595–603, Oct. 2004.

[68] S. Kamath, M. L. Chen, and a M. Chakrabarty, “Secretion of nucleoside

diphosphate kinase by mucoid Pseudomonas aeruginosa 8821: involvement

of a carboxy-terminal motif in secretion.,” Journal of bacteriology, vol.

182, pp. 3826–31, Jul. 2000.

[69] B. a Chopade, S. Shankar, G. W. Sundin, S. Mukhopadhyay, and a M.

Chakrabarty, “Characterization of membrane-associated Pseudomonas

aeruginosa Ras-like protein Pra, a GTP-binding protein that forms

complexes with truncated nucleoside diphosphate kinase and pyruvate

kinase to modulate GTP synthesis.,” Journal of bacteriology, vol. 179, pp.

2181–8, Apr. 1997.

[70] O. Zaborina, N. Misra, J. Kostal, S. Kamath, V. Kapatral, M. E. El-Idrissi,

B. S. Prabhakar, and a M. Chakrabarty, “P2Z-Independent and P2Z

receptor-mediated macrophage killing by Pseudomonas aeruginosa isolated

from cystic fibrosis patients.,” Infection and immunity, vol. 67, pp. 5231–

42, Oct. 1999.

[71] J. Sun, X. Wang, A. Lau, T.-Y. A. Liao, C. Bucci, and Z. Hmama,

“Mycobacterial nucleoside diphosphate kinase blocks phagosome

maturation in murine RAW 264.7 macrophages.,” PloS one, vol. 5, pp. 1–

12, Jan. 2010.

[72] H. H. Dar, D. Prasad, G. C. Varshney, and P. K. Chakraborti, “Secretory

nucleoside diphosphate kinases from both intra- and extracellular

pathogenic bacteria are functionally indistinguishable.,” Microbiology, vol.

157, pp. 3024–35, Nov. 2011.

[73] J. H. Miller, P. Funchain, W. Clendenin, T. Huang, A. Nguyen, E. Wolff,

A. Yeung, J. Chiang, L. Garibyan, M. M. Slupska, and H. Yang,

“Escherichia coli strains (ndk) lacking nucleoside diphosphate kinase are

powerful mutators for base substitutions and frameshifts in mismatch-

repair-deficient strains,” Genetics, vol. 162, pp. 5–13, Jun. 2002.

[74] K. T. Wright, R. Seabright, a Logan, a J. Lilly, F. Khanim, C. M. Bunce,

and W. E. B. Johnson, “Extracellular Nm23H1 stimulates neurite

outgrowth from dorsal root ganglia neurons in vitro independently of nerve

growth factor supplementation or its nucleoside diphosphate kinase

activity.,” Biochemical and biophysical research communications, vol. 398,

pp. 79–85, Jul. 2010.

Page 109: Discovering and exploiting bacterial proteins as ...repositorio.ul.pt/bitstream/10451/9665/1/ulfc103147_tm_goncalo_sil… · Discovering and exploiting bacterial proteins as anticancer

85

[75] I. Andolfo, D. De Martino, L. Liguori, G. Petrosino, G. Troncone, N. Tata,

A. Galasso, C. Roma, F. Chiancone, S. Zarrilli, G. Arrigoni, S. Staibano, C.

Imbimbo, and M. Zollo, “Correlation of NM23-H1 cytoplasmic expression

with metastatic stage in human prostate cancer tissue.,” Naunyn-

Schmiedeberg’s archives of pharmacology, vol. 384, pp. 489–98, Oct.

2011.

[76] P. S. Steeg, L. Kopper, U. P. Thorgeirsson, E. Talmadge, L. A. Liotta, and

M. E. Sobep, “Tumor metastasis, the process by which cells leave a tumor

and colonize distant sites, is the major cause of death,” Journal of the

Nation Cancer Institute, vol. 88, pp. 200–204, Apr. 1988.

[77] A. J. Lilly, F. L. Khanim, R. E. Hayden, and Q. T. Luong, “Nm23-H1

Indirectly Promotes the Survival of Acute Myeloid Leukemia Blast Cells

by Binding to More Mature Components of the Leukemic Clone Nm23-H1

Indirectly Promotes the Survival of Acute Myeloid Leukemia Blast Cells

by Binding to More Mature,” Cancer research, vol. 71, pp. 1177–1186,

Feb. 2011.

[78] I. L. O. Buxton and N. Yokdang, “Extracellular Nm23 signaling in breast

cancer: Incommodus verum,” Cancers (Basel), vol. 3, pp. 1–12, Sep. 2011.

[79] B.-G. Han, K.-S. Min, B.-I. Lee, and S.-H. Lee, “Refined Structure of

Human NM23-H1 from a Hexagonal Crystal,” Bulletin of the Korean

Chemical Society, vol. 31, pp. 1397–1399, May 2010.

[80] I. M. C. van Amsterdam, M. Ubbink, M. van den Bosch, F. Rotsaert, J.

Sanders-Loehr, and G. W. Canters, “A new type 2 copper cysteinate azurin.

Involvement of an engineered exposed cysteine in copper binding through

internal rearrangement.,” The Journal of biological chemistry, vol. 277, pp.

44121–30, Nov. 2002.

[81] B. N. Taylor, R. R. Mehta, T. Yamada, F. Lekmine, K. Christov, A. M.

Chakrabarty, A. Green, L. Bratescu, A. Shilkaitis, C. W. Beattie, and T. K.

Das Gupta, “Noncationic peptides obtained from azurin preferentially enter

cancer cells.,” Cancer research, vol. 69, pp. 537–46, Jan. 2009.

[82] N. Bernardes, A. S. Ribeiro, S. Abreu, B. Mota, R. G. Matos, C. M.

Arraiano, R. Seruca, J. Paredes, and A. M. Fialho, “The Bacterial Protein

Azurin Impairs Invasion and FAK/Src Signaling in P-Cadherin-

Overexpressing Breast Cancer Cell Models.,” PloS one, vol. 8, pp. 1-8, Jul.

2013.

[83] K. Kushiro and N. Núñez, “Ethanol inhibits B16-BL6 Melanoma

Metastasis and Cell phenotypes associated with metastasis,” In Vivo, vol.

26, pp. 47–58, Feb. 2012.

Page 110: Discovering and exploiting bacterial proteins as ...repositorio.ul.pt/bitstream/10451/9665/1/ulfc103147_tm_goncalo_sil… · Discovering and exploiting bacterial proteins as anticancer

86

[84] M. Xu, G. Chen, W. Fu, M. Liao, J. a Frank, K. a Bower, S. Fang, Z.

Zhang, X. Shi, and J. Luo, “Ethanol disrupts vascular endothelial barrier:

implication in cancer metastasis.,” Toxicological sciences : an official

journal of the Society of Toxicology, vol. 127, pp. 42–53, May 2012.

[85] J.-Y. Yang, C. S. Zong, W. Xia, Y. Wei, M. Ali-Seyed, Z. Li, K. Broglio,

D. a Berry, and M.-C. Hung, “MDM2 promotes cell motility and

invasiveness by regulating E-cadherin degradation.,” Molecular and

cellular biology, vol. 26, pp. 7269–82, Oct. 2006.

[86] N. Tikhmyanova and E. a Golemis, “NEDD9 and BCAR1 negatively

regulate E-cadherin membrane localization, and promote E-cadherin

degradation.,” PloS one, vol. 6, pp.1-10, Jan. 2011.

[87] S. Ramachandran, S. Sarkar, A. Mazumadar, and M. Mandal, “Azurin

Synthesis from Pseudomonas Aeruginosa MTCC 2453, Properties,

Induction of Reactive Oxygen Species, and p53 Stimulated Apoptosis in

Breast Carcinoma Cells,” Journal of Cancer Science & Therapy, vol. 3, pp.

104–111, Mar. 2011.

[88] H. Jung, H.-A. Seong, and H. Ha, “NM23-H1 tumor suppressor and its

interacting partner STRAP activate p53 function.,” The Journal of

biological chemistry, vol. 282, pp. 35293–307, Nov. 2007.

[89] S.-L. Chen, Y.-S. Wu, H.-Y. Shieh, C.-C. Yen, J.-J. Shen, and K.-H. Lin,

“P53 is a regulator of the metastasis suppressor gene Nm23-H1.,”

Molecular carcinogenesis, vol. 36, pp. 204–14, Apr. 2003.

[90] T. Desvignes, P. Pontarotti, C. Fauvel, and J. Bobe, “Nme protein family

evolutionary history, a vertebrate perspective.,” BMC evolutionary biology,

vol. 9, pp. 1–25, Jan. 2009.

[91] J. Yoon, P. Singh, D. Lee, J. Qiu, S. Cai, R. O. Connor, Y. Chen, B. Shen,

and G. P. Pfeifer, “Characterization of the 3’ to 5' exonuclease activity

found in human nucleoside diphosphate kinase 1 (NDK1) and several of its

homologues,” Biochemistry, vol. 44, pp. 15774–15786, 2005.

[92] D. Perina, M. H. Bosnar, R. Bago, A. Mikoč, M. Harcet, M. Deželjin, and

H. Cetković, “Sponge non-metastatic Group I Nme gene/protein - structure

and function is conserved from sponges to humans.,” BMC evolutionary

biology, vol. 11, p. 87, Jan. 2011.

[93] E. Suzuki, T. Ota, K. Tsukuda, A. Okita, K. Matsuoka, M. Murakami, H.

Doihara, and N. Shimizu, “nm23-H1 reduces in vitro cell migration and the

liver metastatic potential of colon cancer cells by regulating myosin light

chain phosphorylation.,” International journal of cancer. Journal

international du cancer, vol. 108, pp. 207–11, Jan. 2004.

Page 111: Discovering and exploiting bacterial proteins as ...repositorio.ul.pt/bitstream/10451/9665/1/ulfc103147_tm_goncalo_sil… · Discovering and exploiting bacterial proteins as anticancer

87

[94] F. Liu, Y. Zhang, X.-Y. Zhang, and H.-L. Chen, “Transfection of the

nm23-H1 gene into human hepatocarcinoma cell line inhibits the

expression of sialyl Lewis X, alpha1,3 fucosyltransferase VII, and

metastatic potential.,” Journal of cancer research and clinical oncology,

vol. 128, pp. 189–96, Apr. 2002.

[95] M. T. Hartsough, D. K. Morrison, M. Salerno, D. Palmieri, T. Ouatas, M.

Mair, J. Patrick, and P. S. Steeg, “Nm23-H1 Metastasis Suppressor

Phosphorylation of Kinase Suppressor of Ras via a Histidine Protein

Kinase Pathway Nm23-H1 Metastasis Suppressor Phosphorylation of

Kinase Suppressor of Ras via a Histidine,” Journal of Biological

Chemistry, vol. 277, pp. 32389–32399, Jun. 2002.

[96] Q. Zhang, J. R. Mccorkle, M. Novak, and M. Yang, “Metastasis Supressor

Function of NM23-H1 Requires its 3’-5' Exonuclease Activity,”

International journal of cancer, vol. 128, pp. 40–50, Jan. 2012.