Understanding the general role of the microRNAs in...

59
Universidade de Lisboa Faculdade de Ciências Departamento de Biologia Animal Understanding the general role of the microRNAs in Drosophila wing development Ana Patrícia de Almeida Ferreira MESTRADO EM BIOLOGIA EVOLUTIVA E DO DESENVOLVIMENTO 2009

Transcript of Understanding the general role of the microRNAs in...

Page 1: Understanding the general role of the microRNAs in ...repositorio.ul.pt/bitstream/10451/1448/1/20902_ulfc080582_tm.pdf · Em Drosophila melanogaster existem dois isotipos desta enzima

Universidade de Lisboa Faculdade de Ciências

Departamento de Biologia Animal

Understanding the general role of the microRNAs in Drosophila wing

development

Ana Patrícia de Almeida Ferreira

MESTRADO EM BIOLOGIA EVOLUTIVA E DO DESENVOLVIMENTO

2009

Page 2: Understanding the general role of the microRNAs in ...repositorio.ul.pt/bitstream/10451/1448/1/20902_ulfc080582_tm.pdf · Em Drosophila melanogaster existem dois isotipos desta enzima

Universidade de Lisboa Faculdade de Ciências

Departamento de Biologia Animal

Understanding the general role of the microRNAs in Drosophila wing

development

Ana Patrícia de Almeida Ferreira

Dissertação de mestrado orientada por:

Doutor Marco Milán – Institute for Research in Biomedicine, Barcelona Doutor Rui Gomes – Faculdade de Ciências da Universidade de Lisboa, Lisboa

MESTRADO EM BIOLOGIA EVOLUTIVA E DO

DESENVOLVIMENTO

2009

Page 3: Understanding the general role of the microRNAs in ...repositorio.ul.pt/bitstream/10451/1448/1/20902_ulfc080582_tm.pdf · Em Drosophila melanogaster existem dois isotipos desta enzima

  

1  

TABLE OF CONTENTS   

ACKNOWLEDGMENTS ................................................................................................................. 3 

I.  SUMÁRIO ................................................................................................................................. 4 

II.  ABSTRACT .............................................................................................................................. 7 

III.    INTRODUCTION ...................................................................................................................... 8 

3.1 Biogenesis of the microRNAs ............................................................................................ 8 

3.2 Regulation of miRNAs biogenesis .................................................................................. 10 

3.3 Mode of Action of the microRNAs .................................................................................. 12 

3.4 miRNAs and growth control ............................................................................................. 13 

3.5 Drosophila wing imaginal disc ......................................................................................... 14 

3.5.1 Growth control in the wing disc ................................................................................. 16 

3.5.2 Wing imaginal disc patterning ................................................................................... 18 

                 Subdivision notum versus wing ............................................................................ 19 

                 Wing hinge patterning ............................................................................................ 19 

IV.    AIM OF THE PROJECT ....................................................................................................... 21 

V.  MATERIALS AND METHODS ............................................................................................ 22 

5.1 DROSOPHILA STRAINS ................................................................................................. 22 

5.1.1 Fly husbandry .............................................................................................................. 22 

5.1.2 Mutant alleles and transgenic lines .......................................................................... 22 

5.2 GENETICS ......................................................................................................................... 22 

5.2.1 The UAS-Gal4 System ............................................................................................... 22 

5.2.2 Genetic mosaic analysis ............................................................................................ 23 

5.2.3 Mitotic recombination.................................................................................................. 24 

5.2.4 Minute technique ......................................................................................................... 24 

5.2.5 Mosaic analysis with a repressive cell marker (MARCM) ..................................... 25 

5.3 IMMUNOHISTOCHEMISTRY ......................................................................................... 26 

5.4 IN SITU HYBRIDIZATION ............................................................................................... 27 

5.5 TUNEL ................................................................................................................................ 27 

5.6 IMAGE ACQUISITION AND ANALYSIS ....................................................................... 27 

5.6.1 Quantification of cell and tissue growth parameters in adult wings and wing discs ......................................................................................................................................... 27 

Page 4: Understanding the general role of the microRNAs in ...repositorio.ul.pt/bitstream/10451/1448/1/20902_ulfc080582_tm.pdf · Em Drosophila melanogaster existem dois isotipos desta enzima

  

2  

VI.    RESULTS ............................................................................................................................... 29 

6.1 Reduced growth in the absence of Dcr-1 ...................................................................... 29 

6.2 Dcr-1 regulates E2F activity and Dap protein levels .................................................... 31 

6.3 dMyc levels are regulated by Dcr-1 activity ................................................................... 33 

6.4 Contribution of dMyc to growth and E2F activity defects caused by Dcr-1 depletion ...................................................................................................................................................... 37 

6.5 A role of bantam miRNA in reducing Mei-P26 and Dap protein levels ..................... 37 

VII.   DISCUSSION ......................................................................................................................... 41 

VIII.  CONCLUDING REMARKS ................................................................................................. 45 

IX.    REFERENCES ....................................................................................................................... 46 

X.  ANNEXES ............................................................................................................................... 51 

 

Page 5: Understanding the general role of the microRNAs in ...repositorio.ul.pt/bitstream/10451/1448/1/20902_ulfc080582_tm.pdf · Em Drosophila melanogaster existem dois isotipos desta enzima

  

3  

ACKNOWLEDGMENTS I want to thank everyone that made this thesis possible:

First of all, to my supervisor Marco, for giving me the change to be part of such a

wonderful project and lab, for guiding, teaching and supporting me. To all the members of

the Development and Growth Control lab for making me feel part of their “familia”. A very

special thanks to Héctor, without him this work would not have been possible. I thank him

for the scientific discussions and specially for his patience in teaching and helping me all

the time that I needed. To Duarte, for teaching me how to deal with flies, for being

available to answer whatever I needed and for the Portuguese discussions. All the people

that I worked with in the lab for the help and for the great environment created. I thank:

Héctor Herranz, Duarte Mesquita, Lara Barrio, Isabelle Becam, Andrés Dekanty, Neus

Rafel Berge, Georgina Sorrosal, Lidia Pérez Daniel Olivieri, Dorien Hermkens, Melissa

Cizeron, Ulla-Maj Fiúza, Florencia Tevy and Laura Boulan. I thank to professor Rui

Gomes, for guide and support. To Isadora Monteiro, João Picão, Pedro Simões and

Soraia Gonçalves for sharing with me so many great moments. Without their friendship,

support and laugh it would have been very difficult. I thank all my friends, for the joy and

happiness they bring to my life. Finally, to my parents and sister for the constant love and

support that made possible to me to get till here.

Page 6: Understanding the general role of the microRNAs in ...repositorio.ul.pt/bitstream/10451/1448/1/20902_ulfc080582_tm.pdf · Em Drosophila melanogaster existem dois isotipos desta enzima

  

4  

I. SUMÁRIO

Pequenos RNAs não-codificantes têm vindo a ser implicados na regulação de genes e

genomas a sensivelmente todos os níveis genómicos. Pequenos RNAs podem ser

categorizados nas seguintes três classes: siRNAs (do inglês short interfering RNAs),

piRNAs (do inglês piwi-interacting RNAs) e microRNAs, a melhor compreendida entre as

três. Desde a sua descoberta em 1993, em Caenorhabditis elegans, que os microRNAs

(miRNAs) se tornaram numa área com um extremo interesse devido às suas

potencialidades em vários processos na biologia do desenvolvimento, tornando-se cada

vez mais apetecível e competitiva. Como consequência, têm vindo a sofrer um

exponencial e exaustivo estudo com vista à compreensão da sua biogénese, função e

regulação. MicroRNAs são endógenos, curtos (~20-23 nucleótidos), RNAs não-

codificantes de cadeia simples que reprimem transcritos alvos através da ligação à sua

3´UTR, conferindo assim um novo nível de regulação pós-transcricional.

A transcrição dos microRNAs é mediada quer pela RNA polimerase II, quer pela RNA

polimerase III, originando transcritos primários de miRNAs (pri-miRNA). Estes têm vários

kilobases de comprimento e são caracterizados por terem muitos ‘stem-loops’. A

maturação dos microRNAs começa com a clivagem ao nível do ‘stem’ da estrutura

‘hairpin’, libertando um pequeno ‘hairpin’, o pré-miRNA, que tem cerca de 70 nucleótidos

de comprimento. O complexo microprocessador, que em Drosophila tem cerca de 500

kDa, é o responsável por esta etapa, que ocorre no núcleo. Este complexo é formado

pela proteína RNase tipo III Drosha juntamente com o seu cofactor Pasha (DGCR8 em

humanos).

Seguidamente, os pré-miRNAs são exportados para o citoplasma por um membro da

família dos receptores nucleares de transporte, a proteína Exportin 5 (EXP5). O último

passo na maturação dos microRNAs é realizado no citoplasma pela enzima Dicer. Os

pré-miRNAs são clivados perto do ‘loop’ terminal, libertando duplexes de miRNAs com

cerca de 22 nucleótidos de comprimento. Assim, durante a maturação dos microRNAs,

Drosha predetermina as sequências maduras dos miRNAs pois gera uma extremidade,

enquanto que Dicer cria a outra extremidade através da medição de 22 nucleótidos a

partir da ponta pré-existente. Dicer é uma proteína extremamente conservada que pode

ser encontrada em praticamente todos os organismos eucarióticos. Em Drosophila

melanogaster existem dois isotipos desta enzima – Dicer1 (Dcr-1), que é fundamental

para a biogénese dos microRNAs, e Dicer2 que funciona na produção de siRNA. Dcr-1

favorece ligações a hélices com emparelhamento imperfeito, de modo a processá-las.

Dicer contém um putativo domínio helicase, um domínio DUF283, um domínio PAZ (Piwi-

Page 7: Understanding the general role of the microRNAs in ...repositorio.ul.pt/bitstream/10451/1448/1/20902_ulfc080582_tm.pdf · Em Drosophila melanogaster existem dois isotipos desta enzima

  

5  

Argonaute-Zwille), dois domínios tandem RNase-III e um domínio dsRBD (do inglês

double stranded) de ligação a RNAs de dupla cadeia. Sabe-se que o domínio PAZ

permite a interacção com a extremidade 3’ que resulta da clivagem por parte de Drosha.

Dicer1 requer Loquacious (LOQS), que contém domínios de ligação a RNAs de cadeia

dupla, para o processamento de pré-miRNAs.

Depois da clivagem por parte de Dicer, a duplex de RNA resultante é carregada numa

proteína Argonaute (Ago) de modo a originar o complexo effector RISC (do inglês RNA-

induced silencing complex). Juntamente com LOQS e AGO1, Dcr-1 forma o complexo

RLC (do inglês RISC loading complex). Uma das cadeias do duplex permanece na

proteína Ago como o miRNA maduro (miRNA), enquanto que a complementar é

degradada (miRNA*). O modo como qual das cadeias é seleccionada para agir como

miRNA é ainda desconhecido.

miRNAs ligam-se, por emparelhamento imperfeito da sua região 5’ (em inglês seed

region), a múltiplas cópias de sítios de ligação presentes na região 3´UTR (do inglês 3’

untranslated region) do RNA mensageiro (mRNA) dos genes alvos. A complementaridade

entre o miRNA e o mRNA alvo é determinante do mecanismo regulatório. Enquanto que

complementaridade perfeita leva à clivagem do mRNA alvo, falhas nessa mesma

complementaridade origina repressão da tradução do mRNA.

O controlo preciso dos níveis de miRNAs é crucial para manter as funções celulares em

homeostasia e a desregulação de miRNAs associada a doenças humanas, como cancro,

já foi reportada. Casos em que miRNAs são produzidos sob o controlo dos seus próprios

alvos que regulam têm sido documentados. Este tipo de ‘feedbacks’ podem, de certo

modo, transmitir robustez ao sistema, face a uma eventual situação de ‘stress’.

Tendo em consideração que a enzima Dicer é uma peça essencial na biogénese e

função dos microRNAs, possíveis efeitos da sua ausência reflectem funções colectivas

de múltiplos miRNAs que são expressos durante o desenvolvimento. Tem sido

demonstrado que a perda de dcr-1 causa defeitos na manutenção das células estaminais

nos ovários de Drosophila e nestas células foi também observado um atraso na transição

da fase G1 para a fase S do ciclo celular. Para além de terem sido envolvidos na

regulação do ciclo celular, também já foram implicados no controlo do crescimento

celular. Os principais efectores que medeiam a actividade de Dcr-1 nestes processos não

foram ainda identificados.

O disco imaginal de Drosophila, uma monocamada epitelial que surge como um grupo de

30‐40 células que proliferam durante os três estádios larvares até atingir um tamanho

final de cerca de 50.000 células, é uma sistema extremamente apropriado para a análise,

a nível celular, do papel dos miRNAs num epitélio proliferativo e identificar os seus

efectores. De modo a atingir esse objectivo, a actividade de Dcr-1 foi afectada, utilizando

Page 8: Understanding the general role of the microRNAs in ...repositorio.ul.pt/bitstream/10451/1448/1/20902_ulfc080582_tm.pdf · Em Drosophila melanogaster existem dois isotipos desta enzima

  

6  

para esse efeito dois alelos mutantes para esta enzima, bem como RNA de interferência

(RNAi) para a mesma.

A diminuição da actividade de Dcr-1 teve efeito ao nível da taxa de crescimento do

primórdio da asa de Drosophila, um epitélio altamente proliferativo. Células mutantes

para dcr-1, ou com reduzida actividade de dcr-1, mostraram ser mais pequenas

comparativamente a células normais e originaram alas mais pequenas. Além disso,

células mutantes para dcr-1 demonstraram ser susceptíveis a competição por células

com crescimento mais rápido in vivo e a maquinaria dos microRNAs mostrou ser capaz

de promover a transição da fase G1 para a fase S do ciclo celular. Os dados obtidos

apresentam evidência de que o miRNA bantam e o proto-oncogene dMyc contribuem

para a actividade de Dcr-1 nestes processos. Na ausência de Dcr-1, os níveis de dMyc

são reduzidos, afectando alvos como E2F, CycE e Dap que estão envolvidos no controlo

do ciclo celular. Foi aqui demonstrado que os níveis proteicos de dMyc são regulados

pela actividade de Dcr-1 através de um duplo mecanismo de repressão mediado pela

proteína Mei-P26, que pertence à família TRIM-NHL. dMyc contribui para a actividade de

Dcr-1 regulando o crescimento, tanto ao nível do tecido bem como ao nível da célula, e

também a actividade de E2F (proteína envolvida no ciclo celular). Neste sistema, tanto

Mei-P26 como Dacapo (inibidor da transição da fase G1 para a fase S, reprimindo a

CycE) são alvos directos de bantam. Estes resultados mostram que a via dos microRNAs

é necessária, num epitélio altamente proliferativo, para manter os níveis de dMyc

suficientes de modo a atingir o crescimento apropriado do tecido.

PALAVRAS-CHAVE: microRNA, Dacapo, bantam, controlo de crescimento, Drosophila, ciclo celular, disco imaginal da asa   

Page 9: Understanding the general role of the microRNAs in ...repositorio.ul.pt/bitstream/10451/1448/1/20902_ulfc080582_tm.pdf · Em Drosophila melanogaster existem dois isotipos desta enzima

  

7  

II. ABSTRACT   MicroRNAs (miRNAs) are short (~20-23 nucleotides long), endogenous, single-stranded

non-coding RNAs that have been shown to repress target transcripts, by binding to their 3’

UTR, and thus conferring a new level of post-transcriptional regulation. Since 1993, the

first description of a miRNA, it has became a very exciting area in research, leading to a

crescent implication of these small RNAs in a large range of processes. They have been

implicated in cell cycle regulation and in some cases show to have a role in tissue growth

control.

Dicer-1 (Dcr-1) is a double-stranded RNaseIII essential for miRNA biogenesis, therefore

impairing its activity offers a way to access the role of the pathway in a particular

biological process. The wing imaginal disc offers a great model to access this issue in a

highly proliferative epithelium. Adult cells lacking dcr-1, or with reduced Dcr-1 activity,

were smaller than normal cells and gave rise to smaller wings. dcr-1 mutants cells showed

evidence of being susceptible to competition by faster growing cells in vivo and the

miRNA machinery was shown to promote G1-S transition. The results obtained also

present evidence that the bantam miRNA and the dMyc proto-oncogene contribute to the

activity of Dcr-1 in these processes. dMyc protein levels are regulated by the activity of

Dcr-1 through a double repression mechanism mediated by the TRIM-NHL protein Mei-

P26. dMyc contributes to the activity of Dcr-1 in regulating cell and tissue growth and E2F

activity. Mei-P26 and the p21/Dacapo CDK inhibitor are direct targets of bantam in this

system. These results define a frame in which the miRNA pathway is required, in a highly

proliferative epithelium, to maintain sufficient levels of dMyc in order to achieve proper

tissue growth.

KEYWORDS: microRNA, Dacapo, bantam, growth control, Drosophila, cell cycle, wing

imaginal disc

   

Page 10: Understanding the general role of the microRNAs in ...repositorio.ul.pt/bitstream/10451/1448/1/20902_ulfc080582_tm.pdf · Em Drosophila melanogaster existem dois isotipos desta enzima

  

8  

III. INTRODUCTION During the last centuries, gene regulation has been shown to have a huge importance in a

batch of aspects in biology, in which physiology and development are included.

Regardless of the importance of the gene regulation at the transcriptional level in

development, the relevance of post-transcriptional gene regulation has considerably

increased during the last years. Small non coding RNAs regulating genes and genomes

have been identified at almost all levels of genome function, including chromatin structure,

chromosome segregation, transcription, RNA processing, RNA stability as well as

translation. Small RNAs can be categorized in the following three classes: short interfering RNAs

(siRNAs), piwi-interacting RNAs (piRNAs) and microRNAs, the best understood among

the three. However, the boundaries of these categories are becoming very difficult to

discriminate, as new non-canonical small RNAs are discovered. Three aspects separate

piRNAs from the other two classes: while the distribution of the miRNAs and siRNAs is

broad in both phylogenetic and physiological terms, piRNAs are only found in animals and

act typically in the germline; piRNAs appear from single-stranded precursors while the

other two classes are precede from double-stranded antecedents1; piRNAs associate with

Piwi effector proteins, whereas miRNAs and siRNAs bind to Argonaute proteins2.

What distinguish miRNAs from siRNAs subsist in three aspects, most of them concerning

their biogenesis, and not their function. In the first place, miRNAs are endogenous

products, derived from the genome of the own organism, whereas siRNAs are thought to

be exogenous, derived from virus, transposon or transgene. Secondly, miRNAs are

processed from stem-loop precursors with incomplete double-stranded character, while

siRNAs are excised twice, by the same enzyme, from long, fully complementary double-

stranded RNAs (dsRNAs)3. Another important difference between these two types of small

RNAs is that miRNAs bind to the target by imperfect complementarity at multiple sites,

although siRNAs habitually form a perfect duplex with their targets at only one site4.

3.1 Biogenesis of the microRNAs

MicroRNAs (miRNAs) are short (~20-23 nucleotides long), endogenous, single-stranded

non-coding RNAs5 that have been shown to repress6 as well as activate7 target

transcripts, thus conferring a new level of post-transcriptional regulation. miRNAs were

first described in the worm Caenorhabditis elegans during the early 1990s8. Since then, a

huge effort has been made in order to understand their biogenesis, function and

regulation.

Page 11: Understanding the general role of the microRNAs in ...repositorio.ul.pt/bitstream/10451/1448/1/20902_ulfc080582_tm.pdf · Em Drosophila melanogaster existem dois isotipos desta enzima

  

9  

The majority of miRNAs are produced individually from separated units, however some of

them are produced from transcription units that make more than one product5. The

transcription of miRNAs is mediated by either RNA polymerase II or RNA polymerase III

into primary miRNA transcripts (pri-miRNAs)9,10 (Fig. 1).

The pri-miRNAs are usually several kilobases long and are characterized for having stem-

loop structures11. MicroRNAs maturation starts with the cleavage at the stem of the hairpin

structure, releasing a small hairpin, the pre-miRNA (~70 nucleotides)12. This step takes

place in the nucleus by the Microprocessor complex, which has ~500 kDa in D.

melanogaster13, formed by the RNase III-type protein Drosha14 together with the cofactor

Pasha protein (DiGeorge syndrome critical region gene 8 (DGCR8) in humans)15 (Fig. 1).

A typical metazoan pri-miRNA consists of a ~33 base pairs (bp) stem, a terminal loop and

flanking ssRNA segments. DGCR8 interacts with pri-miRNAs through the ssRNA

segments and the ~33 bp stem, and assists Drosha to cleave the substrate ~11 bp away

from the ssRNA-dsRNA junction16. It was recently shown that Drosha can cleave, not only

pri-miRNAs, but also mRNAs that contain longer hairpins17. Drosha negatively regulates

its own cofactor, DGCR8, by cleaving the hairpins in the second exon of the DGCR8

mRNA17.

After processing in the nucleus, pre-miRNAs are exported to the cytoplasm by a member

of the nuclear transport receptor family, Exportin-5 (EXP5) (Fig. 1)18. EXP5 promotes the

release of pre-miRNAs from Drosha in the nucleus, as the level of RanGTP is high, then

in the cytoplasm, where the RanGTP level is low, it releases the pre-miRNAs for further

processing. It is known that EXP5 recognizes the >14-bp dsRNA stem along with a short

3´overhang (1-8nt)19.

The last step in the maturation of the microRNAs is performed in the cytoplasm by the

enzyme Dicer. The pre-miRNAs are cleaved near the terminal loop, releasing ~22 nt

miRNAs duplexes20 (Fig. 1). Therefore, during the maturation process Drosha

predetermines mature miRNAs sequences by generating one end, while Dicer creates the

other end by measuring ~22 nt from the pre-existing terminus11. Dicer is an extremely

conserved protein that can be found in almost all eukaryotic organisms11. In Drosophila

melanogaster there are two isotypes of this enzyme – Dicer-1 (Dcr-1), that is required for

miRNAs biogenesis, and Dicer-2, that functions in siRNA production. While Dcr-1 favors

binding to imperfectly paired helices, participating in the pre-miRNAs processing, Dcr-2

prefers dsRNA with perfect complementary in order to process them21. Dicer contains a

putative helicase domain, a DUF283 domain, a PAZ (Piwi-Argonaute-Zwille) domain, two

tandem RNase-III domains and a dsRNA-binding domain (dsRBD)22. It is known that the

PAZ domain allows the interaction with the 2-nucleotide 3´overhangs that result from

Page 12: Understanding the general role of the microRNAs in ...repositorio.ul.pt/bitstream/10451/1448/1/20902_ulfc080582_tm.pdf · Em Drosophila melanogaster existem dois isotipos desta enzima

  

10  

Drosha cleavage23. D. melanogaster Dicer-1 requires Loquacious (LOQS), which have

dsRNA-binding domains (dsRBDs), for pre-miRNA processing24 (Fig. 1).

After Dicer cleavage, the RNA duplex is loaded into an Argonaute (Ago) protein in order to

give rise to a ribonucleoprotein effector complex, the RISC, mediating post-transcriptional

gene silencing25. The evolutionary conserved Argonaute (AGO) proteins are defined by

having a Piwi domain and a PAZ domain26, demonstrated to bind characteristically to 2

nucleotides 3’ overhangs typically generated by Dicer-like enzymes27. Others proteins

have been identified as RISC-associated proteins, but whether they are RISC core

components or only accessory proteins to provide functional specificity has not been

clarified yet4. Altogether with LOQS and AGO1, Dcr-1 composes the RISC loading

complex (RLC)28. One strand of the duplex remains in Ago as a mature miRNA (guide

strand or miRNA), while the other (passenger strand or miRNA*) is degraded11 (Fig. 1).

The way of determinate which of the strand is going to be selected to act as miRNA is not

well known, nevertheless studies in the miRNA precursors indicated that the

determination is related with the relative thermodynamic stability of the two ends of the

duplex29. However some hairpins produce miRNAs from both strands at comparable

frequencies11. Studies have shown that removal of the passenger strand of siRNA and of

some miRNA duplexes is responsibility of the endonucleolytic enzymatic activity (slicer

activity) of Ago protein30. However, as the majority of miRNAs have central mismatches,

and as some Ago proteins do not have slicer activity, an helicase is thought to mediate

unwinding and removal of the unselected strand of the miRNA duplex11.

In D. melanogaster, the determination of small RNA sorting is based in the structure of the

precursor31. While miRNAs duplexes with mismatches are preferentially sorted in AGO1,

perfectly matching siRNAs duplexes are loaded into AGO211.

3.2 Regulation of miRNAs biogenesis

Studies on expression profiles indicate that miRNAs are under control of developmental

and/or tissue specific signaling32. Precise control of miRNA levels is crucial to maintain

normal cellular functions and deregulation of miRNA have been shown to be associated

with human diseases, such as cancer33.

Transcription is one point of regulation in miRNAs biogenesis, given the fact that both

RNA polymerases are regulated in a different way and recognize specific promoter and

terminator elements, this provides a broad range of regulatory options10. Several Pol II-

associated transcription factors are involved in transcriptional control of miRNAs genes,

namely tumour suppressors and oncogenes. The tumour suppressor p53 have been

shown to activate one family of miRNAs34, whereas the oncogenic protein MYC

Page 13: Understanding the general role of the microRNAs in ...repositorio.ul.pt/bitstream/10451/1448/1/20902_ulfc080582_tm.pdf · Em Drosophila melanogaster existem dois isotipos desta enzima

  

11  

transactivates or represses miRNAs that are involved in the cell cycle and apoptosis35.

There are reported cases in which the epigenetic control, namely DNA methylation, also

contributes to miRNA gene regulation36.

Figure 1 – miRNAs biogenesis pathway. Canonical microRNAs genes are transcribed by RNA polymerase II / III (Pol II / III) to generate the primary transcripts (pri-miRNAs). The initiation step (cropping) is mediated by the Drosha-DGCR8 (Pasha in D. melanogaster) complex, generating ~70 nucleotides pre-miRNAs. A pre-miRNA has a short stem and a ~2-nt 3’ overhang that is recognized by the Exportin-5-Ran-GTP, which translocates it to the cytoplasm. Then, the cytoplasmic RNase III Dicer, together with TRBP (LOQS in Drosophila), catalyzes the second processing step (dicing) to produce miRNAs duplexes. In flies, Dicer-1 and AGO1 (Ago2 in the scheme) mediate the assembly of the miRNA duplex into the RISC. One strand of the duplex, the mature miRNA, remains on the Ago protein, while the other strand is send to degradation. (Adapted from Ref. 25)

The number of miRNAs controlled at post-transcriptional level is increasing, although the

mechanisms of such regulation are yet poorly understood. All steps of miRNA maturation

are possible points for post-transcriptional regulation. Nuclear RNA-binding proteins with

influence in miRNA processing through specific interactions with pri-miRNAs were

described37. For example, the RNA-binding protein LIN28 has been shown to have a role

in suppression of let-7 biogenesis38. The mechanisms proposed for this suppression

include blockage of Drosha processing, interference with Dicer processing and terminal

uridylation of pre-let-7. Another possibility for regulation is the turnover of miRNA, taking

Page 14: Understanding the general role of the microRNAs in ...repositorio.ul.pt/bitstream/10451/1448/1/20902_ulfc080582_tm.pdf · Em Drosophila melanogaster existem dois isotipos desta enzima

  

12  

into account that RNA decay enzymes might target not only mature miRNAs, but also the

precursors (pri-mRNAs and pre-miRNAs)39. The RNA editing can also change the target

specificity of the miRNA if it occurs in miRNA sequences40.

miRNA biogenesis has been shown to be controlled by multiple levels of feedback loops

involving the biogenesis factors, the miRNAs themselves and their targets. It has been

shown that Drosha and Dicer are controlled by single negative feedbacks to maintain

homeostasis of miRNA production17,41. Another studies have shown that double-negative

feedback control is frequently used as an effective genetic switch of a specific miRNA

during a particular stage of development38. Cases in which miRNAs genes are produced

under the control of the targets that they regulated have also been documented42.

3.3 Mode of Action of the microRNAs

miRNAs typically bind, through their 5´ end, to their multiple copies of binding sites

present in the 3’-untranslated regions (UTR) of the target messenger RNA by imperfect

base pairing43 (Fig. 2). Despite of the mismatches, a mean aspect of the recognition step

involves Watson-Crick base pairing of miRNA seed region (nucleotides 2-8)2.

Figure 2 – Base-pairing between microRNA and target mRNA. The seed sequence (2 to 8 bases) of the 5’ end of the microRNA (shown by an arrow) make almost perfect base-pairing within the 3’ UTR sequences of the target gene (dotted region). (Adapted from Ref. 43)

One key determinant of the regulatory mechanism is the degree of miRNA-mRNA

complementarity. While perfect match consent to Ago-catalyzed cleavage of the mRNA

strand, central mismatches encourage repression of mRNA translation by the Ago protein

(Fig. 1). Similar to let-7 in worms44, in flies, the anti-apoptotic miRNA bantam, binds to the

3’UTR of its targets and negatively regulate their translation45.

Although in animal kingdom the majority of miRNAs repress target translation, some

cases of miRNA-mRNA interactions are accompanied by a reduction in mRNA abundance

because of deadenylation, decapping, and exonucleolytic digestion of the mRNA leading

to an increase in mRNA degradation46. As example, mir-196 directs mRNA cleavage of its

target, Hoxb847. In order to try to explain why there are some targets that are degraded

Page 15: Understanding the general role of the microRNAs in ...repositorio.ul.pt/bitstream/10451/1448/1/20902_ulfc080582_tm.pdf · Em Drosophila melanogaster existem dois isotipos desta enzima

  

13  

and others not, it has been proposed that the nature, position and amount of mismatches

between the miRNA and the mRNA could have responsibility in triggering or degradation

or translation arrest48.

3.4 miRNAs and growth control

Most of the miRNAs characterized so far seem to regulate aspects of development.

MicroRNAs have been implicated in cell cycle regulation49,50,51,52,53,54 and in some cases

were shown to have a role in tissue and cell growth control35,45,55,56.

In Drosophila the first microRNA associated with growth control, named bantam, was

shown to generate smaller flies in loss-of-function situations57. The wings of bantam

mutants were characterized by having fewer, but normal sized cells57. Later, it was shown

that, besides the role of bantam in tissue growth by stimulating cell proliferation, it was

also involved in the prevention of cell death blocking hid-induced apoptosis45. Afterward,

evidence was published that the Hippo pathway regulates bantam to control tissue

growth, namely cell proliferation and apoptosis, in Drosophila58.

Mutations in Dicer disrupt the miRNAs due to the role of this enzyme in their biogenesis

and cause diverse developmental defects already documented in C. elegans, A. thaliana,

zebrafish and mice. Because Dicer is an essential piece in miRNA biogenesis and

function, the defects certainly reflect the collective functions of multiple miRNAs that are

expressed during early development. Loss of dcr-1 causes defects in Drosophila and

vertebrate stem cell maintenance and a delay in G1-S transition is also observed in these

cells50,59,60. It was proposed that miRNAs are essential to bypass the G1-S transition

checkpoint by repressing, directly, its inhibitor Dacapo (Dap) in Drosophila61,62. Dacapo,

the Drosophila p21/p27 (Cdkn1a/Cdkn1b) ortholog, is a CDK inhibitor, that traps the

Cyclin E/CDK2 complex in a stable but inactive form63, 64 (Fig. 3). Dacapo was

documented to have miRNA binding sites in the 3’ UTR65. This checkpoint is also inhibited

by the Retinoblastoma family proteins, Rbf1 and Rbf2, which interact and negatively

regulate E2F activity. E2F proteins are necessary for a homeostatic mechanism that

coordinates the rates of G1-S and G2-M progression and that allows cells to accelerate

progression during G2 when progression through G1 is retarded, or vice versa, probably

by targeting Cyclin E66. It was suggested that, miRNA- and Dap-based cell cycle

regulation, in the ovarian stem cells, might be under the control of the Insulin (InR)

signaling pathway, showing a crosstalk between intrinsic and extrinsic regulators in

division control53. Moreover, in ovarian stem cells, a binding partner of Argonaute-1, the

TRIM-NHL protein Mei-P2667, was shown to inhibit the microRNA pathway, restricting

Page 16: Understanding the general role of the microRNAs in ...repositorio.ul.pt/bitstream/10451/1448/1/20902_ulfc080582_tm.pdf · Em Drosophila melanogaster existem dois isotipos desta enzima

  

14  

growth and proliferation68. Furthermore, in the developing mouse limb, loss of dcr-1 leads

to growth deficiencies69.

Figure 3 – Cell cycle. The miRNA pathway modulates the cell cycle of the stem cells by repressing p21/Dap and thus affecting the G1-S transition. (Adapted from Ref. 50)

The majors effectors mediating the activity of Dcr-1 in these processes have not been

identified so far. The wing imaginal disc of Drosophila is an extremely appropriate system

to analyze, at a cellular level, the role of miRNAs in a proliferative epithelium and identify

such effectors. Since Dcr-1 is a crucial component in miRNAs biogenesis, impairing its

activity offers a way to access the role of microRNAs in a particular biological process.

3.5 Drosophila wing imaginal disc The appendages of Drosophila have proven to be ideal models for the investigation of a

vast range of aspects of limb development, namely cell and tissue growth. Like the

appendages of vertebrates, they develop as outgrowths of the body wall that are initially

inside the larva. Drosophila developing adult tissues are sac‐like structures, called

imaginal discs, that will give rise namely to wings, legs and eyes of the adult fly (imago)

(Fig. 4). The wing primordium is a continuous epithelial monolayer that arises from the late

embryonic ectoderm as a group of 30‐40 cells and proliferates during the three larval

stages to reach a final size of around 50.000 cells70. This growth happens in 5 days of

continued division (cell proliferation) and the control of tissue size is intrinsic to the disc70.

As Drosophila undergoes complete metamorphosis the primordium extends along its

proximal-distal axis by turning inside out through the lumen (appendages come outside of

Page 17: Understanding the general role of the microRNAs in ...repositorio.ul.pt/bitstream/10451/1448/1/20902_ulfc080582_tm.pdf · Em Drosophila melanogaster existem dois isotipos desta enzima

  

15  

the body wall after disc eversion)71. In particular, wing imaginal discs have been used for

the analysis of combinatorial effects, of many conserved signaling molecules, in the

specification of positional values for differentiation and growth.

Figure 4 – Imaginal disc and the structures they give rise to. (a) A third-larva instar showing the position of the imaginal discs (colored according to the adult structure that they will develop into). Wing, haltere and leg disc photographs are shown. (b) Body of an adult fly indicating the various appendages. Mouthparts and analia are also considered appendages. Appendages are segment specific structures; the second thoracic segment develops a wing in dorsal and the second leg in the ventral region, while in the third segment a haltere and the third leg appear. (Adapted from Ref. 72)

The greater part of the wing disc consists of a columnar epithelium, that will give rise to

most adult cuticular structures, covered by a squamous epithelium named the peripodial

Page 18: Understanding the general role of the microRNAs in ...repositorio.ul.pt/bitstream/10451/1448/1/20902_ulfc080582_tm.pdf · Em Drosophila melanogaster existem dois isotipos desta enzima

  

16  

membrane. After disc eversion, the luminal face forms the external surface of the adult

structures. The outmost region of the disc constitutes the thoracic structures (notum and

pleura), the next ‘ring’ will make the wing hinge and the central pouch gives rise to the

wing blade73 (Fig. 5).

Figure 5 – Fate determination along the proximal-distal axis. This determination is believed to result from complex interactions between the anterior-posterior and dorso-ventral morphogen-mediated signaling, leading to the specification of the wing blade, the wing hinge and the notum, that will give rise to the adult hemithorax. (Adapted from Ref. 74)  

 3.5.1 Growth control in the wing disc

During the growth and development of most organs, a multitude of cellular processes are

orchestrated to give rise to an organ of proper size, pattern and proportion. Specifically,

rates of cell growth and cell division are coordinated so that cell size does not change

much over time. Cell growth (increase in cell mass), cell division, cell proliferation

(increase in cell number) and also cell death all play important roles in morphogenesis

and growth of Drosophila. Namely, the morphogenetic patterning mechanisms that specify

cell fate across a field of cells must take into consideration the increasing size of the field.

Moreover, cell movements, such as migration, must also deal with this increasing in the

size of the field and must be coordinated with demands placed on the cytoskeleton by the

mitotic apparatus of dividing cells.

Each imaginal disc has a distinctive shape and size, indicating that its growth has been

regulated before the onset of its morphogenesis75. In the Drosophila wing disc, cell growth

can be uncoupled from cell division. When cell division is slowed or blocked, cells

continue to accumulate mass, and hence increase in size76. Cell division rates do not

Page 19: Understanding the general role of the microRNAs in ...repositorio.ul.pt/bitstream/10451/1448/1/20902_ulfc080582_tm.pdf · Em Drosophila melanogaster existem dois isotipos desta enzima

  

17  

drive growth, however it is unclear whether the converse is true and cell growth is

sufficient to drive cell division77. The rate of cellular growth is determined by the identity of

a particular organ, the position of a cell within this organ and its interactions with

neighboring cells76. In addition to cell proliferation, apoptosis, that takes place through

local cell‐cell interactions, also provides a mechanism for the generation of cell territories

with continuous positional values, relevant to size and shape developmental control78.

Genes involved in many aspects of growth at the organism and tissue levels can be

divided in two distinct classes: the first one includes genes that control synthesis of

proteins and other metabolic processes, affecting both the rate of growth and the organ

final size (these genes respond to extrinsic signals like environmental cues); the second

class comprises genes that determine identity, pattern, structure and final size of imaginal

discs, although their effect on growth rates is not so clear (disc intrinsic signals)76.

Therefore, developmental programs must be sufficiently robust and flexible to allow for

changes in the size of a developing organ or organism, whether over developmental time,

through evolution, or in response to external cues. Understanding how growth is

controlled and how it is integrated with other cellular processes is of critical importance to

the developmental biology field.

One known signaling pathway that regulate these processes, in response to growth

factors and nutrients, is the Insulin/PI3K/TOR pathway. When the pathway is inactive it

causes a strongly decrease in cell and organ size, and the other way around. FoxO is a

target of the pathway, that is downregulated when the pathway is active79. The well known

Hippo (Hpo) pathway controls organ size by coordinately regulation of cell growth,

proliferation and apoptosis. The pathway consists of a cascade of kinases, starting with

Hpo, that leads to the transcriptional control of many targets. The pathway nuclear effector

Yorkie (Yki), a transcriptional coactivator, is upstream of the targets. This pathway has a

variety of target genes, including the cell cycle regulator Cyclin E and the cell death

inhibitor Diap1. Expanded (Ex), an upstream regulator of Hpo, functions as an adaptor

protein that links transmembrane proteins to the cytoskeleton or cytoskeleton-associated

proteins80.

Myc protein have been described to have functions in a wide range of fields, including cell

biology, cell cycle, apoptosis, development, signal transduction, transcriptional and post-

transcriptional regulatory mechanisms, non-coding RNAs, stem cell biology and cancer81.

dMyc protein contain a C-terminal basic-helix-loop-helix-zipper (bHLHZ) which mediates

DNA-binding and dimerization82. This regions allows for its heterodimerization with the

protein Max, leading to DNA binding and consequently stimulating transcription83. Within

the N-terminal is a short motif called Myc-box II (MBII), essential for most biological

functions of dMyc. The antagonist of the Myc’s ‘network’ is dMnt, that was shown to also

Page 20: Understanding the general role of the microRNAs in ...repositorio.ul.pt/bitstream/10451/1448/1/20902_ulfc080582_tm.pdf · Em Drosophila melanogaster existem dois isotipos desta enzima

  

18  

bind dMax82. dMyc has been proven to be a master regulator of growth, since mutations in

dMyc lead to a decrease in cell size, body size and viability. Many of these defects are

also characteristic of mutations in genes encoding ribosomal proteins (the Minute class of

mutations) and other components of ribosome biogenesis82. dMyc mutants also showed a

small increase in the fraction of cells in the G1 phase of the cell cycle. On the other hand,

overexpressing dMyc increased cell size by promoting G1-S transition and thus

accelerating cellular growth83. All these growth defects in dMyc mutant cells caused these

cells to become subject to cell competition when surrounded by normal cells in the

Drosophila wing. Cell competition is a phenomenon in which slowly growing cells are

eliminated from the epithelium83. This occurrence was first described in Minutes, a class of

mutations with similar phenotypes of dMyc mutants, including developmental delay and

small body size84. Cells lost in competition die by apoptosis, but how the apoptotic

program is initiated is not clear85. Therefore, cell competition might contribute to a size-

control mechanism. For example, although dMyc overexpressing clones in the developing

wing proliferate more than wild type cells, they do not change the final size of the adult

wing. Overgrowth is prevented because the competitive elimination of wild type cells

compensates for the increased growth of dMyc-expressing cells82. Overall, the conclusion

is that dMyc is strongly required in vivo for cellular growth, and that it acts in a dose-

sensitive manner.

3.5.2 Wing imaginal disc patterning

The establishment of the three major axes and the definition of compartments of the

appendages takes place during imaginal disc development. The primordium receives the

patterning cues needed to generate an anterior‐posterior, dorsal‐ventral and

proximal‐distal axes. Diffusible signals termed morphogens86 provide cells within the

epithelium with positional information87. Distinct properties, like differences in adhesion88,

make anterior and posterior cells immiscible populations and thus defining a compartment

boundary, the A-P boundary89. This has been shown after, given a growth advantage to

some cells, observe that they still cannot cross the boundary, subsequently remaining

within their compartment71. Compartments are defined as subdomains of a tissue that are

separated by a boundary of cell lineage restriction with organizing activity, that pattern the

surrounding tissue and divide up tissues in terms of cell fate/identity71.

Early in larval development, an invaginating cell cluster that constitutes the leg and wing

common precursor, inherits the stripe pattern of Engrailed (En) and Wingless (Wg)

expression from the anterior-posterior (A-P) boundary of an embryonic parasegment. The

wing disc derives from the dorsal part of this common precursor, therefore taking with you

Page 21: Understanding the general role of the microRNAs in ...repositorio.ul.pt/bitstream/10451/1448/1/20902_ulfc080582_tm.pdf · Em Drosophila melanogaster existem dois isotipos desta enzima

  

19  

a portion of the En stripe that will specify the posterior compartment of the wing, but not

the Wg expression90.

Subdivision notum versus wing

The first step in wing development is to define the region where the wing will form. During

second larval instar, 24-48h after hatching, wg starts to be expressed in the ventral region.

This wg expression is controlled by the Hh signaling pathway, which is also required to

establish the A-P organizing center91. On the other hand, the formation of the notum

region is dependent on the activity of the EGFR pathway, where activation of the pathway

is done by the protein Vein92. wg suppresses Vein expression in the ventral region,

restricting Vein expression to the dorsal part of the disc. Vein is also capable of

suppressing wg expression in this dorsal region92. This antagonist relationship between

these two pathways leads to the division of the disc into notum and wing regions.

Wing hinge patterning

As explained above, the first subdivision in the wing primordium is between an anterior (A)

and a posterior (P) compartment, innate from the embryonic ectoderm, by the restricted

expression and activity of the homeodomain protein Engrailed in P cells89, 93. hedgehog

(hh) expression, promoted in P cells by En93, acts as a short-range morphogen to specify

decapentaplegic (dpp) expression in a narrow stripe of cells adjacent to the A-P

boundary91(Fig. 8). Dpp is a member of the TGF-β family and operates in a concentration

dependent manner to promote patterning, inducing target  genes like spalt (sal),

optomotor-blind (omb) and vestigial (vg)94, 95. Besides giving genetic positional values

along the medial-to-lateral wing, the Dpp activity gradient seems to indirectly promote

wing growth96, being counterbalanced by the brinker transcriptional repressor, which is

expressed in a complementary lateral-to-medial fashion, being itself repressed by Dpp

activity97. The important contribution of Dpp signaling to the proximal-distal patterning of

the wing disc is further completed by intersection of signals coming from the dorsal-ventral

(D-V) boundary of the wing disc. During the early stages of larval development, when the

disc consists of 100 cells, the D-V boundary is established as a result of apterous (ap)

expression in the dorsal region of the disc92, 98 (Fig. 6). This is a consequence of the

antagonist relationship between EGFR and Wg signaling explained above. Ap, through a

complex regulation of the Notch ligands Serrate and Delta, is able to activate Notch at the

D-V boundary99. Afterward, Notch directly activates transcription of genes at the D-V

boundary, such as wg and cut, which restrict expression of Notch ligands, fine-tuning

Notch activity to a narrow stripe of cells100.

Page 22: Understanding the general role of the microRNAs in ...repositorio.ul.pt/bitstream/10451/1448/1/20902_ulfc080582_tm.pdf · Em Drosophila melanogaster existem dois isotipos desta enzima

  

20  

 

Figure 6 – Territorial subdivision of the imaginal wing disc. (a) The first subdivision between anterior (A) and posterior (P) is defined by Engrailed in the P compartment. Signaling from posterior to anterior cells by Hedgehog (Hh) causes A cells near the boundary to express Dpp, which diffuses into both compartments, acting as a morphogen and inducing activation of distinct targets in diverse domains. The second subdivision is along the proximal-distal axis and carried out by three signaling pathways. In the first, Wingless (Wg) is expressed in the distal part of the disc, defining the wing primordium by activating wing-specific genes. Second, by downregulating EGFR signaling, that becomes restricted to the proximal region, defines the notum through activation of the Iroquois complex (Iro-C). In the third, Dpp, that in early stages is only active in distal regions, confines Iro-C expression to the proximal part. (b) A dorsal-ventral subdivision is achieved by the expression of Apterous (Ap) in D compartment (comprises the notum and the dorsal part of the wing). In the third instar, Dpp subdivides the notum into lateral and medial domains by activating pannier (pnr). Pnr represses Iro-C and provides identity to the medial notum. The lateral notum is defined by the continuing expression of Iro-C. (Adapted from Ref. 101)

In addition, in the wing blade, scalloped (sc) and vestigial (vg) are activated by

combination of Notch, Wg and Dpp signaling102, providing the organizer functions of the D-

V boundary. Functions that include promoting growth along the proximal-distal axis103, and

specifying a gradient of cell-affinities that discriminates the blade from the hinge104.

In the end, this complex circuit of signaling molecules seems to drive the establishment

and maintenance of region specific cell architecture, giving rise to a wing properly

patterned.

Page 23: Understanding the general role of the microRNAs in ...repositorio.ul.pt/bitstream/10451/1448/1/20902_ulfc080582_tm.pdf · Em Drosophila melanogaster existem dois isotipos desta enzima

  

21  

IV. AIM OF THE PROJECT

The importance of the microRNAs, as post-transcriptional regulators, in developmental

processes is increasing exponentially nowadays. In Drosophila most studies in this area

have been reported in ovarian and neural stem cells. The fact that the wing disc is a highly

proliferative epithelium makes it an excellent model to analyze the general role of

microRNAs. As principal aim of this project, we wanted to characterize the phenotype of

depletion of Dicer-1 in a proliferative epithelium. Understand its consequences both at the

level of adult structures, as at an early phase in development by looking at wing imaginal

discs. Once characterize, identify which are the effectors mediating the role of miRNAs in

wing disc development. In addition, investigate whether bantam miRNA, by itself or acting

redundantly with other miRNAs, has a role in the wing and concretely if is targeting the

effectors of the miRNA machinery on its all.

Page 24: Understanding the general role of the microRNAs in ...repositorio.ul.pt/bitstream/10451/1448/1/20902_ulfc080582_tm.pdf · Em Drosophila melanogaster existem dois isotipos desta enzima

  

22  

V. MATERIALS AND METHODS

5.1 DROSOPHILA STRAINS

5.1.1 Fly husbandry

All fly stocks were raised at 25ºC, according to standard conditions105. Crosses were

cultured in small vials containing a yeast-glucose-agar medium.

5.1.2 Mutant alleles and transgenic lines

The mutant alleles for Dicer-1 utilized in this work were the dcr-1Q1147X and dcr-1d102 50.

E2F1 responsive element106, in order to monitor E2F activity ; UAS-dMyc, hs-dMyc83; tub-

dMyc107; bantam1, UAS-bantam-GFP, bantam sensor (JB20)45; UAS-CycE108, UAS-mei-

P26RNAi-7553, UAS-mei-P26RNAi-106754, UAS-dMycRNAi, UAS-dcr-1RNAi (Vienna Drosophila

RNAi Center); hs-bantam68; dap4 and Rbfsls15; en-gal4, ptc-gal4, UAS-GFP and other

stocks are described in Flybase.

5.2 GENETICS

5.2.1 The UAS-Gal4 System

Expression of UAS constructs was performed using the Gal4 system at 29ºC, unless

mentioned otherwise. Gal4 protein is a yeast transcription factor, identified in

Saccharomyces cerevisiae, that regulates the Gal genes in the metabolism of glucose.

Gal4 gene has been cloned in a variety of organisms, such as Drosophila, and is used in

genetic manipulation of genes. UAS sequences, promoter sequences containing a

consensus binding site for activating Gal factors, have been introduced in eukaryotic

expression vectors and thus allowing the production of transgenic constructs that are

conditionally expressed upon the control of the Gal4109. Therefore, in Drosophila, a

parental stock carrying a Gal4 line can be cross to another stock with a UAS-construct,

and as a result, expression of the later is driven under the pattern of expression of the

Gal4 in the progeny109 (Fig. 1). Several well characterized enhancer sequences have

been trapped by the Gal4 coding gene to yield spatio-temporal specific drivers of gene

expression110. The Gal4 drivers used in this work are represented in Table 1.

Page 25: Understanding the general role of the microRNAs in ...repositorio.ul.pt/bitstream/10451/1448/1/20902_ulfc080582_tm.pdf · Em Drosophila melanogaster existem dois isotipos desta enzima

  

23  

Table 1 – Gal4 drivers used for this work.

Gal-4 drivers Spatio-temporal domain of activity in the wing disc

engrailed-Gal4 (en-Gal4) From end of embryogenesis to end of larval stage in the posterior compartment

patched-Gal4 (ptc-Gal4) From the early embryogenesis to end of larval stage along the A-P boundary

engrailed-gal4 or patched-gal4 virgins were crossed with males carrying different UAS

transgenes and allowed to lay eggs for 24 hours at 25ºC. Resulting larvae developed at

29ºC until adulthood (temperature maximal for Gal4 activity109).

Figure 1 – Gal4-UAS system in Drosophila. When females carrying the UAS construct (UAS-GFP) are mated to males that carry a Gal4 driver, progeny containing both elements are produced. The presence of Gal4 in an alternating segmental pattern in the depicted embryos then drives expression of the UAS element in the corresponding pattern (central panel). (Adapted from Ref. 108)

5.2.2 Genetic mosaic analysis

Genetic mosaic techniques, crucial for our current understanding of many developmental

processes in Drosophila, are those that induce genetic changes in a subset of cells in an

individual organism. Such techniques are very important since they provide a way of

examining homozygous situations that would be embryonic lethal if applied to the entire

Page 26: Understanding the general role of the microRNAs in ...repositorio.ul.pt/bitstream/10451/1448/1/20902_ulfc080582_tm.pdf · Em Drosophila melanogaster existem dois isotipos desta enzima

  

24  

organism. Another advantage is that genetic mosaics juxtapose wild type (control) and

genetically modified cells, and this can be used to test the cell-autonomy of a mutant

phenotype.

5.2.3 Mitotic recombination

The FLP/FRT system was used to generate mitotic clones. The Flipase (FLP)

recombinase belongs to the family of integrases from S. cerevisiae. Members of this

family are known to direct site-specific recombination of two DNA strands in cis, which

occurs through the recognition of Flipase Recombination Target (FRT) sites111. Several

stocks have been generated with FRTs inserted close to the centromere of each

chromosome, through the p-element insertion, as well as a transgene encoding FLP

driven by the hsp70 heat-shock inducible promoter (hs-FLPase)112. So, if a fly have two

FRTs in the equivalent position in both chromosomes, heat-shock induced expression of

the FLPase can cause recombination between the two sites. As a result, in a

heterozygous setup, recombination between nonsister chromatids results in the

generation of a daughter cell homozygous for the mutation distal to the FRT site.

Succeeding cell division of this homozygous mutant cell give rise to a mutant clone that

can be analyzed phenotypically. On the other hand, from the same mitotic recombination

event, a wild type homozygous sibling cell is generated, giving rise to the twin spot of the

mutant clone. Usually, the wild type FRT chromatid contains a cell marker, such as GFP,

under control of a ubiquitous promoter, or the lacZ gene under the control of armadillo.

This permits the generation of negatively marked mutant clones (Fig. 2).

The following genotypes were used to generate loss-of-function clones by the classic

FLP/FRT system in order to analyze twin/clone: hs-FLP/+; FRT82 dcrQ1147X/FR82 Ubi-

GFP, hs-FLP/+; FRT82 dcrQ1147X/FR82 arm-lacZ, hs-FLP/+; FRT82 dcrd102/FR82 Ubi-GFP;

hs-FLP/+; FRT82 dcrd102/FR82 arm-lacZ and hs-FLP/+; bantam-sensor-GFP; FRT82

dcrQ1147X/FR82 arm-lacZ. Larvae were grown at 25ºC, treated for 45 minutes at 37ºC

during early second instar and dissected 72 hours later. Mutant cells were marked by the

absence of GFP or lacZ (antibody to β-gal) expression.

5.2.4 Minute technique

The FLP/FRT system was combined with the Minute technique (Minute mutations are

defective in ribosomal proteins, and in heterozygous conditions induce a delay in growth

rates84) to induce clones covering large areas of the wing. These clones were wild type for

the Minute mutation and had a growth advantage over the neighboring Minute/+

Page 27: Understanding the general role of the microRNAs in ...repositorio.ul.pt/bitstream/10451/1448/1/20902_ulfc080582_tm.pdf · Em Drosophila melanogaster existem dois isotipos desta enzima

  

25  

heterozygous wing cells. This technique can also increase the chances that a mutant

clone survives. As mutant clones in imaginal discs are often lost because of a

phenomenon called cell competition, where abnormal slow-growing cells are eliminated in

some way by the surrounding wild type cells, giving the clone a growth advantage using

the Minute technique often rescues it112.

Minute clones in adult wings were generated in the following genotype: hs-FLP; FRT 82

P(forked+) M(3)95A2/FRT82 dcr-1Q1147X. Larvae were grown at 25ºC, treated for 15

minutes at 37ºC during early second instar and developed back at 25ºC until adulthood.

Mutant cells were marked by the absence of the P(forked+) rescue construct.

Minute clones in wing imaginal discs were generated in the following genotypes: hs-FLP/+;

FRT82 dcrQ1147X/FR82 M(3)95A2 Ubi-GFP, hs-FLP/+; FRT82 dcrQ1147X/FR82 M(3)95A2

arm-lacZ, hs-FLP/+; FRT82 dcrd102/FR82 M(3)95A2 Ubi-GFP, hs-FLP/+; FRT82 /FR82

M(3)95A2 Ubi-GFP, hs-FLP/+; FRT82 /FR82 M(3)95A2 arm-lacZ, hs-FLP; M(3L) ubi-GFP

FRT80B/ban∆1 FRT80B. Larvae were grown at 25˚C, treated for 3 minutes at 37˚C during

first instar and dissected 96 hours later. The heat-shock treatment was short to reduce the

frequency of clones per wing and avoid fusion of clones. Mutant cells were marked by the

absence of GFP or β-gal.

5.2.5 Mosaic analysis with a repressive cell marker (MARCM)

The MARCM technique is used to generate positively marked loss-of-function clones

simultaneously expressing diverse transgenes in those clones113. This technique makes

combined use of Gal4 together with its repressor Gal80, arranged in a manner such that,

FLP-mediated mitotic recombination causes genetic loss of Gal80 and the associated de-

repression of Gal4. Such de-repression can be set to happen in a mutant clone in a way

that allows the expression of a certain UAS-marker, like UAS-GFP (Fig. 2)113.

The following genotypes were used to generate MARCM clones: hs-FLP tub-Gal4 UAS-

GFP; UAS-dMyc/+; FRT82 dcrQ1147X / FRT82 Gal80, hs-FLP tub-Gal4 UAS-GFP;; FRT82

dcrQ1147X / FRT82 Gal80 and hs-FLP tub-Gal4 UAS-GFP; UAS-CycE/+; FRT82 dcrQ1147X /

FRT82 Gal80. Larvae were grown at 25ºC, treated for 45 minutes at 37ºC during first instar

and dissected 96 h later. Mutant cells were marked by the presence of GFP.

Page 28: Understanding the general role of the microRNAs in ...repositorio.ul.pt/bitstream/10451/1448/1/20902_ulfc080582_tm.pdf · Em Drosophila melanogaster existem dois isotipos desta enzima

  

26  

Figure 2 – Genetic mosaic techniques. (A) Mitotic recombination induced by the FLP recombinase during the cell cycle G2 stage results in the loss of heterozigosity at the locus coding for the mutation (black star) and a ubiquitously expressed cell marker (green box). As a consequence, one of the daughter cells will become the precursor of the mutant clone, while the other will become the complementary wild type population (twin spot) positively marked with a cell marker, such as GFP. (B) In the MARCM technique, Gal80 represses the expression of the UAS-construct (green box), such as UAS-GFP, but loss of heterozigosity at the Gal80 locus, as induced by FLP mediated mitotic recombination, results in the expression of the construct in the homozygous mutant clone.  

5.3 IMMUNOHISTOCHEMISTRY

Third larvae instar imaginal discs were dissected in PBS and fixed in 4% formaldehyde in

PBS, for 20 minutes at room temperature. Larvae were then washed and permeabilized in

PBT (PBS 1x, 0,2% Triton X-100) 3 times, 10 minutes each. Afterwards they were blocked

in BBT (PBS 1x, 5mM NaCl, 0,2% Triton X-100, Bovine Serum Albumin - BSA) for 45

minutes and incubated with the primary antibody in BBT overnight at 4ºC. Then, larvae

were blocked in BBT 3 times, 15 minutes each, and incubated in the dark with the

secondary antibody (and/or DAPI) diluted in BBT for 1 hour and 30 minutes at room

temperature. Next, they were washed 4 times, 15 minutes each, in PBT and mounted in

mounting medium (nPG 50% in EtOH, PBS 10x, Glycerol anhydrous, pH 8,5/9).

For the treatment of imaginal discs with MG-132, a proteosome inhibitor, en-gal; UAS-

mei-P26 third instar wing discs were cultured in cl-8 cell medium containing 40μM MG132

(experimental discs) or an equal volume of DMSO (control discs) for three hours before

fixation and staining.

Page 29: Understanding the general role of the microRNAs in ...repositorio.ul.pt/bitstream/10451/1448/1/20902_ulfc080582_tm.pdf · Em Drosophila melanogaster existem dois isotipos desta enzima

  

27  

The primary antibodies used were: Guinea-pig anti-dMyc114 (1:100); rabbit anti-Mei-P2668

(1:250); mouse anti-Dap63 (1:25); rabbit anti-CycE (1:50), rabbit anti-CycB (1:100) and

mouse anti-PCNA (1:50; Santa Cruz Biotechnology); mouse anti-CycB (1:100; described

in the Developmental Studies Hybridoma Bank) and rabbit anti-βgal (1:100; Cappel). The

secondary antibodies used were made in Goat (1:200; Molecular Probes). To mark the

nuclei DAPI (1:200) staining was performed.

5.4 IN SITU HYBRIDIZATION

Third larvae instar imaginal discs were dissected in PBS and fixed, first in 4%

formaldehyde in PBS and then in 4% formaldehyde in PBT for 20 minutes each fixation

step at room temperature. Antisense Digoxigenin-labeled RNA probes of dMyc were used

and in situ hybridization was performed as described in Ref. 115.

5.5 TUNEL

To visualize dead cells in the wing imaginal discs TUNEL staining, which labels

fragmented DNA in dying cells, was carried out as described in Ref. 116, using an in situ

cell death detection kit (Roche).

5.6 IMAGE ACQUISITION AND ANALYSIS Fluorescent images were obtained on a Leica TCS SP2 confocal microscope, in situ and

adult wings images were acquired on a Nikon Elipse E600 by ACT 2007® software.

Further image processing was achieved with Adobe Photoshop 7.0® software.

5.6.1 Quantification of cell and tissue growth parameters in adult wings and wing discs

dcr-1RNAi expression: Flies were collected in SH (1 Glycerol: 5 EtOH) overnight and

mounted, at least ten adult wings per genotype, in Fauré medium (H2O, Glycerin, Chloral

Hydrate, Arabic rubber). Size of the engrailed and patched domains was measured using

Image J Software® (NIH, USA). Cell density in these domains and in the neighboring

ones was measured as the number of hairs (each wing cell differentiates a hair) per

defined area. Using the Adobe Photoshop 7.0® software the following areas were used to

measure cell densities: (1) two conserved regions between veins L4 and L5 (engrailed

domain) and veins L3 and L4 (neighboring tissue) in the engrailed-gal4 experiments and

Page 30: Understanding the general role of the microRNAs in ...repositorio.ul.pt/bitstream/10451/1448/1/20902_ulfc080582_tm.pdf · Em Drosophila melanogaster existem dois isotipos desta enzima

  

28  

(2) two conserved regions between veins L3 and L4 (patched domain) and veins L4 and

L5 (neighboring tissue) in the patched-gal4 experiments. Final area and cell density

values were normalized as a percent of the control engrailed-gal4, UAS-GFP or patched-

gal4, UAS-GFP (Fig. 3).

Figure 3 – The several veins of a Drosophila adult wing.

dcr-1 twin/clone size analysis: in hs-FLP/+; FRT82 dcrQ1147X/FR82 arm-lacZ and hs-FLP/+;

FRT82 dcrd102/FR82 arm-lacZ, clone and twin size was measure using Image J Software®

(NIH, USA) and the values were presented in arbitrary units.

dcr-1 Minute(+) clones frequency and size analysis: in hs-FLP/+; FRT82 dcrQ1147X/FR82

Minute(3R)w124 Ubi-GFP and hs-FLP/+; FRT82 /FR82 Minute(3R)w124 Ubi-GFP larvae,

clone and cell frequency was measured. Cells were counted by the nuclei stained by

DAPI. Also clone size was calculated using Image J Software® (NIH, USA). Values were

normalized as a percent of the size of the wild type Minute(+) clones.

dcr-1 clones expressing dMyc or CycE (MARCM clones): in hs-FLP tub-Gal4 UAS-GFP;;

FRT82 arm-lacZ / FRT82 Gal80; hs-FLP tub-Gal4 UAS-GFP; UAS-dMyc/+; FRT82

dcrQ1147X / FRT82 Gal80; and hs-FLP tub-Gal4 UAS-GFP;; FRT82 dcrQ1147X / FRT82 Gal80,

frequency of clones and cells was achieved. Moreover, size of the clones (in number of

cells) in the wing pouch region was measured.

Using Microsoft-Excel Software®, average values and the corresponding standard

deviations were calculated and t-test analysis was carried out.

Page 31: Understanding the general role of the microRNAs in ...repositorio.ul.pt/bitstream/10451/1448/1/20902_ulfc080582_tm.pdf · Em Drosophila melanogaster existem dois isotipos desta enzima

  

29  

VI. RESULTS

6.1 Reduced growth in the absence of Dcr-1

Making use of the UAS-Gal4 system, a RNAi construct of dcr-1 (dcr-1RNAi) was expressed

in specific territories within the developing wing, in order to reduce Dcr-1 activity. To test

whether dcr-1RNAi expression was able to significantly down-regulate miRNA levels in wing

cells, we monitored the activity levels of bantam miRNA, known to be required for wing

growth45. To monitor its activity we used a bantam sensor that expresses GFP under the

control of an ubiquitously active tubulin promoter and has two perfect bantam target sites

in the 3’ UTR45. Expression of dcr-1RNAi in wing cells induced a reduction in bantam activity

(upregulation of the sensor) comparing to wild type (Fig. 1A-C). Nevertheless, the

magnitude of this effect was less than the one observed in clones of cells mutant for a null

allele of dcr-1, the dcr-1Q1147X allele21 (Fig. 1D). This demonstrates that, dcr-1RNAi

expression leads to an intermediate condition where Dcr-1 activity and miRNA levels are

reduced, but not eliminated.

Figure 1 – Dcr-1 activity quantification. (A) Wild type disc labeled to visualize expression of the bantam sensor (white). (B, C) Expression of the bantam sensor (white) in wing discs expressing dcr-1RNAi in the patched (B, red arrowheads) or engrailed (C, red brackets) domains. Note in B and C increased expression of bantam sensor in the ptc and en domain when compared with wild type in A. (D) Wing disc with clones of cells lacking Dcr-1 activity marked by the absence of β-gal (red) and labeled in order to see expression of the bantam sensor (green). Genotype: hs-FLP; bantam-sensor-GFP/+; FRT 82 lacZ /FRT82 dcr-1Q1147X. Clones were induced 72 hours before dissection. Note increased expression of bantam sensor in mutant cells (red arrowheads).

Page 32: Understanding the general role of the microRNAs in ...repositorio.ul.pt/bitstream/10451/1448/1/20902_ulfc080582_tm.pdf · Em Drosophila melanogaster existem dois isotipos desta enzima

  

30  

Expression of dcr-1RNAi caused an autonomous decrease in cell size in the adult wing

(higher cell density in the directly affected domain) (Fig. 2C and D; see also Sup Table1).

Consequently, the tissue size in dcr-1RNAi expression domain is also decreased (Fig. 2A, B

and E; see also Table S1). All these effects were completely rescued by co-expression of

a dcr-1 construct (Fig. 2B, C and E). Decreasing the amount of dcr-1 by 50% (dcr-1Q1147X

in heterozygosis) enhanced the size effects already seen with dcr-1RNAi expression (Fig.

2E).

Figure 2 – Reduced cell growth in the absence of Dcr-1 activity. (A-C) Histograms plotting the size (A, B) and the cell density values (C) of the ptc and en domains expressing the transgenes. Values were normalized as a percent of the control GFP-expressing wings. Error bars indicate standard error. Only adult males were analyzed. (A) In ptc>dcr-1RNAi, a significant decrease in the size of the ptc domain was observed when compared to ptc>GFP wings (p<10-5). (B, C) In en>dcr-1RNAi, a significant decrease in the size of the en domain was observed when compared to en>GFP wings (p<10-7), and a significant increase in cell density was observed (p<10-3). Co-expression of dcr-1 rescued tissue size (p<10-10) and cell size (p<10-2) defects. Tissue size values: ptc>GFP=100±8,4 (n of wings=10); ptc>dcr-1RNAi=75±5,1 (n=12); en>GFP=100±5,7 (n=12); en>dcr-1RNAi=75±5,1 (n=10); en>dcr-1RNAi>dcr-1=110±5,8 (n=12). Cell density values: en>GFP=100±7,6 (n=10); en>dcr-1RNAi=123±9,4 (n=10); en>dcr-1RNAi>dcr-1=95±2,6 (n=10). (D) en>dcr-1RNAi adult wing cells. The red line labels the boundary between anterior cells (do not express dcr-1RNAi) and posterior cells (do express dcr-1RNAi). Note the reduced size of posterior cells. (E) Images of adult wings expressing GFP or dcr-1RNAi in the ptc or en domains (labeled in blue), a wing co-expressing dcr-1 in the en domain and a wing with a dcr-1 mutant background (dcr-1Q1147X) in which en>dcr-1RNAi was expressed.

Page 33: Understanding the general role of the microRNAs in ...repositorio.ul.pt/bitstream/10451/1448/1/20902_ulfc080582_tm.pdf · Em Drosophila melanogaster existem dois isotipos desta enzima

  

31  

To further study the requirement of Dcr-1 in tissue growth, clones of dcr-1Q1147X mutant

cells in the wing primordium were induced and compared in size with their wild type twin

spots (Fig. 3A and B). In dcr-1Q1147X clones analyzed 72 hours after induction, Dcr-1

activity was reduced, monitored by the reduced activity of bantam microRNA (Fig. 1D).

These clones were smaller than their corresponding wild type twin clones ((Fig. 3B), see

also Ref. 117, 118). Cell size defects were also observed in clones of dcr-1 mutant cells (dcr-

1Q1147X, Fig. 3C). With another allele of dcr-1, dcr-1d102, clones of mutant cells gave similar

results in terms of clone size (Fig. 3A). It is noticeable that these mutant clones usually

were broken (Fig. 3D), pointing to a possible engulfment from wild type cells, typical in cell

competition situations. After 96 hours of induction many clones were eliminated from the

wing disc (Fig. 3E), 31,5% of the wild-type twin clones were lacking their corresponding

dcr-1 mutant clones (n of twin clones = 4,1; n of dcr-1 clones = 2,4; n of discs = 10). In

order to check if these mutant clones were dying, a TUNEL staining was carried out. Most

of dcr-1 mutant clones were positive for the staining (Fig. 3F). All of these observations

described above suggested that dcr-1 mutant cells were being eliminated through cell

competition, a process by which slower growing cells are detected and removed by

apoptosis107,84,85,119,120. With the purpose of confirm this hypothesis, we used the Minute

technique to generate clones of mutant cells with a relative growth advantage when

comparing with the wild type cells (mutants for Minute). In a Minute/+ background, dcr-1

mutant clones were recovered at the same frequency as wild type control clones, 96

hours after induction (n clones/wing disc [dcr-1 M(+)] = 3,2, n clones = 32; n clones/wing

disc [M(+)] = 3, n clones = 30) rescuing the cell competition phenotype seen in FRT

clones. Despite that, clones were still smaller that wild type control clones (Fig. 3G and H).

Altogether, these results indicate that activity of Dcr-1 is required for cell and tissue

growth.

6.2 Dcr-1 regulates E2F activity and Dap protein levels

Given the fact that, in Drosophila and Vertebrate stem cells it has been reported that the

lost of dcr-1 causes a delay in G1-S transition50,59,60, we analyzed the cell cycle profile in

mutant cells of the wing disc by staining against some cell cycle markers (Fig. 4 and Fig.

S1). Once more, the Minute technique was used to generate bigger dcr-1Q1147X mutant

clones in order to become possible a greater visualization of the effect. In cells mutant for

dcr-1, as well as cells expressing dcr-1RNAi, expression of Cyclin B (CycB) protein, which

accumulates during the G2 phase of the cell cycle121, was reduced (Fig. 4A and B) when

compared to wild type expression (Fig. S1 D and J).

Page 34: Understanding the general role of the microRNAs in ...repositorio.ul.pt/bitstream/10451/1448/1/20902_ulfc080582_tm.pdf · Em Drosophila melanogaster existem dois isotipos desta enzima

  

32  

 

Figure 3 – Dcr-1 and cell competition. (A, B) Imaginal discs with clones of cells, induced 72 hours before dissection, lacking dcr-1 activity marked by the absence of GFP (white). Note reduced size of the mutant clones (in black) when compared with the control wild type twins (in white). The corresponding graphics plotting the relative sizes (clone areas in arbitrary units) of individual pairs of dcr-1-/- clones (black bars) and dcr-1+/+ twins (grey bars). Two different alleles of dcr-1 were used: dcr-1d102 (A) and dcr-1Q1147X (B). Genotypes: hs-FLP; FRT 82 Ubi-GFP/FRT82 dcr-1Q1147X, and hs-FLP; FRT 82 Ubi-GFP/FRT82 dcr-1d102. (C) Adult wings with clones of cells lacking dcr-1 activity. The mutant tissue in adult wings (genotype: hs-FLP; FRT 82 P(forked+) M(3)95A2/FRT82 dcr-1Q1147X) was marked by the absence of the P(forked+) rescue construct. In the left image, the red bar labels the mutant bristles and blue arrows label the wild type bristles. The red line in the right image labels the boundary between wild type and mutant tissue. Wild type and M(3)95A2/+ adult wings show a similar cell size122. (D-F) Wing discs with clones of cells lacking dcr-1 activity marked by the absence of GFP (white in D and E, green in F), induced 72 hours (D, F) or 96 hours (E) before dissection. Note mutant clones tend to break (red arrowheads in D) and enter apoptosis (labeled by TUNEL staining in red, F) 72 hours after induction and are frequently lost from the epithelium 96 hours after induction (E). (G) Wing discs with clones of cells lacking dcr-1 activity (right panels) or wild type for dcr-1 (left panels) and generated with the Minute technique to give clone cells a growth advantage. Clones were labeled by the absence of GFP expression (white) and induced 96 hours before dissection. The genotypes were: hs-FLP; FRT 82 Ubi-GFP M(3)95A2/FRT82 dcr-1Q1147X; and hs-FLP; FRT 82 Ubi-GFP M(3)95A2/FRT82. (H) Histogram plotting the size of dcr-1 (n clones= 21) and wild type Minute(+) clones (n = 18) induced 96 hours before dissection. Clone size was normalized as a percent of the wild type Minute (+) clone size. Error bars indicate standard error. The difference between both genotypes was statistically significant (p<10-8).

Page 35: Understanding the general role of the microRNAs in ...repositorio.ul.pt/bitstream/10451/1448/1/20902_ulfc080582_tm.pdf · Em Drosophila melanogaster existem dois isotipos desta enzima

  

33  

Cyclin E (CycE) protein, which accumulates during the G1 phase108 was increased (Fig.

4C and D) when compared to the wild type situation (Fig. S1 B and H). Wild type control

clones generated in a Minute heterozygous background did not show any change in CycB

and CycE levels (Fig. S1 H and J). These results point towards a delay in G1-S transition

in the cell cycle when dcr-1 is absent. Consistently, Dap, a inhibitor of the G1-S transition

checkpoint63,64, is increased in cells with reduced Dcr-1 activity (Fig. 4I and J) compared to

wild type (Fig. S1 E). Concerning E2F activity, we checked expression of the dE2F1-

responsive reporter ORC1-GFP106 and also expression of a dE2F target, PCNA. We

observed a decreased in the reporter levels (Fig. 4E and F) as well as in PCNA protein

levels (Fig. 4G and H) when compared to wild type expression (Fig. S1 F, G and I).

Overall, E2F activity is reduced in cells lacking Dcr-1 activity. PCNA protein levels were not

changed in control wild-type clones generated in a Minute heterozygous background (Fig.

S1 I). These results suggest that microRNAs, by increasing E2F activity and limiting Dap

expression, promote cell division.

Some cell cycle regulators, such as Dap, Rbf and CycE, have been stated to not exert a

direct effect on tissue growth in wing discs and adult wings77 (see also Table S1). With the

aim of verifying if this also occurs in a reduced miRNAs activity condition, we analyzed if

increasing CycE or reducing Dap or Rbf proteins cells we could overcome the

consequences of dcr-1 depletion. In all the three situations, cell size was rescued

(decreased cell density) (Fig. 4M) and the size of the wing territory was increased (Fig. 4K

and L), overcoming the effect of decreased Dcr-1 activity. All the results described above

raise the possibility that the influence of cell cycle regulators on tissue and cell growth

could be somehow modulated by the microRNA pathway. Despite the fact that co-

expressing cell cycle regulators like CycE was sufficient to rescue tissue and cell size in

adult wings (Fig. 4K, L and M), the effects of reduced clonal recovery rate and size

caused by the impairment of Dcr-1 activity were not overcome by expression of CycE

(compare Fig. S2 A with Fig. 6B, C and E). These results suggest that the miRNA

machinery have an impact on the activity of a more general growth regulator than those

involved in the cell cycle.

6.3 dMyc levels are regulated by Dcr-1 activity

The proto-oncogene dMyc is known to regulate aspects like cell growth, tissue growth and

G1-S transition and moreover differences in dMyc expression induce cell competition in

Drosophila83,85,107. Taking that into account, and also the fact that reduced dMyc

expression levels leads to phenotypes that remind some aspects of reduced Dcr-1 activity

Page 36: Understanding the general role of the microRNAs in ...repositorio.ul.pt/bitstream/10451/1448/1/20902_ulfc080582_tm.pdf · Em Drosophila melanogaster existem dois isotipos desta enzima

  

34  

Figure 4 – Dcr-1 promotes G1-S transition. (A-J) Wing discs with cells lacking dcr-1 activity (A, D, E, G, I; genotype: hs-FLP; FRT 82 lacZ M(3)95A2/FRT82 dcr-1Q1147X) clones marked by the absence of β-gal (green), or expressing dcr-1RNAi (B, C, F, H. J) in the patched (ptc) domain (red arrowheads), labeled to visualize in pink or white Cyclin B (CycB, A, B), Cyclin E (CycE, C, D), dE2F activity (an E2F1 responsive reporter ORC1-GFP was used, antibody to GFP, E, F), PCNA (G, H) and Dacapo (Dap, I, J) protein expression. Clones were induced 72 h before dissection. (K) Adult wings expressing dcr-1RNAi in the engrailed (en) domain in several genetic backgrounds. (L, M) Histograms plotting the size (L) and cell density (M) of en domain in adult wings expressing dcr-1RNAi in different genetic backgrounds. Tissue size and cell density values were normalized as a percent of the values of control GFP-expressing wings. Error bars indicate standard error. In en>dcr-1RNAi, a significant decrease in the size of the en domain was observed when compared with en>GFP wings (p<10-7 in males and p<10-3 in females). Halving the dose of dap or Rbf, or co-expression of CycE significantly rescued this phenotype (p(dap4)<10-5, p(Rbfsls5)<10-5, p(CycE)<10-8). In en>dcr-1RNAi males, a significant increase in the cell density of the en domain was observed when compared with en>GFP wings (p<10-3). Halving the dose of dap or co-expression of CycE significantly rescued the cell size defects (p(dap4)<10-3, p(CycE)<10-3). Tissue size values (males): en>GFP=100±5,7 (n=12); en>dcr-1RNAi=75±5,1 (n=10); en>dcr-1RNAi; dap4/+= 122±7,3 (n=7) ; en>dcr-1RNAi >CycE=111±4,5 (n=12). Tissue size values (females): en>GFP=100±7,4 (n=12); en>dcr-1RNAi=66±1,7 (n=12) ; Rbfsls5/+ ;en> dcr-1RNAi=80±3,1 (n=12). Cell density values (males): en>GFP=100±7,6 (n=10); en>dcr-1RNAi=123±9,4 (n=10); en> dcr-1RNAi; dap4/+=105±4,5 (n=10); en>dcr-1RNAi>CycE=106±4,5 (n=10).

Page 37: Understanding the general role of the microRNAs in ...repositorio.ul.pt/bitstream/10451/1448/1/20902_ulfc080582_tm.pdf · Em Drosophila melanogaster existem dois isotipos desta enzima

  

35  

that we have seen so far, we checked for dMyc levels in this situation. Both dMyc protein

and mRNA levels were reduced in cells expressing dcr-1RNAi as well as in dcr-1Q1147X

mutant cells when compared to the wild type (compare Fig. 5A-F with S1 J). In order to be

confident that this role of miRNAs regulating dMyc protein levels is specific, we check for

the activity of two other pathways involved in growth control in Drosophila, PI3K and hippo

pathways79,80 and their activity was not affected (Fig. S2). Then, these results reveal a

novel and specific role of the miRNA pathway in regulating dMyc protein levels.

The TRIM-NHL protein TRIM32 has been recently reported to through binding c-Myc

ubiquinate and send it to degradation123. We tested if Mei-P26, the Drosophila TRIM32

homolog, known to regulate cell growth and proliferation in stem cells68, had a similar role

in the wing disc. In Mei-P26 overexpressing cells, dMyc protein levels were strongly

reduced, while dMyc mRNA levels had a milder reduction (Fig. 5G and H). To determine

whether dMyc protein might be degraded by Mei-P26 in a proteasome-dependent

manner, we incubated wing discs overexpressing Mei-P26 with the proteasome inhibitor

MG-132. In drug treated wing discs, dMyc protein degradation was prevented (Fig. 5G’)

and mRNA levels were still reduced (data not shown), while in control solvent treated

discs, overexpression of Mei-P26 still induced a strong reduction in dMyc protein levels

(data not shown).

We next used the Gal4-UAS system to express two distinct RNAi constructs of mei-P26

(mei-P26RNAi-7553 and mei-P26RNAi-106754) to analyze dMyc protein levels in a situation of

reduced Mei-P26 activity. Both constructs were able to reduce protein levels in the wing

disc (Fig. S3 A and B). Co-expressing both mei-P26RNAi did cause a slightly increase in

dMyc protein levels (Fig. 5I), suggesting that endogenous levels of Mei-P26 activity are not

high enough to limit dMyc protein levels, but high levels of Mei-P26, as shown above, might

reason a fall in dMyc levels. A possible explanation is that miRNAs control dMyc protein

levels through the regulation of Mei-P26 activity. Interestingly, mei-P26 is predicted to be a

target for regulation by miRNAs124. In both dcr-1Q1147X mutant cells and dcr-1RNAi expressing

cells, Mei-P26 protein levels were increased but not mei-P26 mRNA levels (Fig. 5J-L). In

order to confirm that, in a reduced Dcr-1 activity situation, the increase in Mei-P26 protein

levels was responsible for the reduction of dMyc expression, we examined dMyc protein

and mRNA levels when both Dcr-1 and Mei-P26 were reduced. In cells expressing mei-

P26RNAi together with dcr-1RNAi, where Mei-P26 was reduced, dMyc protein and mRNA

levels were totally rescued (Fig. 5M, N and Fig. S3 C). Overall, these results show that the

miRNA pathway represses Mei-P26 protein production that consequently regulates dMyc.

Page 38: Understanding the general role of the microRNAs in ...repositorio.ul.pt/bitstream/10451/1448/1/20902_ulfc080582_tm.pdf · Em Drosophila melanogaster existem dois isotipos desta enzima

  

36  

 

Figure 5 – Dcr-1 regulates dMyc by repressing Mei-P26 protein levels. (A-F) Wild type wing discs (E, F) or wing discs with reduced dcr-1 activity (A-D) labeled to visualize dMyc protein (red or white, A, B, D, E) or mRNA (purple, C, F) expression. In A, clones of cells were generated (genotype: hs-FLP; FRT 82 Ubi-GFP M(3)95A2/FRT82 dcr-1Q1147X) and marked by the absence of GFP (green). In B-D, dcr-1RNAi was expressed in the patched (ptc, red arrowheads in B, C) or engrailed (en, red brackets in D) domains. The en domain was also labeled in D by the expression of GFP. (G-I) Wing discs overexpressing mei-P26 (G, G’ H) or expressing two mei-P26RNAi constructs (I) in the engrailed (en, red brackets) domain and labeled to visualize dMyc protein (white) and dMyc mRNA (purple). In G’, wing discs were cultured in the presence of MG132, a proteosome inhibitor. (J-L) Wing discs with reduced dcr-1 activity labeled to visualize Mei-P26 protein (red or white) or mei-P26 mRNA (purple) expression. In J, clones of dcr-1 mutant cells were generated (genotype: hs-FLP; FRT 82 lacZ M(3)95A2/FRT82 dcr-1Q1147X) and marked by the absence of β-Gal (green). In K and L, dcr-1RNAi was expressed in the patched (ptc, red arrowheads) domains. (M, N) Wing discs expressing dcr-1RNAi and mei-P26RNAi-7553 in the patched (ptc, red arrowheads) domain and labeled to visualize dMyc (green or white) and Mei-P26 (red or white) protein expression in M, and dMyc mRNA (purple) expression in N.

Page 39: Understanding the general role of the microRNAs in ...repositorio.ul.pt/bitstream/10451/1448/1/20902_ulfc080582_tm.pdf · Em Drosophila melanogaster existem dois isotipos desta enzima

  

37  

6.4 Contribution of dMyc to growth and E2F activity defects caused by Dcr-1 depletion

Reduction in Dcr-1 activity leads to phenotypes that look like reduced dMyc activity

phenotypes83,85,107. We decided to check the contribution of dMyc to the defects caused by

Dcr-1 depletion on cell and tissue growth and G1-S transition. In order to achieve the

answer, we analyzed tissue and cell size in wing cells with a dcr-1RNAi background and

restored dMyc expression by three different experimental settings. First, the Gal4-UAS

system was used to express high levels of dMyc in the same territory as dcr-1RNAi.

Second, two different promoters (heat-shock and tubulin) were used to drive endogenous

levels of dMyc expression in the disc. Third, mei-P26RNAi was co-expressed with dcr-1RNAi

to decrease Mei-P26 protein and rescue endogenous dMyc levels. In all of the three

different settings, the tissue and cell size defects in adult wings caused by the absence of

Dcr-1 activity were rescued (Fig. 6A).

As have been shown above, dcr-1 mutant clones have fewer cells than wild type control

clones and 96 hours after induction these clones were eliminated from the wing disc (Fig.

3D and E; see also Fig. 6B, C and E). In these clones, expressing dMyc entirely rescued

their recovery rates (Fig. 6D and E), but clone size was not totally recovered (Fig. 6E).

We next looked to cell cycle regulators expression in dMyc-recued dcr-1 mutant clones or

dcr-1RNAi expressing cells with restored dMyc expression by the three distinct means

described above. CycB and CycE levels were restored to wild type levels (compare Fig. 7A

and C, S4 B and D with Fig. 4A-D). The elevated level of Dap protein expression observed

in dcr-1 mutant cells, remained high in dMyc-recued dcr-1 mutant cells or dcr-1RNAi

expressing cells (Fig. 7D and S4 E). Despite of that, E2F activity was totally rescued to wild

type levels (Fig. 7B and S4 A and C). Therefore, some effects of Dcr-1 activity on cell cycle

regulators can be explained by the reduced level of dMyc protein, but not all of these

effects.

6.5 A role of bantam miRNA in reducing Mei-P26 and Dap protein levels

bantam is a miRNA that was identified as a growth promoter and as an inhibitor of

apoptosis in Drosophila45. bantam inhibits apoptosis by negatively regulate the pro-

apoptotic gene hid45, although the target genes that mediate its role in promoting growth

have not been identified so far. Consistently with the growth promoter role of bantam, mei-

P26 (negative regulator of dMyc) and dap (negative regulator of G1-S transition) are

predicted to be bantam targets124 (see also Fig. S5). We proceeded to the analysis of

bantam capacity to target these genes in the wing disc and to restore the growth defects

Page 40: Understanding the general role of the microRNAs in ...repositorio.ul.pt/bitstream/10451/1448/1/20902_ulfc080582_tm.pdf · Em Drosophila melanogaster existem dois isotipos desta enzima

  

38  

Figure 6 – dMyc contributes to the activity of Dcr-1 in regulating cell and tissue growth. (A) Histograms plotting the size and cell density values of the en domain expressing different transgenes. Tissue size and density values were normalized as a percent of the values of control GFP-expressing wings. Error bars indicate standard deviation. Only adult wing males were analyzed. In en>dcr-1RNAi, a significant decrease in size of the en domain was observed when compared with en>GFP wings (p<10-7). Co-expression of dMyc, hs-dMyc, tub-dMyc or mei-P26RNAi significantly rescued this phenotype (p(dMyc)<10-9, p(hs-dMyc)<10-5, p(tub-dMyc)<10-9 and p(mei-P26RNAi-106754)<10-4). In en>Dcr-1RNAi, a significant increase in cell density of the en domain was observed when compared with en>GFP wings (p<10-3). Co-expression of dMyc, hs-dMyc, tub-dMyc or mei-P26RNAi significantly rescued the cell size defects (p(dMyc)<10-5; p(hs-dMyc)<10-6, p(tub-dMyc)<10-3 and p(mei-P26RNAi-106754)<10-4 ). Tissue size values: en>GFP=100±5,7 (n=12); en>dcr-1RNAi=75±5,1 (n=10); en>dcr-1RNAi>dMyc=118±9,5 (n=12) ; en>dcr-1RNAi;hs-dMyc=99±13,3 (n=10); en>dcr-1RNAi;tub-dMyc=108±5,8 (n=10); en>dcr-1RNAi;mei-P26RNAi-106754=86±11,2 (n=10). Cell density values: en>GFP=100±7,6 (n=10); en>dcr-1RNAi=123±9,4 (n=10); en>dcr-1RNAi>dMyc=105±2,98 (n=10); en>dcr-1RNAi;hs-dMyc=89±3,5 (n=10); en>dcr-1RNAi;tub-dMyc=111±17,1 (n=10); en>dcr-1RNAi;mei-P26RNAi-

106754=105±2,0 (n=10). (B-D) Wing discs with clones of wild type cells (B), mutant cells for dcr-1 (C), and mutant cells for dcr-1 and expressing dMyc (D). (genotypes: hs-FLP tub-Gal4 UAS-GFP; FRT82/ FRT82 Gal80; hs-FLP tub-Gal4 UAS-GFP; FRT82 dcrQ1147X / FRT82 Gal80; hs-FLP tub-Gal4 UAS-GFP; UAS-dMyc/+; FRT82 dcrQ1147X / FRT82 Gal80 respectively). Clones were marked by the presence of GFP and induced 96 hours before dissection. Wing disc in C and D were labeled to visualize dMyc protein expression in red. (E) Frequency and size (in number of cells) of clones shown in B-D.

Page 41: Understanding the general role of the microRNAs in ...repositorio.ul.pt/bitstream/10451/1448/1/20902_ulfc080582_tm.pdf · Em Drosophila melanogaster existem dois isotipos desta enzima

  

39  

Figure 7 – dMyc contributes to the activity of Dcr-1 in regulating E2F1 activity. (A-D) Wing discs with clones of cells mutant for dcr-1 and expressing GFP and dMyc (genotype: hs-FLP tub-Gal4 UAS-GFP; UAS-dMyc/+; FRT82 dcrQ1147X / FRT82 Gal80). Clones were induced 72 hours before dissection. Cyclin B (CycB, A), PCNA (B), Cyclin E (CycE, C) and Dacapo (Dap, D) protein expression was visualized in red or white. For exception of Dap, the expression of the cell cycle regulators were rescued to wild type levels.

caused by the Dcr-1 reduction. Expressing bantam in a dcr-1RNAi background was able to

rescue cell and tissue size deficiencies originated by dcr-1RNAi in adult wings (Fig. 8A), to

restore Dap (compare Fig. 8B with Fig. 4J) and also Mei-P26 endogenous levels (data not

shown). As a consequence of Mei-P26 levels decrease, dMyc levels are also rescued to

normal (data not show). Consistently with this, dMyc expression is induce when bantam is

overexpressed114. In bantam overexpression, Mei-P26 protein levels were lower, when

compared to wild type situation (compare Fig. S1 A with Fig. 8C), as well as Dap protein

levels (Fig. 8D). In sum, all these data indicate that both Mei-P26 and Dap are direct

targets of the bantam miRNA contributing to its role in promoting tissue growth. However,

clones of bantam mutant cells did not show increased Dap or Mei-P26 protein levels (Fig.

8E and F), meaning that possibly other miRNAs might play a redundant role in their

regulation. According with this possibility, another miRNAs have been recently reported to

contribute to Dap regulation53.

Page 42: Understanding the general role of the microRNAs in ...repositorio.ul.pt/bitstream/10451/1448/1/20902_ulfc080582_tm.pdf · Em Drosophila melanogaster existem dois isotipos desta enzima

  

40  

Figure 8 – bantam role in the regulation of Mei-P26 and Dacapo levels. (A) Histograms plotting the size and cell density values of the en domain expressing different transgenes. Tissue size and density were normalized as a percent of the values of control GFP-expressing wings. Error bars indicate standard deviation. Only adult wing males were analyzed. In en>dcr-1RNAi, a significant decrease in the size of the en domain was observed when compared with en>GFP wings (p<10-7). Co-expression of hs-bantam significantly rescued this phenotype (p(hs-bantam)<10-11). In en>Dcr-1RNAi, a significant increase in the cell density of the en domain was observed when compared with en>GFP wings (p<10-3). Co-expression of hs-bantam significantly rescued the cell size defects (p(hs-bantam)<10-4). Tissue size values: en>GFP=100±5,7 (n=12); en>dcr-1RNAi=75±5,1 (n=10); en>dcr-1RNAi;hs-bantam=112±4,4 (n=12). Cell density values: en>GFP=100±7,6 (n=10); en>dcr-1RNAi=123±9,4 (n=10); en>dcr-1RNAi;hs-bantam=105±1,8 (n=10). (B-F) Wing discs labeled to visualize Dacapo (Dap, in red or white, B,D,E), Mei-P26 (in red or white, C,F), Gal4 or GFP (in green) protein expression in ptc-Gal4; UAS-dcr-1RNAi, UAS-bantam-GFP (B); ptc-Gal4; UAS-bantam-GFP (C, D) larvae, or in clones of cells mutant for bantam (genotype: hs-FLP; M(3L) ubi-GFP FRT80B/ban∆1 FRT80B) and marked by the absence of GFP (green) (E, F). The patched (ptc) domain is labeled by a red arrowhead.

Page 43: Understanding the general role of the microRNAs in ...repositorio.ul.pt/bitstream/10451/1448/1/20902_ulfc080582_tm.pdf · Em Drosophila melanogaster existem dois isotipos desta enzima

  

41  

VII. DISCUSSION

Since the first identification of a microRNA in 1993 in C. elegans, the field has become

very competitive leading to an explosion in the number of miRNA identified in all

organisms nowadays. Some studies have focus on understanding the role of the miRNA

pathway in development, but most of them do not clarify which are the main effectors in

the processes they are involved in. Along this work, we have analyzed the general role of

the microRNA pathway in Drosophila wing development, a highly proliferative epithelium,

using as tool the RNAi for Dicer-1 enzyme and the mutant alleles available with the mean

of disrupting Dicer-1 activity. Dcr-1 is an important enzyme for microRNAs biogenesis, so

its disruption causes failure in the production of microRNAs.

Depletion of Dc-1 activity originated a reduction in cell and tissue size (see Results Fig. 2),

a delay in G1-S transition in the cell cycle (see Results Fig. 4) and when mutant cells for

dcr-1 were confronted with wild type cells they were lost from the epithelium by the

mechanism of cell competition (see Results Fig. 3). Some of these consequences of

reduction in Dcr-1 activity have been shown previously in other systems. We wanted to

go further in the identification of the molecules that concretize miRNA function. In that

direction, we have concluded that the miRNA bantam, redundantly with other miRNAs not

identified in this work, and the dMyc proto-oncogene contribute to the role of Dcr-1 in

these processes of growth control. Their expression was able to rescue not only the cell

and tissue size defects, but also the defects on cell cycle regulators caused by depletion

of Dcr-1 activity. The cell competition phenotype observed was also rescued when dMyc

was expressed in cells deficient for Dcr-1 activity. It would be extremely interesting to

check if bantam is also able to overcome the cell competition deficiencies of these cells.

Based on the results, we purpose a model (Fig. 1) in which dMyc protein levels are

regulated by a double repression mechanism mediated by the direct activity of miRNAs on

the mei-P26 gene. bantam is a miRNA involved in this regulation, but since in its absence

mei-P26 is not upregulated, other miRNAs might be acting together and redundantly with

bantam. Consistently with our model, in mouse, the Mei-P26 homolog TRIM32 has been

reported to bind, ubiquitinate and degrade cMyc123. In fact, through the use of a

proteosome inhibitor, we confirm that Mei-P26 sends dMyc to degradation, since dMyc is

no longer downregulated (see Results Fig. 5). So it seems that the mechanism by which

Mei-P26 is able to repress Myc is conserved among these species, and we can speculate

that this might be a general role of the TRIM-NHL proteins.

 

Page 44: Understanding the general role of the microRNAs in ...repositorio.ul.pt/bitstream/10451/1448/1/20902_ulfc080582_tm.pdf · Em Drosophila melanogaster existem dois isotipos desta enzima

  

42  

Figure 1 – Purposed model for the mechanism by which Dcr-1 acts in the wing imaginal disc development in Drosophila. dMyc protein levels are regulated by a double repression mechanism mediated by the direct activity of miRNAs on the mei-P26 gene. Through bantam and other microRNAs, Dcr-1 represses both Mei-P26 and Dacapo (Dap). This has consequences on cell cycle regulators, as dMyc represses Dap and promotes E2F activity, thus affecting G1-S transition. We can speculate about two possible feedback loops that miRNAs and Myc proto-oncogene use with the purpose of regulate their own activity or expression levels, respectively (dashed lines). The first possibility involves the fact that, by targeting mei-P26, miRNAs could be targeting a positive modulator of the miRISC complex and then modulating it own activity (purple dashed line). The second possibility is that Myc is restricting its own expression levels through transcriptional repression of miRNAs (red dashed line).   Evidence was presented that Dcr-1 keeps the levels of Mei-P26 under a protein

concentration threshold, by repressing it, with the purpose of alleviate repression of dMyc

protein. In what concerns cell cycle regulation, we observed that, in a Dcr-1 depletion

condition, E2F activity is compromised and Dacapo levels are upregulated (see Results

Fig. 4). However, dMyc expression rescued some of the defects in cell cycle regulators,

namely E2F activity and CycE, but not Dap levels (see Results Fig. 7). Therefore, some

defects of the reduction of Dcr-1 activity on cell cycle regulators can be explained by the

reduced level of dMyc protein levels, but not all of these effects. Moreover, the fact that in

these cells, Dap protein levels still high but CycE protein levels are rescued to normal

suggests somehow that Dap might not play a major role in regulation of CycE protein

levels in a Dcr-1 activity depletion situation and restored dMyc expression. Alternatively,

the influence of cell cycle regulators on tissue growth might be normally limiting, and it

could be that they are revealed under conditions where the requirement for their activity is

sensitized. It is interesting to note, in this context, that expression of cell cycle regulators

was recently shown to promote tissue growth in early wing primordia and to rescue the

growth defects caused by a reduction in Notch signaling125. Additionally, the observation

that Dap levels were not rescue in dMyc-recued dcr-1 deficiency cells, can also be

explained by the fact that Dap is a direct target of bantam in this system (as shown by its

downregulation in cells overexpressing bantam (see Results Fig. 8)) and other miRNAs

are likely to be regulators of Dap as revealed by the fact that this protein has miRNAs

Page 45: Understanding the general role of the microRNAs in ...repositorio.ul.pt/bitstream/10451/1448/1/20902_ulfc080582_tm.pdf · Em Drosophila melanogaster existem dois isotipos desta enzima

  

43  

binding sites in its 3´UTR53. All these results define a frame by which the miRNA pathway

is required, in a growing epithelium, to maintain sufficient levels of dMyc expression in

order to achieve proper tissue growth.

Deregulated expression of the proto-oncogene Myc frequently occurs in human

malignancies81. Likewise, some miRNAs are known to have abnormal expression in human

disease126. So, it is evident that tight regulation of their expression or activity levels, of both

Myc and microRNAs, is crucial for human health. Based on the results presented in this

work and in other published observations it is possible to speculate about two different

feedback loops that the miRNA machinery and the Myc proto-oncogene are using with the

purpose of regulate their own activity or expression levels, respectively (Fig. 1). First,

Drosophila Mei-P26 and its murine and C. elegans orthologs, TRIM32 and nhl-2, have

been shown to bind to components of the miRISC complex and regulate the activity of the

miRNA machinery68,123,127. These results, together with our observations on the negative

regulation of mei-P26 by microRNAs, suggest that a feedback loop might be used by the

miRNA machinery in order to regulate tightly its own activity levels. They might exert this

action by targeting mei-P26, a positive modulator of the miRISC complex. Consistent with

this notion, we have observed that overexpressed Mei-P26 positively modulates the activity

of the bantam miRNA (data not show). Alternatively, the vertebrate Myc has been shown

to promote widespread repression of miRNA expression35 and we also presented evidence

that the miRNA machinery regulates dMyc protein levels by targeting its repressor mei-

P26. In this way, a negative feedback loop might be used by Myc to restrict its own

expression levels through transcriptional repression of those miRNAs that target mei-P26

(Fig.1). In this sense, when dMyc levels are too low, microRNAs are not repressed any

longer and can repress Mei-P26, thus alleviating repression of dMyc.

The gene regulatory relationships between miRNAs, master growth regulators and genes

controlling cell cycle progression, described in this work also reveal some underlying

mechanisms used to confer robustness to the process of tissue growth in order to work

properly during development. The role of Mei-P26 is very illustrative of this robustness

feature. Although endogenous levels of Mei-P26 protein are not limiting in wild type

situations, Mei-P26 is being used to reduce dMyc protein levels in a situation of reduced

miRNA activity, thus buffering the effects of high levels of dMyc protein. This shows that a

hidden mechanism can be used in a stress situation, in this case of Dcr-1 activity depletion,

buffering the system, in order to achieve proper growth. Single miRNAs, like bantam, can

also be playing roles on the road to confer robustness to the regulatory network involved in

tissue growth or even in other processes in development.

In Drosophila there is at least three known proteins members of the TRIM-NHL proteins

family – Mei-P26, Brat and Dappled128. Brat (Brain Tumour) was shown to be a

Page 46: Understanding the general role of the microRNAs in ...repositorio.ul.pt/bitstream/10451/1448/1/20902_ulfc080582_tm.pdf · Em Drosophila melanogaster existem dois isotipos desta enzima

  

44  

transcriptional repressor129. Besides its role in controlling growth in central neural system

development, specifically in neuroblasts130,131, Brat proteins has also been documented to

negatively regulate cell growth and ribosomal synthesis in the wing imaginal disc of the fruit

fly132. Taking all that into account and also the fact that the protein Brat is an homolog of

Mei-P26128, it would be very exciting to investigate in the near future if Brat is somehow

connected to the mechanism described in this thesis. Intriguingly, Brat protein does not

have the RING domain, characteristic of the members of this family and that confers the

ubiquitin ligase activity128, that sends Myc to degradation as shown for Mei-P26 in this

system. Curiously, in differentiating neuroblasts Brat is thought to repress Myc, through an

unknown mechanism, in order to cells stop growing, exit the cell cycle and avoid self-

renewal, thus allowing cells to enter differentiation131.

Interestingly, bantam was reported to be a target of the Hippo signaling pathway58,133.

bantam expression is increased by Yki overexpression, and Yki-induced overproliferation is

suppressed by loss of bantam. In Yki mutants bantam overexpression is able to rescue the

growth defects80. The Hpo pathway regulates the transcription of bantam independently of

CycE and diap1133. In this sense, it would be exciting to see if the Hpo pathway targeting

microRNAs is a more general feature, and also if it could be somehow related to the

mechanism provided in this work.

An additional interesting point to explore focus on the other proteins involved in the

microRNAs biogenesis, like Pasha (Drosha) and Argonaute1 proteins. It would be

appealing to check if they also cause the same defects consequents of the Dcr-1 activity

depletion. Knowing that Mei-P26, and also its orthologs in mouse and C. elegans, were

documented to bind Ago168,123,127 and modulating somehow the microRNA pathway, it

would be interesting to clarify if that interaction occurs in the wing disc and if it could enable

the utilization of Mei-P26 by the miRNA machinery to regulate its own activity levels, as

speculated above.

Another crucial point would be the identification of others microRNAs that might be working

along with bantam in the mechanism described here, and also to clarify if this mechanism

is a general system utilized by all organisms, and in all type of tissues, across the animal

kingdom. Since the number of microRNAs connected to human health is growing in an

exponential way, and given the fact that Myc has been already associated with a batch of

malignancies, understanding the entire process can be useful in the comprehension, not

only of growth control as a whole, but also of several diseases.

Page 47: Understanding the general role of the microRNAs in ...repositorio.ul.pt/bitstream/10451/1448/1/20902_ulfc080582_tm.pdf · Em Drosophila melanogaster existem dois isotipos desta enzima

  

45  

VIII. CONCLUDING REMARKS

On the whole, the work presented in this thesis has clarified the role of the microRNAs

machinery in wing disc development. First of all, through the characterization of the

microRNAs depletion phenotype, it was shown that miRNAs have an important role in

growth control and cell cycle regulation during wing disc development. Furthermore this

work has clarified the effectors that mediate that role of the microRNAs in growth in the

wing imaginal disc. In addiction this work pointed to bantam microRNA, and probably

other miRNAs, targeting such described effectors.

The results propose the existence of a feedback loop involving a double repression

mechanism between the miRNA machinery and the Myc proto-oncogene in order to

regulate their own activity or expression levels.

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

Page 48: Understanding the general role of the microRNAs in ...repositorio.ul.pt/bitstream/10451/1448/1/20902_ulfc080582_tm.pdf · Em Drosophila melanogaster existem dois isotipos desta enzima

  

46  

IX. REFERENCES

1. Malone, C.D. & Hannon, G.J. Small RNAs as Guardians of the Genome. Cell 136, 656-668 (2009). 2. Carthew, R.W. & Sontheimer, E.J. Origins and Mechanisms of miRNAs and siRNAs. Cell 136, 642-

655 (2009). 3. Tomari, Y. & Zamore, P.D. Perspective: machines for RNAi. Genes & Development 19, 517-529

(2005). 4. He, L. & Hannon, G.J. Micrornas: Small RNAs with a big role in gene regulation. Nature Reviews

Genetics 5, 522-531 (2004). 5. Bartel, D.P. MicroRNAs: Genomics, biogenesis, mechanism, and function. Cell 116, 281-297 (2004). 6. Filipowicz, W., Bhattacharyya, S.N. & Sonenberg, N. Mechanisms of post-transcriptional regulation

by microRNAs: are the answers in sight? Nature Reviews Genetics 9, 102-114 (2008). 7. Vasudevan, S., Tong, Y. & Steitz, J.A. Switching from repression to activation: microRNAs can up-

regulate translation. Science 318, 1931-4 (2007). 8. Lee, R.C., Feinbaum, R.L. & Ambros, V. The C. elegans heterochronic gene lin-4 encodes small

RNAs with antisense complementarity to lin-14. Cell 75, 843-854 (1993). 9. Lee, Y. et al. MicroRNA genes are transcribed by RNA polymerase II. Embo Journal 23, 4051-4060

(2004). 10. Borchert, G.M., Lanier, W. & Davidson, B.L. RNA polymerase III transcribes human microRNAs.

Nature Structural & Molecular Biology 13, 1097-1101 (2006). 11. Kim, V.N., Han, J. & Siomi, M.C. Biogenesis of small RNAs in animals. Nature Reviews Molecular

Cell Biology 10, 126-139 (2009). 12. Lee, Y., Jeon, K., Lee, J.T., Kim, S. & Kim, V.N. MicroRNA maturation: stepwise processing and

subcellular localization. Embo Journal 21, 4663-4670 (2002). 13. Denli, A.M., Tops, B.B.J., Plasterk, R.H.A., Ketting, R.F. & Hannon, G.J. Processing of primary

microRNAs by the Microprocessor complex. Nature 432, 231-235 (2004). 14. Lee, Y. et al. The nuclear RNase III Drosha initiates microRNA processing. Nature 425, 415-419

(2003). 15. Landthaler, M., Yalcin, A. & Tuschl, T. The human DiGeorge syndrome critical region gene 8 and its

D-melanogaster homolog are required for miRNA biogenesis. Current Biology 14, 2162-2167 (2004). 16. Han, J.J. et al. Molecular basis for the recognition of primary microRNAs by the Drosha-DGCR8

complex. Cell 125, 887-901 (2006). 17. Han, J. et al. Posttranscriptional Crossregulation between Drosha and DGCR8. Cell 136, 75-84

(2009). 18. Bohnsack, M.T., Czaplinski, K. & Gorlich, D. Exportin 5 is a RanGTP-dependent dsRNA-binding

protein that mediates nuclear export of pre-miRNAs. Rna-a Publication of the Rna Society 10, 185-191 (2004).

19. Lund, E., Guttinger, S., Calado, A., Dahlberg, J.E. & Kutay, U. Nuclear export of microRNA precursors. Science 303, 95-98 (2004).

20. Hutvagner, G. et al. A cellular function for the RNA-interference enzyme Dicer in the maturation of the let-7 small temporal RNA. Science 293, 834-838 (2001).

21. Lee, Y.S. et al. Distinct roles for Drosophila Dicer-1 and Dicer-2 in the siRNA/miRNA silencing pathways. Cell 117, 69-81 (2004).

22. Bernstein, E., Caudy, A.A., Hammond, S.M. & Hannon, G.J. Role for a bidentate ribonuclease in the initiation step of RNA interference. Nature 409, 363-366 (2001).

23. Lingel, A., Simon, B., Izaurralde, E. & Sattler, M. Structure and nucleic-acid binding of the Drosophila Argonaute 2 PAZ domain. Nature 426, 465-469 (2003).

24. Saito, K., Ishizuka, A., Siomi, H. & Siomi, M.C. Processing of pre-microRNAs by the Dicer-1-Loquacious complex in Drosophila cells. Plos Biology 3, 1202-1212 (2005).

25. Winter, J., Jung, S., Keller, S., Gregory, R.I. & Diederichs, S. Many roads to maturity: microRNA biogenesis pathways and their regulation. Nat Cell Biol 11, 228-34 (2009).

26. Cerutti, L., Mian, N. & Bateman, A. Domains in gene silencing and cell differentiation proteins: the novel PAZ domain and redefinition of the Piwi domain. Trends in Biochemical Sciences 25, 481-482 (2000).

27. Peters, L. & Meister, G. Argonaute proteins: Mediators of RNA silencing. Molecular Cell 26, 611-623 (2007).

28. Tomari, Y., Matranga, C., Haley, B., Martinez, N. & Zamore, P.D. A protein sensor for siRNA asymmetry. Science 306, 1377-1380 (2004).

29. Khvorova, A., Reynolds, A. & Jayasena, S.D. Functional siRNAs and rniRNAs exhibit strand bias. Cell 115, 209-216 (2003).

30. Diederichs, S. & Haber, D.A. Dual role for argonautes in MicroRNA processing and Posttranscriptional regulation of MicroRNA expression. Cell 131, 1097-1108 (2007).

31. Forstemann, K., Horwich, M.D., Wee, L., Tomari, Y. & Zamore, P.D. Drosophila microRNAs are sorted into functionally distinct argonaute complexes after production by Dicer-1. Cell 130, 287-297 (2007).

Page 49: Understanding the general role of the microRNAs in ...repositorio.ul.pt/bitstream/10451/1448/1/20902_ulfc080582_tm.pdf · Em Drosophila melanogaster existem dois isotipos desta enzima

  

47  

32. Landgraf, P. et al. A mammalian microRNA expression atlas based on small RNA library sequencing. Cell 129, 1401-1414 (2007).

33. Jiang, Q.H. et al. miR2Disease: a manually curated database for microRNA deregulation in human disease. Nucleic Acids Research 37, D98-D104 (2009).

34. He, L., He, X.Y., Lowe, S.W. & Hannon, G.J. microRNAs join the p53 network - another piece in the tumour-suppression puzzle. Nature Reviews Cancer 7, 819-822 (2007).

35. Chang, T.C. et al. Widespread microRNA repression by Myc contributes to tumorigenesis. Nature Genetics 40, 43-50 (2008).

36. Bueno, M.J. et al. Genetic and epigenetic silencing of microRNA-203 enhances ABL1 and BCR-ABL1 oncogene expression. Cancer Cell 13, 496-506 (2008).

37. Michlewski, G., Guil, S., Semple, C.A. & Caceres, J.F. Posttranscriptional Regulation of miRNAs Harboring Conserved Terminal Loops. Molecular Cell 32, 383-393 (2008).

38. Newman, M.A., Thomson, J.M. & Hammond, S.M. Lin-28 interaction with the Let-7 precursor loop mediates regulated microRNA processing. Rna-a Publication of the Rna Society 14, 1539-1549 (2008).

39. Kennedy, S., Wang, D. & Ruvkun, G. A conserved siRNA-degrading RNase negatively regulates RNA interference in C-elegans. Nature 427, 645-649 (2004).

40. Kawahara, Y. et al. Frequency and fate of microRNA editing in human brain. Nucleic Acids Research 36, 5270-5280 (2008).

41. Forman, J.J., Legesse-Miller, A. & Coller, H.A. A search for conserved sequences in coding regions reveals that the let-7 microRNA targets Dicer within its coding sequence. Proceedings of the National Academy of Sciences of the United States of America 105, 14879-14884 (2008).

42. Li, X. & Carthew, R.W. A microRNA mediates EGF receptor signaling and promotes photoreceptor differentiation in the Drosophila eye. Cell 123, 1267-1277 (2005).

43. Behura, S.K. Insect microRNAs: Structure, function and evolution. Insect Biochemistry and Molecular Biology 37, 3-9 (2007).

44. Reinhart, B.J. et al. The 21-nucleotide let-7 RNA regulates developmental timing in Caenorhabditis elegans. Nature 403, 901-906 (2000).

45. Brennecke, J., Hipfner, D.R., Stark, A., Russell, R.B. & Cohen, S.M. bantam encodes a developmentally regulated microRNA that controls cell proliferation and regulates the proapoptotic gene hid in Drosophila. Cell 113, 25-36 (2003).

46. Lim, L.P. et al. Microarray analysis shows that some microRNAs downregulate large numbers of target mRNAs. Nature 433, 769-773 (2005).

47. Yekta, S., Shih, I.H. & Bartel, D.P. MicroRNA-directed cleavage of HOXB8 mRNA. Science 304, 594-596 (2004).

48. Aleman, L.M., Doench, J. & Sharp, P.A. Comparison of siRNA-induced off-target RNA and protein effects. Rna-a Publication of the Rna Society 13, 385-395 (2007).

49. Cloonan, N. et al. The miR-17-5p microRNA is a key regulator of the G1/S phase cell cycle transition. Genome Biology 9 (2008).

50. Hatfield, S.D. et al. Stem cell division is regulated by the microRNA pathway. Nature 435, 974-978 (2005).

51. O'Donnell, K.A., Wentzel, E.A., Zeller, K.I., Dang, C.V. & Mendell, J.T. c-Myc-regulated microRNAs modulate E2F1 expression. Nature 435, 839-43 (2005).

52. Shcherbata, H.R. et al. The MicroRNA pathway plays a regulatory role in stem cell division. Cell Cycle 5, 172-175 (2006).

53. Yu, J.Y. et al. Dicer-1-dependent Dacapo suppression acts downstream of Insulin receptor in regulating cell division of Drosophila germline stem cells. Development 136, 1497-507 (2009).

54. Brosh, R. et al. p53-Repressed miRNAs are involved with E2F in a feed-forward loop promoting proliferation. Mol Syst Biol 4, 229 (2008).

55. He, L. et al. A microRNA polycistron as a potential human oncogene. Nature 435, 828-833 (2005). 56. Aguda, B.D., Kim, Y., Piper-Hunter, M.G., Friedman, A. & Marsh, C.B. MicroRNA regulation of a

cancer network: consequences of the feedback loops involving miR-17-92, E2F, and Myc. Proc Natl Acad Sci U S A 105, 19678-83 (2008).

57. Hipfner, D.R., Weigmann, K. & Cohen, S.M. The bantam gene regulates Drosophila growth. Genetics 161, 1527-1537 (2002).

58. Thompson, B.J. & Cohen, S.M. The hippo pathway regulates the bantam microRNA to control cell proliferation and apoptosis in Drosophila. Cell 126, 767-774 (2006).

59. Jin, Z.G. & Xie, T. Dcr-1 maintains Drosophila ovarian stem cells. Current Biology 17, 539-544 (2007).

60. Wang, Y.M., Medvid, R., Melton, C., Jaenisch, R. & Blelloch, R. DGCR8 is essential for microRNA biogenesis and silencing of embryonic stem cell self-renewal. Nature Genetics 39, 380-385 (2007).

61. deNooij, J.C., Letendre, M.A. & Hariharan, I.K. A cyclin-dependent kinase inhibitor, dacapo, is necessary for timely exit from the cell cycle during Drosophila embryogenesis. Cell 87, 1237-1247 (1996).

62. Lane, M.E. et al. Dacapo, a cyclin-dependent kinase inhibitor, stops cell proliferation during Drosophila development. Cell 87, 1225-1235 (1996).

Page 50: Understanding the general role of the microRNAs in ...repositorio.ul.pt/bitstream/10451/1448/1/20902_ulfc080582_tm.pdf · Em Drosophila melanogaster existem dois isotipos desta enzima

  

48  

63. de Nooij, J.C., Letendre, M.A. & Hariharan, I.K. A cyclin-dependent kinase inhibitor, Dacapo, is necessary for timely exit from the cell cycle during Drosophila embryogenesis. Cell 87, 1237-47 (1996).

64. Lane, M.E. et al. Dacapo, a cyclin-dependent kinase inhibitor, stops cell proliferation during Drosophila development. Cell 87, 1225-35 (1996).

65. Stark, A., Brennecke, J., Russell, R.B. & Cohen, S.M. Identification of Drosophila MicroRNA targets. Plos Biology 1, 397-409 (2003).

66. van den Heuvel, S. & Dyson, N.J. Conserved functions of the pRB and E2F families. Nat Rev Mol Cell Biol 9, 713-24 (2008).

67. Page, S.L., McKim, K.S., Deneen, B., Van Hook, T.L. & Hawley, R.S. Genetic studies of mei-P26 reveal a link between the processes that control germ cell proliferation in both sexes and those that control meiotic exchange in Drosophila. Genetics 155, 1757-1772 (2000).

68. Neumuller, R.A. et al. Mei-P26 regulates microRNAs and cell growth in the Drosophila ovarian stem cell lineage. Nature 454, 241-U72 (2008).

69. Harfe, B.D., McManus, M.T., Mansfield, J.H., Hornstein, E. & Tabin, C.J. The RNaseIII enzyme Dicer is required for morphogenesis but not patterning of the vertebrate limb. Proceedings of the National Academy of Sciences of the United States of America 102, 10898-10903 (2005).

70. Garcia-Bellido, A. & Merriam, J.R. Parameters of the wing imaginal disc development of Drosophila melanogaster. Developmental Biology 24, 61-87 (1971).

71. Weihe U, Milán M & SM, C. in Comprehensive Insect Physiology, Biochemistry, Pharmacology and Molecular Biology (eds. Gilbert L, Gill S & K, I.) (Developmental Biology Programme - EMBL, Heidelberg (Germany), 2004).

72. Morata, G. How Drosophila appendages develop. Nature Reviews Molecular Cell Biology 2, 89-97 (2001).

73. Cohen, B., Simcox, A.A. & Cohen, S.M. Allocation of the thoracic imaginal primordia in the Drosophila embryo. Development 117, 597-608 (1993).

74. Cohen, S.M. Imaginal disc development. ed. Martinez-Arias, A. & Bate, M. in The develepment of Drosophila melanogaster, Vol.II, CHL Press, New York (1993).

75. Su, T.T. & O'Farrell, P.H. Size control: cell proliferation does not equal growth. Curr Biol 8, R687-9 (1998).

76. Johnston, L.A. & Gallant, P. Control of growth and organ size in Drosophila. Bioessays 24, 54-64 (2002).

77. Neufeld, T.P., de la Cruz, A.F.A., Johnston, L.A. & Edgar, B.A. Coordination of growth and cell division in the Drosophila wing. Cell 93, 1183-1193 (1998).

78. Milan, M., Campuzano, S. & GarciaBellido, A. Developmental parameters of cell death in the wing disc of Drosophila. Proceedings of the National Academy of Sciences of the United States of America 94, 5691-5696 (1997).

79. Neufeld, T.P. Body building: regulation of shape and size by PI3K/TOR signaling during development. Mech Dev 120, 1283-96 (2003).

80. Pan, D. Hippo signaling in organ size control. Genes Dev 21, 886-97 (2007). 81. Meyer, N. & Penn, L.Z. Reflecting on 25 years with MYC. Nat Rev Cancer 8, 976-90 (2008). 82. de la Cova, C. & Johnston, L.A. Myc in model organisms: A view from the flyroom. Seminars in

Cancer Biology 16, 303-312 (2006). 83. Johnston, L.A., Prober, D.A., Edgar, B.A., Eisenman, R.N. & Gallant, P. Drosophila myc regulates

cellular growth during development. Cell 98, 779-790 (1999). 84. Morata, G. & Ripoll, P. Minutes: mutants of drosophila autonomously affecting cell division rate.

Developmental Biology 42, 211-221 (1975). 85. de la Cova, C., Abril, M., Bellosta, P., Gallant, P. & Johnston, L.A. Drosophila Myc regulates organ

size by inducing cell competition. Cell 117, 107-116 (2004). 86. Turing, A. The chemical basis of morphogenesis. Philos Trans R Soc Lond 237, 37-72 (1952). 87. Wolpert, L. Positional information and the spatial pattern of cellular differentiation. Journal of

Theoretical Biology 25 (1969). 88. McNeill, H. Sticking together and sorting things out: Adhesion as a force in development. Nature

Reviews Genetics 1, 100-108 (2000). 89. Zecca, M., Basler, K. & Struhl, G. SEQUENTIAL ORGANIZING ACTIVITIES OF ENGRAILED,

HEDGEHOG AND DECAPENTAPLEGIC IN THE DROSOPHILA WING. Development 121, 2265-2278 (1995).

90. Klein, T. Wing disc development in the fly: the early stages. Current Opinion in Genetics & Development 11, 470-475 (2001).

91. Basler, K. & Struhl, G. COMPARTMENT BOUNDARIES AND THE CONTROL OF DROSOPHILA LIMB PATTERN BY HEDGEHOG PROTEIN. Nature 368, 208-214 (1994).

92. Wang, S.H., Simcox, A. & Campbell, G. Dual role for Drosophila epidermal growth factor receptor signaling in early wing disc development. Genes & Development 14, 2271-2276 (2000).

93. Tabata, T., Schwartz, C., Gustavson, E., Ali, Z. & Kornberg, T.B. CREATING A DROSOPHILA WING DE-NOVO, THE ROLE OF ENGRAILED, AND THE COMPARTMENT BORDER HYPOTHESIS. Development 121, 3359-3369 (1995).

Page 51: Understanding the general role of the microRNAs in ...repositorio.ul.pt/bitstream/10451/1448/1/20902_ulfc080582_tm.pdf · Em Drosophila melanogaster existem dois isotipos desta enzima

  

49  

94. Lecuit, T. et al. Two distinct mechanisms for long-range patterning by decapentaplegic in the Drosophila wing. Nature 381, 387-393 (1996).

95. Nellen, D., Burke, R., Struhl, G. & Basler, K. Direct and long-range action of a DPP morphogen gradient. Cell 85, 357-368 (1996).

96. Rogulja, D. & Irvine, K.D. Regulation of cell proliferation by a morphogen gradient. Cell 123, 449-461 (2005).

97. Martin, F.A., Perez-Garijo, A., Moreno, E. & Morata, G. The brinker gradient controls wing growth in Drosophila. Development 131, 4921-4930 (2004).

98. Diazbenjumea, F.J. & Cohen, S.M. INTERACTION BETWEEN DORSAL AND VENTRAL CELLS IN THE IMAGINAL DISC DIRECTS WING DEVELOPMENT IN DROSOPHILA. Cell 75, 741-752 (1993).

99. Milan, M. & Cohen, S.M. A re-evaluation of the contributions of Apterous and Notch to the dorsoventral lineage restriction boundary in the Drosophila wing. Development 130, 553-562 (2003).

100. deCelis, J.F. & Bray, S. Feed-back mechanisms affecting Notch activation at the dorsoventral boundary in the Drosophila wing. Development 124, 3241-3251 (1997).

101. Gomez-Skarmeta, J., Campuzano, S. & Modolell, J. Half a century of neural prepatterning: The story of a few bristles and many genes. Nature Reviews Neuroscience 4, 587-598 (2003).

102. Zecca, M., Basler, K. & Struhl, G. Direct and long-range action of a wingless morphogen gradient. Cell 87, 833-844 (1996).

103. Delanoue, R. et al. The Drosophila wing differentiation factor Vestigial-Scalloped is required for cell proliferation and cell survival at the dorso-ventral boundary of the wing imaginal disc. Cell Death and Differentiation 11, 110-122 (2004).

104. Liu, X.F., Grammont, M. & Irvine, K.D. Roles for scalloped and vestigial in regulating cell affinity and interactions between the wing blade and the wing hinge. Developmental Biology 228, 287-303 (2000).

105. Standen, D.B.R.a.G.N. The elements of Drosophila biology and genetics. (Oxford Science Publishing, Oxford, 2003).

106. Asano, M. & Wharton, R.P. E2F mediates developmental and cell cycle regulation of ORC1 in Drosophila. EMBO J 18, 2435-48 (1999).

107. Moreno, E. & Basler, K. dMyc transforms cells into super-competitors. Cell 117, 117-129 (2004). 108. Knoblich, J.A. et al. Cyclin E controls S phase progression and its down-regulation during Drosophila

embryogenesis is required for the arrest of cell proliferation. Cell 77, 107-20 (1994). 109. Duffy, J.B. GAL4 system in Drosophila: a fly geneticist's Swiss army knife. Genesis 34, 1-15 (2002). 110. Brand, A.H. & Perrimon, N. TARGETED GENE-EXPRESSION AS A MEANS OF ALTERING CELL

FATES AND GENERATING DOMINANT PHENOTYPES. Development 118, 401-415 (1993). 111. Xu, T. & Rubin, G.M. ANALYSIS OF GENETIC MOSAICS IN DEVELOPING AND ADULT

DROSOPHILA TISSUES. Development 117, 1223-1237 (1993). 112. Blair, S.S. Genetic mosaic techniques for studying Drosophila development. Development 130, 5065-

72 (2003). 113. Lee, T.M. & Luo, L.Q. Mosaic analysis with a repressible cell marker (MARCM) for Drosophila neural

development. Trends in Neurosciences 24, 251-254 (2001). 114. Herranz, H., Perez, L., Martin, F.A. & Milan, M. A Wingless and Notch double-repression mechanism

regulates G1-S transition in the Drosophila wing. Embo Journal 27, 1633-1645 (2008). 115. Milan, M., Campuzano, S. & GarciaBellido, A. Cell cycling and patterned cell proliferation in the

Drosophila wing during metamorphosis. Proceedings of the National Academy of Sciences of the United States of America 93, 11687-11692 (1996).

116. Wang, S.L., Hawkins, C.J., Yoo, S.J., Muller, H.A.J. & Hay, B.A. The Drosophila caspase inhibitor DIAP1 is essential for cell survival and is negatively regulated by HID. Cell 98, 453-463 (1999).

117. Friggi-Grelin, F., Lavenant-Staccini, L. & Therond, P. Control of antagonistic components of the hedgehog signaling pathway by microRNAs in Drosophila. Genetics 179, 429-39 (2008).

118. Martin, R. et al. A Drosophila pasha mutant distinguishes the canonical microRNA and mirtron pathways. Mol Cell Biol 29, 861-70 (2009).

119. Li, W. & Baker, N.E. Engulfment is required for cell competition. Cell 129, 1215-25 (2007). 120. Moreno, E., Basler, K. & Morata, G. Cells compete for Decapentaplegic survival factor to prevent

apoptosis in Drosophila wing development. Nature 416, 755-759 (2002). 121. Whitfield, W.G., Gonzalez, C., Maldonado-Codina, G. & Glover, D.M. The A- and B-type cyclins of

Drosophila are accumulated and destroyed in temporally distinct events that define separable phases of the G2-M transition. EMBO J 9, 2563-72 (1990).

122. Morata, G. & Ripoll, P. Minutes: mutants of drosophila autonomously affecting cell division rate. Dev. Biol. 42, 211-221 (1975).

123. Schwamborn, J.C., Berezikov, E. & Knoblich, J.A. The TRIM-NHL Protein TRIM32 Activates MicroRNAs and Prevents Self-Renewal in Mouse Neural Progenitors. Cell 136, 913-925 (2009).

124. Stark, A., Brennecke, J., Bushati, N., Russell, R.B. & Cohen, S.M. Animal microRNAs confer robustness to gene expression and have a significant impact on 3 ' UTR evolution. Cell 123, 1133-1146 (2005).

125. Rafel, N. & Milan, M. Notch signalling coordinates tissue growth and wing fate specification in Drosophila. Development 135, 3995-4001 (2008).

126. McManus, M.T. MicroRNAs and cancer. Semin Cancer Biol 13, 253-8 (2003).

Page 52: Understanding the general role of the microRNAs in ...repositorio.ul.pt/bitstream/10451/1448/1/20902_ulfc080582_tm.pdf · Em Drosophila melanogaster existem dois isotipos desta enzima

  

50  

127. Hammell, C.M., Lubin, I., Boag, P.R., Blackwell, T.K. & Ambros, V. nhl-2 Modulates MicroRNA Activity in Caenorhabditis elegans. Cell 136, 926-938 (2009).

128. Loedige, I. & Filipowicz, W. TRIM-NHL Proteins Take on miRNA Regulation. Cell 136, 818-820 (2009).

129. Sonoda, J. & Wharton, R.P. Drosophila brain tumor is a translational repressor. Genes & Development 15, 762-773 (2001).

130. Bello, B., Reichert, H. & Hirth, F. The brain tumor gene negatively regulates neural progenitor cell proliferation in the larval central brain of Drosophila. Development 133, 2639-2648 (2006).

131. Betschinger, J., Mechtler, K. & Knoblich, J.A. Asymmetric segregation of the tumor suppressor brat regulates self-renewal in Drosophila neural stem cells. Cell 124, 1241-1253 (2006).

132. Frank, D.J., Edgar, B.A. & Roth, M.B. The Drosophila melanogaster gene brain tumor negatively regulates cell growth and ribosomal RNA synthesis. Development 129, 399-407 (2002).

133. Nolo, R., Morrison, C.M., Tao, C.Y., Zhang, X.W. & Halder, G. The bantam microRNA is a target of the hippo tumor-suppressor pathway. Current Biology 16, 1895-1904 (2006).

Page 53: Understanding the general role of the microRNAs in ...repositorio.ul.pt/bitstream/10451/1448/1/20902_ulfc080582_tm.pdf · Em Drosophila melanogaster existem dois isotipos desta enzima

  

51  

X. ANNEXES

Table S1 – Dcr-1 and tissue and cell growth

GENOTYPE TISSUE SIZE1,3 CELL DENSITY1

Males ptc domain

ptc>GFP 100 ± 8,4 (n=10) 2 --

ptc>dcr-1RNAi 75 ± 5,1 (n=12) --

en domain en domain4 anterior compartment5

en>GFP 100 ± 5,7 (n=12) 100 ± 7,6 (n=10) 100 ± 10,7 (n=10)

en> dcr-1RNAi 75 ± 5,1 (n=10) 123 ± 9,4 (n=10) 108 ± 15,4 (n=10)

en> dcr-1RNAi>dcr-1 110 ± 5,8 (n=12) 95 ± 2,6 (n=10) 113 ±5,0 (n=10)

en> dcr-1RNAi; dap4/+ 122 ± 7,3 (n=7) 105 ± 4,5 (n=10) 107 ± 6,7 (n=10)

en> dcr-1RNAi >CycE 111 ± 4,5 (n=12) 106 ± 4,5 (n=10) 110 ± 4,7 (n=10)

en> dcr-1RNAi>dMyc 118 ± 9,5 (n=12) 105 ± 2,98 (n=10) 109 ± 2,2 (n=10)

en> dcr-1RNAi; hs-dMyc 99 ± 13,3 (n=10) 89 ± 3,5 (n=10) --

en> dcr-1RNAi; tub-dMyc 108 ± 5,8 (n=10) 111 ± 17,1 (n=10) --

en> dcr-1RNAi;mei-P26RNAi-

106754 86 ± 11,2 (n=10) 105 ± 3,0 (n=10) --

en> dcr-1RNAi; hs-bantam 112 ± 4,4 (n=12) 105 ± 1,8 (n=10) 107 ± 4,1 (n=10)

dap4/+ 6 112 ± 6,8 (n=13) 96 ± 4,1 (n=10) 105 ± 8,6(n=10)

en>CycE 6 100 ± 6,3 (n=13) 156 ± 6,6 (n=10) 102 ± 2,4 (n=10)

en> dMyc 6 136 ± 7,9 (n=13) 81 ± 3,2 (n=10) 92 ± 4,8 (n=10)

en>mei-P26RNAi-106754 6 97 ± 6,5 (n=10) 93 ± 2,3 (n=10) --

hs-bantam 6 119 ± 6,3 (n=12) 94 ± 4,2 (n=10) 97 ± 2,7 (n=10)

Females

en>GFP 100 ± 7,4 (n=12) -- --

en> dcr-1RNAi 66 ± 1,7 (n=12) -- --

Rbfsls5/+ ; en> dcr-1RNAi 80 ± 3,1 (n=12) -- --

--

Rbfsls5/+ 6 101 ± 8,1 (n=12) -- --

1 Tissue size and cell density values were measured as a ratio (in percentage) with respect to the GFP expressing cells.

Page 54: Understanding the general role of the microRNAs in ...repositorio.ul.pt/bitstream/10451/1448/1/20902_ulfc080582_tm.pdf · Em Drosophila melanogaster existem dois isotipos desta enzima

  

52  

2 Numbers in parentheses indicate the number of adult wings analyzed for each genotype. 3 In ptc>dcr-1RNAi, a significant decrease in the size of the ptc domain was observed when compared with ptc>GFP wings (p<10-5). In en>dcr-1RNAi, a significant decrease in the size of the en domain was observed when compared with en>GFP wings (p<10-7 in males and p<10-3 in females). Halving the dose of dap or Rbf, co-expression of dcr-1, CycE or dMyc and expression of hs-dMyc, tub-dMyc, mei-P26RNAi-106754 or hs-bantam significantly rescued this phenotype (p(dap4)<10-5, p(Rbfsls5)<10-5, p(dcr-1)<10-10, p(CycE)<10-8, p(dMyc)<10-9, p(hs-dMyc)<10-5, p(tub-dMyc)<10-9, p(mei-P26RNAi (106754))<10-4 and p(hs-bantam)<10-9). 4 In en>dcr-1RNAi, a significant increase in the cell density of the en domain was observed when compared with en>GFP wings (p<10-3). Halving the dose of dap, co-expression of dcr-1, CycE or dMyc and expression of hs-dMyc, tub-dMyc, mei-P26RNAi (106754) or hs-bantam significantly rescued the cell size defects (p(dap4)<10-3, p(dcr-1)<10-2 , p(CycE)<10-3, p(dMyc)<10-5 p(hs-dMyc)<10-6, p(tub-dMyc)<10-2, p(mei-P26RNAi (106754))<10-4 and p(hs-bantam)<10-4). 5 Cell densities in the anterior compartment were not significantly changed when compared to en>GFP wings (p(dcr-1RNAi)=0,25; p(dcr-1RNAi;dcr-1)=0,03; p(dcr-1RNAi;dap4/+)=0,58; p(dcr-1RNAi;CycE)=0,05; p(dcr-1RNAi dMyc)=0,04; and p (en>Dcr-1RNAi;hs-bantam)=0,06. 6 Size of the en domain, when compared to en>GFP wings, was significantly increased in en>dMyc (p<10-10), dap4/+ (p<10-4) and hs-bantam (p<10-5) wings, but not in en>CycE (p=0,9), en>mei-P26RNAi (106754) (p=0,6) or Rbfsls5/+ (p=0,9) wings. Cell density, when compared to en>GFP wings, was significantly decreased in en>dMyc (p<10-5) and in en>mei-P26RNAi (106754) (p<10-3) wings, increased in en>CycE (p<10-8) wings and remained unchanged in dap4/+ (p=0,2) and hs-bantam (p=0,06) wings.

Page 55: Understanding the general role of the microRNAs in ...repositorio.ul.pt/bitstream/10451/1448/1/20902_ulfc080582_tm.pdf · Em Drosophila melanogaster existem dois isotipos desta enzima

  

53  

Figure S1 – Cell cycle markers in wild type and different mutant conditions. (A-L) Wing discs labeled to visualize expression of Mei-P26 (A, in red) Cyclin E (CycE, B, C, H, and L in red, white or green), Cyclin B (CycB, D and J, red or white), Dacapo (Dap, E, red), PCNA (F, I and L, red or white), dMyc (K and L, red, white or blue) and GFP (C, H-K, green) protein expression and dE2F activity (an E2F1 responsive reporter ORC1-GFP was used, antibody to GFP, G, red) of the following genotypes: wild type (A, B, D-G); ptc-Gal4; UAS-CycE (C), hs-FLP;;FRT82 arm-lacZ/ FRT82 M(3)95A2 Ubi-GFP (H-K) and ptc-Gal4; UAS-dMyc (L).

 

Page 56: Understanding the general role of the microRNAs in ...repositorio.ul.pt/bitstream/10451/1448/1/20902_ulfc080582_tm.pdf · Em Drosophila melanogaster existem dois isotipos desta enzima

  

54  

Figure S2 – Pathways involved in growth control (A) Wing discs with mutants clones for dcr-1 expressing CycE. Number of clones per disc and number of cells per clone showed no rescue of the dcr-1 depletion phenotype. Genotype: hs-FLP tub-Gal4 UAS-GFP; UAS-CycE/+; FRT82 dcrQ1147X / FRT82 Gal80 (B-D) Wing discs labeled to visualize expression of FOXO (in red or white, B), CycE-lacZ (in red or white, C), ex-lacZ (in red or white, D) and GFP or Ptc in green of the following genotypes: hs-FLP; FRT 82 lacZ M(3)95A2/FRT82 dcr-1Q1147X; ptc>dcr-1RNAi and wild type. (E) Wing discs labeled for Diap1-lacZ (in red or white) and Ptc in green in ptc>dcr-1RNAi and wild type flies. FoxO is used as a read out of the Insulin/PI3K/TOR pathway. CycE-lacZ, ex-lacZ and Diap1-lacZ are used as read out the Hippo pathway.

Page 57: Understanding the general role of the microRNAs in ...repositorio.ul.pt/bitstream/10451/1448/1/20902_ulfc080582_tm.pdf · Em Drosophila melanogaster existem dois isotipos desta enzima

  

55  

  

 Figure S3 – RNAi constructs for mei-P26. (A, B) Wing discs stained for Mei-P26 (in red) in two different RNAi lines for mei-P26 showing a decrease in the levels of the protein (white brackets in B). Genotypes: ptc>mei-P26RNAi (106754) and en>mei-P26RNAi (7553). (C) Wing disc labeled to visualize Mei-P26 (in red or white) and dMyc (in blue or white) in ptc>dcr-1RNAi, mei-P26RNAi (106754).                             

Page 58: Understanding the general role of the microRNAs in ...repositorio.ul.pt/bitstream/10451/1448/1/20902_ulfc080582_tm.pdf · Em Drosophila melanogaster existem dois isotipos desta enzima

  

56  

  

  

Figure S4 – (A, B) ptc>dcr-1RNAi wing discs in which dMyc was expressed under the tubulin promoter stained for PCNA (A) and CycE (B) visualized in red or white. (C-E) Imaginal discs with cells expressing doubled RNAi for dcr-1 and mei-P26, in order to restore dMyc levels, labeled to visualize PCNA (C), CycE (D), Dap (E) visualized in red or white and also for dMyc (in blue or white, C) and Mei-P26 (in green or white, C).

                         

Page 59: Understanding the general role of the microRNAs in ...repositorio.ul.pt/bitstream/10451/1448/1/20902_ulfc080582_tm.pdf · Em Drosophila melanogaster existem dois isotipos desta enzima

  

57  

  

  

Figure S5 – bantam target sites. (A, B) Conservation of the predicted bantam target sites in the mei-P26 (A) and dap 3’UTR (B) among 12 Drosophila species. Sequences complementary to bantam seed region are boxed in red.