Você está na página 1de 64

10/06/20

ENDOCRINOLOGIA+
MOLECULAR
Profa.+Isis+Hara+Trevenzoli,+PhD
haraisis@yahoo.com.br

Conteúdo(do(Módulo
• Aspectos(gerais(e(compar.mentalização(celular
• Estrutura(e(função(do(DNA(e(cromossomos
• Replicação,(Reparo(e(Recombinação(gênica
• Transcrição,(tradução(e(função(de(proteínas
• Mutações,(Polimorfismos,(Heranças(gené.cas
• Controle(da(expressão(gênica
• Epigené.ca
• Bases(gené.cas(de(doenças(endócrinas

QUAL(A(IMPORTÂNCIA(DE(SE(COMPREENDER(MECANISMOS(
MOLECULARES(EM(ENDOCRINOLOGIA???
2

1
10/06/20

QUAL%A%IMPORTÂNCIA%DE%SE%COMPREENDER%MECANISMOS%
MOLECULARES%EM%ENDOCRINOLOGIA???

• Diagnós(co: patologias/endócrinas/de/origem/gené(ca,/congênita/ou/adquirida

• Tratamento:/alvos/moleculares/para/terapias/farmacológicas,/alvos/para/nutraceu(cos,/etc

• Pesquisa:/estudos/de/prevalência,/impactos/sobre/a/saúde/pública,/estratégias/de/prevenção

Sistêmico Celular

Sistema%Endócrino
Tecidual Molecular

Sistêmico Celular

Sistema(Endócrino
Tecidual Molecular

• Biossíntese
TIREÓCITO • Metabolismo
• Ação/Efeitos

2
10/06/20

Compar'mentalização0Celular

DNA DNA

! DNA$nuclear
! DNA$mitocondrial
5

Organização+do+Núcleo

A"conformação"do"material
gené3co"depende"do"estado
funcional"da"célula"

Estágio"do"ciclo"celular
6

3
10/06/20

Nucleossomos
! 140$pares$de$bases
! Histonas$ao$centro

Cariótipo&Humano
Ciclo&Celular

1 a 22
ordem de tamanho

23 e 24
cromossomos sexuais

8
Molecular Biology of The Cell, 4th ed, 2002

4
10/06/20

Carió&po)Humano

Pares&homólogos:&1/ 22&(idên6cos)
Cromossomos&autossomos
Cromossomos&Paternos&e&Maternos&

Padrão de&bandas de&cada


Cromossomo é ÚNICO

Par&heterólogo:&1&par
Cromossomos&sexuais&X +&Y
Cromossomos&Paternos&e&Maternos&

Estrutura'dos'Cromossomos

Telômeros

Braço curto (braço p)

Centrômeros

Braço longo (braço q)

10

5
10/06/20

Alterações*na*Estrutura*dos*Cromossomos

• Análise Citogenética
• Detecção alterações numéricas (Trissomia 21)
• Não disjunção durante a>mitose ou meiose
• Perda ou duplicação de>Regiões cromossômicas (Prader Willi)
• Câncer e>rearranjo cromossômico (Câncer Tireóide)

11

Síndrome de Patau (trissomia do 13) Síndrome de Edwards (trissomia do 18)

Deleção'do'braço'curto'do'cromossomo'5' Síndrome de Down (trissomia do 21)


12

6
10/06/20

Síndrome)de)Klinefelter Síndrome)47

Síndrome)do)X) Síndrome)de)Turner
13

Função'e'Estrutura'do'DNA
• DNA$=$ácido$desoxirribonucleico
• Molécula$que$contém$INFORMAÇÃO$– código$genéBco
• A$informação$de$uma$caracterísBca$é$codificada$em$genes
• Cada$cromossomo$contém$diversos$genes
• Cada$gene$é$composto$por$regiões$codificantes$(EXONS)$e$nãoQcodificantes$(INTRONS)
• A$informação$dos$genes$de$uma$célula$é$herdável$para$as$célulasQfilhas$(mitose$e$meiose)

Transcrição$

DNA Tradução$

RNA Função$

PROTEINA
FENÓTIPO

14

7
10/06/20

Estrutura'do'DNA
• Dupla&fita&em&forma&de&Hélice&– cadeias&de&nucleotídeos&complementares
• Nucleotídeos:&Adenina&(A),&Citosina&(C),&Guanina&(G)&e&Timina&(T)

• Pareamento&complementar&por&pontes&de&hidrogênio:& A:T e&&&C:G

15

Estrutura'e'Caracterís,cas'do'DNA
• Direcionamento de,leitura:,5’,para 3’;,,,,Fitas an7paralelas
• Replicação precisa e,semi;conserva7va >,código gené7co das,células somá7cas
mães para células filhas na divisão celular
• Projeto genoma humano (2003/2004),>,25,mil,genes
• Para,um,determinado gene,X,,os individuos de,uma mesma espécie apresentam
basicamente a,mesma sequência,,com,pequenas diferenças (polimorfismos)
• Apenas gêmeos univitelinos tem,sequencias iden7cas

3’
5’

3’ 5’ 16

8
10/06/20

Replicação+do+DNA+
• Para$dividir é preciso replicar antes!!$Importância da$fase S

17

Replicação+do+DNA+
• Para$dividir é preciso replicar antes!!
• Processo celular “preciso”$comandado pela DNA$polimerase (I$e$III)
• Origem de$Replicação – regiões ricas em A:T

DNA$pol$I
III
FORQUILHA+DE+REPLICAÇÃO

Fita
Descontínua

Fita
Contínua
III
DNA$Pol:$5’$$>>>$$3’

18

9
10/06/20

Reparo'do'DNA'

• Mecanismo inicial >,,checagem da,DNA,polimerase III


• Segundo,mecanismo >,exonuclease de,correção 3’=5’
• Se,não funcionarem >,Sistema de,reparo de,pareamento incorreto
• Falhas de,replicação sem reparo >>,mutações &,seleção natural
Ex.:,BRCA=2,>,câncer de,mama,e,ovários >,maquinaria reparo defeituosa

19

Transcrição+do+DNA+em+RNA+
“A#leitura do#código gené2co”

DNA

?
Expressão Gênica
Diferencial

CONJUNTOS+DE+
20
PROTEINAS+DISTINTOS

10
10/06/20

RNA$3 Acido$Ribonucleico
Como$o$DNA$se$diferencia$do$
RNA$???
O"açúcar do"DNA"é"a"desoxiribose
enquanto"que"o"do"RNA"é"a"ribose."

O"DNA"contém"a"base timina e"o"RNA"


possui"a"base uracila."

O"DNA"é"um"filamento"duplo e"o"RNA"é"
um"monofilamento."

O"DNA"apresenta"uma"molécula"longa,"
bastante"estável e"o"RNA"uma"molécula"
curta,"com"meiaAvida"variável

21

Transcrição+do+DNA+em+RNA+
! Baseada em complementariedade de1bases

Pareamento:
DNA RNA
A U
T A
C G
G C

22

11
STRUCTURE OF A GENE ENCODING A are implicated in the regulation of gene transcription (see
POLYPEPTIDE HORMONE “Epigenetic Inheritance of Phenotypic Traits”).
The histones of open-configured chromatin (i.e., euchro-
Structural analyses of gene sequences have resulted in at matin) are heavily acetylated and methylated; this loosens
least three major discoveries that are important for under- their association with DNA and allows the access of tran-
standing the expression of peptide-encoding genes. First, scription factors to the promoter regions of expressed10/06/20
sequences of almost all of the known biologically active genes. Conversely, the histones of closed chromatin (i.e.,
hormonal peptides are contained within larger precursors heterochromatin) are underacetylated and undermethyl-
that often encode other peptides, many of which are of ated, adhere tightly to DNA, and prevent access of tran-
unknown biologic activity. Second, the transcribed regions scription factors to the promoters of transcriptionally silent
of genes (called exons) are interrupted by sequences (called genes.
introns) that are transcribed but subsequently cleaved A typical protein-encoding gene consists of two func-
from the initial RNA transcripts during their nuclear pro- tional units (Fig. 3-8). One is a transcriptional region, and
cessing and assembly into specific mRNAs. Third, specific the other is a promoter or regulatory region.
Transcrição+do+DNA+em+RNA+
DNA-binding proteins

Sinais
RNA Transcription
Repressores Ativadores Metabolismo initiation site
Pol II
Celular
CP
5´ TSS TSE MRE Exon Intron Exon 3´
(TATA-box)

Metabolic Basal ATG TGA


Tissue-specific
silencers/enhancers response constitutive Transcription unit
element promoter
Promoter region
Figure 3-8 Structure of a consensus gene encoding a prototypical polypeptide hormone. A consensus gene typically consists of a promoter region and a
transcription unit. The transcription unit is the region of DNA composed of exons and introns that is transcribed into a messenger ribonucleic acid (mRNA)
precursor. Transcription begins at the cap site sequence in DNA and extends several hundred bases beyond the poly(A) addition site in the 3′ region. During
Região+promotora+no+DNA+>>+porção+regulatória+da+transcrição
post-transcriptional processing of the RNA precursor, the 5′ end of mRNA is capped by the addition of methylguanosine residues. The transcript is then
cleaved at the poly(A) addition site approximately 20 bases 3′ to the AATAAA signal sequence, and the poly(A) tract is added to the 3′ end of the RNA.
Unidade+de+transcrição+no+DNA+>>+vai+dar+origem+ao+RNA+precursor+e+depois+ao+RNAm
Introns are cleaved from the RNA precursor, and exons are joined together. Dinucleotides GT and AG are invariably found at the 5′ and 3′ ends of introns.
Translation of mRNA starts with the codon ATG for methionine. Translation is terminated when the polyribosome reaches the stop codon TGA, TAA, or
TAG. The promoter region >>+promotores+alterna<vos
of the gene located 5′ to the cap site contains numerous short regulatory DNA sequences that are targets for interactions with
specific DNA-binding proteins. These sequences consist of the basal constitutive promoter (TATA box), metabolic response elements that modulate transcrip-
tion (e.g., in response to cyclic adenosine monophosphate [cAMP], steroid hormone receptors, or thyroid hormone receptors), and tissue-specific enhancers
and silencers that respectively permit or prevent transcription of the gene. The enhancer and silencer elements direct expression
23 of specific subsets of genes
to cells of a given phenotype. Whether a gene is or is not expressed in a particular cellular phenotype depends on complex interactions of the various
DNA-binding proteins among themselves and, most importantly, with the TATA box proteins of the basal constitutive promoter.

Transcrição+do+DNA+em+RNA+
! A"transcrição da"sequencia de"DNA"em RNA"é contínua???

GENETIC CONTROL OF PEPTIDE HORMONE FORMATION 39

E1 IA E2 IB E3 IC E4 ID E5 IE E6
Gene 3´ 5´

Gene+
glucagon M
Q
H
K
R
K K
R R
R R
R R
K
K
mRNA 5´ 3´
UN-TX S N Gluc GLP-I GLP-II UN-TX
24
IP-I IP-II
Figure 3-9 The pancreatic glucagon gene and its encoded messenger RNA (mRNA): complementary DNA. In the glucagon gene, exons precisely encode
separate functional domains. The gene consists of six exons (E1 through E6) and five introns (1A through 1E). The mRNA encoding pre-proglucagon, the
protein precursor of glucagon, consists of 10 specific regions: from left to right, a 5′ untranslated sequence (UN-TX, open), a signal sequence (S, stippled), an
amino-terminal extension sequence (N, hatched), glucagon (Gluc, shaded), a first intervening peptide (IP-I, hatched), a first glucagon-like peptide (GLP-I, shaded),
a second intervening peptide (IP-II, hatched), a second glucagon-like peptide (GLP-II, shaded), a dilysyl dipeptide (hatched) after the GLP-II sequence, and an
untranslated region (UN-TX, open). Exons from left to right encode the 5′ untranslated region, signal sequence, glucagon, GLP-I, GLP-II, and 3′ untranslated
sequence. Letters shown above the mRNA denote amino acids located at positions in the pre-proglucagon molecule that are cleaved during cellular process-
ing of the precursor. The amino acid methionine (M) marks the initiation of translation of mRNA into pre-proglucagon. H, histidine; K, lysine; Q, glutamine;
R, arginine.

12
post-transcriptional processing of mRNA, translation, or directly or indirectly to enhance the efficiency of initiation
post-translational processing. In different endocrine cells, of translation of proinsulin mRNA.52
one or more levels may serve as specific control points for Rapid metabolic regulation at the level of post-
regulation of the production of a hormone (see “Biologic transcriptional processing of mRNA precursors has
Diversification”). not been clearly established. However, alternative exon
10/06/20

Processamento"do"RNA"

! A"extremidade"5 do"RNA"é"protegida"imediatamente"após"emergir"do"DNA:"adição"
do CAP

! Após"o"reconhecimento"do"sinal"de"término"de"transcrição,"o"RNA"é"clivado."É"
adicionada"a cauda"poliCA (150"a"200"A)

! Os"introns são"removidos"por"um"processo"altamente"controlado"denominado
SPLICING

PROMOTOR exon intron exon intron exon DNA

exon intron exon intron exon RNA primário

25

Processamento+do+RNA+
Gene$IGF'1$$>$splicing$alternativo

Cromossomo$12

26
Front.$Endocrinol.,$04$February$2013$

13
10/06/20

Processamento+do+RNA+
Genes%Receptores de%hormônio tireoideano >%splicing%alternativo
Especificidade tecidual

Gene+THRA Gene+THRB

DBD%=%domínio%ligação%ao%DNA
LBD%=%domínio%ligação%ao%ligante%(hormônio)
27
Clinical Diabetes(and Endocrinology20173:8

Processamento+do+RNA+
Receptores de*Leptina >*splicing*alternativo

28

14
10/06/20

Tradução)do)RNAm)para)Peptídeo/Ptns

RNA)maduro

•4)nucleotídeos)!códons)3)nucleotídeos)! 64)combinações

• 61 códons)codificam)para)os)aminoácidos

• 3)códons)de)"parada")marcam)o)polipeptídeo como)terminado

29
• 1)códon)AUG)é)um)sinal)de)"início")para)começar)a)tradução)(metionina)

Tradução)do)RNA)para)Peptídeo/Ptns
Segunda'Letra
Codons
mRNA

Fim
Primeira'Letra

Terceira'Letra

Início

30

15
10/06/20

Um#único#gene#pode#dar#origem
A#vários#hormônios#peptídicos
diferentes

Mecanismos#Transcricionais
Ou
Pós>Transcricionais
Ou#
Pós>Traducionais

31

32 Transcrição,,Tradução,e,Função,Biológica
GENETIC CONTROL OF PEPTIDE HORMONE FORMATION

Nucleus

Nucleosome Chromatin
(DNA + protein) DNA

RNA polymerase Transcription


mRNA precursor

Excision of introns Pre-mRNA


Rejoining
Post-transcriptional
processing

mRNA
“Cap”
AAAAA3´ mRNA
Me7 GPPP
Cytoplasm
Translation
tRNA, amino acids
mRNA
Ribosomes Golgi region Preprotein

Cisterna of Secretion Modification


CHO
endoplasmic
reticulum Post-
Mature transcriptional
Pre-prohormone Prohormone hormone Protein processing

Cotranslational Post-translational Transport —Presecretory

—Postsecretory
Biologic action 32

Figure 3-2 Cellular synthesis of polypeptide hormones. Steps that take place within the nucleus include transcription of genetic information into a messenger
ribonucleic acid precursor (pre-mRNA) followed by post-transcriptional processing, which includes RNA cleavage, excision of introns, and rejoining of exons,
resulting in formation of mRNA. Ends of mRNA are modified by addition of methylguanosine caps at the 5′ end and poly(A) tracts at the 3′ ends. The cyto-
plasmic mRNA is assembled with ribosomes. Amino acids, carried by amino-acylated transfer RNAs (tRNAs), are then polymerized into a polypeptide chain.
The final procesess in protein synthesis take place during growth of the nascent polypeptide chain (cotranslational) and after release of the completed chain
(post-translational). They include proteolytic cleavages of the polypeptide chain (conversion of pre-prohormones or prohormones to hormones), derivatiza-
tions of amino acids (e.g., glycosylation, phosphorylation), and cross-linking and assembly of the polypeptide chain into its conformed structure. Post-
translational synthesis and processing of a typical secreted polypeptide require vectorial or unidirectional transport of the polypeptide chain across the
membrane bilayer of the endoplasmic reticulum, resulting in sequestration of the polypeptide in the cisterna of the endoplasmic reticulum, a first step in the
export of proteins destined for secretion from the cell (see Fig. 3-6). Most translational processing occurs within the cell (presecretory); in some instances,
it occurs outside the cell, when further proteolytic cleavages or modifications of the protein take place (postsecretory). CHO, carbohydrate.
16
phosphorylation, acetylation, amidation, lipidation, sul- packaging into secretory granules and export from the cell.
fation),5 any one of which may affect the conformation or Most smaller hormones and regulatory peptides are pro-
function of the protein, a single gene may ultimately duced as a consequence of post-translational cleavages of
encode a wide variety of specific proteins as a result of the precursors within the Golgi complex of secretory cells.
10/06/20

Fatores(de(Transcrição(na(organogênese
De(sistemas(endócrinos

Duas(famílias(de(proteínas(regulatórias(orquestradas(durante(o(desenvolvimento
! Família(de(receptores(nucleares((TRs,(GR,(RXR...)
! Família(de(proteínas(de(homeodomínio ligadoras de(DNA((fatores(de(transcrição(com
homeobox de(60(aminoácidos(que(se(liga(ao(DNA)

1J Agenesia(hipofisária(parcial((POU1F1(ou(PitJ1)
2J Agenesia(pancreática((PDX1)
3J Agenesia(adrenal(e(gonadal((SF1(e(NROB1/DAX1)

33

Agenesia#hipofisária#parcial#(POU1F1#ou#Pit;1)

NATURE'REVIEWS' |'
ENDOCRINOLOGY'VOLUME'7' |'
DECEMBER'2011' |'34
727#

17
10/06/20

Agenesia(pancreática((PDX1)
Mutação(no(“pancreas duodenum homeobox 1”

Homozigose)– prejuízo)desenvolvimento
Heterozigose)– risco)de)DM2

35
Pancreatic(Stem(Cells:(Unresolved(Business,(DOI:(10.5772/23760

Agenesia(adrenal(and(gonadal((SF1(e(DAX1)
Família'de'receptores'nucleares:
• SF1'– fator'esteroidogênico'que'se'liga'a'elemento'responsivo'ao'estrogênio'(ERE)
• DAX1B fator'transcrição'que'se'liga'a'elemento'responsivo'ao'ácido'retinóico'(RAR)
B HIPOPLASIA'ADRENAL'CONGÊNITA
B HIPOGONADISMO'HIPOGONADOTRÓFICO

36

18
10/06/20

QUESTÃO(TEEM

O"dogma"central"da"biologia"pode"ser"entendido"como:
a)#Mecanismo#pelo#qual#todos#organismos#utilizam#sequências#semelhantes#de#DNA#
para#cada#aminoácido,#ao#longo#da#evolução.
b)#Propriedade#de#determinados#aminoácidos#serem#codificados#por#mais#de#uma#
trinca#de#nucleotídeos.
c)#Capacidade#de#proteínas#poderem#contra!regular#a#expressão#gênica,#como#
ocorre#em#vários#receptores#hormonais.
d)#Fluxo#unidirecional#da#informação#genética,#quando#o#DNA#especifica#a#síntese#de#
RNA#que,#por#sua#vez,#determina#a#síntese#de#polipeptídeos,#que#formarão#
proteínas.
e)#Capacidade#de#um#único#gene#poder#formar#várias#proteínas#distintas,#através#de#
mecanismos#como#o#“splicing” alternativo,#como#no#caso#das#isoformas alfa#e#beta#
do#receptor#glicocorticóide.

37

QUESTÃO(TEEM

Em relação ao processo de incorporação de aminoácidos na cadeia peptídica


para a síntese de proteínas, assinale a alternativa CORRETA:
a) Cada tríade de bases (códon) deve codificar um aminoácido diferente.
b) Alguns códons podem codificar mais de um aminoácido.
c) Alguns aminoácidos podem ser codificados por mais de um códon.
d) O stop códon codifica o aminoácido metionina.

38

19
10/06/20

QUESTÃO(TEEM

Em relação aos mecanismos de ação hormonais, marque a CORRETA:


a. São hormônios peptídeos ou neurotransmissores, que agem através dos receptores
acoplados à proteína G: TSH, GH, epinefrina e PTH.
b. Os receptores de fatores de crescimento como IGF1 e insulina tem a capacidade
de se autofosforilar, exercendo seus efeitos biológicos através de diferentes cascatas
de sinalização.
c. Os receptores esteroides, como receptor da progesterona e receptor do estrogênio,
são acoplados a proteínas chaperônicas citoplasmáticas e carreados ao núcleo onde
exercem seus efeitos.
d. Mutações ativadoras no gene do receptor sensor de cálcio acoplado à proteína Gq
são descritas em pacientes portadores de hipercalcemia hipocalciúrica familiar.

39

QUESTÃO(TEEM

Sobre os receptores hormonais, marque a INCORRETA:


a. Os PPAR gama e alfa tem, como ligantes, os ácidos graxos circulantes.
b. Existem diferentes receptores para os ácidos trans!retinoico e o cis-
retinoico.
c. Os receptores da aldosterona também tem afinidade pelo cortisol
d. O subtipo alfa dos receptores do hormônio tireoidiano e do receptor
estrogênico são codificados por um gene único.

40

20
10/06/20

MUTAÇÕES,*
POLIMORFISMOS*E*
HERANÇA*GENÉTICA
Profa.*Isis*Hara*Trevenzoli,*PhD
haraisis@yahoo.com.br

41

Locus Gênicos
Os#cromossomos#existem#aos#pares#nas#células#somáticas.
Cada#gene#ocupa#um#lugar#definido#no#cromossomo.
Esse#lugar#definido#é#denominado#locus gênico

Genes+Alelos
Os#genes#que#ocupam#o#mesmo#locus em#
cromossomos#homólogos#são#denominados#
genes#alelos.#

Versões alterna@vas da#


seqüência de#DNA#no#locus#
são chamadas de#alelos

42

21
10/06/20

• Alelo do&'po selvagem:&presente em mais da&metade dos&


indivíduos em uma população

• Outras versões do&gene&são alelos variantes (ou mutantes)&

• Se&houver dois ou mais alelos rela'vamente comuns (definidos


por convenção como tendo uma freqüência alélica >1%)&em um&
locus&na população,&dizIse&que esse locus&apresenta
polimorfismo

43

MUTAÇÕES
! Erros na replicação,6recombinação e6reparo do6DNA
! Contribuição para6evolução e6aperfeiçoamento de6caracteristicas
! A6maioria das6mutações são “neutras”,6nem benéficas nem maléficas

1" Mutações pontuais ou point&mutations&>&substituição de2um2par2de2bases

2" Mutações em larga escala ou large"scale2genome2rearrangements2>2deleções,2


duplicações,2inversões e2translocação de2DNA2entre2cromossomos

3" Elementos2“móveis”2de2DNA2ou2transposons >&sequencias2moveis2que2se2inserem2em2


diversas2regiões2genômicas

44

22
10/06/20

CLASSIFICAÇÃO'DAS'MUTAÇÕES

! Quanto'à'origem:
" espontâneas
" induzidas'(agentes'mutagênicos:'agentes'químicos,'radiação,'vírus)

! Quanto'ao'tipo'celular:
" Somáticas'(mosaicismo,'câncer)
" Germinativas'(doenças'genéticas'clássicas'hereditárias)

podem'ser'transmitidas'à'prole,'mas'as'somáticas'não.
! Quanto'à'adaptabilidade:
" Benéficas
polimorfismos
" Neutras
" Deletérias'(doenças'genéticas,'câncer)
! Quanto'ao'nível:
alterações'numéricas Inversão,'deleção,'
" Cromossômicas
alterações'estruturais'(>5Mb) translocação
"Gênicas mutações'pontuais
deleções'e'inserções 45
members.cox.net/amgough/Fanconi-genetics-gene

MUTAÇÕES)PONTUAIS
! Substituições*de*nucleotídeos*(mutações*pontuais)*" mais*frequentes

" Tipos*de*substituição

Transições:*purina por*purina ou*pirimidina por*pirimidina


A!G C!T*******
Transversões:*purina por*pirimidina ou*pirimidina por*purina

A→C C→A
A→T C→G
G→C T→ A
G→T T→G

Mais)comuns:)transições

46

23
10/06/20

MUTAÇÕES)PONTUAIS
Mutações)Silenciosas:
São%mutações%que%ocorrem%em%locais)que)não)modificam)a)função)do)genoma.%Estas%
mutações%%ocorrem%em%regiões%não6codificantes%do%DNA%(intergênicas)ou)íntrons).%Estas%
regiões%compõem%98.5%%do%genoma%humano.

Mutações)neutras/samesense:
Mutações%que%ocorrem)nas)sequências)codificantes)e%resultam%na%geração%de%um%códon%
que%codifica%o%mesmo)aminoácido)do)códon)original)(não%mutado).%Ocorrem%geralmente%
no%terceiro%nucleotídeo%do%códon.

Mutações)Missense (sentido)trocado):
Que%levam%à%substituição)de)um)aminoácido)por)outro,%alterando%a%sequência%da%
proteína.%Frequentemente%não%alteram%a%função%biológica%da%proteína,%a%maioria%tolera%
mudança%de%poucos%aminoácidos.

Mutações)Nonsense (sem)senKdo):
A%mutação%cria)um)códon)de)terminação,%fazendo%com%que%a%sequência%da%proteína%
termine%prematuramente.%O%efeito%da%mutação%nonsense depende%do%quanto%da%proteína%
é%perdida.%Geralmente%é%catastrófica%e%gera%proteína%não6funcional.

47

MUTAÇÕES)PONTUAIS

Exemplos)de
Mutações)em
Sequencias)
codificantes

48

24
10/06/20

DELEÇÕES'E'INSERÇÕES
! A"mutação"na"forma"de"deleção é"a"perda"do"material"genético.
! A"mutação"na"forma"de"inserção"é"a"adição"de"nucleotídeos"com"ganho"de"material"genético
! Pode"ocorrer"em:
;um"ou"mais"cromossomos"ou"parte"destes
;um"gene"inteiro"ou"em"parte"do"gene
;em"um"único"códon"(3"nucleotídeos)"ou"
múltiplos"CODONS"(in;frame)
;1,"2,"4,"5....."nucleotídeos"(frameshift) não"múltiplo"de"3

Normalmente'geram'aberrações'cromossômicas
Podendo'ser'detectadas'com'análise'do'cariótipo

49

QUESTÃO(TEEM

Qual%das%definições%abaixo%é%INCORRETA:

a) Mutações*silenciosas*são*aquelas*onde*o*códon*mutado*codifica*o*mesmo*aminoácido*
que*o*códon*normal.

b)*Mutações*INDEL*(inserções*/*deleções)*sempre*alteram*a*matriz*de*leitura,*sendo*assim*
podem*ser*consideradas*mutações*um*tipo*de*mutação*“frameshift”.

c)*Mutações*“frameshift” são*aquelas*onde*a*matriz*de*leituras*dos*codons é*alterada,*


gerando*uma*sequência*de*aminoácidos*anômala*a*partir*de*sua*ocorrência.

d)*Mutações*“nonsense” (ou*sem!sentido)*são*aquelas*com*substituição*nãoKsinônima*
resultando*na*substituição*de*um*códon*que*especifica*um*aminoácido*por*um*códon*de*
terminação.

e)*Mutações*“missense” determinam*a*codificação*de*aminoácido*diferente*que*o*original.

50

25
10/06/20

QUESTÃO(TEEM

As#mutações#gênicas#correspondem#a#uma#das#alterações#que#
determinam#o#aparecimento#de#fenótipos#patológicos.#Qual#dentre#
os#tipos#de#mutação#abaixo#é#o#mais#freqüente em#humanos?

a)#Inserções/duplicações.
b)#Missense/nonsense.
c)#Splicing.
d)#Rearranjos.
e)#Variação#do#número#de#repetições.

51

QUESTÃO(TEEM

Com$relação$às$mutações,$MARQUE$A$CORRETA:

a)#Mutações#são#modificações#súbitas#que#ocorrem#no#material#genético#e#
sua#taxa#de#ocorrência#independe#da#presença#ou#não#de#agentes#
mutagênicos.
b)#Mutações#sempre#são#deletérias.
c)#O#término#prematuro#da#tradução#protéica não#interfere#na#função#e#nem#
na#eficiência#da#proteína.
d)#A#troca#de#um#nucleotídeo#não#muda#obrigatoriamente#o#aminoácido#que#
será#incluído#na#proteína.
e)#Quanto#ocorrem#em#células#germinativas,#não#serão#transmitidas#aos#
descencentes.

52

26
10/06/20

QUESTÃO(TEEM

Em#relação#aos#genes,#MARQUE#A#INCORRETA:

a)#A#troca#de#um#nucleotídeo#do#RNAm#não#muda#obrigatoriamente#o#aminoácido#
incluído#na#proteína
b)#Mutações#nonsense são#aquelas#que#determinam#a#troca#de#nucleotídeos#na#
proteína,#porém#mantendo#as#mesmas#características#físico@químicas#do#
aminoácido
c)#Quando#ocorrem#inserções#ou#deleções#de#um#único#nucleotídeo#em#uma#
sequência#codificadora#de#um#gene,#ocorrerá#alteração#no#quadro#de#leitura#
(frameshift)
d)#Um#pseudogene#é#uma#sequência#de#nucleotídeos#semelhante#a#um#gene#
normal,#incapaz#de#codificar#uma#proteína#com#mesma#função#do#gene#referência#
expresso,#em#virtude#das#mutações#presentes#em#sua#sequência.
e)#Os#genes#estão#presentes#em#todas#as#células#do#organismo,#com#exceção#
daquelas#que#perderam#o#núcleo#durante#a#diferenciação#celular.

53

QUESTÃO(TEEM

Qual das seguintes situações mais provavelmente


determina perda da função de um gene?
Assinale a CORRETA:

a. Uma mutação missense


b. O desaparecimento de um stop códon
c. Uma troca de um T por um C na região promotora
d. Uma mutação do tipo frameshift
e. Uma mutação silenciosa

54

27
10/06/20

QUESTÃO(TEEM

Dentre os quadros abaixo, qual é causado por mutação


inativadora do receptor sensor de cálcio extracelular.
Assinale a CORRETA:

a. Hipocalcemia autossômica dominante


b. Hiperparatiroidismo terciário
c. Neoplasia endócrina múltipla tipo 1
d. Neoplasia endócrina múltipla tipo 2 A
e. Hiperparatiroidismo neonatal grave

55

QUESTÃO(TEEM

Várias doenças endócrinas tem sua origem em mutações. As mutações podem ter
efeitos diferentes na saúde dependendo onde elas ocorrem e se alteram a função
de proteínas essenciais. Assinale a alternativa onde o tipo de mutação esta
conceituado de forma incorreta:

a) Mutação missense: mudança em um par de bases do DNA resultando na substituição


de um aminoácido por outro e gerando uma proteína diferente.
b) Mutação nonsense: mudança em um par de bases do DNA, resultando em sequencia
de DNA que causa uma parada prematura da síntese daquela proteína.
c) Mutação frameshift: adição ou perda de base leva a mudanças na leitura do gene e na
codificação dos aminoácidos, resultando usualmente em uma proteína não funcional.
d) Mutação defense: mutação proposital em uma célula para causar apoptose da mesma.

56

28
10/06/20

QUESTÃO(TEEM

Em relação à Síndrome de Sensibilidade Reduzida ao Hormônio Tireoidiano, marque a


CORRETA:

a. Mutação no gene TRβ; T4 livre, T3 livre e T3-reverso elevados, TSH normal a elevado;
paciente com bócio, taquicardia e déficit de atenção/hiperatividade.

b. Defeito no transportador celular do hormônio tireoidiano; T4 livre e T3 livre baixos, com


TSH e T3-reverso elevados; paciente feminino com severo retardo psicomotor.

c. Defeito no metabolismo do hormônio tireoidiano; T4 livre, T3 livre, T3-reverso e TSH


aumentados; bócio e taquicardia.

d. Defeito no transportador celular do hormônio tireoidiano; T4 livre alto e T3 livre baixo com
TSH e T3-reverso elevados; paciente masculino com severo retardo psicomotor.

REVIEWS 57

G protein-coupled receptors: mutations


and endocrine diseases
Gilbert Vassart and Sabine Costagliola
Abstract | Over the past 20 years, naturally occurring mutations that affect G protein-coupled receptors (GPCRs)
have been identified, mainly in patients with endocrine diseases. The study of loss-of-function or gain-of-function
mutations has contributed to our understanding of the pathophysiology of several diseases with classic
hypophenotypes or hyperphenotypes of the target endocrine organs, respectively. Simultaneously, study of the
mutant receptors ex vivo was instrumental in delineating the relationships between the structure and function of
these important physiological and pharmacological molecules. Now that access to the crystallographic structure
of a few GPCRs is available, the mechanics of these receptors can be studied at the atomic level. Progress in
Transportador
the fields of cell biology, molecular pharmacology and proteomics has also widened our view of GPCR functions.

REVIEWSInitially considered simply as guanine nucleotide exchange factors capable of activating G protein-dependent
regulatory cascades, GPCRs are now known to display several additional characteristics, each susceptible to
alterations by disease-causing mutations. These characteristics include functionally important basal activity
of the receptor; differential activation of various G proteins; differential activation of G protein-dependent and

G protein-coupled receptors: mutations


independent effects (biased agonism); interaction with proteins that modify receptor function; dimerization-
dependent effects; and interaction with allosteric modulators. This Review attempts to illustrate how natural
and endocrine diseases
mutations of GPCR could contribute to our understanding of these novel facets of GPCR biology.
Vassart, G. & Costagliola, S. Nat. Rev. Endocrinol. 7, 362–372 (2011); published online 8 February 2011; doi:10.1038/nrendo.2011.20
Gilbert Vassart and Sabine Costagliola
Introduction
Abstract | Over the past 20 years, naturally occurring mutations that affect G protein-coupled receptors (GPCRs)
Thebeen
have G protein-coupled
identified, mainly inreceptors (GPCRs)
patients with endocrinecomprise
diseases. Thedescribing individual diseases
study of loss-of-function in detail, this Review will
or gain-of-function
58
mutations has contributed to our understanding of the pathophysiology of several diseases with classic whether known or still
one of the largest protein families in both invertebrates illustrate how GPCR mutations,
and vertebrates.
hypophenotypes All GPCRs haveofathe
or hyperphenotypes shared
targetstructure of
endocrine organs, to be discovered,
respectively. might contribute
Simultaneously, study of theto our understand-
seven receptors
mutant transmembraneex vivo wasα instrumental
helical domains, hence the
in delineating their ing ofbetween
relationships the diverse facets ofand
the structure GPCRs involved
function of in the field
otherimportant
these popularphysiological
name—7TM andreceptors. For reasons
pharmacological that Now of
molecules. thatendocrinology.
access to the crystallographic structure
ofare unclear,
a few GPCRsthe three-dimensional
is available, structure
the mechanics achieved
of these receptors bycan be studied at the atomic level. Progress in
insertion
the fields ofof cellseven α helices
biology, into
molecular a cell membrane
pharmacology seems has
and proteomics ThealsoGwidened
protein-coupled
our view of GPCRreceptors
functions.
to be particularly
Initially considered simply suited
as to the formation
guanine nucleotide of biological
exchange The classic
factors capable mode Gofprotein-dependent
of activating action of GPCRs has been defined
sensors. 7TM
regulatory cascades,proteins
GPCRssimilar to known
are now GPCRstofirst evolved
display severalinadditional
fromcharacteristics,
dissection ofeach susceptible tocoupling activation29
the mechanism
alterations by disease-causing mutations. These
prokaryotes as bacteriorhodopsin and sensory rhodopsin characteristics include functionally important
of the β2 adrenergic receptor basalto
activity
generation of the second
of(light
the receptor; differential activation of various
sensors). The family of GPCR molecules then
1 G proteins; differential activation of G protein-dependent
messenger cyclic AMP (cAMP) (Figure 1). and 7,8
The acti-
independent effects (biased agonism); interaction
diversified, probably by convergent evolution, into sensors with proteins that modify receptor function; dimerization-
vated receptors function as guanyl nucleotide exchange
that kept the wild-type
■ G protein-coupled receptors (GPCRs) are the largest family of transmembrane
ral data obtained from
receptors
cted to mimic the active
■ GPCRs are key factors in endocrinology, as they are the main sensors of the
activation that involves 10/06/20
internal environment
ons of the ligand-binding
■ Hereditary and congenital forms of classic endocrine diseases that display
ociated with a dramatic
hypophenotyes or hyperphenotypes of the target endocrine organs are
helix (TM) 6 secondary
c lock. The result of this
cavity between the cyto-
REVIEWS attributable to loss-of-function or gain-of-function mutations of GPCRs,
respectively
■ In addition to their canonical role as guanine nucleotide exchange factors,
nd TM6 enabling inter- GPCRs have a series of G protein-independent effects that might be the cause
he G protein.18,19 Crystals
d β2 adrenergic receptor G protein-coupled receptors: mutations
of many endocrine diseases
■ Endocrine phenotypes resulting from mutations that affect noncanonical
(nanobody) that behaves and endocrine diseases
functions of GPCRs remain to be identified
onfirmed and extended
Gilbert Vassart and Sabine Costagliola
onist or activating muta-
stabilizing interactions, | Over the past 20 years, naturally occurring mutations
AbstractLigands Effectorsthat affect G protein-coupled receptors (GPCRs)
rier and enabling transi- have been identified,
Sensory mainly
stimuli in
(light, patients
odor, with
taste) endocrine diseases.
Adenylyl cyclases The study(of )loss-of-function or gain-of-function
mutations ions,hasneurotransmitters,
contributed to our understanding chemokines,of the pathophysiology
Phospholipase of ()
Cβseveral diseases with classic
active to the active state, hormones, prescribed drugs K+, CIorgans,

, Na+ channels ( )Simultaneously,
GIRK ( ) study of the
hypophenotypes or hyperphenotypes of the target endocrine respectively.
with the G protein.20 mutant receptors VSCC ( )
N ex vivo was instrumental in delineating the relationships between the structure and function of
PI3 kinase ( )
or activation involved these important physiological GPCR and pharmacological molecules. Now that access to the crystallographic structure
an equilibrium from an of a few GPCRs is available, the mechanics of these receptors can be studied at the atomic level. Progress in
Extracellular
ve conformation. How-21 the fields of cell biology, molecular pharmacology and proteomics has also widened our view of GPCR functions.
Initially considered simply as guanine nucleotide Membrane
exchange factors capable of activating G protein-dependent
view, a series of concepts
regulatory cascades, GPCRs are now known to display several additional characteristics, each susceptible to
Intracellular
st be considered to fully alterations by disease-causing mutations. These γ characteristics
Off αinclude functionally important γ basal activity
α
tions in the genes that of the receptor; differential activation of various β G proteins; differential activation of G protein-dependent
β and
C On GTP
w that multiple inactive independent effects (biased agonism); interaction with proteins that modify receptor function; dimerization-
GDP
GPCRs do exist and that dependent effects; and interaction with allosteric modulators. This Review attempts to illustrate how natural
mutations of GPCR could contribute to our understanding of these cAMP novel facets of GPCR biology. K+ current
ze active conformations G proteins
G , Gq, Gi/o, G12/13 IP , DAG PI3 59 kinase
nt preferences for G pro- Vassart, G. & Costagliola, S. Nat. Rev. Endocrinol. 7,s362–372 (2011); published online38 February 2011; doi:10.1038/nrendo.2011.20

t regulatory cascades).22 FigureIntroduction


1 | Binding of its agonist to a GPCR causes a conformational change that
only in desensitization The G protein-coupled
activates the guanine receptors nucleotide (GPCRs)exchange comprise function describing
of theindividual
receptor diseases
towards in detail,
onethis of Review will
ivation, β arrestins are theone of the largest
possible protein families
interacting in both invertebrates
heterotrimeric Gαβγ proteins. illustrate Thehowresult
GPCR is mutations,
the whether known or still
and vertebrates. All GPCRs have a shared structure of to be discovered, might contribute to our understand-
ar scaffolds that control REVIEWS replacement of GDP by GTP on the α subunit, which causes its activation and the
seven transmembrane α helical domains, hence their ing of the diverse facets of GPCRs involved in the field
atory cascades.22 Biased dissociation
other popularof the βγ subunits.
name—7TM receptors. The activated
For reasons that α subunit,
of endocrinology. classically considered to
dissociate from the receptor, and the βγ subunit activate downstream effectors.
e ability of some agonists Table are unclear, the three-dimensional structure achieved by
1 | Loss-of-function mutations of G protein-coupled receptors causing endocrine diseases
These
Receptorinsertioneffectors
of seven are αDiseasenumerous
helices into a celland depend
membrane seems on theThe
Mechanism nature of theModesubunits,
G protein-coupled of inheritance for
receptors
Reference
between coupling to dif- Argine example αs and αsuited will stimulate and inhibit adenylyl cyclase, respectively; α
to be particularly to the formation of biological The classic mode of action of GPCRsq willhas been defined
vasopressin receptor 2 Nephrogenic diabetes insipidus Simple loss of function or X-linked recessive Fujiwara & Bichet 79

i
activation of G protein- Melanocortin sensors.
activate 7TM proteins
phospholipase
2 receptor similar
Familial C,towhereas
GPCRs
glucocorticoid first
deficiency βγevolved
type 1can for
constitutive desensitization
in instance
Simple from
loss dissection
of function activate of the
GIRK
Autosomal mechanism
channels
recessive coupling
Bailey and activation
et al. 60

endent cascades.22 The Luteinizing PI3prokaryotes


kinase. as
The
hormone receptor bacteriorhodopsin
intrinsic GTPase
Familial hypogonadism and sensoryactivity rhodopsin
of the of
Simpleαloss the
subunit
of β2 adrenergic
function causes receptor
therecessive
Autosomal to generation
system to & of the second
Themmen
(lightto sensors). 1
The family of GPCR
Leydig cell hypoplasia (males)
molecules thento a messenger cyclic
theAMP (cAMP) Huhtaniemi
(Figure 1).7,8 The acti-
34

to function as scaffolds, return the inactive Primary state,


amenorrheawith GDP
(females) bound subunit of reconstituted
Folliclediversified, probablySperm-related
by convergent evolution,
(males) into sensors vated receptors functionrecessive as guanyl nucleotide exchange
y or targeting effects, has Gαβγ
receptor
heterotrimer.
stimulating hormone
of both external stimuli
Notdysgenesis
illustrated
hypofertility
(such as light,
Ovarian
on this scheme
odors, pheromones
(females)
Simple loss
is the basalAutosomal
of function
activity displayed
factors (GEFs), with subsequent stimulation
Themmen
Huhtaniemi
&
by
of effectors 34

some
se effects mainly involve Gonadotropin-releasing receptors
and flavors) and
Centralthe
and internal stimuli G (for
protein-independent
hypogonadotropic example, hormones,
hypogonadism Simple effects
byofthe
loss α of
function GPCRs.
or βγ subunits
Autosomal Abbreviations:
of the G proteins.
recessive Bédécarrats &According to
GPCR,
hormone receptor
G protein-coupled receptor; GIRK, G protein-regulated inwardly rectifying
Kaiser 37

l of GPCRs. Many non- KiSS 1neuropeptides, receptor


bioactive amines and lipids, nucleotides
Central hypogonadotropic hypogonadism
their canonical mode
Simple loss of function
of action, GPCRs
Autosomal recessive de Roux
could be viewed 40

potassium;
and ions). HumansPI3, phosphatidyl
have
Central~750 GPCRs, inositol
of which3; ~300VSCC,are voltage
as ofdevices sensitive
function that transmit
calcium
Autosomalan extremely wideet al.
range of signals
l activity, which makes NK3R (TACR3) hypogonadotropic hypogonadism Simple loss recessive Tapaloglu 102

channels.
Prokineticin receptor Adapted
nonolfactory 2 in function. with
Central Thepermission
GPCRhypogonadism
hypogonadotropic family from © Vilardaga,
is divided Unknown through J.-P. et Autosomal
the cell al. J. Cell
membrane, which
recessive* 123,
Sci. Sarfati
results
et al.in activation of
regulatory cascades, in
48

into five subfamilies


4215–4220 (2010). with106little,(Kallmann
and anosmia
if any, shared
syndrome)
homology in a limited numberCodominant* of cytoplasmic regulatory cascades that
Digenic*
nate agonist. GPCRs have their
Institut de Recherche Relaxin receptor primary structure. 2
The GPCRs
Cryptorchidism in mice implicated in endo- areofcontrolled
Simple loss function byUnknown
G proteins. Feng et al. 103

r heterodimerize 25,26
anden
Interdisciplinaire
Biologie Humaine et
crine functions belong to subfamilies
Unknown in humans A (prototypes: rho- Unknown The crystallographic structure of a handful of GPCRs
dopsin and β2 adrenergic
Thyrotropin-releasing hormone
receptor), B (prototype: secretin
Central hypothyroidism
belonging to subfamily
Simple loss of function
A has been
Autosomal recessive
determined,9–12 pro-
Collu et al. 38

s have sometimes been


Moléculaire Loss-of-function
(IRIBHM), receptor mutations
Faculty of Medicine, receptor) and C (prototype: metabotropic glutamate viding templates for realistic modeling
Vassartof members of this
ernary structure. Simple
TSH receptor
loss of function
Euthyroid hyperthyrotropinemia Simple loss of function Autosomal dominant 104

Université Libre de
Bruxelles (ULB),
receptors), with the vast majority
Congenital in
hypothyroidismfamily A. subfamily in their inactive
Autosomal state.
recessive A wide range of artificial
par excellence 808 Route
in which In endocrinology,
The aim of this Review
Growth-hormone-releasing
de Lennik, hormone loss is toof GPCR
provide function
an update
Short stature (growth hormone deficiency)
on the is associated
and natural mutations
Simple loss of function
in the genes
Autosomal recessive
that encode GPCRs
Martari &
Salvatori 36

1070 Brussels,
between loss-of-function with
Ghrelin field of GPCR mutations
global hypophenotypes
receptor and
Short stature endocrine diseases,
of3–6the target which have
Loss oftissues; been studied
basal activity for Dominant* over the past 20 years.
Pantel et al. These studies
13–15
52
Belgium (G. Vassart,
has previously
4 receptor beenExtreme reviewedobesityelsewhere. Rather than led activity
to theories onCodominant
how GPCRs areSrinivasan activated; however, the
yper) phenotypes of the example,
Melanocortin
hypothyroidism, hypogonadism, short
Loss of basal
stature, et al. 49
S. Costagliola).
Parathyroid hormone and Bloomstrand chondrodysplasia direct
Simple loss structuralAutosomal
of function data to recessive
confirm or refute
Thakker et al.these theories 105

ons of the GPCRs that areto: parathyroid


Correspondence diaCompeting
betes
relatedinsipidus
protein and hypocorticism (Table 1). are
interests Benign familial hypocalciuric hypercalcemia
Theonly clini-
just becoming available. 10 The first activat-
G. Vassart Calcium-sensing receptor Simple loss of function Autosomal dominant Riccardi & Brown 45

eir target tissues are rare


gvassart@ulb.ac.be calThe aspects of these
authors declare conditions
noNeonatal
competing severeinterests. are convincingly ing
primary hyperparathyroidism explained
mutation identified in the β2 adrenergic receptor
Autosomal recessive
60
mplicit ‘brake or acceler- by a decrease in the activity of the GPCR-positive cells,
All receptors are members of subfamily A apart from parathyroid hormone receptor and parathyroid related protein receptor (subfamily B), growth-hormone-releasing hormone receptor
(subfamily B) and calcium sensor (subfamily C). *Mode of inheritance is uncertain.

ology of loss-of-function
362 | JUNE 2011 | VOLUME which7 is directly related to the severity of the mutation. As www.nature.com/nrendo
respectively, must now expected fortheloss-of-function © 2011ofMacmillan
residual activity mutations,
the alleles Publishers
present in theAllcorrespond-
Limited.
individualrights reserved
from the past 5 years on the activation mechanisms in
homozygous or compound heterozygous patients. homodimeric or heterodimeric receptors might provide
he variety of functional ing phenotypes are mainly
Loss-of-function mutationstransmitted
of GPCRs have aas autosomal
diverse an explanationor for this observation: activation would
CRs in addition to their X-linked recessive range of traits (Table 1). The severity
mechanistic consequences (Figure 2).
know that GPCRs can exist as dimers or oligomers, one
As we now induceof the asymmetry
a functional
one protomer activated.
in the dimers, with only
Loss of function of one of the
41–43

disease might mightextend over dominant-negative


expect to observe a wide range, depending
effects in protomers on(by mutation or interaction with an inverse
some heterozygous individuals. Such effects have been
described in obese patients with mutations in the gene
agonist) might even favor activation of the G protein by
the dimer.44
30
that encodes melanocortin 4 receptor (MC4R)27,28 and, The dominant transmission of benign familial hypo-
outside the endocrine field, in forms of retinitis pigmen- calciuric hypercalcemia, caused by mutation of the
OGY tosa caused by mutations in the gene that encodes rho- calcium sensor, might7be| explained
VOLUME JUNE by 2011 | 363
the unusual inverse
dopsin.29All
© 2011 Macmillan Publishers Limited. Thisrights
dominant-negative
reserved effect has been related effect of activation of this receptor on target-tissue func-
to defective routing of the complex formed between the tion (parathyroid hormone secretion). In this particu-
45
10/06/20

REVIEWS

Table 2 | Gain-of-function mutations of GPCRs causing endocrine diseases


Receptor Disease Mechanism Mode of inheritance References
Germ line mutations
Arginine vasopressin receptor 2 Nephrogenic syndrome of inappropriate Increased constitutive activity X-linked dominant Feldman
antidiuresis et al.66
Luteinizing hormone receptor Male-limited precocious puberty Increased constitutive activity Autosomal dominant Shenker73
Follicle stimulating hormone In females: spontaneous ovarian Broadening of specificity Autosomal dominant Smits et al.90
receptor hyperstimulation syndrome and constitutivity Vasseur et al.91
TSH receptor Nonautoimmune familial hyperthyroidism Increased constitutive activity Autosomal dominant Vassart104
Familial pregnancy-limited hyperthyroidism Broadening of specificity One dominant germ Rodien et al.89
line family described
KiSS 1 receptor Precocious puberty Decreased desensitization One isolated case Teles et al.78
described
Parathyroid hormone and Jansen metaphyseal chondrodysplasia Increased constitutive activity Autosomal dominant Thakker
parathyroid related protein receptors et al.105
Calcium-sensing receptor Familial hypocalcemic hypercalciuria Increased sensitivity to calcium Autosomal dominant Riccardi
(autosomal dominant hypoparathyroidism) & Brown45
Bartter syndrome type V Fully activated at physiological Autosomal dominant Vargas-Poussou
calcium concentrations et al.84
α2A-Adrenergic receptor Associated with type 2 diabetes mellitus Increased expression in islets Associated with type 2 Rosengren
controls negatively insulin secretion diabetes mellitus et al.82
Somatic mutations
Luteinizing hormone receptor Leydig cell adenomas with precocious Increased constitutive activity NA Shenker73
puberty
TSH receptor Autonomous thyroid adenomas Increased constitutive activity NA Vassart104
(rare carcinomas)
All receptors are members of subfamily A apart from parathyroid hormone receptor and parathyroid-related protein receptor (subfamily B) and the calcium-sensing receptor (subfamily C).
Abbreviations: GPCRs, G protein-coupled receptors; NA, not applicable.
61

hormone regulation mean that gain-of-function muta- Increased sensitivity to modulators


tions of the calcium sensor cause hypofunction of the The allosteric model for GPCR activation predicts that
parathyroid gland. constitutively active mutants would display increased
sensitivity to their normal agonists.21 Mutations of the
Increased GPCR expression calcium-sensing receptor provide a nice illustration
For GPCRs with low or absent basal activity, increased of this phenomenon: in familial hypocalcemic hyper-
receptor expression is expected to increase the sensiti- calciuria (quite a benign condition) the phenotype results
vity of the tissue to the agonist, hence to lower the steady from an increased sensitivity of the mutant receptor to
state concentration of the agonist, if the agonist–receptor plasma levels of calcium, rather than from its basal acti-
couple is part of a well-controlled chemostat. Differences vity.83 In the more severe Bartter syndrome type V, the
POLIMORFISMOS
in the strength of individual GPCR alleles might thus con-
tribute to the distribution of normal circulating agonist
receptor is almost fully active even when exposed to very
low concentrations of calcium.84
concentrations in the population. In other diseases, the contribution that increased
! A"sequência
An interesting de"DNA"nuclear"é
observation has been made in an example aproximadamente 99,5%"idên;ca
sensitivity of the mutant entre"dois
receptor to its normal agonist seres
where the GPCR involved is not implicated in a simple makes to the phenotype is difficult to appreciate, as it is
chemostat.humanos não aparentados."
The α2A-adrenergic receptor (ADRA2) is dominated by the hormone-independent effects of the
known, among many other roles, to control insulin secre- mutations and obliterated by the negative feedback.
tion.81 !In congenic strains of the
Genome@wide diabetic Goto-Kakizaki
associa;on studies (GWAS)Outside human endocrinology, an observation from
rat, an Adra2 allele has been identified that is associated a study published in 2010 is worth mentioning as it is
with overexpression of the receptor and reduced insulin compatible with the existence of mutations unmasking
! Ocorrem"em"exons
production e"introns
by isolated islets of Langerhans. 82
This dis- stimulation by allosteric modulators. In a genome-wide
covery led to identification in a cohort of patients with search for mutations associated with domestication of
diabetes mellitus of a homologous human ADRA2 risk- the chicken, Rubin et al. identified a single amino acid
allele, and Podem"modular"a"função"genica"e"contribuir"para"doenças"poligenicas
! demonstration that the variant receptor causes substitution in the TSH receptor as the strongest candi-
a similar in vitro phenotype when tested in human islets date.85 TSH receptor expressed in ependymal cells has a
! Iden;ficação"de"marcadores"gené;cos"
of Langerhans. However, it must be stressed that con- key role in the adaptation of birds and mammals to the
82

trary to disease-causing mutants, this ADRA2 allele is length of the day.86,87 As a consequence, it is tempting to
present in the human population as a relatively frequent hypothesize that domestication has selected an allele of
! Respostas"diferenciais"de"individuos
polymorphism (allele frequency ~15%),82 which is simply a"tratamentos"farmacologicos
the TSH receptor with increased sensitivity to an allo-
statistically associated with type 2 diabetes mellitus. steric modulator, or agonist, which would be present only

NATURE REVIEWS | ENDOCRINOLOGY VOLUME 7 | JUNE 2011 | 369


Ponto%de%corte © 2011 Macmillan Publishers Limited. All rights reserved

@ para"ser"classificado"como"polimorfismo,"o"alelo"variante"deve"existir"em"
mais"de"1%"da"população."Se"a"frequência é"menor,"denomina@se"mutação.

62

31
10/06/20

Polimorfismo)ou)Mutação?
Qual%a%diferença?

Mutação
. pode%ser%definida%como%uma%mudança%na%sequência%do%DNA%que%difere%do%normal;
. existe%um%alelo%normal%que%é%prevalente%na%população;%a%mutação%altera%este%alelo%
para%uma%variante%rara%e%anormal.%

Polimorfismo
. é%uma%variação%na%sequência%do%DNA%que%é%comum%na%população;
. neste%caso%não%há%um%alelo%padrão,%há%duas%ou%mais%alternativas%igualmente%aceitas.

63

Tipos&de&Polimorfismos
! Single'Nucleotide Polymormorphism (SNP)
! Microssatélite:;de;2;a;10;nucleotideos repetidos;em;tandem
! Minissatélite;ou;Variable Number of Tandem;Repeats (VNTRs):;10'70;nucl
! Alterações;em;sequências;de;DNA;repetitivo;disperso
Short;Interspersed;Repeat;Sequences;(SINEs):
−;Sequências;repetitivas;até;300;pb
46 Long;Interspersed;Repeat;Sequences;(LINEs):
THOMPSON &; THOMPSON GENÉTICA MÉDICA

−;Sequências;repeQQvas;até;6500;pb
TABELA 4-2 Variação Comum no Genoma Humano
Tipo de Variação Extensão do Tamanho (Aprox.) Base para o Polimorfismo Número de Alelos
Polimorfismos de 1 pb Substituição de um ou outro par de bases em uma Geralmente dois
nucieotídeo único localização específica no genoma
Inserção/deleções 1 pb a > 100 pb Simples: Presença ou ausência de um pequeno segmento Simples: 2
(indels) de DNA de 100-1.000 pb de comprimento Microssatélites:
Microssatélítes: Geralmente, uma unidade de 2, 3 tipicamente 5 ou
ou 4 nucleotídeos repetida em tandem 5-25 vezes mais
Variantes no número lOkba > 1 Mb Tipicamente a presença ou ausência de segmentos de DNA 2 ou mais
de cópias de 200 pb a 1,5 Mb, embora a duplicaçãoem tandem
de 2, 3, 4 ou mais cópias também possaocorrer
Inversões Poucos pb a > 1 Mb Um segmento de DNA presente em qualquer uma das 2
duas orientações com respeito ao DNA circundante 64
pb, par de bases;kb, par de quitobases; Mb, par de mcgabases

5 10 15 20
I I I I
Seqüência de referência ...GGATTTCT AGGT AACTGAGTCGA...

SNP
Aieioi ...GGATTTCT AGGT AACTGAGTCGA... 32
AIBI02 ...GG ATTTC0AGGT AACTC AGTCG A...
Aieid ...GG ATTTCT AGGT A ACTC AGTCG A...
Indel A
A!eio2 ...GGATTTCTAG G0T AACTGAGTCG A...
10/06/20

POLIMORFISMOS
SNP*+ single*nucleotide polymorphism

65

POLIMORFISMOS$e$MUTAÇÃO$podem$afetar$ptns

DNA$normal
Seq aa"normal PTN$normal

Troca&1&nucleotídeo&DNA
Sequencia"de"aa"alterada PTN&
anormal

Troca&1&nucleotídeo&no&DNA PTN$normal
Sem&alteração&de&aa

Troca&1&nucleotídeo&no&DNA PTN$funciona
Sequencia$de$aa$alterada normal

66

33
10/06/20

POLIMORFISMOS)
• Indels podem ser mul,alélicas,2devido a2números variáveis de2um2segmento de2DNA2
que é inserido em tandem2em um2determinado locus2genico

• trechos de2DNA2compostos por unidades de2dois,2três ou quatro nucleo>deos,2como


TGTGTG,2CAACAACAA2ou AAATAAATAAAT,2repe,dos entre2uma e2algumas dúzias de2
vezes em um2local2específico no2genoma

• Um2locus2de2micro/2minissatélite freqüentemente possui muitos alelos (n.2de2repe,ção)

“GTATACACACATATACATATATAT”2– 252repetições 67

Microsatélites
2 a 6 pb

CA repeat – mais comum

individuo 1

(CA) 13

individuo 2

(CA) 15
68

34
Aeb 2

Figura 4-2 Exemplos de polimorfismos no genoma humano maiores que SNPs. No sentido horário da
direita superior: O locus de microssatélite possui três alelos, com quatro, cinco ou seis cópias de uma
repetição trinucleotídica CAA. O polimorfismo de inversão possui dois alelos correspondentes às duas
10/06/20
orientações (indicados pelas setas) do segmento genômico mostrado em verde; tais inversões podem
envolver regiões deaté muitas mcgabascs de DNA. Asvariantes de número decópias envolvem deleção ou
duplicação decentenas de pares dequilobases até mais de uma megabase de DNA genômico. No exemplo
mostrado, o alelo 1 contém uma cópia única, enquanto o alelo 2 contém três cópias do segmento cromos-
sômicoque contémos genesFe G; outros alelos possíveis com zero, duas, quatro ou maiscópiasde F e G
não são mostrados. O polimorfirsmo de inserção por elemento móvel possui dois alelos, um com e outro
sem inserção de um retroelemento repetido LINE deaproximadamente 6 kb;a inserção do elemento móvel
altera o espaçamento entre os dois genes e pode alterar a expressão gênica na região.
POLIMORFISMOS)* Microssatélite

Indivíduos não aparentados Membros da família

Mãe Pai Criança 1 Criança 2 Criança 3

Figura 4-3 Esquema de um marcadorde microssatélite hipotético no DNAhumano.Os alelos de tamanho


diferentes (numerados de 1 a 7)correspondem aos fragmentos de DNA genômico contendodiferentes números
de cópiasde uma repetição de microssatélites, c os seustamanhos relativos são determinados separando-os
por eietroforese em gel. O alelo mais curto (alelo 1) migra em direção à parte inferior do gel, enquanto o
alelo mais longo (alelo 7)permanece mais próximo dotopo. Àesquerda, Para este microssatélite multialélico,
cada um dos seis indivíduos não aparentados possuidois alelosdiferentes. A direita. Dentro de uma família,
a herança dos alelos pode ser seguida a partir de cada um dos pais para cada uma das três crianças.

69
de microssatélites polimórficos são conhecidas ao longo do Bureau of Investigation (FBI) nos Estados Unidos utiliza
genoma humano. atualmente uma coleção de alelos em 13 desses loci para
Os microssatélites são um grupo particularmente útil o seu painel de impressão digital de DNA. É imprová
de indels. A determinação dos alelos nos múltiplos loci de vel que dois indivíduos (exceto gêmeos monozigóticos)
microssatélites é atualmente o método de escolha para a tenham exatamente os mesmos alelos em todos os 13 loci
impressão digital de DNA (DNA fíngerprinting) utiliza para os quais o painel determinará em definitivo se duas
da para o teste de identificação. Por exemplo, o Federal amostras vieram de um mesmo indivíduo. A informação

POLIMORFISMOS)* Microssatélite

70

35
10/06/20

POLIMORFISMOS)* EXEMPLOS
CAPÍTULO 4 — DIVERSIDADE GENÉTICA HUMANA: MUTAÇÃO E POLIMORFiSMO 47

Polimorfismo de microssatélite
Polimorfismo de inserção AlelO 1 ...GGATTTlCAAlCAACAAlCÃÃlGGTAACTCAGTCGA-
de elemento móvel
Alelo 2 -..GGATTTICAAICAAICAACAACAAICAÃIGGTAACTCAGTCGA-.
Alel0 3 ...GGATTTÍCAACAACAACAACAAiGGTAACTCAGTCGA...
Aleb 1

Alelo 2
L NE

Po imorfismo de inversão

Variante de numero de copias

Alelo 1 ABCDEFGH

Aeb 2

Figura 4-2 Exemplos de polimorfismos no genoma humano maiores que SNPs. No sentido horárioda
direita superior: O locus de microssatélite possui três alelos, com quatro, cinco ou seiscópias de uma
repetição trinucleotídica CAA. O polimorfismo de inversão possui dois alelos correspondentes às duas
71
orientações (indicados pelas setas) do segmento genômico mostrado em verde; tais inversões podem
envolver regiões deaté muitas mcgabascs deDNA. Asvariantes de número decópias envolvem deleção ou
duplicação decentenas de pares dequilobases até mais de uma megabase deDNA genômico. No exemplo
mostrado, o alelo 1 contém uma cópia única, enquanto o alelo 2 contém três cópias do segmento cromos-
sômicoque contémos genesFe G; outros alelos possíveis com zero,duas, quatro ou maiscópiasde F e G
não são mostrados. O polimorfirsmo de inserção por elemento móvel possui dois alelos, um com e outro
sem inserção de umretroelemento repetido LINE deaproximadamente 6 kb;a inserção do elemento móvel
altera o espaçamento entre os dois genes e pode alterar a expressão gênica na região.

Farmacogené,ca-Farmacogenômica
Indivíduos não aparentados Membros da família

Mãe Pai Criança 1 Criança 2 Criança 3

! marcadores2
gené,cos
! efe-vidade%clínica
! METABOLIZAÇÃO
! efeitos%tóxicos%

Figura 4-3 Esquema de um marcadorde microssatélite hipotético no DNAhumano.Os alelos de tamanho


diferentes (numerados de 1 a 7)correspondem aos fragmentos de DNA genômico contendodiferentes números
de cópiasde uma repetição de microssatélites, c os seustamanhos relativos são determinados separando-os
por eietroforese em gel. O alelo mais curto (alelo 1) migra em direção à parte inferior do gel, enquanto o
alelo mais longo (alelo 7)permanece mais próximo dotopo. Àesquerda, Para este microssatélite multialélico,
cada um dos seis indivíduos não aparentados possuidois alelosdiferentes. A direita. Dentro de uma família,
a herança dos alelos pode ser seguida a partir de cada um dos pais para cada uma das três crianças.

de microssatélites polimórficos são conhecidas ao longo do Bureau of Investigation (FBI) nos Estados Unidos utiliza
genoma humano. atualmente uma coleção de alelos em 13 desses loci para
Os microssatélites são um grupo particularmente útil o seu painel de impressão digital de DNA.
72 É imprová
Issa,%Nature'Reviews'Drug'Discovery 1,%300(308,%2002
de indels. A determinação dos alelos nos múltiplos loci de vel que dois indivíduos (exceto gêmeos monozigóticos)
microssatélites é atualmente o método de escolha para a tenham exatamente os mesmos alelos em todos os 13 loci
impressão digital de DNA (DNA fíngerprinting) utiliza para os quais o painel determinará em definitivo se duas
da para o teste de identificação. Por exemplo, o Federal amostras vieram de um mesmo indivíduo. A informação

36
10/06/20

Exemplo(esquemático(das(possíveis(alterações(de(perfil(metabólico de(indivíduos(
com(diferentes(características(genéticas((polimorfismos)(em(enzimas(metabolizadoras

! dose administrada
! processos farmacociné7cos
! processos farmacodinâmicos

73
FARMACOGENÉTICA:(PRINCÍPIOS,(APLICAÇÕES(E(PERSPECTIVAS,(2006

QUESTÃO(TEEM

Em relação à variação genética gerada por polimorfismos e mutações,


assinale a INCORRETA:
SNP = single nucleotide polymorphism = polimorfismo de um único nucleotídeo
SRTP = short tandem repeat polymorphism = polimorfismo de repetições curtas
em sequência = microsatélites

a) SNPs geralmente são causadores de doença.


b) Polimorfismos são relacionados com a resposta aos medicamentos.
c) Mutações são quase sempre patológicas quando geram ganho ou perda de
função de um gene.
d) SRTPs são usados para estudos de ancestralidade e de paternidade.

74

37
10/06/20

QUESTÃO(TEEM

Em relação às doenças poligênicas, assinale a alternativa CORRETA:


a) São menos frequentes que as doenças monogênicas.
b) São determinadas quase que exclusivamente por fatores genéticos, sofrendo pouca
influência de fatores ambientais.
c) Podem ocorrer em consequência de polimorfismos presentes em indivíduos não afetados.
d) São autossômicas dominantes.

75

QUESTÃO(TEEM

Em relação aos testes genéticos, o que caracteriza os microsatelites?


a) Repetições do RNA mensageiro.
b) Região do DNA onde ocorre repetições de nucleotídeos.
c) Repetições dos microRNA.
d) Regiões não codificadoras dos genes.

76

38
10/06/20

QUESTÃO(TEEM

Em relação aos fatores capazes de alterar as concentrações e a função da 25(OH)-


D, marque a CORRETA:
(25OHD = 25(OH)-vitamina D; DBP = proteína ligadora de vitamina D)

a. Etnias caucasianas e asiáticas têm maior risco de concentrações reduzidas.


b. Salva!vidas que trabalham em praias têm grande risco de exibir concentrações
tóxicas.
c. Obesos ingerem mais vitamina D dietética, por isso têm menor risco de apresentarem
deficiência.
d. Polimorfismos da DBP que reduzem sua afinidade se acompanham de menor
concentração de 25OHD.

77

Como&que&estes&“conceitos”&aparecem&em&testes&gené6cos??

78

39
10/06/20

Como&que&estes&“conceitos”&aparecem&em&testes&gené6cos??

79

80

40
10/06/20

81

Heranças(
Gené+cas

82

41
10/06/20

Conceitos/Gerais

Genótipo e Fenótipo
O"genótipo de"uma pessoa é a"sua constituição genética."
O"fenótipo é a"expressão observável de"um"genótipo como um"caracter
morfológico,"bioquímico ou molecular."

www.icb.ufmg.br"
83

Conceitos)Gerais

Homozigoto)ou)Puro
! indivíduo(que(apresenta(alelos(
iguais(para(um(ou(mais(caracteres

Heterozigoto)ou)Híbrido
! indivíduo(que(apresenta(alelos(
diferentes(para(um(ou(mais(caracteres

84

42
10/06/20

Conceitos%Gerais
ALELO Dominante
! aquele&que&sempre&que&está&presente&se&manifesta

Dominância&→&Propriedade&de&um&alelo&(dominante)&de&produzir&o&mesmo&
fenó>po&tanto&em&condição&homozigó>ca&quanto&heterozigó>ca.&

CoDdominância&→&Propriedade&do&alelo&de&um&gene&
expressarDse&sem&encobrir&ou&mesmo&mesclar&sua&
expressão&com&a&de&seu&outro&alelo,&em&indivíduos&
heterozigó>cos.&! Sistema&ABO

ALELO%Recessivo
! aquele&que&só&se&manifesta&na&ausência&do&
dominante;&&fenótipo&mascarado&pelo&gene&
dominante
85

Conceitos)Gerais
Penetrância
! É,a,probabilidade,de,um,gene,ter,qualquer,expressão,fenotípica.

Completa ) todos,os,indivíduos,portadores,de,uma,cópia,do,gene,
dominante,ou,ser,homozigoto,para,um,gene,recessivo,
apresentam,a,determinada,característica.,Ex.,Acondroplasia
(nanismo),) completa

Incompleta / apesar,de,possuir,um,determinado,gene,,o,indivíduo,
não,manifesta,o,fenótipo.,Ex.:,Retinoblastoma ) incompleta,,pois,
20%,das,pessoas,que,possuem,o,gene,não,manifestam,a,doença,,
porém,passam,o,gene,para,geração,futura.

86

43
10/06/20

HERANÇAS&MONOGÊNICAS
Herança'biológica'determinada'por'um'gene'apenas,'apresentando'genó6pos'e'
fenó6pos'distribuídos'conforme'padrões'caracterís6cos.

Os'distúrbios'monogênicos,'denominados'mendelianos,'caracterizamFse'por'seus'
padrões'de'transmissão'nas'famílias.'A'fim'de'estabelecer'o'padrão'de'transmissão,'a'
primeira'etapa'é'obter'informações'sobre'a'história'familiar'do'paciente'e'resumir'os'
detalhes'na'forma'de'um'heredograma,'por'meio'de'sinais'e'símbolos'padronizados.'

Tipos&de&Herança&
Monogênica: Autossomos
Herança'Autossômica'Dominante' cromossomos'não sexuais
Herança'Autossômica'Recessiva'
Herança'Dominante'Ligada'ao'X
Herança'Recessiva'Ligada'ao'X
87

HERANÇAS(POLIGÊNICAS

Tipo%de%herança%em%que%uma%caracterís4ca%é%codificada%por%dois%ou%mais%genes,%
cujos%alelos%exercem%efeitos%cumula4vos%sobre%a%intensidade%da%caracterís4ca%
(peso,%altura,%pigmentação%da%pele...).

A%maioria%dos%distúrbios%comuns%na%idade%adulta%é%
considerada%herança%poligênica%mul4fatorial.
A%manifestação%ou%não%dos%distúrbios%mul4fatoriais%
estará%sempre%na%dependência%das%associações%entre
fatores%ambientais e%a%base%gené4ca.

88

44
10/06/20

DOENÇAS(POLIGÊNICAS

A.base.gené9ca.desses.distúrbios.é.complexa (vários.loci.com.vários.alelos)
! tendência.a.serem.recorrentes.dentro.de.família
! não.é.possível.observar.nos.heredogramas os.padrões.de.transmissão
! os.genes.não.determinam.o.aparecimento.do.distúrbio.e.sim.conferem.um
maior.grau.de.suscep9bilidade ao.desenvolvimento.do.distúrbio

Exemplos:
Doença.coronariana
Asma
Diabetes.Mellitus

89

QUESTÃO(TEEM

Em relação às doenças poligênicas, assinale a alternativa CORRETA:

a. São menos frequentes que as doenças monogênicas


b. São determinadas quase que exclusivamente por fatores genéticos, sofrendo pouca
influência de fatores ambientais
c. São determinadas por alterações genéticas semelhantes nas diversas populações
acometidas
d. Podem ocorrer em consequência de polimorfismos presentes em indivíduos não
afetados
e. O diabetes do tipo MODY é um exemplo de doença poligênica

90

45
10/06/20

Herança'Autossômica'Dominante

! Fenó%po(é(expresso(da(mesma(maneira(em(homozigotos(e(heterozigotos
! Toda(pessoa(afetada(em(um(heredograma possui(um(genitor(afetado,(que(por(sua(vez(
possui(um(genitor(afetado.
! Um(indivíduo(não(afetado(nunca transmite(a(caracterís%ca.(
! Qualquer(filho(de(genitor(afetado(tem(um(risco(de(50%(de(herdar(o(fenó%po.(
! Familiares(feno%picamente(normais(não(transmitem(o(fenó%po(para(seus(filhos.(
! Homens(e(Mulheres(têm(a(mesma(probabilidade(de(transmi%r(o(fenó%po(aos(filhos(de(
ambos(os(sexos. 91

Doença'Autossômica'Dominante

Para$a$maioria$das$doenças$autossômicas$dominantes,$o$genó4po$heterozigoto$m/+
possui$probabilidade$de$apresentar$a$doença$<100%.
Isto$ocorre$em$virtude$da$penetrância'incompleta – possuir$o$alelo$mutante$não$
significa$que$terá$o$fenó4po$da$doença.

Exemplos$de$doenças$que$mostram$penetrância$incompleta:
Hipercalcemia$Hipocalciuria$Familiar$(gene$sensor$cálcio):$± 90%
Diabetes$4po$I$(MODY):$exibe$± 30%$de$penetrância
Câncer$de$Mama:$± 85%$
92

46
10/06/20

Doença'Autossômica'Dominante
2 Biomarker Insights

Table 1. The major conditions and features of MEN (multiple endocrine neoplasia).

MEN TYPE GENE CONDITIONS (FEATURES)

MEN 1 (Wermer) MEN 1 (menin) Hyperparathyroidism (95%)


Pancreas tumors (30%-80%)
Pituitary gland tumors (30%-42%)
Rarely (facial angiofibromas, collagenomas, lipomas, meningiomas, ependymomas)

MEN2A (Sipple) RET (specially codon 634) Thyroid gland tumors (specifically medullary carcinoma) (95%)
Pheochromocytoma (tumor of the adrenal glands) (40%-50%)
Hyperparathyroidism (10%-20%)

MEN2b (multiple mucosal RET (specially codon 918) Neuromas (99%)


neuroma syndrome) Physical characteristics similar to those in people with Marfan syndrome (99%)
Thyroid gland tumors (specifically medullary carcinoma) (95%)

MEN4 CDNK1B Parathyroid and anterior pituitary tumors (possibly associated with adrenal, renal,
and reproductive organ tumors)

Figure 1. MEN1 gene in genomic location: bands according to ensemble (http://www.genecards.org/cgi-bin/carddisp.pl?gene=MEN1).

pancreatic neuroendocrine tumors with size more than 3 cm Menin can be activated in fibrinogenes via TGF-beta.54 It has
should be taken into consideration.26,27 been shown that menin inhibits gene transcription through
MEN1 disease is a consequence of the MEN1 gene mutation different chromatin-modifying enzymes or posttranscription-
93
whose genetic locus is chromosome 11q13 (Figure 1).28,29 The ally acting. What is more is G2-M phase transition stopping
role of MEN1 gene is a tumor suppressor confirmed by micros- through cyclin B2 expression.55,56 It also plays its role through
atellite analysis in cancerous tissues of MEN1 patients.30–33 JunD-mediated gene transcription and other mecha-
Germ line mutation in the MEN1 gene resulting in loss of nisms.55,57-60 More than that menin in a straight line interacts
heterozygosity (LOH) at both alleles of MEN1 in the endo- with the p65 subunit of NF-κB to repress NF-κB–dependent
crine tumor and can be extent throughout the coding region of transcription.61,62 There are some suggesting interactions with
the gene.34,35 The protein product of MEN1 comprises 610 the PTN gene as a pro-proliferative receptor in lung cancer
residues and is completely consensus from Drosophila mela- cells for the inhibition of complex 2 (PRC2) attachment to the
nogaster to humans,36 contrary to yeast or Caenorhabditis ele- PTN gene promoter in addition to enhancing the suppressive
gans, signifying its new evolutionarily origin.36 Despite the fact chromatin spot H3K27me3.41
that during mouse embryogenesis, MEN1 gene is ubiquitously Menin is able to induce posttranscriptional modification
expressed in countless tissues and organs during mouse embry- through increasing the microRNA expression like microRNA-
onic development,37–39 its role is completely restricted and tis- 26a (miR-26a) which is crucial for osteoblastic differentia-
sue specific in the way that even exhibiting contrasting function tion.63 More than nuclear localization of menin, it is present in
between different organs.38 In endocrine organs, MEN1 sup- cytoplasm or even extracellular spaces suggesting that it has
presses tumorigenesis in some organs, such as lung, prostate, additional role in control of multiple signaling pathways, rang-
and breast, and it makes worse diabetes in mouse models.40–46 ing from Ras to Akt to Hedgehog signaling (Figure 2). It was
Interestingly, there are some reports over the role of MEN1 shown that with supporting of the transforming growth
function of further organs such as liver and bone.47–50 In mouse factor type β signaling pathway, cell proliferation inhibition
mesenchymal and osteoblastic cells, it is related to β-catenin, removed.65 For Wnt signaling and glyco-kipoprotein emission
cell-cell adhesion, and gene transcription factor, which is essen- the trascriptional co-activator btea-caten controls homeostasis
tial for osteoblast differentiation.49 Also, MEN1 protein in embryonic and adult development.66 Menin holds back
(menin) is considered to maintain bone morphogenetic protein extracellular regulated protein kinase-1/2 (ERK-1/2) mitogen-
2 (BMP-2), TGF-β super family of proteins, and Runt-related activated protein kinase pathway which is a downstream target in
transcription factor 2 (Runx2), resulting in mesenchymal cells Ras pathway.67–69 The correlation of menin with reduced activity
to osteoblasts differentiation.51 Overexpression of MEN1 of protein kinase Akt1 in cultured cells and mouse pancreatic
repressed the ALP activity induced by JunD. Actually, it has denoted that translocation of Akt1 to the cell membrane is
been recommended that menin destroys the maturation of inhibited by menin.70 Further studies had shown that the tran-
osteoblast, through stopping the differentiation of JunD.52,53 scription factor FOXO1 in the cytoplasm of hepatocytes

94

47
10/06/20

REVIEWS

MEN1 MEN2 (below 1%), and it has not been widely recognized as
inherent in MEN1 REF. 21. Third, the mice develop giant
hyperplasia of the pancreatic islets. This is a precursor
Pituitary Nerve (MEN2B)
stage for the monoclonal insulinoma. The hyperplastic
precursor is polyclonal by the criteria of retention of
Parathyroid the wild-type Men1 allele and retention of menin by
Thyroid C-cell immunohistology10. By contrast, a hyperplastic tumour
precursor stage has not been documented in any tissue
in patients with MEN1 REF. 22.
Bronchial carcinoid
THE RET gene and its mutations
Enteropancreatic
The rearranged during transfection (RET) protoonco-
gene is near the centromere of chromosome 10. It spans
60 kb with 21 exons and encodes a protein of approxi-
mately 1,100 amino acids. The protein is a transmem-
Adrenal chromaffin
brane receptor tyrosine kinase (RTK), termed RET23–25.
Almost all of the germline RET mutations in MEN2
Metabolic expressions
are missense; the rest are small deletions or insertions
that also preserve the RET open reading frame. MEN2
Similarities Differences is likely to be all from the RET gene, as no MEN2 fam-
• High penetrance for hormonal tumour • Distinct variants only in MEN2 ily has been identified that excludes the RET locus.
• Parathyroid adenomas in each • Unique organ grouping for each MEN The RET germline mutations characterized in MEN2
• Many good treatments available for most
are concentrated in only a small fraction of the open
symptoms
• Includes life-threatening hormone/metabolic reading frame (FIG. 2); most are in the cysteine-rich
expression (i.e. gastrin in MEN1 or catecholamines portion of the extracellular domain. Their limitation
in MEN2) that now is well managed to missense codons and their highly focused distribu-
Malignant expressions
tion are characteristic of mutations that cause a gain of
function, in this case in the RET RTK activity25. This
Similarities Differences contrasts with the loss-of-function RET mutations that
• Usually indolent • Unique organ grouping for each MEN cause many cases of familial HIRSCHSPRUNG DISEASE; such
• Cancer is potentially lethal in 30% of all carriers • Good prevention or cure for an mutations are generally of the stop-codon type and
if not cured associated cancer by early surgery in
show a much broader distribution across the RET open
MEN2 only
reading frame5,26. Hirschsprung disease, or megacolon
Figure 1 | Endocrine tumours expressed in multiple endocrine neoplasia types 1 and 2. with aganglionosis of the colon, is a phenotype remotely
Only the most important tumours are depicted. Similarities and differences of clinical features are reminiscent of MEN2 only insofar as either can affect
tabulated. The difference in the curability of cancers determines the central differences in clinical the intestinal nerves95 . Occasional MEN2 families have
26,27
management. A benign tumour is shown in green. One with high malignant potential is shown in
several affected members that surprisingly also express
yellow. For multiple endocrine neoplasia type 1 (MEN1), only the commonest tumours are shown.
Hirschsprung disease. Loss of function at the RET pro-
moter or its interactors has been suggested to cause the
cells results in the growth of insulinoma or of parathy- Hirschsprung part of this two-component rarity28.
roid adenoma10,16,17. However, conditional knockout
of Men1 in the hepatocyte has no effect, emphasizing RET-gene sequencing in patients. Oncogenic RET
the tissue selectivity of neoplasia from the menin-null mutations show striking and important correlations
status18. Germline heterozygosity for Men1 mutation with the variant of the MEN2 phenotype (FIGS 2,4). The
results in a normal conceptus, and these animals develop commonest phenotypic variant is termed MEN2A;
PENETRANCE endocrine tumours after the age of 9 months10,19. The this has combinations of CCELL cancer (also termed
The frequency with which commonest tumours in these mutant mice are prolac- MEDULLARY THYROID CANCER ), pheochromocytoma
individuals who carry a given tinoma, insulinoma and parathyroid adenoma. As these and hyperparathyroidism. 90% of the RET muta-
mutation show the tumours are commonly seen in patients with MEN1, tions in MEN2A occur in only 6 cysteines in a small
Herança'Autossômica'Recessiva
manifestations associated with
that mutation. If the penetrance
of a disease allele is 100%, then
this is an excellent model of MEN1. Three important
differences between mouse and human MEN1 have
25-amino-acid domain in the extracellular region1.
MEN2B is a distinct variant in which C-cell cancer
all individuals carrying that been informative. First, the prevalence of MEN1- begins at a much earlier age and is more aggres-
allele will express the associated associated gastrinoma is lower in the mouse (0−10%) sive than in MEN2A. Almost all RET mutations in
phenotype.
than in man (40%); this is unexplained, other than by MEN2B are confined to one cytoplasmic amino
PHEOCHROMOCYTOMA species or strain specificity. In this regard, three large acid — Met918Thr. A third MEN2 variant is termed
A neuroendocrine tumour that human kindreds with MEN1 also show an unusually FAMILIAL ISOLATED MEDULLARY THYROID CANCER (FMTC);
typically arises in the adrenal low (5%) and otherwise unexplained PENETRANCE for most of its heterozygous RET mutations are in the same
medulla. These tumours can be gastrinoma20. Second, the Men1-heterozygous mice amino acids as those in MEN2A. Other mutations
benign or malignant. Symptoms
often relate to the ability of these
develop PHEOCHROMOCYTOMA (7%), supporting this as a seemingly specific to FMTC are in amino acid 533 of
tumours to secrete weakly penetrant MEN1 tumour. The penetrance of the extracellular domain30 and amino acids 791−891 of
catecholamines. pheochromocytoma in human MEN1 is even lower the cytoplasmic domain1. As a modest-sized kindred

NATURE REVIEWS | C ANCER VOLUME 5 | MAY 2005 | 369


© 2005 Nature Publishing Group

GENITOR NORMAL
CARREADOR
! Não$está$presente$em$todas$as$gerações.
GENITOR NORMAL
CARREADOR

! O$risco$de$ocorrência$na$progênie$é$de$1$em$4.$

! Os$filhos$dos$indivíduos$afetados$nem$sempre$são$atingidos$

(não$é$comumente$transmitida$para$a$próxima$geração)

! Ambos$os$sexos$têm$a$mesma$probabilidade$de$serem$afetados. 96

48
10/06/20

Doença'Autossômica'Recessiva
+*alelo*normal
m*alelo*mutado

m/m

! Indivíduos*que*carregam*a*caracterís3ca*em*
heterozigose*não*desenvolvem*a*doença*
9 são*assintomá3cos.
! São*considerados*carreadores.

97

Herança'Dominante'Ligada'ao'X

Interprete este heredograma:

! Homens'afetados'não'transmitem'a'característica'aos'filhos.'
! Homens'afetados'tem'todas'filhas'afetadas.'
! Em'média,'metade'dos'filhos'e'das'filhas'de'mulher'heterozigota'serão'afetados.
! Há maior número de'mulheres afetadas.
98

49
10/06/20

Herança'Recessiva'Ligada'ao'X

! A"incidência"do"fenó-po"é"muito"mais"alta"em"homens"do"que"em"mulheres."
! O"gene"responsável"pela"afecção"é"transmi-do"de"um"pai"afetado para"todas"as"suas"
filhas,"mas"nunca"para"os"filhos"homens."
! Uma"mãe"heterozigota"carreadora"do"gene,"tem"50%"de"chance"de"transmi-Flo"para"
suas"filhas"e"filhos.
! Filhos"homens"que"recebem"o"gene"da"mãe"serão"doentes"(mesmo"em"heterozigose).
! As"mulheres"heterozigó-cas"não"são"afetadas,"são"carreadoras.
99

Doença'Recessiva'Ligada'ao'X

Síndrome)de)insensibilidade)(resistência))a)androgênios
Mutação)no)receptor)para)andrógenos.
Hemofilia)A
Distúrbio)recessivo)ligado)ao)X)clássico.)
Causado)por)mutações)no)gene)que)codifica)o)fator)VIII.
Distrofia)Muscular)de)Duchene)(DMD)
O)defeito)básico)é)uma)anormalidade)do)gene)estrutural)da)proteína)distrofina)
causando)níveis)nulos)ou)bastante)reduzidos)de)distrofina)no)músculo.)
100
hMp://www.nlm.nih.gov/MEDLINEPLUS/ency/imagepages/19095.htm

50
10/06/20

Padrões(não,clássicos de(herança
Herança Mitocondrial
! É"caracterizado"por"uma"Herança"Materna.
! A"mãe"transmite"seu"DNA"a"toda"prole."Suas"filhas"o"transmitem,"mas"seus"filhos"não.
! O"ovócito"é"bem"suprido"de"mitocôndrias,"mas"o"espermatozóide"contém"poucas"e"
mesmo"essas"poucas"não"persistem"na"progênie.

101

The&role&of&the&mitochondrial&genome&in&energy&genera:on

a This%highlights%the%importance%of%the%mitochondrial%genome%in%contribu5ng%polypep5de%subunits%to%the%five%enzyme%complexes%that%
comprise%the%oxida5ve%phosphoryla5on%(OXPHOS)%system%within%the%inner%mitochondrial%membrane%— the%site%of%ATP%synthesis.%The%
reoxida5on%of%reducing%equivalents%(NADH%(reduced%flavin%adenine%dinucleo5de))%that%are%produced%by%the%oxida5on%(reduced%
nico5namide%adenine%dinucleo5de)%and%FADH2 of%carbohydrates%(the%tricarboxylic%acid%(TCA)%cycle)%and%faMy%acids%(βOoxida5on)%is%
coupled%to%the%genera5on%of%an%electrochemical%gradient%across%the%inner%mitochondrial%membrane,%which%is%harnessed%by%the%ATP%
synthase%to%drive%the%forma5on%of%ATP.%

b A%map%of%the%human%mitochondrial%genome.%The&genes&that&encode&the&subunits&of&complex&I (ND1–ND6 and%ND4L)%are%shown%in%


blue;%cytochrome&c&oxidase&(COI–COIII)%is%shown%in%red;%cytochrome%b of%complex&III&is%shown%in%green;%and%the%subunits%of%the%ATP&
synthase&(ATPase16 and%8)%are%shown%in%yellow.%The%two%ribosomal%RNAs%(rRNAs;%12S and%16S,%shown%in%purple)%and%22%tRNAs,%
indicated%by%black%lines%and%denoted%by%their%single%leMer%code,%which%are%required%for%mitochondrial%protein%synthesis%are%also%shown.%
The%displacement%loop%(DOloop),%or%nonOcoding%control%region,%contains%sequences%that%are%vital%for%the%ini5a5on%of%both%mtDNA
replica5on%and%transcrip5on,%including%the%proposed%origin%of%heavyOstrand%replica5on%(shown%as%OH).%The%origin%of%lightOstrand%
102
replica5on%is%shown%as%OL.

51
10/06/20

202
Heranças gené*cas em Endocrinologia
PITUITARY PHYSIOLOGY AND DIAGNOSTIC EVALUATION

TABLE 8-6
Hereditary Pituitary Deficiency Caused by Transcription Factor Mutations*
Associated Inheritance
Gene Chromosome Pituitary Deficiency MRI Findings Malformations Mode
POU1F1† 3p11 GH, PRL, ± TSH Normal or hypoplastic anterior Recessive, dominant
pituitary
PROP1 ‡
5q35 GH, PRL, TSH, LH, FSH, ± ACTH Normal, hypoplastic, hyperplastic, Recessive
or cystic anterior pituitary
HESX1§ 3p21 GH, PRL, TSH, LH, FSH, ACTH, Hypoplastic or hyperplastic Septo-optic dysplasia Recessive
posterior defects anterior pituitary
PITX2 4q25 GH, PRL, TSH, FSH, LH Normal or ectopic posterior Rieger syndrome Dominant
pituitary
LHX3 9q34 GH, PRL, TSH, LH, FSH Hypoplastic or hyperplastic Stubby neck with rigid Recessive
anterior pituitary cervical spine
LHX4 1q25 GH, TSH, ACTH Hypoplastic anterior pituitary, Dominant
ectopic posterior pituitary
TPIT 1q23 ACTH Normal Recessive
OTX2 GH, TSH, ACTH Hypoplastic anterior pituitary, Eye malformations Dominant/Negative
ectopic posterior pituitary
SIX6 14q22 Hypoplastic pituitary, absent Brachio-otorenal and Haplo-insufficiency
chiasm oculoauriculo-vertebral
syndromes
SOX2 3q26 GH, FSH, LH Anterior pituitary hypoplasia, Anophthalmia,
midbrain defects esophageal atresia
SOX3 Xq27 GH, TSH, ACTH, FSH, LH Anterior pituitary hypoplasia, X-linked recessive
ectopic posterior pituitary
*Genes involved in pituitary development or in maintaining the integrity of the hypothalamic-pituitary axis. Functional defects include missense mutations or
frameshifts leading to truncated or deleted protein, DNA binding abnormality, inactivated protein, or impaired coactivation.

POU1F1 mutations result in varying phenotypes of early growth failure with or without hypothyroidism.

PROP1 mutations may be fully manifest only in adulthood. 103
§
HESX1 is critical for corpus development and is associated with structural brain defects.
ACTH, adrenocorticotropic hormone; FSH, follicle-stimulating hormone; GH, growth hormone; LH, luteinizing hormone; PRL, prolactin; TSH, thyroid-stimulating
hormone.
(Adapted from Netchine I, Léger J, Rappaport R. Magnetic resonance imaging of the hypothalamic-pituitary region in nontumoral hypopituitarism. In: Rappaport
R, Amselem S, eds. Hypothalamic Pituitary Development. Basel, Switzerland: Karger; 2001:94-108; and Romero CJ. Molecular basis of hypopituitarism. Trends
Endocrinol Metab. 2009;20:506.)

ethylenediaminetetra-acetic acid (EDTA) and stored below (0.3 U/kg or more) to achieve symptoms of glucopenia,
−20° C for transport. Morning (8 a.m.) plasma ACTH levels including sweating, hunger, palpitations, and tremors.364
range from 8 to 25 ng/L as measured by IRMA. Episodic Venous samples are collected at −15, 0, 15, 30, 45, 60, 90,
secretion and short plasma half-life result in wide and rapid and 120 minutes for measurement of glucose, ACTH, and
fluctuation of plasma measurements. Cortisol values at 4 cortisol levels. GH can also be measured. After the test, oral
p.m. are about half those of morning levels, and at 11 p.m. glucose should be administered. Intraindividual variations
DOENÇAS(ENDÓCRINAS(DE(ORIGEM(GENÉTICA(
levels are usually less than 5 µg/dL.
Random ACTH values do not on their own provide an
in blood glucose levels attained after a given dose of insulin,
as well as fluctuations in central sensitivity to glucose and
>"Casos Clínicos
accurate assessment of HPA function unless concurrent
cortisol levels are obtained. Therefore, an integrated assess-
activation of catecholamines, can lead to difficulties in
reproducibility. The test is contraindicated in subjects with
ment of both hormone levels is required for interpreting a history of seizures, active coronary or cerebral ischemia,
the significance of an appropriately obtained ACTH value. and pregnancy. If pronounced adrenal insufficiency is
Enzima
Often, measurement of the cortisol level alone provides a likely, insulin injection may provoke an adrenal crisis due
useful surrogate end point for assessing ACTH action and to inadequate adrenal reserve, and hydrocortisone (100 mg)
Metaboliz
HPA axis integrity. Plasma ACTH levels fluctuate broadly should be available for urgent intravenous use, if required.
within the same individual and are highly sensitive to Metyrapone blocks cortisol synthesis 104 by inhibiting
stress, time of collection, and gender. Pregnant women adrenal 11β-hydroxylase. The drug releases the HPA axis
have higher ambient ACTH levels, possibly because of pla- from negative feedback by cortisol, which normally results
cental CRH secretion.363 in an ACTH surge and elevated levels of 11-deoxycortisol
(compound S). A single oral dose (2 to 3 g) is given at
Dynamic Testing for ACTH Reserve midnight, and serum levels of ACTH, 11-deoxycortisol,
Hypothalamic Testing. Insulin hypoglycemia is a potent and cortisol are measured at 8 a.m. the following morning.
endogenous stressor that evokes ACTH secretion.222 Insulin
Proteina
The test is only valid in the face of documented suppressed
(0.1 to 0.15 U/kg) is injected intravenously after an over- cortisol levels lower than 10 µg/dL. Ligadora
In normal subjects,
night fast to achieve symptomatic hypoglycemia and a peak ACTH values higher than 200 ng/L are achieved. Side
blood glucose level of less than 40 mg/dL. This test must effects include nausea, gastrointestinal upset, and insom-
be performed under supervision. A normal HPA response nia.365 False-positive results may be obtained when phe-
to this stressor evokes cortisol levels higher than 20 µg/dL. nytoin is being administered because the drug prevents
Receptor(
Because hypoglycemia acts centrally, a normal response adequate enzymatic blockade. This test should be per-
implies integrity of all three tiers of HPA axis control. Up formed under observation in the hospital because acute
Citoplasma
to 20% of patients may require greater amounts of insulin adrenal insufficiency may ensue.

Receptor(
Tirosina Cinase

52
10/06/20

Esteroidogenese Adrenal

105

599
Esteroidogenese Ovariana
PHYSIOLOGY AND PATHOLOGY OF THE FEMALE REPRODUCTIVE AXIS

21 20 22
23
18 26
24
12 17 25
11 13 16 27
HO 19 C 14 D 15
Cholesterol 1 9
2 10 8
StAR 3 A 5 B 7
4 6

CH3
CYP11A1 CH3
C O
C O
Mitochondrion HSD3B2

HO O
Pregnenolone
Progesterone

CH3 CYP17A1 CH3


CYP17A1
C O C O
OH HSD3B2 OH

HO O
17-Hydroxypregnenolone 17-Hydroxyprogesterone
CYP17A1 CYP17A1
O O OH
HSD3B2 HSD17B1

HO O O
Dehydroepiandrosterone Androstenedione Testosterone
CYP19A1 CYP19A1
(aromatase) O (aromatase) OH
HSD17B1

106
HO HO
Estrone Estradiol
Figure 17-18 Steroidogenic pathway in the human ovary. The biologically active steroids progesterone and estradiol are produced primarily in the ovary of
a woman of reproductive age. Estradiol production requires the activity of six steroidogenic proteins, including StAR, and six enzymatic steps.
17-Hydroxylase/17,20-lyase, the product of the CYP17A1 gene, catalyzes two enzymatic reactions. The four rings of the cholesterol molecule and its derivative
steroids are identified by the first four letters in the alphabet, and the carbons are numbered in the sequence shown in the insert. CYP17A1, 17-hydroxylase/17,20-
lyase; CYP19A1, aromatase; HSD17B1, 17β-hydroxysteroid dehydrogenase type 1; HSD3B2, 3β-hydroxysteroid dehydrogenase-∆5,4 isomerase type 2; StAR,
steroidogenic acute regulatory protein.

active steroids is orchestrated in the follicle and corpus into the mitochondrion, which is regulated by the StAR
luteum in a cell-specific manner that is under the control protein encoded by the STAR gene (see Fig. 17-18).124 This
of LH and FSH.
Steroids formed by the ovary and other steroid-producing
movement is followed by conversion of cholesterol to preg-
nenolone, which is catalyzed by the mitochondrial side- 53
organs are derived from cholesterol (see Fig. 17-18). Several chain cleavage enzyme complex consisting of CYP11A1,
sources of cholesterol can provide the ovary with substrate adrenodoxin, and flavoprotein. LH induces steroidogenesis
for steroidogenesis, including plasma lipoprotein choles- by increasing intracellular cAMP, which increases the con-
terol, cholesterol synthesized de novo within the ovary, version of cholesterol to pregnenolone in two distinct
and cholesterol from intracellular stores of cholesterol ways: acute regulation, which occurs over minutes through
esters within lipid droplets. In the human ovary, LDL- phosphorylation of preexisting StAR and rapid synthesis of
10/06/20

“ACBiologiaCporCtrásCdoCgênero”

h%ps://www.ted.com/talks/emily_quinn_the_way_we_
think_about_biological_sex_is_wrong?language=pt

107

EPIGENÉTICA
Organismos++++++X++++Ambiente++++X+++Gené3ca

?? Meio
Interno
Meio
Externo

108

54
10/06/20

FENÓTIPO

109
Kanherkar(et(al.(Front(Cell(Dev(Biol.(2014;(2:(49

Regulação-da-diferenciação-celular

DNA

?
110

55
10/06/20

Modificações6epigenéticas

DNA

Cromatina

Nucleossomo
111

Metilação/DNA/e/modificações/em/histonas

Histonas H2A,%H2B,%
H3,%and%H4

Metilação/de/DNA
CH3

C/N:terminal/
!Metilação/(Lisina,/arginina)
!Acetilação/(Lisina)
!Fosforilação(Serina,/treonina)
Nucleossomo
!Ubiquitinação/(Lisina)
Joss$Moore(LA(and(Lane(RH((2011).(Neoreviews,(12(9),(e498($e505.
112

56
10/06/20

EPIGENÉTICA

Alterações*herdáveis*na*função gênica*que*ocorrem*sem*modificação*na*
sequência*de*nucleotídeos – “on top*of genetics”

Tipos*de*alterações* CaracterísAcas

!Me,lação2de2citosinas "Preservação*na*mitose*e*meiose

!HidroximeAlação "Herança*transgeracional*

!Ace,lação2de2histonas "Reversibilidade

!MeAlação*de*histonas

!microRNAs (Goldberg*et*al.,*2007)* 113

EPIGENÉTICA:+Metilação

! Ilhas&CpGs
! Regiões&promotoras&de&genes
! Padrões&tecido7específicos
! Padrão&célula7específico
! Recrutamento&de&deacetilases de&histonas

DNMT&1/2:&manutenção&do&padrão&de&meNlação
DNMT&3a&/3b:&meNlação&de#novo

Bird,&2002
114
Barros&&&Offenbacher,&2009

57
10/06/20

PERSPECTIVES
International Journal of Ace$lação de+histonas
Molecular Sciences
were mitochondrial protein
Reader
Review diction was made that mito
Regulatory Roles of MicroRNAs
Ac in Diabetes
Ac Ac
Ac
represent a unique environ
lation is abundant. In the sa
Acetylated
Juan Fengchromatin
1 , Wanli Xing 1,2, * and Lan Xie 1,2, *
Open and transcriptionally active
Gene On mitochondrial acetylated p
1 Medical Systems Biology Research Center, School of Medicine, Tsinghua University, Beijing 100084, China; by SIRT3 was identified —
2
fengjuansdu@126.com thetase81,82 — demonstratin
National Engineering Research Center for Beijing Biochip Technology, Beijing 102206, China
* Correspondence: wlxing@tsinghua.edu.cn (W.X.); xielan@tsinghua.edu.cn (L.X.);
role of protein acetylation i
Chromatin metabolism.
Writer (L.X.)
Tel.: +86-10-6277-3427 (W.X.); +86-10-6279-5218 Eraser
remodelling
Academic Editor: Martin Pichler Three years later, Mann
Received: 17 August 2016; Accepted: 9 October 2016; Published: 17 October 2016 used high-resolution mass
Abstract: MicroRNAs
to look deeper into the cel
Deacetylated chromatin (miRNAs), a class of endogenous small noncoding RNAs in eukaryotes,
have been recognized as significant
repressedregulators of gene expression through They identified 3,600 Lys a
Compact and transcriptionally Genepost-transcriptional
mechanisms. To date, >2000 miRNAs have been identified in the human genome, and they orchestrate 1,750 proteins83, and obser
a variety of biological and pathological processes. Disruption of miRNA levels correlates with
Figure 4 | Histone acetylation, chromatin condensation and gene expression. Acetylation targets tion was particularly prom
many diseases, including diabetes mellitus, a complex multifactorial metabolic disorder affecting
Lys residues in the amino-terminal tails of core histone proteins.pathogenesis
A string of nucleosomes is shown with macromolecular complexe
“Writers”+–
>400 HATs+–
million people histone+acetyltransferases
worldwide. miRNAs are involved in the Nature Reviewsof| Molecular
diabetes mellitus by
Cell Biology
the tails protruding when acetylated. Acetylation of the tail domains inhibits the folding of nucleo- range of cellular activities,
“Erasers”+–
affecting HDAC+–
pancreatic -cellhistone+deacetilases
functions, insulin resistance, or both. In this review, we summarize the
someinvestigations
arrays into secondary and tertiary
roles chromatin structures, with acetylation ofas
histones H2B and H4 remodelling, the cell cycle
of the regulatory of important miRNAs Nature'Reviews'Molecular'Cell'Biology
in diabetes, as well the potential of
having the greatest effect on tertiary structure formation 103–105
circulating miRNAs as diagnostic markers for diabetes mellitus. . Thus, histone tail acetylation115results in
16,"258–264"(2015)
transport and actin nuclea
chromatin decondensation, thereby allowing access to transcription factors and other transcription large number of acetylated
co-activators.
Keywords: miRNAs; diabetes; -cell function; insulin resistance; circulating miRNAs in numerous cellular proce
acetylation was finally esta
globally important PTM.
Soon
1. after, Guarente
MicroRNAs (miRNAs) and colleagues
and Diabetes example, SIRT3 (REFS 78,79)), suggested that
elegantly demonstrated that yeast Sir2 and acetylation might be more broadly distributed Targeting acetylation wit
Diabetes mellitus is a progressive metabolic disease that is characterized by high blood sugar and
mouse SIRT2
is a great proteins
threat to human are HDACs, the
health. According than had
to the World been
Health anticipated.there
Organization, Thisareprediction
currently The discovery of protein ac
activity
>400 of which
International
million is Journal
people uniquely of
suffering dependent
from diabetes worldwide, became andpartly validated
that number will inreach
2006552 by the first
million the proteins that write, eras
+ Molecular Sciences
on by
NAD 2030 [1]. Diabetes
(REF. 74) . During deacetylation,
was first reported by the ancient reported
Egyptians proteomic
nearly 3000 survey
yearsof protein
ago acetyla- groups within proteins has
[2]. In 1936,
NAD the+ distinction
is cleaved,between releasing type 1 diabetes (T1DM) and tion.
nicotinamide type 2Using
diabetes (T2DM)
a new was clearly
enrichment defined [3].
approach based cation of many novel epige
Review
Among patients diagnosed with diabetes, T1DM accounts for 5%–10% with the other ~90% having
and the ADP–ribose covalently linked to the on acetylation-specific antibodies, Zhao and The first FDA-approved ac
Regulatory
T2DM. T1DM is a form
removed acetyl group
Roles of MicroRNAs
of diabetes
(acetyl–ADP–ribose)
mellitus in which not inenough
colleagues
Diabetes insulin is produced by islet cells
identified 388 acetylation sites in ing agent was the HDAC in
in the pancreas and subsequently results in high blood sugar levels. However, the exact cause of
(BOX 1)
T1DM
Juan .Feng
Based on
1 , Wanli
is still previous work
Xing 1,2,In* and
unknown. most showing
cases,
Lan 1,2,
Xie it is* an autoimmune195 proteins
disease, a.condition
80
Thus, ininone single
which the experi-
immune (also known as suberaniloh
thatsystem
1 a gain-of-functio
Medicalmistakenly
Systems Biology
n mutation
attacks Research
of Sir2 was cellsment,
the insulin-producing
Center, School of Medicine, themore
inTsinghua
pancreas. acetylated
University,T1DM isproteins
Beijing mostlyChina;
100084,
had beenin
diagnosed (SAHA)), initially identifie
associated
children, with an increased
adolescents,
fengjuansdu@126.com or young lifespan
adults [3].in T2DM
yeast, is featured
identified withthanhigh inbloodthesugar,
preceding insulin40 years.
resistance inducing the differentiation
2
Guarente
(IR), andfurther
National relative
* Correspondence:
proposed
deficiency
Engineering Research
that
of Center
wlxing@tsinghua.edu.cn
the
insulin. depend-
for The
Beijingoccurrence
(W.X.);
Remarkably
of T2DMBeijing
Biochip Technology,
and
results unexpectedly,
from
102206, a combination
China
many of these
of genetic, cells in vitro and subsequen
environmental, and behavioral risk factors [4].xielan@tsinghua.edu.cn
Different from T1DM, (L.X.);
T2DM is often adult onset.
ency ofTel.:
Sir2 on NAD +
for its enzymatic
+86-10-6277-3427 (W.X.); +86-10-6279-5218 (L.X.) activ-
MicroRNAs (miRNAs) are short, non-coding RNAs with a size of ~22 nt [5]. In most cases,
ity enabled
Academicacts
miRNA
it to
Editor: sense theregulators
Martin Pichler
as negative
energy status of
at the post-transcriptional level by inhibiting mRNA translation or
thedegrading
cell. Indeed,
Received: 17 AugustNAD levels
2016;+ Accepted: increase
9 October when
the mRNA by complementary binding to its
2016; Published: 17 October
Box 2016
1 | The
30 -untranslated interface
region (3between
0 -UTR) via the protein
seed acetylation and metabolism
cellu lar
sequence nutrient levels
at the are
5 restrictively
0 end of the miRNA. low,
Abstract: MicroRNAs (miRNAs), a class of endogenous small noncoding RNAs in eukaryotes, 30% of
region miRNAs are estimated to affect approximately
Metabolism can influence both protein acetylation
and by changes in the cellular concent
andprotein
thisbeen
have activates
coding Sir2 [6].
genes
recognized and Another
aberrant
as significant sirtuins,
expression
regulators of miRNAs
of gene expression interferes
through with physiological
post-transcriptional
mechanisms. To date, >2000 miRNAs have been identified acetyl-coenzyme
in the human genome, A (acetyl-CoA).
and they orchestrateFor example, during fasting the relative concen
thereby transducing
pathological processes. a metabolic signal to
a variety
Sinceofthe biological
discovery andofpathological
miRNAs, an processes.
increasing increases,
Disruption
number leading
ofofmiRNA
them tobeen
levels
have an increase
correlates
foundwith in the enzymatic
involved in activity of sirtuins and the dea
various
many proteins,
diseases, including
including histones,
diabetes by adea-
mellitus, complex targets
multifactorial(see the
metabolicfigure). In
disorder contrast
affecting to kinases, the enzymatic activity of which is l
diabetes mellitus pathogenesis [7]. Dysregulation of miRNA can lead to profound impairment of
cetylating
>400 million
glucose
them
metabolism
(BOX 1)
people . At the miRNAs
worldwide.
[8]. miRNA
time ofare
expression
its involved
pro-
profiles in the pathogenesis
ofindependent
various tissues of diabetes mellitus
of(e.g.,
fluctuations
pancreas,inadipose
ATP byconcentrations, the activity of acetyltransfera
tissue,
posal, this model
affecting pancreatic that-cell
chromatin-modifying
functions, insulin resistance, or both. In this review, we summarize the
function of acetyl-CoA concentrations. When nutrient abundance increases, the
investigations of the regulatory roles of important miRNAs in diabetes, as well as the potential of
proteins sense changes in the environment concentration of acetyl-CoA increases, leading to an increase in acetyltransferas
circulating miRNAs as diagnostic markers for diabetes mellitus.
through their
Int. J. Mol. effect
Sci. 2016, on intermediary
17, 1729; metabo-
doi:10.3390/ijms17101729 target protein acetylation. www.mdpi.com/journal/ijms

litesKeywords:
was a bold idea, one
miRNAs; which
diabetes; isfunction;
-cell currently insulin resistance; circulating miRNAs
116
gaining much experimental support for ↓ Nutrients
acetylation and other chromatin PTMs
(reviewed
1. MicroRNAs in REF. 75
(miRNAs)). and Diabetes
↑ NAD + Ac C
Diabetes mellitus is a progressive metabolic disease that is characterized by high blood sugar and
Lys
Acetylation goes
is a great threat global
to human health. According to the World Health Organization, there are currently
>400 million people suffering from diabetes worldwide, and that number will reach 552 million
With the exception of tubulin, most acetylated Protein
by 2030 [1]. Diabetes was first reported by the ancient Egyptians nearly 3000 years ago [2]. In 1936,
proteins appeared
the distinction to be
between nuclear
type and(T1DM)
1 diabetes associ- Sirtuin
and type 2 diabetes (T2DM) was clearly defined [3].
Acetyltransferase
58
ated withpatients
Among transcription
diagnosedregulation. In T1DM
with diabetes, 2000,accounts for 5%–10% with the other ~90% having
Kouzarides
T2DM. T1DM wondered indiabetes
is a form of a review whether
mellitus in which not enough insulin is produced by islet cells
in the pancreas and subsequently results in high blood sugar levels. However, the exact cause of
“acetylation was a regulatory modification to
T1DM is still unknown. In most cases, it is an autoimmune disease, a condition in which the immune
Nicotinamide Lys ↑
rival phosphorylation” (REF. 76). Soon after,
10/06/20

International Journal of
Molecular Sciences

Review
Int. J. Mol. Sci. 2016, 17, 1729 4 of 12
Regulatory Roles of MicroRNAs in Diabetes
Int. J. Mol. Sci. 2016, 17, 1729 4 of 12
Juan Feng 1 , Wanli Xing 1,2, * and Lan Xie 1,2, *
1 Medical Systems Biology Research Center, School of Medicine, Tsinghua University, Beijing 100084, China;
fengjuansdu@126.com
2 National Engineering Research Center for Beijing Biochip Technology, Beijing 102206, China
* Correspondence: wlxing@tsinghua.edu.cn (W.X.); xielan@tsinghua.edu.cn (L.X.);
Tel.: +86-10-6277-3427 (W.X.); +86-10-6279-5218 (L.X.)

Academic Editor: Martin Pichler


Received: 17 August 2016; Accepted: 9 October 2016; Published: 17 October 2016
Int. J. Mol. Sci. 2016,
Abstract: 17, 1729 (miRNAs), a class of endogenous small noncoding RNAs in eukaryotes,
MicroRNAs 3 of 12
have been recognized as significant regulators of gene expression through post-transcriptional
mechanisms. To date, >2000 miRNAs have been identified in the human genome, and they orchestrate
for pancreatic -cell differentiation
a variety of biological and pathological and development
processes. Disruption of[22],
miRNAand in vitro
levels forced
correlates with expression of miR-7
many diseases, including diabetes mellitus, a complex multifactorial metabolic disorder affecting
or miR-375 helps to differentiate hPSCs into IPCs [23,24]. In addition, a number of other miRNAs,
>400 million people worldwide. miRNAs are involved in the pathogenesis of diabetes mellitus by
including miR-30d,
affecting let-7e,
pancreatic -cell miR-21, and miR-376,
miR-9,resistance,
functions, insulin or both. Inare
thisalso implicated
review, we summarize inthe
human pancreatic islet
investigations
differentiation andof the regulatory roles[25–27].
development of important miRNAs in diabetes, as well as the potential of
circulating miRNAs as diagnostic markers for diabetes mellitus.
Aside from the above, many other miRNAs are involved in pancreatic function, especially insulin
secretion (e.g., miRNAs;
Keywords: miR-375, -184, -cell
diabetes; -33,function;
-187, insulin and -30a)
-29a, resistance; [28–37].
circulating For example,117
miRNAs although miR-29 is
mentioned above in regard to its role in regulating -cell proliferation, it has also been shown in
multiple
Figurereports to protein
1. Major negatively
cascade regulate insulin
and miRNAs secretion
in insulin by directly
signaling pathway.targeting Stx-1a, a t-SNARE
Red arrow—activation; T protein
Figure 1. Major protein cascade
1. MicroRNAs (miRNAs) and
and miRNAs in insulin signaling pathway. Red arrow—activation;
Diabetes
involved in insulin
arrow—direct exocytosis
inhibition (blue);[31], andinhibition
indirect Mct1, which (black).may affect insulin
Abbreviations: secretionreceptor;
INSR—insulin [32]. miR-124a is
T arrow—direct inhibition
Diabetes mellitus
IRS—insulin (blue); PI3K—phosphoinositide
receptorissubstrate;
a progressive
indirect inhibition (black).
metabolic disease that is3-kinase;
Abbreviations:
characterized by high blood
INSR—insulin
1;sugar and
receptor;
increased in type 2 diabetic human pancreatic islets andSIRT1—sirtuin
negatively PTEN—phosphatase
regulates insulin secretion by
IRS—insulin
and receptor
is a great threat
tensin tosubstrate;
human
homolog; PI3K—phosphoinositide
health. According to the World
PIP3—phosphatidylinositol 3-kinase;
Health SIRT1—sirtuin
Organization,
3,4,5-triphosphate; 1; 2;PTEN—phosphatase
there are currently
MFN2—mitofusin AKT—AKT
directly
>400 targeting thesuffering
million people GTPase from diabetes[33]
Rab27a and Foxa2
worldwide, andlike[34],
that which contribute to -cell dysfunction in
and tensin homolog;
serine/threonine PIP3—phosphatidylinositol
kinase; ORP8—oxysterol binding protein 8;number
3,4,5-triphosphate; HNF1 will reach 552 million
MFN2—mitofusin
β—hepatocyte 2; AKT—AKT
nuclear factor 1-β;
T2DM. Taken
by 2030 [1]. together,
Diabetes wasmiRNAs
first reported and their
by the target
ancient genes
Egyptians constitute
nearly 3000 yearsaago
complex regulatory network in
[2]. In 1936,
serine/threonine kinase;
GLUT4—glucose ORP8—oxysterol
transporter 4. binding protein like 8; HNF1b—hepatocyte
the distinction between type 1 diabetes (T1DM) and type 2 diabetes (T2DM) was clearly defined [3]. nuclear factor 1-b;
pancreatic -cell functions, and the dysregulation of miRNAs may lead to diabetes mellitus.
GLUT4—glucose transporter
Among patients diagnosed 4.with diabetes, T1DM accounts for 5%–10% with the other ~90% having
3.1. Insulin
T2DM. T1DM Receptor is a (INSR)
form of diabetes mellitus in which not enough insulin is produced by islet cells
in the pancreas Table Principle microRNAs
and1.subsequently results in high(miRNAs)
blood sugarinvolved in pancreatic
levels. However, the exact cause-cell of
function.
The
T1DMligand-receptor
3.1. Insulin Receptor unknown. Ininteraction
is still(INSR) most cases, itis is the first step of
an autoimmune insulin
disease, signaling.
a condition in whichMice thelacking
immune the INSR gene
suffersystem
from hyperglycemia
mistakenly
Cell Processes and
attacksmiRNA
the hyperinsulinemia,
insulin-producing andpancreas.
cells in the
Putative Targets aCells
large number
T1DM
or Tissues is of studies
mostly
Studied diagnosed reveal
in a decrease
Species in
References
TheINSR
ligand-receptor
children,
in T2DM adolescents,
patients interaction
or[38].
miR-577
adultsisfindings
youngThese the
[3]. T2DMfirst
Fgf-21
is step
featured
support of
with
the insulin
high blood
importance signaling.
sugar,
INS-1 cells of insulinMice
INSR lacking
resistance
for maintaining
rat the INSR
insulin [11] gene
(IR),survival/apoptosis
-cell and relative deficiency of insulin. The occurrence
miR-200a/b/c - of T2DM resultsislets offrom a combination
diabetic mice of genetic,
mouse [12]
suffer from environmental,
hyperglycemia
sensitivity. Previous and
studies
and behavioral hyperinsulinemia,
also demonstrate
risk factors [4].
miR-34a
that andT1DM,
Different from
Bcl-2
miR-195a large and
T2DM
MIN-6 number
miR-15b
is often adult
cells of onset.
are studiesmouse
both reveal a decrease
increased in the
[13] in
livers of MicroRNAs
obese T2DM model animals, accompanied by the downregulation of INSR, and further
INSR in T2DM patients (miRNAs) [38]. These
miR-375 findings
are short, non-coding
Cadm1 support
RNAs the
with
islets a importance
size of
of miR-375 KO~22 ntINS-1E
mice; [5]. ofInINSR for
most cases,
cells maintaining
mouse; rat insulin
[14,15]
analysis
miRNA confirms the direct
acts as negative binding
regulators
miR-181a at theofpost-transcriptional
these
Pdgfra miRNAs level toand
3- theby 3′-UTR
inhibitingof
12-month-old ratINSR,
mRNA resulting
isletstranslation in impairment
or rat [16]
sensitivity. Previous
degrading
studiesbyalso
-cell proliferation
the mRNA
demonstrate
miR-17
complementary Meninthat
binding
miR-195 and
to its 30 -untranslated MIN-6 miR-15b
cells(30 -UTR)
region
are both increased
via the seed mouse in the[17] livers
of insulin signaling in hepatocytes
miR-24
[39,40].
Hnf1a; Neurod1 islets of obese mice mouse [19]
of obese T2DM model
sequence region animals, accompanied
at the 50 end of
miR-29a
the miRNA. miRNAs
- by are
the downregulation
estimated of INSR,
to affect approximately
INS-1E cells
30% ofand further analysis
rat [20]
protein coding genes [6]. An aberrant expression of miRNAs 0 interferes with both physiological and
confirms3.2.
the direct
Insulin binding
Receptor of
Substrate
pathological processes.
these
1/2
miR-375 miRNAs
(IRS-1/2)
HNF1bto the 3 -UTR
hPSCsof INSR,
differentiated resulting
IPCs in impairment
human of
[21]insulin
miR-7 PAX6 hPSCs differentiated IPCs human [21]
signaling inInsulin
hepatocytes
Sincereceptor [39,40].
the discovery of miRNAs,
miR-34a
substrate an increasing
1 (IRS-1) -
serves number
as of
the key them
hPSCs have been
differentiated
molecule found
in IPCs involved
the insulin in
human pathway
signaling [21]
diabetes mellitus pathogenesis [7]. Dysregulation
miR-146a - of miRNA hPSCs
can lead to profound
differentiated IPCsimpairment of
human [21]
in peripheral tissues by transmitting
-cell differentiation
glucose metabolism [8]. miRNA miR-30dexpressionthe RFX6
signals from thetissues
INSR
profiles of various hPSCs
to the downstream
differentiated
(e.g., pancreas,
enzymes.
IPCs adipose tissue,
human The IRS- [25]
3.2. Insulin Receptorlevel
1 expression Substrate
is lower 1/2 in (IRS-1/2)
let-7e
the skeletal RFX6 hPSCs differentiated
muscle of obese-type T2DMIPCs mice and humans, human indicating [25]
miR-21 SOX6; RBJ pancreatic progenitor cells human [26]
that the downregulation ofmiR-9 IRS-1 is associated - with IR and T2DM.
human In saturated fattyhuman
fetal islets acids and high [27]
InsulinInt.
receptor substrate
J. Mol. Sci. 2016, 1 (IRS-1) serves
17, 1729; doi:10.3390/ijms17101729
-
as the key molecule in the insulin signaling pathway
human fetalwww.mdpi.com/journal/ijms
islets
in
fat diet-induced insulin-resistant miR-376 L6 myocytes, miR-29a expression is increased andhuman represses IRS-1 [27]
peripheral tissues by transmitting
expression via the direct targeting miR-375 the signals
of theMtpn from the INSR
3′-UTR of IRS-1 [41]. to the
MIN-6 cells downstream
Similarly, in another cellular enzymes.
mouse
IR model The
[28] IRS-1
miR-184 Ago 2 MIN-6 cells mouse [29]
expression
(C2C12level is lowerpretreated
myoblasts in the skeletal
miR-7a with fatty muscle
- acids),of overexpression
obese-type T2DM
-cells of diabetic micedownregulates
ofmicemiR-7 andmousehumans,IRS-1 indicating
[30]
miR-29a Stx-1a; Mct1 rat islets; mouse islets rat; mouse [31,32]
that theexpression
downregulation via a direct
Insulin secretion of IRS-1 is
interaction
miR-187
associated
through
HIPK3
with IR
binding and
to T2DM.
its 3′-UTR
human islets from T2DM patients
In saturated
[42]. In IR fatty
resulting
human
acidsfromand
[33]
high
fat diet-induced insulin-resistant L6 myocytes, miR-29a expression is increased and represses IRS-1
mitochondrial dysfunction, significant
miR-30a 2; upregulation
NeuroD1 of
rat miR-96
islets and is
INS-1 found
cells in SK-Hep1 rat cells and,[34]
miR-124 Rab27a; Foxa2 human islets; MIN-6 cells human; mouse [35,36]
consequently,
expression via the impairs insulin
direct targeting miR-33
signaling
of the through
Abca130 -UTRtargeting
of mouse
IRS-1 the 3′-UTR
[41].
islets;
of IRS-1 [43].in
Similarly,
MIN-6 cells
In maternal diet-
another
118
mouse cellular
[37] IR
induced obesity of offspring and IR in later life, miR-126 plays a negative role via targeting IRS-1 [44].
model (C2C12 myoblasts pretreated with fatty acids), overexpression of miR-7 downregulates IRS-1
Aside from the miRNAs that directly bind to IRS-1, a class of miRNAs indirectly inhibits the
3. miRNAs
expression via a direct andinteraction
Insulin Resistance through (IR) 30 -UTR
expression of IRS-1. For example, in binding
livers and to its
adipose [42]. of
tissues In diet-induced
IR resulting obesity
from mitochondrial
mice,
dysfunction, IRsignificant
refers to
overexpression of theupregulation
impaired
miR-103/107 of miR-96
cellular
negatively is found
response
regulates in signaling
to insulin
insulin SK-Hep1 andbythe cells and, Cav-1,
inability
targeting consequently,
of normal
a caveolae impairs
amounts
insulinofsignaling 0 -UTR
insulin
protein tothrough
that achieve insulin
activates targeting
normalsignaling the 3by
glucose of IRS-1
homeostasis,
stabilizing thewhich[43].isIn
interaction maternal
a hallmark
between of diet-induced
T2DM.and
caveolae obesity
In IRS-1.
this of
process,
offspring
theand
Thus, IR in
enhanced
insulin later life,
signalingmiR-103/107 miR-126
pathway levels plays
are
plays a negative
concomitant
a central role. Itrole
with is avia
a targeting
decreased
highly stability
complicated of [44].
IRS-1 network
IRS-1 [45]. that is initiated
Aside
Insulin
by insulin
frombinding
receptor
the miRNAs tosubstrate
the insulin 2 (IRS-2)
receptor
that directly
is the alternative
(INSR)
bind the substrate
toonIRS-1, cellasurface, of the INSR inbyIRS-1-deficient
class offollowed miRNAs the insulin receptor
indirectly inhibits 59
conditions. In skeletal muscle C2C12 cells, miR-135a targets Irs-2 by binding to its 3′-UTR, and this
substrate
the expression of IRS-1. For example, in livers and adipose tissues of diet-induced obesity(PI3K),
launching downstream signaling cascades including phosphoinositide 3-kinase mice,
interaction negatively regulates insulin signaling [46].
AKT serine/threonine
overexpression kinase
of miR-103/107 (AKT), and
negatively glucose transporter
regulates 4 (GLUT4).
insulin signaling This process
by targeting is illustrated
Cav-1, in
a caveolae
Figure 1. A growing body of evidence indicates that IR is associated with defects in insulin signaling.
protein that activates insulin signaling by stabilizing the interaction between caveolae and IRS-1. Thus,
10/06/20

Marcas epigenéticas(em atuação “switch”

!Permanente(ON(ou(OFF
" Estática3(especificidade(tecidual(

!Alternativa(ON/OFF
" Dinâmica3(regulação(fisiológica(desenvolvimento

!Volume(para(ON
" Moderação(de(intensidade3(programação(de(doenças
" Influências(ambientais

“Key%Points”
1" Onde'as'marcas'podem'acontecer'e'suas'consequências
2" Estabelecer'causa"consequência
119

EPIGENÉTICA:
Níveis(de(Regulação
Modificadores (enzimas)
EpigenéAcos
Transcrição
Frio

Metilação/
Acetilação Secreção
Calor

Níveis(de(regulação
epigenéAca Ação(celular

Poluentes Oferta(de(Nutrientes

120
Zang(&(Ho,(2011

60
10/06/20

NIH Public Access


Author Manuscript
Nutr Rev. Author manuscript; available in PMC 2010 February 16.

NIHEpigené(ca+&+Obesidade
Public Access
Published in final edited form as:
NIH-PA Author Manuscript

Nutr Rev. 2008 August ; 66(Suppl 1): S7–11. doi:10.1111/j.1753-4887.2008.00056.x.

Author Manuscript
The agouti mouse model:
Nutr an
Rev.epigenetic biosensor
Author manuscript; for nutritional
available in PMC 2010 February 16.
and environmental alterations
Published inon
finalthe fetal
edited epigenome
form as:
NIH-PA Author Manuscript

Nutr Rev. 2008 August ; 66(Suppl 1): S7–11. doi:10.1111/j.1753-4887.2008.00056.x.


Dana C Dolinoy
Department of Radiation Oncology, Duke University Medical Center, Durham, North Carolina,
USA

The agouti mouse model: an epigenetic biosensor for nutritional


The ability of environmental factors to shape health and disease involves epigenetic
and environmental alterations on the fetal epigenome
mechanisms that mediate gene-environment interactions. Epigenetic gene regulation
comprises the heritable changes in gene expression that occur in the absence of changes to
the DNA sequence itself. Epigenetic mechanisms include chromatin folding and attachment
to the nuclear matrix,Dana
packaging of DNA around nucleosomes, covalent modifications of
C Dolinoy
histone tails (e.g. acetylation, methylation, phosphorylation), and DNA methylation. The
Department of Radiation Oncology, Duke University Medical Center, Durham, North Carolina,
NIH-PA Author Manuscript

"AgRP influence of regulatory small RNAs and micro RNAs on gene transcription is also
USA
increasingly recognized as a key mechanism of epigenetic gene regulation.
!AgRP
Conventional gene-environment interaction studies strive to understand how individuals
The environmental
with different genotypes respond to various ability of environmental
factors and how factors to shape health and disease involves epigenetic
these responses
change over time. Such research effortsmechanisms
have highlighted that
the mediate
important gene-environment Neuropep'deo AgRP
contribution of both interactions. Epigenetic gene regulation
genetic and environmental variability in human diseases. However, it has been argued that a
comprises the heritable changes in gene expression that occur in the absence of changes to
full understanding of gene-environment interactions requires that epigenetic mechanisms be
the DNA sequence itself. Epigenetic
Antagonista MC4R
mechanisms include chromatin folding and attachment
taken into account. Therefore, the interdisciplinary field of environmental epigenomics
emphasizes the potential for nutritionalto
andthe nuclear matrix,
environmental packaging
factors to of DNA
influence fetal, adult, around nucleosomes, covalent modifications of
and transgenerational epigenetic gene regulation, resulting
histone tails (e.g.in acetylation,
numerous phenotypic
methylation, phosphorylation), and DNA methylation. The
NIH-PA Author Manuscript

consequences.1
influence of regulatory small RNAs and micro RNAs on gene transcription is also
increasingly
The viable yellow agouti (Avy) mouse model, recognized
in which coat as a iskey
color variation mechanism
correlated to of epigenetic gene regulation.
epigenetic marks established early in development, has been used to investigate the impacts Efeito Orexigênico no
of nutritional and environmental influences on the fetal epigenome
Conventional (Fig. 1A and interaction
gene-environment
wild-type murine Agouti gene encodes a paracrine signaling molecule that produces either
B). The Hipotálamo (SNC)
studies strive to understand how individuals
with different genotypes respond to various environmental factors and how these responses
black eumelanin (a) or yellow phaeomelanin (A). Both A and a transcriptions are initiated
change overpromoter
time. Such research Reduz pigmentação
efforts have highlighted the important contribution of both
NIH-PA Author Manuscript

from a developmentally regulated hair-cycle-specific in exon 2 (Fig. 1A).


Transient A expression in hair follicles genetic and environmental
during a specific variability
stage of hair growth results in in
a human diseases. However, it has been argued that a
sub-apical yellow band on each black hairfullshaft, causing the brown
understanding PeleEeEanexos
121mechanisms be
agouti coat color of interactions requires that epigenetic
of gene-environment
vy
wild-type mice.2 The A metastable epiallele resulted from the insertion of an intracisternal
taken into account. Therefore, the interdisciplinary field of environmental epigenomics
A particle (IAP) murine retrotransposon upstream of the transcription start site of the Agouti
gene (Fig. 1A).2,3 A cryptic promoter in emphasizes
the proximal the
end ofpotential forpromotes
the Avy IAP nutritional and environmental factors to influence fetal, adult,
constitutive ectopic Agouti transcriptionand
not transgenerational epigenetic
only in hair follicles, but throughoutgene regulation, resulting in numerous phenotypic
all cells,
leading to yellow fur, as well as adult-onset obesity, diabetes, and tumorigenesis.4,5
consequences.1
vy
Interestingly, CpG methylation in the A IAP correlates inversely with ectopic Agouti
expression. The degree of methylation within the 5′ IAP long terminal repeat (LTR) varies
The viable yellow agouti (Avy) mouse model, in which coat color variation is correlated to
dramatically among individual isogenic Avy/a mice, causing a wide variation in coat color
epigenetic marks established
ranging from yellow (unmethylated) to pseudoagouti (methylated) (Fig. 1B). early in development, has been used to investigate the impacts
of nutritional and environmental influences on the fetal epigenome (Fig. 1A and B). The
wild-type murine Agouti gene encodes a paracrine signaling molecule that produces either
Correspondence: DC Dolinoy, Box 3433, Duke University Medical black eumelanin
Center Durham, NC 27710(a) ordcd@duke.edu,
USA. yellow phaeomelanin
Phone: (A). Both A and a transcriptions are initiated
+1-919-684-6203, Fax: +1-919-684-5584.
NIH-PA Author Manuscript

Declaration of interest. The author declares no competing financial from


interests.a developmentally regulated hair-cycle-specific promoter in exon 2 (Fig. 1A).

“O#Caso do#gene#Agouti”
Transient A expression in hair follicles during a specific stage of hair growth results in a
sub-apical yellow band on each black hair shaft, causing the brown agouti coat color of
wild-type mice.2 The Avy metastable epiallele resulted from the insertion of an intracisternal
A particle (IAP) murine retrotransposon upstream of the transcription start site of the Agouti
gene (Fig. 1A).2,3 A cryptic promoter in the proximal end of the Avy IAP promotes
constitutive ectopic Agouti transcription not only in hair follicles, but throughout all cells,
leading to yellow fur, as well as adult-onset obesity, diabetes, and tumorigenesis.4,5
Interestingly, CpG methylation in the Avy IAP correlates inversely with ectopic Agouti
expression. The degree of methylation within the 5′ IAP long terminal repeat (LTR) varies
dramatically among individual isogenic Avy/a mice, causing a wide variation in coat color
ranging from yellow (unmethylated) to pseudoagouti (methylated) (Fig. 1B).

Correspondence: DC Dolinoy, Box 3433, Duke University Medical Center Durham, NC 27710 USA. dcd@duke.edu, Phone:
+1-919-684-6203, Fax: +1-919-684-5584.
Declaration of interest. The author declares no competing financial interests.
Gestação

NIH Public Access


Author Manuscript
Nutr Rev. Author manuscript; available in PMC 2010 February 16.
Published in final edited form as:
122
NIH-PA Author Manuscript

Nutr Rev. 2008 August ; 66(Suppl 1): S7–11. doi:10.1111/j.1753-4887.2008.00056.x.

The agouti mouse model: an epigenetic biosensor for nutritional


and environmental alterations on the fetal epigenome
Dana C Dolinoy
Department of Radiation Oncology, Duke University Medical Center, Durham, North Carolina,
USA 61
The ability of environmental factors to shape health and disease involves epigenetic
mechanisms that mediate gene-environment interactions. Epigenetic gene regulation
comprises the heritable changes in gene expression that occur in the absence of changes to
the DNA sequence itself. Epigenetic mechanisms include chromatin folding and attachment
10/06/20

Os#doadores#de#grupo#metil:#
importância#da#dieta

DNA$methylation$and$
histone$methylation$

123

REGULAÇÃO*EPIGENÉTICA*DA*ADIPOGÊNESE

Tecido Adiposo Branco


(TAB)
Cristancho+&+Lazar
Nature'Reviews'Molecular'Cell'Biology'12,+7222734+
124
(November+2011)

62
10/06/20

Epigenetics and endocrinology . A F FLEISCH and others R63

imprints in oocytes and male-specific imprints in Epigene–environment interaction


spermatozoa. The second phase of epigenetic erasure
and reprogramming occurs during pre-implantation Epigenetic dysregulation can result from environ-
when the genome, with the possible exception of mental exposures including dietary factors, physical
imprinted genes and some retrotransposons, becomes activity, social stressors, and environmental toxicants
demethylated. After implantation, DNA methylation is (Mathers et al. 2010, Alegria-Torres et al. 2011).
restored de novo and rapidly ETIOLOGIA(DA(OBESIDADE
acquires cell lineage- However, there is a paucity of human studies that
specific patterns to drive cell differentiation. This is document the causal pathway from environmental
the basis for the tissue-specific gene methylation pattern exposure to epigenetic modification to clinical
! Fatores(genéticos(/(epigenéticos
seen after birth and through adulthood. (Reik et al. outcome (Fig. 3).
2001, Shi & Wu 2009, Perera & Herbstman 2011). Folate We have chosenEpigenetics to demonstrate these
and endocrinology . A themes by
F FLEISCH and others R63
and vitamin B12 serve as exogenous ! Nutrição
methyl donors, and reviewing three broad examples where environmental
their influence on DNA methylation duringand
imprints in oocytes themale-specific
in utero epigenetics
imprints in has impacted endocrinology.
Epigene–environment These include
interaction
time period will be discussed in !
spermatozoa.
more Estilo(de(vida((sedentarismo(x
The
and reprogramming
second
detail phase
later in of epigenetic
this 1) effect
occurs during pre-implantation Epigenetic atividade)
erasureof early-life nutritional exposures on future
dysregulation can result from environ-
manuscript. when the genome, with the possible exception obesity of and mental
insulinexposures
resistance, 2) effect
including dietary of lifetime
factors, physical
The prenatal erasure and genes
reprogramming of environmental exposures such as ionizing radiationtoxicants
on
DNA methylation patterns
imprinted
! Ambiente(perinatal((Programação(metabólica)
demethylated.
makes
and some retrotransposons,
After
the in implantation,
utero time
becomes
endocrine
DNA methylation
activity,
is cancer
(Mathers
social stressors,
risk,etand
al. 2010,
and environmental
3) potential for endocrine
Alegria-Torres et al. 2011).
However, there is a paucity of human studies that
period a window of restored potentialde novo and rapidly acquires
vulnerability for cell lineage- compounds
disrupting to affect endocrine endpoints
document the causal pathway from environmental
specific patterns to drive cell differentiation. This is
epigenetic dysregulation from environmental through
the basis for the tissue-specific gene methylation pattern
epigenetic modifications. We review
exposure to epigenetic modification to clinicalavailable
exposures. This is particularly relevant
seen after birth adulthood. data
in endo-
and through (Reikandet al.suggest avenues
outcome for future research.
(Fig. 3).
crinology where there 2001, Shi & Wu 2009,evidence
is burgeoning Perera & Herbstman
that 2011). Folate We have chosen to demonstrate these themes by
and vitamin B12 serve as exogenous methyl donors, and reviewing three broad examples where environmental
the fetal environment may program adult outcomes epigenetics has impacted endocrinology. These include
their influence on DNA methylation during the in utero
such as obesity and type time 2 diabetes
period mellitusin (Law
will be discussed more detailNutritional
later in this status
1) effect ofand epigenetic
early-life changes
nutritional exposures on future
et al. 1992, Stocker et al.manuscript.
2005). obesity and insulin resistance, 2) effect of lifetime
Other potential vulnerable The windows
prenatal erasure “New%insights%into%ending%chronic%disease”
and reprogramming
for epigenetic of
Poor nutrition environmental exposures such
during pregnancy has as ionizing
been radiation on
associated
DNA methylation patterns makes the in utero time endocrine cancer risk, and 3) potential for endocrine
dysregulation that might affect endocrine systems with DNA hypomethylation in offspring,
disrupting compounds to affect endocrine125 which may
endpoints
period a window of potential vulnerability for
include puberty, during epigenetic
which timedysregulation
there is an overall result from athrough
from environmental decrease in dietary
epigenetic sources
modifications. We of methyl
review available
rapid increase in DNA turnover
exposures.andThis
cellisgrowth, as well
particularly relevantgroup donorsdata
in endo- suchandassuggest
folate, methionine,
avenues for future and choline
research.
as old age, which has been crinology where with
associated there progressive
is burgeoning evidence that
in conjunction with decreased availability of B vitamins
age-related changes in the DNA fetal environment may program adult outcomes
methylation (Bjornsson (B2,
such as obesity and type 2 diabetes mellitus (Law
B6, and B12; Mathers et al. 2010). Plasma
Nutritional status and epigenetic changes
et al. 2008; Fig. 2). et al. 1992, Stocker et al. 2005). homocysteine level, an inverse marker of folate
Other potential vulnerable windows for epigenetic Poor nutrition during pregnancy has been associated
dysregulation that might affect endocrine systems with DNA hypomethylation in offspring, which may
include puberty, during which time there is an overall result from a decrease in dietary sources of methyl
rapid increase in DNA turnover and cell growth, as well group donors such as folate, methionine, and choline
as old age, which has been associated with progressive in conjunction with decreased availability of B vitamins
age-related changes in DNA methylation (Bjornsson (B2, B6, and B12; Mathers et al. 2010). Plasma
et al. 2008; Fig. 2). homocysteine level, an inverse marker of folate
EPIGENETICS

Figure 2 Exposures that occur preconceptionally, in utero, in early life and in adult life may result in epigenetic
dysregulation. Figure 2 Exposures that occur preconceptionally, in utero, in early life and in adult life may result in epigenetic
dysregulation.
126
www.endocrinology-journals.org Journal of Molecular Endocrinology (2012) 49, R61–R67
www.endocrinology-journals.org Journal of Molecular Endocrinology (2012) 49, R61–R67

63
10/06/20

Como'a'Epigenética'Molda'a'nossa'vida
e'da'nossa'descendência???

https://www.ted.com/talks/moshe_szyf_how_early_li
fe_experience_is_written_into_dna?language=ptGbr

Por'Moshe Szyf

127

128

64

Você também pode gostar