Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Nat Commun ; 13(1): 2325, 2022 04 28.
Artigo em Inglês | MEDLINE | ID: mdl-35484123

RESUMO

During embryonic development, epithelial cell blocks called somites are periodically formed according to the segmentation clock, becoming the foundation for the segmental pattern of the vertebral column. The process of somitogenesis has recently been recapitulated with murine and human pluripotent stem cells. However, an in vitro model for human somitogenesis coupled with the segmentation clock and epithelialization is still missing. Here, we report the generation of human somitoids, organoids that periodically form pairs of epithelial somite-like structures. Somitoids display clear oscillations of the segmentation clock that coincide with the segmentation of the presomitic mesoderm. The resulting somites show anterior-posterior and apical-basal polarities. Matrigel is essential for epithelialization but dispensable for the differentiation into somite cells. The size of somites is rather constant, irrespective of the initial cell number. The amount of WNT signaling instructs the proportion of mesodermal lineages in somitoids. Somitoids provide a novel platform to study human somitogenesis.


Assuntos
Células-Tronco Pluripotentes , Somitos , Animais , Desenvolvimento Embrionário , Humanos , Mesoderma , Camundongos , Receptores Notch
2.
Science ; 367(6482)2020 03 06.
Artigo em Inglês | MEDLINE | ID: mdl-32054698

RESUMO

Sex determination of germ cells is vital to creating the sexual dichotomy of germ cell development, thereby ensuring sexual reproduction. However, the underlying mechanisms remain unclear. Here, we show that ZGLP1, a conserved transcriptional regulator with GATA-like zinc fingers, determines the oogenic fate in mice. ZGLP1 acts downstream of bone morphogenetic protein, but not retinoic acid (RA), and is essential for the oogenic program and meiotic entry. ZGLP1 overexpression induces differentiation of in vitro primordial germ cell-like cells (PGCLCs) into fetal oocytes by activating the oogenic programs repressed by Polycomb activities, whereas RA signaling contributes to oogenic program maturation and PGC program repression. Our findings elucidate the mechanism for mammalian oogenic fate determination, providing a foundation for promoting in vitro gametogenesis and reproductive medicine.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Oócitos/fisiologia , Oogênese/genética , Proteínas Repressoras/fisiologia , Diferenciação Sexual/genética , Fatores de Transcrição/fisiologia , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/fisiologia , Animais , Proteínas Morfogenéticas Ósseas/metabolismo , Feminino , Feto/citologia , Masculino , Meiose/genética , Camundongos , Camundongos Knockout , Oócitos/citologia , Proteínas do Grupo Polycomb/metabolismo , Proteínas Repressoras/genética , Processos de Determinação Sexual , Transdução de Sinais , Fatores de Transcrição/genética , Transcriptoma , Tretinoína/fisiologia
3.
Cell Stem Cell ; 21(4): 517-532.e5, 2017 10 05.
Artigo em Inglês | MEDLINE | ID: mdl-28985527

RESUMO

Germline specification underlies human reproduction and evolution, but it has proven difficult to study in humans since it occurs shortly after blastocyst implantation. This process can be modeled with human induced pluripotent stem cells (hiPSCs) by differentiating them into primordial germ cell-like cells (hPGCLCs) through an incipient mesoderm-like cell (iMeLC) state. Here, we elucidate the key transcription factors and their interactions with important signaling pathways in driving hPGCLC differentiation from iPSCs. Germline competence of iMeLCs is dictated by the duration and dosage of WNT signaling, which induces expression of EOMES to activate SOX17, a key driver of hPGCLC specification. Upon hPGCLC induction, BMP signaling activates TFAP2C in a SOX17-independent manner. SOX17 and TFAP2C then cooperatively instate an hPGCLC transcriptional program, including BLIMP1 expression. This specification program diverges from its mouse counterpart regarding key transcription factors and their hierarchies, and it provides a foundation for further study of human germ cell development.


Assuntos
Evolução Biológica , Linhagem da Célula , Células Germinativas/citologia , Células-Tronco Pluripotentes/citologia , Transdução de Sinais/genética , Transcrição Gênica , Animais , Linhagem da Célula/genética , Implantação do Embrião/genética , Embrião de Mamíferos/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Camundongos , Modelos Biológicos , Células-Tronco Pluripotentes/metabolismo , Primatas , Fatores de Transcrição/metabolismo , Transcriptoma/genética , Via de Sinalização Wnt/genética
4.
EMBO J ; 36(21): 3100-3119, 2017 11 02.
Artigo em Inglês | MEDLINE | ID: mdl-28928204

RESUMO

The mechanism for sex determination in mammalian germ cells remains unclear. Here, we reconstitute the female sex determination in mouse germ cells in vitro under a defined condition without the use of gonadal somatic cells. We show that retinoic acid (RA) and its key effector, STRA8, are not sufficient to induce the female germ-cell fate. In contrast, bone morphogenetic protein (BMP) and RA synergistically induce primordial germ cells (PGCs)/PGC-like cells (PGCLCs) derived from embryonic stem cells (ESCs) into fetal primary oocytes. The induction is characterized by entry into the meiotic prophase, occurs synchronously and recapitulates cytological and transcriptome progression in vivo faithfully. Importantly, the female germ-cell induction necessitates a proper cellular competence-most typically, DNA demethylation of relevant genes-which is observed in appropriately propagated PGCs/PGCLCs, but not in PGCs/PGCLCs immediately after induction. This provides an explanation for the differential function of BMP signaling between PGC specification and female germ-cell induction. Our findings represent a framework for a comprehensive delineation of the sex-determination pathway in mammalian germ cells, including humans.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Morfogenéticas Ósseas/farmacologia , Regulação da Expressão Gênica no Desenvolvimento , Células-Tronco Embrionárias Murinas/efeitos dos fármacos , Oócitos/efeitos dos fármacos , Tretinoína/farmacologia , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Proteínas Morfogenéticas Ósseas/genética , Proteínas Morfogenéticas Ósseas/metabolismo , Diferenciação Celular , Feminino , Feto , Perfilação da Expressão Gênica , Genes Reporter , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismo , Camundongos , Camundongos Endogâmicos ICR , Células-Tronco Embrionárias Murinas/citologia , Células-Tronco Embrionárias Murinas/metabolismo , Oócitos/citologia , Oócitos/crescimento & desenvolvimento , Oócitos/metabolismo , Fator 1 de Ligação ao Domínio I Regulador Positivo , Prófase , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Processos de Determinação Sexual , Transdução de Sinais , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
5.
Trends Biochem Sci ; 39(6): 289-98, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24811060

RESUMO

PRDM14 belongs to the PR domain-containing (PRDM) transcriptional regulators. Among the PRDM family members, PRDM14 shows specific expression in preimplantation embryos, primordial germ cells (PGCs), and embryonic stem cells (ESCs) in vitro, and accordingly plays a key role in the regulation of their pluripotency and epigenetic reprogramming, most notably, genome-wide DNA demethylation. The function of PRDM14 appears to be conserved between mice and humans, but it shows several characteristic differences between the two species. A precise understanding of the function of PRDM14 in mice and humans would shed new light on the regulation of pluripotency and the epigenome in these two species, providing a foundation for better control of stem cell fates in a broader context.


Assuntos
Metilação de DNA , Epigênese Genética , Células-Tronco Pluripotentes/citologia , Células-Tronco Pluripotentes/metabolismo , Proteínas Repressoras/metabolismo , Fatores de Transcrição/metabolismo , Animais , Proteínas de Ligação a DNA , Humanos , Camundongos , Proteínas de Ligação a RNA
6.
Nature ; 501(7466): 222-6, 2013 Sep 12.
Artigo em Inglês | MEDLINE | ID: mdl-23913270

RESUMO

The germ-cell lineage ensures the continuity of life through the generation of male and female gametes, which unite to form a totipotent zygote. We have previously demonstrated that, by using cytokines, embryonic stem cells and induced pluripotent stem cells can be induced into epiblast-like cells (EpiLCs) and then into primordial germ cell (PGC)-like cells with the capacity for both spermatogenesis and oogenesis, creating an opportunity for understanding and regulating mammalian germ-cell development in both sexes in vitro. Here we show that, without cytokines, simultaneous overexpression of three transcription factors, Blimp1 (also known as Prdm1), Prdm14 and Tfap2c (also known as AP2γ), directs EpiLCs, but not embryonic stem cells, swiftly and efficiently into a PGC state. Notably, Prdm14 alone, but not Blimp1 or Tfap2c, suffices for the induction of the PGC state in EpiLCs. The transcription-factor-induced PGC state, irrespective of the transcription factors used, reconstitutes key transcriptome and epigenetic reprogramming in PGCs, but bypasses a mesodermal program that accompanies PGC or PGC-like-cell specification by cytokines including bone morphogenetic protein 4. Notably, the transcription-factor-induced PGC-like cells contribute to spermatogenesis and fertile offspring. Our findings provide a new insight into the transcriptional logic for PGC specification, and create a foundation for the transcription-factor-based reconstitution and regulation of mammalian gametogenesis.


Assuntos
Diferenciação Celular , Linhagem da Célula , Células Germinativas/citologia , Células Germinativas/metabolismo , Fatores de Transcrição/metabolismo , Animais , Diferenciação Celular/genética , Linhagem da Célula/genética , Proteínas de Ligação a DNA , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Epigênese Genética , Feminino , Fertilidade , Perfilação da Expressão Gênica , Camadas Germinativas/citologia , Masculino , Mesoderma/citologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos ICR , Camundongos Transgênicos , Fator 1 de Ligação ao Domínio I Regulador Positivo , Proteínas de Ligação a RNA , Espermatogênese , Fator de Transcrição AP-2/genética , Fator de Transcrição AP-2/metabolismo , Fatores de Transcrição/genética
7.
J Exp Med ; 208(2): 395-407, 2011 Feb 14.
Artigo em Inglês | MEDLINE | ID: mdl-21300912

RESUMO

Stimulatory and inhibitory co-receptors play fundamental roles in the regulation of the immune system. We describe a new mouse model of spontaneous autoimmune disease. Activation-induced cytidine deaminase-linked autoimmunity (aida) mice harbor a loss-of-function mutation in the gene encoding lymphocyte activation gene 3 (LAG-3), an inhibitory co-receptor. Although LAG-3 deficiency alone did not induce autoimmunity in nonautoimmune-prone mouse strains, it induced lethal myocarditis in BALB/c mice deficient for the gene encoding the inhibitory co-receptor programmed cell death 1 (PD-1). In addition, LAG-3 deficiency alone accelerated type 1 diabetes mellitus in nonobese diabetic mice. These results demonstrate that LAG-3 acts synergistically with PD-1 and/or other immunoregulatory genes to prevent autoimmunity in mice.


Assuntos
Antígenos CD/metabolismo , Antígenos de Superfície/metabolismo , Proteínas Reguladoras de Apoptose/metabolismo , Autoimunidade/imunologia , Modelos Animais de Doenças , Ativação Linfocitária/imunologia , Animais , Antígenos CD/genética , Antígenos de Superfície/genética , Proteínas Reguladoras de Apoptose/deficiência , Proteínas Reguladoras de Apoptose/genética , Autoimunidade/genética , Primers do DNA/genética , Diabetes Mellitus Tipo 1/genética , Citometria de Fluxo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Repetições de Microssatélites/genética , Miocardite/genética , Polimorfismo de Fragmento de Restrição , Polimorfismo de Nucleotídeo Único , Receptor de Morte Celular Programada 1 , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Linfócitos T Reguladores/imunologia , Proteína do Gene 3 de Ativação de Linfócitos
8.
Int Immunol ; 22(6): 443-52, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20410257

RESUMO

The deficiency of programmed cell death 1 (PD-1, Pdcd1), a negative immuno-receptor belonging to the CD28/cytotoxic T lymphocyte antigen 4 (CTLA-4) family, can support various tissue-specific autoimmune conditions. Here, we analyzed the effect of PD-1 deficiency in MRL mice that is genetically predisposed to systemic autoimmunity. MRL-Pdcd1(-)(/-) mice developed a fatal myocarditis, which is reminiscent of CTLA-4-deficient (Ctla4(-)(/-)) mice. Massive infiltration of CD4(+) and CD8(+) T cells and myeloid cells was found in hearts of MRL-Pdcd1(-)(/-) mice concomitant with the production of high-titer auto-antibodies against cardiac myosin. In contrast to Ctla4(-)(/-) mice in which most of the CD4(+) T cells are non-specifically activated and invade various organs, T cells in the heart but not in the spleen and lymph nodes are activated in MRL-Pdcd1(-)(/-) mice, suggesting that myocarditis is mediated by antigen-specific autoimmune response. Heart infiltrating myeloid cells strongly suppressed the allogenic response of T cells in vitro, suggesting that these Mac1(+)Gr1(+) myeloid cells are phenotypically similar to myeloid suppressor cells, which can be found in tumor-bearing hosts. These findings unravel the hidden heart-specific autoimmune predisposition of MRL mice and provide MRL-Pdcd1(-)(/-) mice as a useful animal model of lymphocytic myocarditis.


Assuntos
Antígenos de Diferenciação/genética , Células Mieloides/metabolismo , Miocardite/metabolismo , Linfócitos T/metabolismo , Animais , Antígenos CD/genética , Antígenos de Diferenciação/metabolismo , Autoanticorpos/biossíntese , Autoanticorpos/genética , Autoanticorpos/imunologia , Antígeno CTLA-4 , Miosinas Cardíacas/imunologia , Predisposição Genética para Doença , Tolerância Imunológica , Inflamação , Teste de Cultura Mista de Linfócitos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos MRL lpr , Camundongos Knockout , Células Mieloides/patologia , Miocardite/genética , Miocardite/imunologia , Especificidade de Órgãos , Receptor de Morte Celular Programada 1 , Linfócitos T/patologia
9.
Int Immunol ; 21(5): 499-509, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19261693

RESUMO

The non-obese diabetic (NOD) mouse strain is prone to developing various autoimmune syndromes including type I diabetes mellitus (T1DM), sialadenitis, thyroiditis and pancreatitis. Although the genetic basis of T1DM has been extensively analyzed, genetic factors that modify the other autoimmune phenotypes are largely unknown. We have recently reported that NOD mice with anti-diabetogenic MHC haplotype (H-2(b)) and programmed cell death 1 (PD-1) deficiency (NOD.H2(b)-Pdcd1(-/-) mice) are protected from T1DM but develop various tissue-specific autoimmune diseases including peripheral neuropathy due to autoimmune neuritis, sialadenitis and gastritis. In the present study, we generated [(C57BL/6 x NOD.H2(b))(F1) x NOD-H2(b)](BC1)-Pdcd1(-/-) mice to screen non-MHC quantitative trait loci (QTLs) that modify autoimmune phenotypes other than T1DM. We identified seven QTLs for peripheral neuropathy and neuritis, one QTL for insulitis, four QTLs for gastritis, two QTLs for sialadenitis and seven QTLs for vasculitis throughout the genome and designated them as Annp loci for autoimmunity due to polymorphisms of non-MHC genes in NOD mice and PD-1 deficiency. Annp1, 5, 6 and 7 overlapped with reported loci for T1DM (Idd3, 9, 15 and 2, respectively), suggesting that these loci modify not only T1DM but also other autoimmune phenotypes. NOD allele was promotive at 9 of 14 Annp loci, while NOD allele was protective at the other loci. Half of Annp loci associated with a single phenotype, while the other seven loci associated with more than two phenotypes. These results indicate that NOD genetic background harbors various QTLs that modify autoimmune phenotypes either by organ-specific or by organ-non-specific manner.


Assuntos
Autoimunidade/genética , Predisposição Genética para Doença , Doenças do Sistema Nervoso Periférico/genética , Locos de Características Quantitativas , Animais , Antígenos de Superfície/genética , Antígenos de Superfície/imunologia , Proteínas Reguladoras de Apoptose/genética , Proteínas Reguladoras de Apoptose/imunologia , Autoimunidade/imunologia , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/imunologia , Modelos Animais de Doenças , Gastrite/genética , Gastrite/imunologia , Gastrite/patologia , Camundongos , Camundongos Endogâmicos NOD , Camundongos Knockout , Neurite (Inflamação)/genética , Neurite (Inflamação)/imunologia , Neurite (Inflamação)/patologia , Pancreatite/genética , Pancreatite/imunologia , Pancreatite/patologia , Doenças do Sistema Nervoso Periférico/patologia , Receptor de Morte Celular Programada 1 , Sialadenite/genética , Sialadenite/imunologia , Sialadenite/patologia , Vasculite/genética , Vasculite/imunologia , Vasculite/patologia
10.
Proc Natl Acad Sci U S A ; 102(33): 11823-8, 2005 Aug 16.
Artigo em Inglês | MEDLINE | ID: mdl-16087865

RESUMO

Mice deficient in programmed cell death 1 (PD-1, Pdcd1), an immunoinhibitory receptor belonging to the CD28/cytotoxic T lymphocyte-associated antigen-4 family, spontaneously develop lupus-like autoimmune disease and autoimmune dilated cardiomyopathy on C57BL/6 and BALB/c backgrounds, respectively. However, how PD-1 deficiency induces different forms of autoimmune diseases on these two strains was unknown. Here, we report that PD-1 deficiency specifically accelerates the onset and frequency of type I diabetes in NOD (nonobese diabetic) mice, with strong T helper 1 polarization of T cells infiltrating into islets. These results suggest that PD-1 deficiency accelerates autoimmune predisposition of the background strain, leading to the induction of different forms of autoimmune diseases depending on the genetic background of the strain. Using NOD-Pdcd1-/- mice as an efficient animal model of type I diabetes, we screened diabetes-susceptible loci by genetic linkage analysis. The diabetic incidence of NOD-Pdcd1-/- mice was controlled by five genetic loci, including three known recessive loci [Idd (insulin-dependent diabetes) 1, Idd17, and Idd20] and two previously unidentified dominant loci [Iddp (Idd under PD-1 deficiency) 1 and Iddp2].


Assuntos
Antígenos de Superfície/genética , Antígenos de Superfície/metabolismo , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/metabolismo , Modelos Animais de Doenças , Deleção de Genes , Animais , Proteínas Reguladoras de Apoptose , Linfócitos T CD8-Positivos/imunologia , Diabetes Mellitus Tipo 1/imunologia , Diabetes Mellitus Tipo 1/fisiopatologia , Ligação Genética , Predisposição Genética para Doença , Incidência , Ilhotas Pancreáticas/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos NOD , Camundongos Knockout , Receptor de Morte Celular Programada 1 , Células Th1/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...