Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
Mol Biol Evol ; 2022 Oct 11.
Artigo em Inglês | MEDLINE | ID: mdl-36219870

RESUMO

Changes in the epigenome can affect the phenotype without the presence of changes in the genomic sequence. Given the high identity of the human and chimpanzee genome sequences, a substantial portion of their phenotypic divergence likely arises from epigenomic differences between the two species. In this study, the transcriptome and epigenome were determined for induced pluripotent stem cells (iPSCs) generated from human and chimpanzee individuals. The transcriptome and epigenomes for trimethylated histone H3 at lysine-4 (H3K4me3) and lysine-27 (H3K27me3) showed high levels of similarity between the two species. However, there were some differences in histone modifications. Although such regions, in general, did not show significant enrichment of interspecies nucleotide variations, gains in binding motifs for pluripotency-related transcription factors, especially POU5F1 and SOX2, were frequently found in species-specific H3K4me3 regions. We also revealed that species-specific insertions of retrotransposons, including the LTR5_Hs subfamily in human and a newly identified LTR5_Pt subfamily in chimpanzee, created species-specific H3K4me3 regions associated with increased expression of nearby genes. Human iPSCs have more species-specific H3K27me3 regions, resulting in more abundant bivalent domains. Only a limited number of these species-specific H3K4me3 and H3K27me3 regions overlap with species-biased enhancers in cranial neural crest cells, suggesting that differences in the epigenetic state of developmental enhancers appear late in development. Therefore, iPSCs serve as a suitable starting material for studying evolutionary changes in epigenome dynamics during development.

2.
Differentiation ; 128: 33-42, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36265360

RESUMO

Non-human primates are important models for investigations of neural development and evolution, and the use of Japanese macaque monkeys has especially contributed to the advancement of neuroscience studies. However, these studies are restricted by the number of animals able to be evaluated and the invasiveness of the methodologies. Induced pluripotent stem cells (iPSCs) can provide an alternative strategy for investigating neural development in vitro. We have established direct neurosphere (dNS) formation cultures of primate iPSCs as an in vitro model of early neurodevelopment in primate species. Here, we used dNS formation and neuronal differentiation cultures established from Japanese macaque iPSCs (jm-iPSCs) to investigate the usefulness of these cells as an in vitro model of early neural development. Time-course analyses of developmental potency and gene expression kinetics were performed during dNS formation culture of jm-iPSCs. During a 1-week culture, jm-iPSC-derived dNSs became neurogenic by day 3 and underwent stepwise expression changes of key developmental regulators along early neural development in a similar manner to chimpanzee dNS formation previously reported. Meanwhile, a subset of genes, including CYP26A1 and NPTX1, showed differential expression propensity in Japanese macaque, chimpanzee, and human iPSC-derived dNSs. Spontaneous upregulation of NOTCH signaling-associated genes HES5 and DLL1 was also observed in neuronal differentiation cultures of Japanese macaque but not chimpanzee dNSs, possibly reflecting the earlier neurogenic competence in Japanese macaques. The use of jm-iPSCs provides an alternative approach to neurological studies of primate development. Furthermore, jm-iPSCs can be used to investigate species differences in early neural development that are key to primate evolution.


Assuntos
Células-Tronco Pluripotentes Induzidas , Animais , Macaca fuscata/genética , Macaca , Haplorrinos , Neurogênese/genética , Diferenciação Celular/genética
3.
Proteins ; 89(3): 361-370, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33146441

RESUMO

Species in the genus Macaca typically live in multimale-multifemale social groups with male macaques exhibiting some of the largest testis: body weight ratios among primates. Males are believed to experience intense levels of sperm competition. Several spermatogenesis genes are located on the Y-chromosome and, interestingly, occasional hybridization between two species has led to the introgression of the rhesus macaque (Macaca mulatta) Y-chromosome deep into the range of the long-tailed macaque (M. fascicularis). These observations have led to the prediction that the successful introgression of the rhesus Y-haplotype is due to functional differences in spermatogenesis genes compared to those of the native long-tailed Y-haplotype. We examine here four Y-chromosomal loci-RBMY, XKRY, and two nearly identical copies of CDY-and their corresponding protein sequences. The genes were surveyed in representative animals from north of, south of, and within the rhesus x long-tailed introgression zone. Our results show a series of non-synonymous amino acid substitutions present between the two Y-haplotypes. Protein structure modeling via I-TASSER revealed different folding patterns between the two species' Y-proteins, and functional predictions via TreeSAAP further reveal physicochemical differences as a result of non-synonymous substitutions. These differences inform our understanding of the evolution of primate Y-proteins involved in spermatogenesis and, in turn, have biomedical implications for human male fertility.


Assuntos
Introgressão Genética/genética , Macaca fascicularis/genética , Macaca mulatta/genética , Proteínas Nucleares , Cromossomo Y/genética , Substituição de Aminoácidos/genética , Animais , Biologia Computacional , Haplótipos/genética , Masculino , Modelos Moleculares , Proteínas Nucleares/química , Proteínas Nucleares/genética , Proteínas de Ligação a RNA/química , Proteínas de Ligação a RNA/genética , Alinhamento de Sequência , Análise de Sequência de Proteína
4.
Differentiation ; 112: 67-76, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32045848

RESUMO

To induce and maintain naïve pluripotency in mouse embryonic and induced pluripotent stem cells (ESCs/iPSCs), chemically defined N2B27 medium with PD0325901, CHIR99021, and leukemia inhibitory factor (2i/LIF) is a classic and simple condition. However, this method cannot be simply extrapolated to human ESCs/iPSCs that are principally stabilized in primed pluripotency and become primitive neuroepithelium-like cells in N2B27+2i/LIF culture. Here, we assessed iPSC reprogramming of fibroblasts from chimpanzee, our closest living relative, in N2B27+2i/LIF culture. Under this condition, chimpanzee cells formed alkaline phosphatase-positive dome-shaped colonies. The colony-forming cells could be stably expanded by serial passaging without a ROCK inhibitor. However, their gene expression was distinct from iPSCs and neuroepithelium. They expressed the OCT3/4 transgene and a subset of transcripts associated with pluripotency, mesenchymal-epithelial transition, and neural crest formation. These cells exhibited a differentiation potential into the three germ layers in vivo and in vitro. The current study demonstrated that iPSC reprogramming in N2B27+2i/LIF culture converted chimpanzee fibroblasts into a multipotent cancerous state with unique gene expression, but not fully pluripotent stem cells.


Assuntos
Diferenciação Celular/genética , Reprogramação Celular/genética , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Multipotentes/citologia , Animais , Benzamidas/farmacologia , Diferenciação Celular/efeitos dos fármacos , Reprogramação Celular/efeitos dos fármacos , Difenilamina/análogos & derivados , Difenilamina/farmacologia , Transição Epitelial-Mesenquimal/genética , Fibroblastos/citologia , Fibroblastos/efeitos dos fármacos , Camadas Germinativas/efeitos dos fármacos , Camadas Germinativas/crescimento & desenvolvimento , Humanos , Fator Inibidor de Leucemia/farmacologia , Camundongos , Células-Tronco Multipotentes/efeitos dos fármacos , Crista Neural/citologia , Pan troglodytes , Células-Tronco Pluripotentes/citologia , Células-Tronco Pluripotentes/efeitos dos fármacos , Piridinas/farmacologia , Pirimidinas/farmacologia
5.
Dev Biol ; 400(1): 43-58, 2015 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-25624265

RESUMO

Mammalian spermatogenesis has been investigated extensively in rodents and a strictly controlled developmental process has been defined at cellular and molecular levels. In comparison, primate spermatogenesis has been far less well characterized. However, important differences between primate and rodent spermatogenesis are emerging so it is not always accurate to extrapolate findings in rodents to primate systems. Here, we performed an extensive immunofluorescence study of spermatogenesis in neonatal, juvenile, and adult testes in the common marmoset (Callithrix jacchus) to determine primate-specific patterns of gene expression that underpin primate germ cell development. Initially we characterized adult spermatogonia into two main classes; mitotically active C-KIT(+)Ki67(+) cells and mitotically quiescent SALL4(+)PLZF(+)LIN28(+)DPPA4(+) cells. We then explored the expression of a set of markers, including PIWIL1/MARWI, VASA, DAZL, CLGN, RanBPM, SYCP1 and HAPRIN, during germ cell differentiation from early spermatocytes through round and elongating spermatids, and a clear program of gene expression changes was determined as development proceeded. We then examined the juvenile marmoset testis. Markers of gonocytes demonstrated two populations; one that migrates to the basal membrane where they form the SALL4(+) or C-KIT(+) spermatogonia, and another that remains in the lumen of the seminiferous tubule. This later population, historically identified as pre-spermatogonia, expressed meiotic and apoptotic markers and were eliminated because they appear to have failed to correctly migrate. Our findings provide the first platform of gene expression dynamics in adult and developing germ cells of the common marmoset. Although we have characterized a limited number of genes, these results will facilitate primate spermatogenesis research and understanding of human reproduction.


Assuntos
Callithrix/crescimento & desenvolvimento , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Espermatogênese/fisiologia , Testículo/crescimento & desenvolvimento , Animais , Apoptose/fisiologia , Western Blotting , Marcadores Genéticos/genética , Masculino , Microscopia de Fluorescência , Microscopia Imunoeletrônica , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Especificidade da Espécie , Testículo/citologia
6.
RNA ; 20(8): 1223-37, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24914035

RESUMO

Small RNAs mediate gene silencing by binding Argonaute/Piwi proteins to regulate target RNAs. Here, we describe small RNA profiling of the adult testes of Callithrix jacchus, the common marmoset. The most abundant class of small RNAs in the adult testis was piRNAs, although 353 novel miRNAs but few endo-siRNAs were also identified. MARWI, a marmoset homolog of mouse MIWI and a very abundant PIWI in adult testes, associates with piRNAs that show characteristics of mouse pachytene piRNAs. As in other mammals, most marmoset piRNAs are derived from conserved clustered regions in the genome, which are annotated as intergenic regions. However, unlike in mice, marmoset piRNA clusters are also found on the X chromosome, suggesting escape from meiotic sex chromosome inactivation by the X-linked clusters. Some of the piRNA clusters identified contain antisense-orientated pseudogenes, suggesting the possibility that pseudogene-derived piRNAs may regulate parental functional protein-coding genes. More piRNAs map to transposable element (TE) subfamilies when they have copies in piRNA clusters. In addition, the strand bias observed for piRNAs mapped to each TE subfamily correlates with the polarity of copies inserted in clusters. These findings suggest that pachytene piRNA clusters determine the abundance and strand-bias of TE-derived piRNAs, may regulate protein-coding genes via pseudogene-derived piRNAs, and may even play roles in meiosis in the adult marmoset testis.


Assuntos
Callithrix/genética , Perfilação da Expressão Gênica , Família Multigênica , RNA Interferente Pequeno/genética , Animais , Proteínas Argonautas/genética , Proteínas Argonautas/metabolismo , Sequência de Bases , Elementos de DNA Transponíveis , Expressão Gênica , Genoma , Masculino , Camundongos , Dados de Sequência Molecular , Conformação de Ácido Nucleico , Mapeamento Físico do Cromossomo , Ligação Proteica , Pseudogenes , RNA Interferente Pequeno/química , RNA Interferente Pequeno/metabolismo , RNA de Transferência/química , RNA de Transferência/metabolismo , Alinhamento de Sequência , Testículo/metabolismo
7.
Mol Reprod Dev ; 81(1): 2-19, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23996404

RESUMO

Induced pluripotent stem cells (iPSCs) are stem cells that can be artificially generated via "cellular reprogramming" using gene transduction in somatic cells. iPSCs have enormous potential in stem-cell biology as they can give rise to numerous cell lineages, including the three germ layers. An evaluation of germ-line competency by blastocyst injection or tetraploid complementation, however, is critical for determining the developmental potential of mouse iPSCs towards germ cells. Recent studies have demonstrated that primordial germ cells obtained by the in vitro differentiation of iPSCs produce functional gametes as well as healthy offspring. These findings illustrate not only that iPSCs are developmentally similar to embryonic stem cells (ESCs), but also that somatic cells from adult tissues can produce gametes in vitro, that is, if they are reprogrammed into iPSCs. In this review, we discuss past and recent advances in the in vitro differentiation of germ cells using pluripotent stem cells, with an emphasis on ESCs and iPSCs. While this field of research is still at a stage of infancy, it holds great promises for investigating the mechanisms of germ-cell development, especially in humans, and for advancing reproductive and developmental engineering technologies in the future.


Assuntos
Bioengenharia/métodos , Diferenciação Celular/fisiologia , Células-Tronco Embrionárias/citologia , Células Germinativas/crescimento & desenvolvimento , Células-Tronco Pluripotentes Induzidas/citologia , Animais , Bioengenharia/tendências , Células Germinativas/citologia , Humanos , Camundongos
8.
In Vitro Cell Dev Biol Anim ; 60(5): 555-562, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38753247

RESUMO

The comparative analysis between humans and non-human primates is an instrumental approach for elucidating the evolutional traits and disease propensity of humans. However, in primates, cross-species analyses of their developmental events have encountered constraints because of the ethical and technical limitations in available sample collection, sequential monitoring, and manipulations. In an endeavor to surmount these challenges, in recent years, induced pluripotent stem cells (iPSCs) have garnered escalating interest as an in vitro tool for cross-species analyses between humans and non-human primates. Meanwhile, compared to humans, there is less information on in vitro differentiation of non-human primate iPSCs, and their genetic diversity including subspecies may cause different eligibility to in vitro differentiation methods. Therefore, antecedent to embarking on a comparative analysis to humans, it is a prerequisite to develop the efficacious methodologies for in vitro differentiation regardless of the intraspecies genetic background in non-human primates. In this study, we executed the in vitro differentiation of cardiomyocytes from four chimpanzee iPSC lines with different subspecies and individual backgrounds. To induce cardiomyocytes from chimpanzee iPSCs, we evaluated our methodology for in vitro cardiac differentiation of human iPSCs. Eventually, with minor alterations, our cardiac differentiation method was applicable to all chimpanzee iPSC lines tested as assessed by the expression of cardiac marker genes and the beating ability. Hence, our in vitro differentiation method will advance iPSC-based research of chimpanzee cardiac development and also hold possible utility to cross-species analyses among primate species.


Assuntos
Diferenciação Celular , Células-Tronco Pluripotentes Induzidas , Miócitos Cardíacos , Pan troglodytes , Células-Tronco Pluripotentes Induzidas/citologia , Animais , Miócitos Cardíacos/citologia , Linhagem Celular , Humanos , Especificidade da Espécie
9.
In Vitro Cell Dev Biol Anim ; 60(5): 544-554, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38386235

RESUMO

As humans' closest living relatives, chimpanzees offer valuable insights into human evolution. However, technical and ethical limitations hinder investigations into the molecular and cellular foundations that distinguish chimpanzee and human traits. Recently, induced pluripotent stem cells (iPSCs) have emerged as a novel model for functional comparative studies and provided a non-invasive alternative for studying embryonic phenomena. In this study, we generated five new chimpanzee iPSC lines from peripheral blood cells and skin fibroblasts with SeV vectors carrying four reprogramming factors (human OCT3/4, SOX2, KLF4, and L-MYC) and characterized their pluripotency and differentiation potential. We also examined the expression of a human-specific non-coding RNA, HSTR1, which is predicted to be involved in human brain development. Our results show that the chimpanzee iPSCs possess pluripotent characteristics and can differentiate into various cell lineages. Moreover, we found that HSTR1 is expressed in human iPSCs and their neural derivatives but not in chimpanzee counterparts, supporting its possible role in human-specific brain development. As iPSCs are inherently variable due to genetic and epigenetic differences in donor cells or reprogramming procedures, it is essential to expand the number of chimpanzee iPSC lines to comprehensively capture the molecular and cellular properties representative of chimpanzees. Hence, our cells provide a valuable resource for investigating the function and regulation of human-specific transcripts such as HSTR1 and for understanding human evolution more generally.


Assuntos
Diferenciação Celular , Células-Tronco Pluripotentes Induzidas , Fator 4 Semelhante a Kruppel , Pan troglodytes , Animais , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/metabolismo , Diferenciação Celular/genética , Humanos , Linhagem Celular , Especificidade da Espécie , Fibroblastos/citologia , Fibroblastos/metabolismo , Reprogramação Celular/genética
10.
Reprod Med Biol ; 12(1): 1-14, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-29699125

RESUMO

In multicellular organisms, germ cells are an extremely specialized cell type with the vital function of transmitting genetic information across generations. In this respect, they are responsible for the perpetuity of species, and are separated from somatic lineages at each generation. Interestingly, in the past two decades research has shown that germ cells have the potential to proceed along two distinct pathways: gametogenesis or pluripotency. Unequivocally, the primary role of germ cells is to produce gametes, the sperm or oocyte, to produce offspring. However, under specific conditions germ cells can become pluripotent, as shown by teratoma formation in vivo or cell culture-induced reprogramming in vitro. This phenomenon seems to be a general propensity of germ cells, irrespective of developmental phase. Recent attempts at cellular reprogramming have resulted in the generation of induced pluripotent stem cells (iPSCs). In iPSCs, the intracellular molecular networks instructing pluripotency have been activated and override the exclusively somatic cell programs that existed. Because the generation of iPSCs is highly artificial and depends on gene transduction, whether the resulting machinery reflects any physiological cell-intrinsic programs is open to question. In contrast, germ cells can spontaneously shift their fate to pluripotency during in-vitro culture. Here, we review the two fates of germ cells, i.e., differentiation and reprogramming. Understanding the molecular mechanisms regulating differentiation versus reprogramming would provide invaluable insight into understanding the mechanisms of cellular reprogramming that generate iPSCs.

11.
Reproduction ; 143(5): 597-609, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22323619

RESUMO

Germ cell development is a fundamental process required to produce offspring. The developmental program of spermatogenesis has been assumed to be similar among mammals. However, recent studies have revealed differences in the molecular properties of primate germ cells compared with the well-characterized mouse germ cells. This may prevent simple application of rodent insights into higher primates. Therefore, thorough investigation of primate germ cells is necessary, as this may lead to the development of more appropriate animal models. The aim of this study is to define molecular signatures of spermatogenic cells in the common marmoset, Callithrix jacchus. Interestingly, NANOG, PRDM1, DPPA3 (STELLA), IFITM3, and ZP1 transcripts, but no POU5F1 (OCT4), were detected in adult marmoset testis. Conversely, mouse testis expressed Pou5f1 but not Nanog, Prdm1, Dppa3, Ifitm3, and Zp1. Other previously described mouse germ cell markers were conserved in marmoset and mouse testes. Intriguingly, marmoset spermatogenic cells underwent dynamic protein expression in a developmental stage-specific manner; DDX4 (VASA) protein was present in gonocytes, diminished in spermatogonial cells, and reexpressed in spermatocytes. To investigate epigenetic differences between adult marmoset and mice, DNA methylation analyses identified unique epigenetic profiles to marmoset and mice. Marmoset NANOG and POU5F1 promoters in spermatogenic cells exhibited a methylation status opposite to that in mice, while the DDX4 and LEFTY1 loci, as well as imprinted genes, displayed an evolutionarily conserved methylation pattern. Marmosets have great advantages as models for human reproductive biology and are also valuable as experimental nonhuman primates; thus, the current study provides an important platform for primate reproductive biology, including possible applications to humans.


Assuntos
Callithrix/genética , Espermatogênese/genética , Espermatozoides/metabolismo , Testículo/metabolismo , Fatores de Transcrição/genética , Fatores Etários , Envelhecimento/genética , Animais , Metilação de DNA , Epigênese Genética , Evolução Molecular , Perfilação da Expressão Gênica/métodos , Regulação da Expressão Gênica no Desenvolvimento , Marcadores Genéticos , Imuno-Histoquímica , Masculino , Camundongos , RNA Mensageiro/metabolismo , Especificidade da Espécie , Testículo/citologia , Fatores de Transcrição/metabolismo
12.
Primates ; 62(1): 113-121, 2021 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-32803510

RESUMO

Non-human primates are our closest relatives and therefore offer valuable comparative models for human evolutionary studies and biomedical research. As such, Japanese macaques (Macaca fuscata) have contributed to the advancement of primatology in both field and laboratory settings. Specifically, Japanese macaques serve as an excellent model for investigating postnatal development and seasonal breeding in primates because of their relatively prolonged juvenile period and distinct seasonal breeding activity in adulthood. Pioneering histological studies have examined the developmental associations between their reproductive states and spermatogenesis by morphological observation. However, a molecular histological atlas of Japanese macaque spermatogenesis is only in its infancy, limiting our understanding of spermatogenesis ontogeny related to their reproductive changes. Here, we performed immunofluorescence analyses of spermatogenesis in Japanese macaque testes to determine the expression of a subset of marker proteins. The present molecular histological analyses readily specified major spermatogonial subtypes as SALL4+ A spermatogonia and Ki67+/C-KIT+ B spermatogonia. The expression of DAZL, SCP1, γH2AX, VASA, and calmegin further showed sequential changes regarding the protein expression profile and chromosomal structures during spermatogenesis in a differentiation stage-specific manner. Accordingly, comparative analyses between subadults and adults identified spermatogenic deficits in differentiation and synchronization in subadult testes. Our findings provide a new diagnostic platform for dissecting spermatogenic status and reproduction in the Japanese macaques.


Assuntos
Macaca fuscata/metabolismo , Espermatogênese/fisiologia , Espermatogônias/citologia , Animais , Cromossomos de Mamíferos/fisiologia , Imuno-Histoquímica , Macaca fuscata/genética , Macaca fuscata/crescimento & desenvolvimento , Masculino , Espermatogônias/metabolismo , Testículo/citologia , Testículo/metabolismo
13.
Mol Reprod Dev ; 77(9): 802-11, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20722049

RESUMO

Pluripotent stem cells can be established by various methods, but they share several cytological properties, including germ cell differentiation in vitro, independently of their origin. Although mouse induced pluripotent stem (iPS) cells can produce functional gametes in vivo, it is still unclear whether or not they have the ability to produce presumptive germ cells in vitro. Here, we show that mouse iPS cells derived from adult hepatocytes were able to differentiate into presumptive germ cells marked by mouse vasa homolog (Mvh) expression in feeder-free or suspension cultures. Embryoid body (EB) formation from iPS cells also induced the formation of round-shaped cells resembling immature oocytes. Mvh(+) cells formed clumps by co-aggregation with differentiation-supporting cells, and increased expression of germ cell markers was detected in these cell aggregates. Differentiation culture of presumptive germ cells from iPS cells could provide a conventional system for facilitating our understanding of the mechanisms underlying direct reprogramming and germline competency.


Assuntos
Células-Tronco Embrionárias/citologia , Hepatócitos/citologia , Células-Tronco Pluripotentes Induzidas/citologia , Animais , Diferenciação Celular , RNA Helicases DEAD-box/análise , RNA Helicases DEAD-box/genética , Corpos Embrioides/citologia , Células Germinativas/citologia , Masculino , Camundongos , Camundongos Transgênicos , Oócitos/citologia , Retroviridae
14.
Stem Cell Res ; 44: 101749, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-32151953

RESUMO

Evolutionary developmental biology of our closest living relative, the chimpanzee (Pan troglodytes), is essential for understanding the origin of human traits. However, it is difficult to access developmental events in the chimpanzee in vivo because of technical and ethical restrictions. Induced pluripotent stem cells (iPSCs) offer an alternative in vitro model system to investigate developmental events by overcoming the limitations of in vivo study. Here, we generated chimpanzee iPSCs from adult skin fibroblasts and reconstructed early neural development using in vitro differentiation culture conditions. Chimpanzee iPSCs were established using straightforward methods, namely, lipofection of plasmid vectors carrying human reprogramming factors, combined with maintenance in a comprehensive feeder-free culture. Ultimately, direct neurosphere formation culture induced rapid and efficient differentiation of neural stem cells from chimpanzee iPSCs. Time course analysis of neurosphere formation demonstrated ontogenetic changes in gene expression profiles and developmental potency along an early neural development path from epiblasts to radial glia. Our iPSC culture system is a potent tool for investigating the molecular and cellular foundation underlying chimpanzee early neural development and better understanding of human brain evolution.


Assuntos
Células-Tronco Pluripotentes Induzidas , Animais , Diferenciação Celular , Reprogramação Celular , Fibroblastos , Humanos , Neurogênese , Pan troglodytes
15.
Insect Biochem Mol Biol ; 38(11): 1001-7, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18835445

RESUMO

The insect brain is the center of developmental control, from which ecdysone governs brain morphogenesis and regulates gene expression cascades associated with molting and metamorphosis. In order to identify the 20-hydroxyecdysone (20E)-inducible genes responsible for molting and metamorphosis, we constructed a 20E-induced subtraction complementary DNA library from the fifth instar larval brain of the silkworm Bombyx mori. We isolated 10 genes, designated as bombeil-1 to bombeil-10, three of which did not show any sequence similarity to previously identified Bombyx genes. Whole-mount in situ hybridization revealed that all of these bombeil messenger RNAs were exclusively located in two pairs of lateral neurosecretory cells in the larval brain, known as prothoracicotropic hormone (PTTH)-producing cells. RNA-interference knockdown targeting bombeil-2 resulted in larval-pupal molt defects, and adult wing and leg malformations. These results, together with the cell-specific co-localization of bombeil transcripts with PTTH, suggest that bombeil genes play important roles during larval-pupal-adult development.


Assuntos
Bombyx/genética , Ecdisterona/farmacologia , Expressão Gênica/efeitos dos fármacos , Proteínas de Insetos/genética , Animais , Bombyx/efeitos dos fármacos , Bombyx/crescimento & desenvolvimento , Encéfalo/metabolismo , Proteínas de Insetos/química , Proteínas de Insetos/fisiologia , Interferência de RNA , RNA Mensageiro/análise , RNA Mensageiro/metabolismo
16.
Sci Rep ; 8(1): 12187, 2018 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-30111816

RESUMO

Non-human primates are our closest relatives and are of special interest for ecological, evolutionary and biomedical research. The Japanese macaque (Macaca fuscata) has contributed to the progress of primatology and neurosciences over 60 years. Despite this importance, the molecular and cellular basis of the Japanese macaque remains unexplored since useful cellular tools are lacking. Here we generated induced pluripotent stem cells (iPSCs) from skin fibroblasts of the Japanese macaque with Sendai virus or plasmid vectors. The Japanese macaque iPSCs (jm-iPSCs) were established under feeder-free culture conditions, but feeder cells turned out to be essential for their maintenance. The jm-iPSCs formed human iPSC-like flat colonies which were positive for pluripotent antigens including alkaline phosphatase, SSEA4, and TRA-1-81. They also expressed endogenous OCT3/4, SOX2, L-MYC, and KLF4 and other pluripotent marker genes. The potential to differentiate into all three germ layers and neural stem cells was confirmed by embryoid body and neurosphere formation, respectively. The jm-iPSCs will provide a robust in vitro tool for investigating the underlying mechanisms of development and physiology studies with the Japanese macaque.


Assuntos
Técnicas de Cultura de Células/métodos , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/metabolismo , Animais , Diferenciação Celular/genética , Células Cultivadas , Reprogramação Celular/fisiologia , Corpos Embrioides/citologia , Células Alimentadoras , Fibroblastos/citologia , Fibroblastos/metabolismo , Camadas Germinativas , Japão , Fator 4 Semelhante a Kruppel , Macaca , Pele/citologia , Pele/metabolismo
17.
Genome Biol Evol ; 9(7): 1963-1970, 2017 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-28810713

RESUMO

Owl monkeys (genus Aotus) are the only taxon in simian primates that consists of nocturnal or otherwise cathemeral species. Their night vision is superior to that of other monkeys, apes, and humans but not as good as that of typical nocturnal mammals. This incomplete night vision has been used to conclude that these monkeys only secondarily adapted to a nocturnal lifestyle, or to their cathemeral lifestyle that involves high night-time activity. It is known that the rod cells of many nocturnal mammals possess a unique nuclear architecture in which heterochromatin is centrally located. This "inverted nuclear architecture", in contrast with "conventional nuclear architecture", provides elevated night vision by passing light efficiently to the outer segments of photoreceptors. Owl monkey rod cells exhibit an intermediate chromatin distribution, which may provide them with less efficient night vision than other nocturnal mammals. Recently, we identified three megasatellite DNAs in the genome of Azara's owl monkey (Aotus azarae). In the present study, we show that one of the three megasatellite DNAs, OwlRep, serves as the primary component of the heterochromatin block located in the central space of the rod nucleus in A. azarae. This satellite DNA is likely to have emerged in the Aotus lineage after its divergence from those of other platyrrhini taxa and underwent a rapid expansion in the genome. Our results indicate that the heterochromatin core in the A. azarae rod nucleus was newly formed in A. azarae or its recent ancestor, and supports the hypothesis that A. azarae, and with all probability other Aotus species, secondarily acquired night vision.


Assuntos
Aotidae/genética , Aotidae/fisiologia , Evolução Biológica , DNA Satélite , Animais , Evolução Molecular , Heterocromatina , Masculino , Visão Noturna , Retina/metabolismo , Células Fotorreceptoras Retinianas Bastonetes/metabolismo , Análise de Sequência de DNA/métodos
18.
BMC Dev Biol ; 6: 34, 2006 Jul 21.
Artigo em Inglês | MEDLINE | ID: mdl-16859545

RESUMO

BACKGROUND: We previously identified a set of genes called ECATs (ES cell-associated transcripts) that are expressed at high levels in mouse ES cells. Here, we examine the expression and DNA methylation of ECATs in somatic cells and germ cells. RESULTS: In all ECATs examined, the promoter region had low methylation levels in ES cells, but higher levels in somatic cells. In contrast, in spite of their lack of pluripotency, male germline stem (GS) cells expressed most ECATs and exhibited hypomethylation of ECAT promoter regions. We observed a similar hypomethylation of ECAT loci in adult testis and isolated sperm. Some ECATs were even less methylated in male germ cells than in ES cells. However, a few ECATs were not expressed in GS cells, and most of them targets of Oct3/4 and Sox2. The Octamer/Sox regulatory elements were hypermethylated in these genes. In addition, we found that GS cells express little Sox2 protein and low Oct3/4 protein despite abundant expression of their transcripts. CONCLUSION: Our results suggest that DNA hypermethylation and transcriptional repression of a small set of ECATs, together with post-transcriptional repression of Oct3/4 and Sox2, contribute to the loss of pluripotency in male germ cells.


Assuntos
Metilação de DNA , Marcadores Genéticos/genética , Espermatozoides/metabolismo , Células-Tronco/metabolismo , Transcrição Gênica/genética , Animais , Células Cultivadas , Imunoprecipitação da Cromatina , Cromossomos de Mamíferos/genética , DNA/genética , DNA/metabolismo , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Fator 4 de Crescimento de Fibroblastos/genética , Fator 4 de Crescimento de Fibroblastos/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/genética , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Masculino , Camundongos , Proteína Homeobox Nanog , Fator 3 de Transcrição de Octâmero/genética , Fator 3 de Transcrição de Octâmero/metabolismo , Proteínas de Transporte de Cátions Orgânicos/genética , Proteínas de Transporte de Cátions Orgânicos/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa/métodos , Fatores de Transcrição SOXB1 , Espermatozoides/citologia , Células-Tronco/citologia , Transativadores/genética , Transativadores/metabolismo
19.
Primates ; 57(1): 129-35, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26530217

RESUMO

Spermatogonia are specialized cells responsible for continuous spermatogenesis and the production of offspring. Because of this biological property, in vitro culture of spermatogonia provides a powerful methodology to advance reproductive biology and engineering. However, methods for culturing primate spermatogonia are poorly established. We have designed a novel method for culturing spermatogonia in the common marmoset (Callithrix jacchus), a small primate. By using our method with a suite of growth factors, adult marmoset testis-derived germ cells could be cultured in the form of a floating sphere for several weeks. Notably, this method could be applied not only to freshly isolated cells but also to cryopreserved cell stocks. The spheres enriched spermatogonia and early spermatocytes, and could be assembled from a C-KIT(+) spermatogonial population. Techniques for culturing spermatogonia could facilitate increased understanding of primate reproduction as well as the preservation of valuable biomaterials from nonhuman primates.


Assuntos
Callithrix/fisiologia , Técnicas de Cultura de Células/métodos , Espermatogênese , Espermatogônias/crescimento & desenvolvimento , Testículo/crescimento & desenvolvimento , Animais , Masculino
20.
PLoS One ; 9(11): e111867, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25365557

RESUMO

In our institute, we have recently found a child Japanese monkey who is characterized by deep wrinkles of the skin and cataract of bilateral eyes. Numbers of analyses were performed to identify symptoms representing different aspects of aging. In this monkey, the cell cycle of fibroblasts at early passage was significantly extended as compared to a normal control. Moreover, both the appearance of senescent cells and the deficiency in DNA repair were observed. Also, pathological examination showed that this monkey has poikiloderma with superficial telangiectasia, and biochemical assay confirmed that levels of HbA1c and urinary hyaluronan were higher than those of other (child, adult, and aged) monkey groups. Of particular interest was that our MRI analysis revealed expansion of the cerebral sulci and lateral ventricles probably due to shrinkage of the cerebral cortex and the hippocampus. In addition, the conduction velocity of a peripheral sensory but not motor nerve was lower than in adult and child monkeys, and as low as in aged monkeys. However, we could not detect any individual-unique mutations of known genes responsible for major progeroid syndromes. The present results indicate that the monkey suffers from a kind of progeria that is not necessarily typical to human progeroid syndromes.


Assuntos
Ciclo Celular , Senescência Celular , Córtex Cerebral , Hipocampo , Progéria , Envelhecimento da Pele , Animais , Córtex Cerebral/diagnóstico por imagem , Córtex Cerebral/metabolismo , Reparo do DNA , Modelos Animais de Doenças , Feminino , Fibroblastos/metabolismo , Fibroblastos/patologia , Hipocampo/diagnóstico por imagem , Hipocampo/metabolismo , Humanos , Macaca , Progéria/diagnóstico por imagem , Progéria/metabolismo , Radiografia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA