Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 76
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cells ; 13(6)2024 Mar 21.
Artigo em Inglês | MEDLINE | ID: mdl-38534402

RESUMO

Non-human primates (NHPs) are pivotal animal models for translating novel cell replacement therapies into clinical applications, including validating the safety and efficacy of induced pluripotent stem cell (iPSC)-derived products. Preclinical development and the testing of cell-based therapies ideally comprise xenogeneic (human stem cells into NHPs) and allogenic (NHP stem cells into NHPs) transplantation studies. For the allogeneic approach, it is necessary to generate NHP-iPSCs with generally equivalent quality to the human counterparts that will be used later on in patients. Here, we report the generation and characterization of transgene- and feeder-free cynomolgus monkey (Macaca fascicularis) iPSCs (Cyno-iPSCs). These novel cell lines have been generated according to a previously developed protocol for the generation of rhesus macaque, baboon, and human iPSC lines. Beyond their generation, we demonstrate the potential of the novel Cyno-iPSCs to differentiate into two clinically relevant cell types, i.e., cardiomyocytes and neurons. Overall, we provide a resource of novel iPSCs from the most frequently used NHP species in the regulatory testing of biologics and classical pharmaceutics to expand our panel of iPSC lines from NHP species with high relevance in preclinical testing and translational research.


Assuntos
Células-Tronco Pluripotentes Induzidas , Animais , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Macaca fascicularis , Diferenciação Celular/fisiologia , Macaca mulatta , Transgenes
3.
Life Sci Alliance ; 7(6)2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38499329

RESUMO

Primordial germ cells (PGCs) are the embryonic precursors of sperm and oocytes, which transmit genetic/epigenetic information across generations. Mouse PGC and subsequent gamete development can be fully reconstituted in vitro, opening up new avenues for germ cell studies in biomedical research. However, PGCs show molecular differences between rodents and humans. Therefore, to establish an in vitro system that is closely related to humans, we studied PGC development in vivo and in vitro in the common marmoset monkey Callithrix jacchus (cj). Gonadal cjPGCs at embryonic day 74 express SOX17, AP2Ɣ, BLIMP1, NANOG, and OCT4A, which is reminiscent of human PGCs. We established transgene-free induced pluripotent stem cell (cjiPSC) lines from foetal and postnatal fibroblasts. These cjiPSCs, cultured in defined and feeder-free conditions, can be differentiated into precursors of mesendoderm and subsequently into cjPGC-like cells (cjPGCLCs) with a transcriptome similar to human PGCs/PGCLCs. Our results not only pave the way for studying PGC development in a non-human primate in vitro under experimentally controlled conditions, but also provide the opportunity to derive functional marmoset gametes in future studies.


Assuntos
Callithrix , Células-Tronco Pluripotentes , Masculino , Camundongos , Animais , Sêmen , Células Germinativas/metabolismo , Células-Tronco Embrionárias/metabolismo
4.
Cell Stem Cell ; 30(7): 938-949.e7, 2023 07 06.
Artigo em Inglês | MEDLINE | ID: mdl-37343565

RESUMO

Differential speeds in biochemical reactions have been proposed to be responsible for the differences in developmental tempo between mice and humans. However, the underlying mechanism controlling the species-specific kinetics remains to be determined. Using in vitro differentiation of pluripotent stem cells, we recapitulated the segmentation clocks of diverse mammalian species varying in body weight and taxa: marmoset, rabbit, cattle, and rhinoceros. Together with mouse and human, the segmentation clock periods of the six species did not scale with the animal body weight, but with the embryogenesis length. The biochemical kinetics of the core clock gene HES7 displayed clear scaling with the species-specific segmentation clock period. However, the cellular metabolic rates did not show an evident correlation. Instead, genes involving biochemical reactions showed an expression pattern that scales with the segmentation clock period. Altogether, our stem cell zoo uncovered general scaling laws governing species-specific developmental tempo.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos , Células-Tronco Pluripotentes , Animais , Camundongos , Humanos , Bovinos , Coelhos , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Relógios Biológicos , Diferenciação Celular , Mamíferos/metabolismo , Regulação da Expressão Gênica no Desenvolvimento
5.
Front Mol Neurosci ; 16: 1154509, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37168680

RESUMO

The evolutionary conserved orphan cytokine receptor-like factor 3 (CRLF3) has been implicated in human disease, vertebrate hematopoiesis and insect neuroprotection. While its specific functions are elusive, experimental evidence points toward a general role in cell homeostasis. Erythropoietin (Epo) is a major regulator of vertebrate hematopoiesis and a general cytoprotective cytokine. Erythropoietic functions mediated by classical Epo receptor are understood in great detail whereas Epo-mediated cytoprotective mechanisms are more complex due to involvement of additional Epo receptors and a non-erythropoietic splice variant with selectivity for certain receptors. In the present study, we show that the human CRLF3 mediates neuroprotection upon activation with the natural Epo splice variant EV-3. We generated CRLF3 knock-out iPSC lines and differentiated them toward the neuronal lineage. While apoptotic death of rotenone-challenged wild type iPSC-derived neurons was prevented by EV-3, EV-3-mediated neuroprotection was absent in CRLF3 knock-out neurons. Rotenone-induced apoptosis and EV-3-mediated neuroprotection were associated with differential expression of pro-and anti-apoptotic genes. Our data characterize human CRLF3 as a receptor involved in Epo-mediated neuroprotection and identify CRLF3 as the first known receptor for EV-3.

6.
Aging (Albany NY) ; 15(5): 1257-1278, 2023 02 27.
Artigo em Inglês | MEDLINE | ID: mdl-36849136

RESUMO

Advanced paternal age is associated with increased risks for reproductive and offspring medical problems. Accumulating evidence suggests age-related changes in the sperm epigenome as one underlying mechanism. Using reduced representation bisulfite sequencing on 73 sperm samples of males attending a fertility center, we identified 1,162 (74%) regions which were significantly (FDR-adjusted) hypomethylated and 403 regions (26%) being hypermethylated with age. There were no significant correlations with paternal BMI, semen quality, or ART outcome. The majority (1,152 of 1,565; 74%) of age-related differentially methylated regions (ageDMRs) were located within genic regions, including 1,002 genes with symbols. Hypomethylated ageDMRs were closer to transcription start sites than hypermethylated DMRs, half of which reside in gene-distal regions. In this and conceptually related genome-wide studies, so far 2,355 genes have been reported with significant sperm ageDMRs, however most (90%) of them in only one study. The 241 genes which have been replicated at least once showed significant functional enrichments in 41 biological processes associated with development and the nervous system and in 10 cellular components associated with synapses and neurons. This supports the hypothesis that paternal age effects on the sperm methylome affect offspring behaviour and neurodevelopment. It is interesting to note that sperm ageDMRs were not randomly distributed throughout the human genome; chromosome 19 showed a highly significant twofold enrichment with sperm ageDMRs. Although the high gene density and CpG content have been conserved, the orthologous marmoset chromosome 22 did not appear to exhibit an increased regulatory potential by age-related DNA methylation changes.


Assuntos
Epigênese Genética , Epigenoma , Humanos , Masculino , Análise do Sêmen , Sêmen , Metilação de DNA , Espermatozoides/metabolismo , Ilhas de CpG
7.
Basic Res Cardiol ; 118(1): 5, 2023 01 26.
Artigo em Inglês | MEDLINE | ID: mdl-36700983

RESUMO

Long non-coding RNAs (lncRNAs) can act as regulatory RNAs which, by altering the expression of target genes, impact on the cellular phenotype and cardiovascular disease development. Endothelial lncRNAs and their vascular functions are largely undefined. Deep RNA-Seq and FANTOM5 CAGE analysis revealed the lncRNA LINC00607 to be highly enriched in human endothelial cells. LINC00607 was induced in response to hypoxia, arteriosclerosis regression in non-human primates, post-atherosclerotic cultured endothelial cells from patients and also in response to propranolol used to induce regression of human arteriovenous malformations. siRNA knockdown or CRISPR/Cas9 knockout of LINC00607 attenuated VEGF-A-induced angiogenic sprouting. LINC00607 knockout in endothelial cells also integrated less into newly formed vascular networks in an in vivo assay in SCID mice. Overexpression of LINC00607 in CRISPR knockout cells restored normal endothelial function. RNA- and ATAC-Seq after LINC00607 knockout revealed changes in the transcription of endothelial gene sets linked to the endothelial phenotype and in chromatin accessibility around ERG-binding sites. Mechanistically, LINC00607 interacted with the SWI/SNF chromatin remodeling protein BRG1. CRISPR/Cas9-mediated knockout of BRG1 in HUVEC followed by CUT&RUN revealed that BRG1 is required to secure a stable chromatin state, mainly on ERG-binding sites. In conclusion, LINC00607 is an endothelial-enriched lncRNA that maintains ERG target gene transcription by interacting with the chromatin remodeler BRG1 to ultimately mediate angiogenesis.


Assuntos
RNA Longo não Codificante , Animais , Humanos , Camundongos , Cromatina , DNA Helicases/genética , DNA Helicases/metabolismo , Células Endoteliais/metabolismo , Camundongos SCID , Proteínas Nucleares/metabolismo , RNA Longo não Codificante/genética , Neovascularização Fisiológica
8.
Nature ; 613(7943): 308-316, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36544022

RESUMO

The testis produces gametes through spermatogenesis and evolves rapidly at both the morphological and molecular level in mammals1-6, probably owing to the evolutionary pressure on males to be reproductively successful7. However, the molecular evolution of individual spermatogenic cell types across mammals remains largely uncharacterized. Here we report evolutionary analyses of single-nucleus transcriptome data for testes from 11 species that cover the three main mammalian lineages (eutherians, marsupials and monotremes) and birds (the evolutionary outgroup), and include seven primates. We find that the rapid evolution of the testis was driven by accelerated fixation rates of gene expression changes, amino acid substitutions and new genes in late spermatogenic stages, probably facilitated by reduced pleiotropic constraints, haploid selection and transcriptionally permissive chromatin. We identify temporal expression changes of individual genes across species and conserved expression programs controlling ancestral spermatogenic processes. Genes predominantly expressed in spermatogonia (germ cells fuelling spermatogenesis) and Sertoli (somatic support) cells accumulated on X chromosomes during evolution, presumably owing to male-beneficial selective forces. Further work identified transcriptomal differences between X- and Y-bearing spermatids and uncovered that meiotic sex-chromosome inactivation (MSCI) also occurs in monotremes and hence is common to mammalian sex-chromosome systems. Thus, the mechanism of meiotic silencing of unsynapsed chromatin, which underlies MSCI, is an ancestral mammalian feature. Our study illuminates the molecular evolution of spermatogenesis and associated selective forces, and provides a resource for investigating the biology of the testis across mammals.


Assuntos
Evolução Molecular , Mamíferos , Espermatogênese , Testículo , Animais , Masculino , Cromatina/genética , Mamíferos/genética , Meiose/genética , Espermatogênese/genética , Testículo/citologia , Transcriptoma , Análise de Célula Única , Aves/genética , Primatas/genética , Regulação da Expressão Gênica , Espermatogônias/citologia , Células de Sertoli/citologia , Cromossomo X/genética , Cromossomo Y/genética , Compensação de Dosagem (Genética) , Inativação Gênica
9.
Cells ; 11(19)2022 10 09.
Artigo em Inglês | MEDLINE | ID: mdl-36231125

RESUMO

The functions of human testicular peritubular cells (HTPCs), forming a small compartment located between the seminiferous epithelium and the interstitial areas of the testis, are not fully known but go beyond intratesticular sperm transport and include immunological roles. The expression of the glucocorticoid receptor (GR) indicates that they may be regulated by glucocorticoids (GCs). Herein, we studied the consequences of the GC dexamethasone (Dex) in cultured HTPCs, which serves as a unique window into the human testis. We examined changes in cytokines, mainly by qPCR and ELISA. A holistic mass-spectrometry-based proteome analysis of cellular and secreted proteins was also performed. Dex, used in a therapeutic concentration, decreased the transcript level of proinflammatory cytokines, e.g., IL6, IL8 and MCP1. An siRNA-mediated knockdown of GR reduced the actions on IL6. Changes in IL6 were confirmed by ELISA measurements. Of note, Dex also lowered GR levels. The proteomic results revealed strong responses after 24 h (31 significantly altered cellular proteins) and more pronounced ones after 72 h of Dex exposure (30 less abundant and 42 more abundant cellular proteins). Dex also altered the composition of the secretome (33 proteins decreased, 13 increased) after 72 h. Among the regulated proteins were extracellular matrix (ECM) and basement membrane components (e.g., FBLN2, COL1A2 and COL3A1), as well as PTX3 and StAR. These results pinpoint novel, profound effects of Dex in HTPCs. If transferrable to the human testis, changes specifically in ECM and the immunological state of the testis may occur in men upon treatment with Dex for medical reasons.


Assuntos
Túbulos Seminíferos , Testículo , Dexametasona/farmacologia , Humanos , Interleucina-6/metabolismo , Interleucina-8/metabolismo , Masculino , Proteoma/metabolismo , Proteômica , RNA Interferente Pequeno/metabolismo , Receptores de Glucocorticoides/metabolismo , Sêmen/metabolismo , Túbulos Seminíferos/metabolismo , Testículo/metabolismo
10.
Front Cell Dev Biol ; 10: 1011109, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36263009

RESUMO

Early forebrain patterning entails the correct regional designation of the neuroepithelium, and appropriate specification, generation, and distribution of neural cells during brain development. Specific signaling and transcription factors are known to tightly regulate patterning of the dorsal telencephalon to afford proper structural/functional cortical arealization and morphogenesis. Nevertheless, whether and how changes of the chromatin structure link to the transcriptional program(s) that control cortical patterning remains elusive. Here, we report that the BAF chromatin remodeling complex regulates the spatiotemporal patterning of the mouse dorsal telencephalon. To determine whether and how the BAF complex regulates cortical patterning, we conditionally deleted the BAF complex scaffolding subunits BAF155 and BAF170 in the mouse dorsal telencephalic neuroepithelium. Morphological and cellular changes in the BAF mutant forebrain were examined using immunohistochemistry and in situ hybridization. RNA sequencing, Co-immunoprecipitation, and mass spectrometry were used to investigate the molecular basis of BAF complex involvement in forebrain patterning. We found that conditional ablation of BAF complex in the dorsal telencephalon neuroepithelium caused expansion of the cortical hem and medial cortex beyond their developmental boundaries. Consequently, the hippocampal primordium is not specified, the mediolateral cortical patterning is compromised, and the cortical identity is disturbed in the absence of BAF complex. The BAF complex was found to interact with the cortical hem suppressor LHX2. The BAF complex suppresses cortical hem fate to permit proper forebrain patterning. We provide evidence that BAF complex modulates mediolateral cortical patterning possibly by interacting with the transcription factor LHX2 to drive the LHX2-dependent transcriptional program essential for dorsal telencephalon patterning. Our data suggest a putative mechanistic synergy between BAF chromatin remodeling complex and LHX2 in regulating forebrain patterning and ontogeny.

11.
Animals (Basel) ; 12(18)2022 Sep 14.
Artigo em Inglês | MEDLINE | ID: mdl-36139275

RESUMO

Non-human primates (NHPs) serve as embryo donors for embryo collection in order to mimic genetic diseases in humans by genetic modification. Reproductive health of the embryo donors is crucial, and chronic distress needs to be avoided. Embryo retrieval rates (ERR), anti-Müllerian hormone (AMH) concentrations, cortisol levels, and body weight fluctuations were assessed as markers for fertility and distress. With regard to successful embryo retrievals (total n = 667), the animals were either used for extended periods (long-term group; LTG) or only for short periods (short-term group; STG). Retrospective evaluation expectedly showed that animals in the LTG had a higher ERR than animals in the STG (p < 0.0001). Importantly, ERR in the LTG remained stable throughout the experimental period, and high embryo rates were already encountered during the first year of experimental use (p = 0.0002). High ERR were associated with high AMH and low cortisol levels, and minimal body weight fluctuations following anesthesia, indicating a superior ability of the LTG animals to handle distress. We conclude that the long-term experimental use of marmosets does not impair their fertility or health status per se, supporting the view that animal reuse can be in accordance with the 3R-principle, implying reduction, replacement, and refinement in animal experimentation.

12.
Primate Biol ; 9(2): 23-28, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36034474

RESUMO

Alfaxan® (alfaxalone) is a steroid general anesthetic widely used in veterinary medicine for induction and maintenance of anesthesia in several species. While the use of alfaxalone in veterinary practice has several benefits compared to the use of other anesthetic agents, the fact that it is derived from progesterone may confound the measurement of the latter in the blood of animals under alfaxalone treatment. In the present case study, we report the measurement of serum progesterone in an individual common marmoset (Callithrix jacchus) during five ovarian cycles in which luteolysis was induced by PGF2 α . Blood samples were usually taken from the awake animal with the exception of the fifth cycle in which the sample was collected under alfaxalone anesthesia in connection with a tooth extraction. In contrast to the previous four cycles in which luteolysis resulted in the expected marked decrease in progesterone concentrations, the - apparent - progesterone level in the cycle under alfaxalone treatment remained unexpectedly high. Cross-reactivity of the non-specific antibody used in the progesterone assay with alfaxalone most likely explains this finding.

13.
Genes (Basel) ; 13(6)2022 06 19.
Artigo em Inglês | MEDLINE | ID: mdl-35741855

RESUMO

Non-ischemic dilated cardiomyopathy (DCM) is one of the most frequent pathologies requiring cardiac transplants. Even though the etiology of this disease is complex, frameshift mutations in the giant sarcomeric protein Titin could explain up to 25% of the familial and 18% of the sporadic cases of DCM. Many studies have shown the potential of genome editing using CRISPR/Cas9 to correct truncating mutations in sarcomeric proteins and have established the grounds for myoediting. However, these therapies are still in an immature state, with only few studies showing an efficient treatment of cardiac diseases. This publication hypothesizes that the Titin (TTN)-specific gene structure allows the application of myoediting approaches in a broad range of locations to reframe TTNtvvariants and to treat DCM patients. Additionally, to pave the way for the generation of efficient myoediting approaches for DCM, we screened and selected promising target locations in TTN. We conceptually explored the deletion of symmetric exons as a therapeutic approach to restore TTN's reading frame in cases of frameshift mutations. We identified a set of 94 potential candidate exons of TTN that we consider particularly suitable for this therapeutic deletion. With this study, we aim to contribute to the development of new therapies to efficiently treat titinopathies and other diseases caused by mutations in genes encoding proteins with modular structures, e.g., Obscurin.


Assuntos
Cardiomiopatia Dilatada , Conectina , Mutação da Fase de Leitura , Cardiomiopatia Dilatada/genética , Conectina/genética , Éxons/genética , Humanos
14.
Nature ; 609(7925): 136-143, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-35709828

RESUMO

Gastrulation controls the emergence of cellular diversity and axis patterning in the early embryo. In mammals, this transformation is orchestrated by dynamic signalling centres at the interface of embryonic and extraembryonic tissues1-3. Elucidating the molecular framework of axis formation in vivo is fundamental for our understanding of human development4-6 and to advance stem-cell-based regenerative approaches7. Here we illuminate early gastrulation of marmoset embryos in utero using spatial transcriptomics and stem-cell-based embryo models. Gaussian process regression-based 3D transcriptomes delineate the emergence of the anterior visceral endoderm, which is hallmarked by conserved (HHEX, LEFTY2, LHX1) and primate-specific (POSTN, SDC4, FZD5) factors. WNT signalling spatially coordinates the formation of the primitive streak in the embryonic disc and is counteracted by SFRP1 and SFRP2 to sustain pluripotency in the anterior domain. Amnion specification occurs at the boundaries of the embryonic disc through ID1, ID2 and ID3 in response to BMP signalling, providing a developmental rationale for amnion differentiation of primate pluripotent stem cells (PSCs). Spatial identity mapping demonstrates that primed marmoset PSCs exhibit the highest similarity to the anterior embryonic disc, whereas naive PSCs resemble the preimplantation epiblast. Our 3D transcriptome models reveal the molecular code of lineage specification in the primate embryo and provide an in vivo reference to decipher human development.


Assuntos
Callithrix , Gastrulação , Útero , Animais , Callithrix/embriologia , Diferenciação Celular , Embrião de Mamíferos/citologia , Embrião de Mamíferos/embriologia , Endoderma/citologia , Endoderma/embriologia , Feminino , Perfilação da Expressão Gênica , Camadas Germinativas/citologia , Camadas Germinativas/embriologia , Humanos , Células-Tronco Pluripotentes/citologia
15.
Neural Regen Res ; 17(9): 1867-1874, 2022 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-35142660

RESUMO

Non-human primates play a key role in the preclinical validation of pluripotent stem cell-based cell replacement therapies. Pluripotent stem cells used as advanced therapy medical products boost the possibility to regenerate tissues and organs affected by degenerative diseases. Therefore, the methods to derive human induced pluripotent stem cell and embryonic stem cell lines following clinical standards have quickly developed in the last 15 years. For the preclinical validation of cell replacement therapies in non-human primates, it is necessary to generate non-human primate pluripotent stem cell with a homologous quality to their human counterparts. However, pluripotent stem cell technologies have developed at a slower pace in non-human primates in comparison with human cell systems. In recent years, however, relevant progress has also been made with non-human primate pluripotent stem cells. This review provides a systematic overview of the progress and remaining challenges for the generation of non-human primate induced pluripotent stem cells/embryonic stem cells for the preclinical testing and validation of cell replacement therapies. We focus on the critical domains of (1) reprogramming and embryonic stem cell line derivation, (2) cell line maintenance and characterization and, (3) application of non-human primate pluripotent stem cells in the context of selected preclinical studies to treat cardiovascular and neurodegenerative disorders performed in non-human primates.

16.
Microorganisms ; 11(1)2022 Dec 21.
Artigo em Inglês | MEDLINE | ID: mdl-36677317

RESUMO

Primate simplexviruses are closely related neurotropic herpesviruses, which are largely apathogenic in their respective host species. However, cross-species transmission of Macacine alphaherpesvirus 1 (McHV1, also termed herpes B virus) from rhesus macaques to humans can cause fatal encephalomyelitis. In contrast, closely related viruses, such as Cercopithecine alphaherpesvirus 2 (CeHV2, also termed simian agent 8) or Papiine alphaherpesvirus 2 (PaHV2, also termed herpesvirus papio 2), have not been linked to human disease and are believed to be largely apathogenic in humans. Here, we investigated whether McHV1, PaHV2 and CeHV2 differ in their capacity to infect human and non-human primate (NHP) cells. For comparison, we included the human simplexviruses HSV1 and HSV2 in our analyses. All five viruses replicated efficiently in cell lines of human and African green monkey origin, and McHV1 and PaHV2 also showed robust replication in rhesus macaque cell lines. In contrast, the replication of CeHV2 and particularly HSV1 and HSV2 in cell lines of rhesus macaque origin were reduced or inefficient. Similarly, McHV1, but not CeHV2, efficiently infected rhesus macaque brain organoids. These results point towards the previously unappreciated partial resistance of certain rhesus macaque cells to HSV1/HSV2/CeHV2 infection and reveal similarities between the cell tropism of McHV1 and PaHV2 that might be relevant for risk assessment.

17.
Methods Mol Biol ; 2454: 717-729, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-33733393

RESUMO

The generation and culture of transgene-free induced pluripotent stem cells (iPSCs) from the common marmoset (Callithrix jacchus) present unique challenges due to the fact that the protocols developed for culture of human or mouse pluripotent cells are not sufficiently optimized for this particular monkey species. Here, we describe the procedures for the reprogramming of marmoset fetal fibroblasts to pluripotency with self-replicating mRNAs using a two-step approach, where intermediate primary colonies generated in the first reprogramming step are converted in the second step to iPSCs with customized marmoset culture medium. The resulting iPSCs are free of transgenes and can be maintained in long-term culture in feeder-free conditions.


Assuntos
Células-Tronco Pluripotentes Induzidas , Animais , Callithrix , Técnicas de Cultura de Células/métodos , Diferenciação Celular/genética , Reprogramação Celular/genética , Fibroblastos , Camundongos , RNA Mensageiro
18.
Methods Mol Biol ; 2454: 697-716, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-33772458

RESUMO

Non-human primates (NHP), and in particular Old World monkeys including macaques and baboons, are key animal models for the late preclinical testing of novel stem cell-based therapies and other advanced therapy medical products (ATMP) for the treatment of degenerative diseases. These pathologies are characterized by the loss of functional cells in an organ, as in Parkinson's disease, age-related macular degeneration, or after myocardial infarction. For preclinically relevant testing of induced pluripotent stem cell (iPSC)-based therapies, robust, and standardized protocols for the generation, characterization, and differentiation of NHP-iPSCs are required. Since the discovery of iPSCs by Takahashi and Yamanaka in 2006, human reprogramming protocols have been continuously refined. However, the generation of integration-free NHP-iPSC lines and a stable feeder- and serum-free long-term culture turned out to be difficult or even impossible with the current protocols established for human iPSCs. Here, we provide a robust protocol for the generation of transgene-free Old World monkey (and human) iPSCs and long-term cultivation under chemically defined conditions. This protocol was successfully applied to generate human, baboon (Papio anubis), rhesus (Macaca mulatta), and cynomolgus macaque (Macaca fascicularis) iPSCs from skin fibroblasts. The resulting NHP-iPSCs provide a valuable resource for the preclinical testing of regenerative therapies in NHP.


Assuntos
Células-Tronco Pluripotentes Induzidas , Animais , Diferenciação Celular , Macaca fascicularis , Papio , Transgenes
19.
Sci Rep ; 11(1): 15439, 2021 07 29.
Artigo em Inglês | MEDLINE | ID: mdl-34326359

RESUMO

Non-human primates (NHPs) are, due to their close phylogenetic relationship to humans, excellent animal models to study clinically relevant mutations. However, the toolbox for the genetic modification of NHPs is less developed than those for other species like mice. Therefore, it is necessary to further develop and refine genome editing approaches in NHPs. NHP pluripotent stem cells (PSCs) share key molecular signatures with the early embryo, which is an important target for genomic modification. Therefore, PSCs are a valuable test system for the validation of embryonic genome editing approaches. In the present study, we made use of the versatility of the piggyBac transposon system for different purposes in the context of NHP stem cell technology and genome editing. These include (1) Robust reprogramming of rhesus macaque fibroblasts to induced pluripotent stem cells (iPSCs); (2) Culture of the iPSCs under feeder-free conditions even after removal of the transgene resulting in transgene-free iPSCs; (3) Development of a CRISPR/Cas-based work-flow to edit the genome of rhesus macaque PSCs with high efficiency; (4) Establishment of a novel protocol for the derivation of gene-edited monoclonal NHP-iPSC lines. These findings facilitate efficient testing of genome editing approaches in NHP-PSC before their in vivo application.


Assuntos
Reprogramação Celular/genética , Elementos de DNA Transponíveis/genética , Edição de Genes/métodos , Células-Tronco Pluripotentes Induzidas/citologia , Macaca mulatta/genética , Animais , Proteína 9 Associada à CRISPR/genética , Sistemas CRISPR-Cas , Linhagem Celular , Feminino , Fibroblastos/citologia , Vetores Genéticos , Proteínas de Fluorescência Verde/genética , Masculino , Camundongos , Filogenia , Pele/citologia , Transfecção , Transgenes , Transposases/genética
20.
Cells ; 10(6)2021 05 24.
Artigo em Inglês | MEDLINE | ID: mdl-34074003

RESUMO

Aging of human testis and associated cellular changes is difficult to assess. Therefore, we used a translational, non-human primate model to get insights into underlying cellular and biochemical processes. Using proteomics and immunohistochemistry, we analyzed testicular tissue of young (age 2 to 3) and old (age 10 to 12) common marmosets (Callithrix jacchus). Using a mass spectrometry-based proteomics approach, we identified 63,124 peptides, which could be assigned to 5924 proteins. Among them, we found proteins specific for germ cells and somatic cells, such as Leydig and Sertoli cells. Quantitative analysis showed 31 differentially abundant proteins, of which 29 proteins were more abundant in older animals. An increased abundance of anti-proliferative proteins, among them CDKN2A, indicate reduced cell proliferation in old testes. Additionally, an increased abundance of several small leucine rich repeat proteoglycans and other extracellular matrix proteins was observed, which may be related to impaired cell migration and fibrotic events. Furthermore, an increased abundance of proteins with inhibitory roles in smooth muscle cell contraction like CNN1 indicates functional alterations in testicular peritubular cells and may mirror a reduced capacity of these cells to contract in old testes.


Assuntos
Envelhecimento/metabolismo , Proteoma/metabolismo , Testículo/metabolismo , Animais , Callithrix , Masculino
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...