Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Front Immunol ; 14: 1296580, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-38149246

RESUMO

Introduction: Lyme disease, the most common tick-borne infectious disease in the US, is caused by a spirochetal pathogen Borrelia burgdorferi (Bb). Distinct host responses are observed in susceptible and resistant strains of inbred of mice following infection with Bb reflecting a subset of inflammatory responses observed in human Lyme disease. The advent of post-genomic methodologies and genomic data sets enables dissecting the host responses to advance therapeutic options for limiting the pathogen transmission and/or treatment of Lyme disease. Methods: In this study, we used single-cell RNA-Seq analysis in conjunction with mouse genomics exploiting GFP-expressing Bb to sort GFP+ splenocytes and GFP- bystander cells to uncover novel molecular and cellular signatures that contribute to early stages of immune responses against Bb. Results: These data decoded the heterogeneity of splenic neutrophils, macrophages, NK cells, B cells, and T cells in C3H/HeN mice in response to Bb infection. Increased mRNA abundance of apoptosis-related genes was observed in neutrophils and macrophages clustered from GFP+ splenocytes. Moreover, complement-mediated phagocytosis-related genes such as C1q and Ficolin were elevated in an inflammatory macrophage subset, suggesting upregulation of these genes during the interaction of macrophages with Bb-infected neutrophils. In addition, the role of DUSP1 in regulating the expression of Casp3 and pro-inflammatory cytokines Cxcl1, Cxcl2, Il1b, and Ccl5 in Bb-infected neutrophils were identified. Discussion: These findings serve as a growing catalog of cell phenotypes/biomarkers among murine splenocytes that can be exploited for limiting spirochetal burden to limit the transmission of the agent of Lyme disease to humans via reservoir hosts.


Assuntos
Borrelia burgdorferi , Doença de Lyme , Camundongos , Humanos , Animais , Borrelia burgdorferi/genética , Transcriptoma , Baço , Análise da Expressão Gênica de Célula Única , Camundongos Endogâmicos C3H , Doença de Lyme/genética
2.
Development ; 150(14)2023 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-37350382

RESUMO

Retinoic acid (RA) is the proposed mammalian 'meiosis inducing substance'. However, evidence for this role comes from studies in the fetal ovary, where germ cell differentiation and meiotic initiation are temporally inseparable. In the postnatal testis, these events are separated by more than 1 week. Exploiting this difference, we discovered that, although RA is required for spermatogonial differentiation, it is dispensable for the subsequent initiation, progression and completion of meiosis. Indeed, in the absence of RA, the meiotic transcriptome program in both differentiating spermatogonia and spermatocytes entering meiosis was largely unaffected. Instead, transcripts encoding factors required during spermiogenesis were aberrant during preleptonema, and the subsequent spermatid morphogenesis program was disrupted such that no sperm were produced. Taken together, these data reveal a RA-independent model for male meiotic initiation.


Assuntos
Testículo , Tretinoína , Animais , Feminino , Masculino , Tretinoína/farmacologia , Espermatogênese/genética , Espermatogônias , Espermatozoides , Meiose/genética , Mamíferos
3.
Methods Mol Biol ; 2656: 1-6, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37249863

RESUMO

Spermatogenesis is maintained throughout adulthood by a pool of adult stem cells termed spermatogonial stem cells (SSCs). Research investigations into spermatogenesis can provide insight into the etiology of certain types of male infertility (e.g., Sertoli cell only syndrome), elucidate means of improving food animal production, reveal new therapeutic avenues to address naturally occurring defects in sperm production, mitigate iatrogenic male infertility (e.g., arising from cancer therapy), and potentially intervene for male contraception. This chapter will serve as a commentary about why studying spermatogenesis is important, including a high-level overview of spermatogonia and SSCs, and make the case for a critical need for use of stringent definitions for SSCs and experimental platforms that allow for clear distinction of the multiple types of spermatogonia that exist in testes of mammals.


Assuntos
Infertilidade Masculina , Células-Tronco , Humanos , Animais , Masculino , Sêmen , Espermatogênese , Espermatogônias/metabolismo , Testículo , Mamíferos
4.
Methods Mol Biol ; 2656: 21-35, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37249865

RESUMO

Single-molecule fluorescence in situ hybridization (smFISH) enables the detection and localization of individual mRNAs in tissue sections with single-molecule resolution while preserving spatial context, and thus, is a useful tool for examining gene expression in biological systems. In particular, the growing reliance on single-cell RNA sequencing (scRNA-seq) to explore cellular heterogeneity has reinvigorated this approach as a validation tool to spatially re-map mRNA expression patterns described in isolated cells to their parent tissue. While use of antibody-based methods, such as indirect immunofluorescence (IIF), remain popular as validation strategies, smFISH often affords superior specificity and maintains congruency with scRNA-seq. Here, we present a detailed protocol that combines multiplexed smFISH using the RNAscope approach with IIF to co-visualize mRNAs and proteins within sections of mouse testes. We provide step-by-step guidelines from testis preparation through visualization that enables mapping of combinations of up to four mRNA/protein targets in frozen sections on the RNAscope platform.


Assuntos
Mamíferos , Testículo , Camundongos , Masculino , Animais , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Testículo/metabolismo , Hibridização in Situ Fluorescente/métodos , Mamíferos/genética , Nanotecnologia
5.
Methods Mol Biol ; 2656: 37-70, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37249866

RESUMO

Robust methods have been developed that leverage next-generation sequencing (NGS) to measure abundance of all mRNAs (RNA-seq) in samples as small as individual cells in order to study the testicular transcriptome in mammals. In this chapter, we present robust options for implementing bioinformatics workflows for the analysis of bulk RNA-seq from aggregate samples of hundreds to millions of cells and single-cell RNA-seq from individual cells. We also provide detailed protocols for using the R packages DESeq2 and Seurat, important parameters for successful implementation, and considerations for drawing conclusions from the results.


Assuntos
Análise da Expressão Gênica de Célula Única , Espermatogônias , Masculino , Animais , Transcriptoma , Testículo , RNA-Seq , Análise de Célula Única/métodos , Análise de Sequência de RNA/métodos , Perfilação da Expressão Gênica/métodos , Mamíferos
6.
Methods Mol Biol ; 2656: 179-193, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37249872

RESUMO

In the mammalian testis, the mitotic complements of spermatogenic cells are spermatogonia, including spermatogonial stem cells (SSCs) which form the basis of life-long spermatogenesis and male fertility. Thus, investigating spermatogonia and subdivisions thereof is essential to increase our understanding of male germline development and infertility. This protocol describes the isolation of spermatogonia from both adult and developing [postnatal day 6 (P6)] mouse testes. Cell suspensions of the adult mouse testis from the Id4-Egfp transgenic mouse line are obtained through a two-step enzymatic digestion and are subjected to Percoll pre-enrichment before spermatogonia are isolated by selecting testis cells that are CD9bright and ID4-EGFP+ through FACS. For P6 mice, the testis is digested using trypsin-DNase, and spermatogonia are isolated by FACS selection of ID4-EGFP+ testis cells. In both cases, nearly pure populations of undifferentiated spermatogonia are obtained that can be further subdivided using additional parameters (e.g., EGFP intensity, cell surface protein immunostaining), and recovered for use in various downstream applications, such as biochemical analyses (e.g., transcriptome/epigenome), functional analyses by SSC transplantation or propagation in vitro.


Assuntos
Espermatogônias , Testículo , Masculino , Camundongos , Animais , Diferenciação Celular , Células-Tronco , Espermatogênese , Camundongos Transgênicos , Mamíferos
7.
Int J Mol Sci ; 24(5)2023 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-36902187

RESUMO

Antineoplastic treatments for cancer and other non-malignant disorders can result in long-term or permanent male infertility by ablating spermatogonial stem cells (SSCs). SSC transplantation using testicular tissue harvested before a sterilizing treatment is a promising approach for restoring male fertility in these cases, but a lack of exclusive biomarkers to unequivocally identify prepubertal SSCs limits their therapeutic potential. To address this, we performed single-cell RNA-seq on testis cells from immature baboons and macaques and compared these cells with published data from prepubertal human testis cells and functionally-defined mouse SSCs. While we found discrete groups of human spermatogonia, baboon and rhesus spermatogonia appeared less heterogenous. A cross-species analysis revealed cell types analogous to human SSCs in baboon and rhesus germ cells, but a comparison with mouse SSCs revealed significant differences with primate SSCs. Primate-specific SSC genes were enriched for components and regulators of the actin cytoskeleton and participate in cell-adhesion, which may explain why the culture conditions for rodent SSCs are not appropriate for primate SSCs. Furthermore, correlating the molecular definitions of human SSC, progenitor and differentiating spermatogonia with the histological definitions of Adark/Apale spermatogonia indicates that both SSCs and progenitor spermatogonia are Adark, while Apale spermatogonia appear biased towards differentiation. These results resolve the molecular identity of prepubertal human SSCs, define novel pathways that could be leveraged for advancing their selection and propagation in vitro, and confirm that the human SSC pool resides entirely within Adark spermatogonia.


Assuntos
Células-Tronco Germinativas Adultas , Espermatogônias , Humanos , Masculino , Animais , Camundongos , Espermatogônias/metabolismo , Testículo , Espermatogênese , Transcriptoma , Primatas
8.
BMC Cancer ; 23(1): 251, 2023 Mar 16.
Artigo em Inglês | MEDLINE | ID: mdl-36922758

RESUMO

BACKGROUND: Currently there are no established fertility preservation options for pre-pubertal boys facing cancer treatment. Granulocyte-colony stimulating factor (G-CSF) treatment has been proposed to be chemoprotective against spermatogonial cell loss in an alkylating chemotherapy model of busulfan treated adult mice. Having previously shown that exposure to the alkylating-like chemotherapy cisplatin resulted in a reduction in germ cell numbers in immature human testicular tissues, we here investigate whether G-CSF would prevent cisplatin-induced germ cell loss in immature human and mouse (fetal and pre-pubertal) testicular tissues. METHODS: Organotypic in vitro culture systems were utilised to determine the effects of clinically-relevant concentrations of G-CSF in cisplatin-exposed immature testicular tissues. Human fetal (n = 14 fetuses) and mouse pre-pubertal (n = 4 litters) testicular tissue pieces were cultured and exposed to cisplatin or vehicle control for 24 hrs and analysed at 72 and 240 hrs post-exposure. Combined G-CSF and cisplatin exposure groups explored varying concentrations and duration of G-CSF supplementation to the culture medium (including groups receiving G-CSF before, during and after cisplatin exposure). In addition, effects of G-CSF supplementation alone were investigated. Survival of total germ cell and sub-populations were identified by expression of AP2γ and MAGE-A4 for human gonocytes and (pre)spermatogonia, respectively, and MVH and PLZF, for mouse germ cells and putative spermatogonial stem cells (SSCs) respectively, were quantified. RESULTS: Exposure to cisplatin resulted in a reduced germ cell number in human fetal and mouse pre-pubertal testicular tissues at 240 hrs post-exposure. Germ cell number was not preserved by combined exposure with G-CSF using any of the exposure regimens (prior to, during or after cisplatin exposure). Continuous supplementation with G-CSF alone for 14 days did not change the germ cell composition in either human or mouse immature testicular tissues. CONCLUSIONS: This study demonstrates that exposure to G-CSF does not prevent cisplatin-induced germ cell loss in immature human and mouse testicular tissues in an in vitro system.


Assuntos
Cisplatino , Testículo , Masculino , Humanos , Animais , Camundongos , Testículo/metabolismo , Cisplatino/farmacologia , Espermatogônias , Fator Estimulador de Colônias de Granulócitos/farmacologia , Fator Estimulador de Colônias de Granulócitos/metabolismo , Fatores Estimuladores de Colônias/metabolismo , Fatores Estimuladores de Colônias/farmacologia , Granulócitos
9.
J Neurochem ; 165(3): 379-390, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-36815399

RESUMO

Dietary lipids, particularly omega-3 polyunsaturated fatty acids, are speculated to impact behaviors linked to the dopaminergic system, such as movement and control of circadian rhythms. However, the ability to draw a direct link between dopaminergic omega-3 fatty acid metabolism and behavioral outcomes has been limited to the use of diet-based approaches, which are confounded by systemic effects. Here, neuronal lipid metabolism was targeted in a diet-independent manner by manipulation of long-chain acyl-CoA synthetase 6 (ACSL6) expression. ACSL6 performs the initial reaction for cellular fatty acid metabolism and prefers the omega-3 polyunsaturated fatty acid, docosahexaenoic acid (DHA). The loss of Acsl6 in mice (Acsl6-/- ) depletes neuronal membranes of DHA content and results in phenotypes linked to dopaminergic control, such as hyperlocomotion, impaired short-term spatial memory, and imbalances in dopamine neurochemistry. To investigate the role of dopaminergic ACSL6 on these outcomes, a dopaminergic neuron-specific ACSL6 knockout mouse was generated (Acsl6DA-/- ). Acsl6DA-/- mice demonstrated hyperlocomotion and imbalances in striatal dopamine neurochemistry. Circadian rhythms of both the Acsl6-/- and the Acsl6DA-/- mice were similar to control mice under basal conditions. However, upon light entrainment, a mimetic of jet lag, both the complete knockout of ACSL6 and the dopaminergic-neuron-specific loss of ACSL6 resulted in a longer recovery to entrainment compared to control mice. In conclusion, these data demonstrate that ACSL6 in dopaminergic neurons alters dopamine metabolism and regulation of light entrainment suggesting that DHA metabolism mediated by ACSL6 plays a role in dopamine neuron biology.


Assuntos
Neurônios Dopaminérgicos , Metabolismo dos Lipídeos , Camundongos , Animais , Neurônios Dopaminérgicos/metabolismo , Dopamina , Gorduras na Dieta , Dieta , Camundongos Knockout , Ácidos Docosa-Hexaenoicos/metabolismo , Coenzima A Ligases/genética , Coenzima A Ligases/metabolismo
10.
Elife ; 122023 01 31.
Artigo em Inglês | MEDLINE | ID: mdl-36719274

RESUMO

Reconstitution of germ cell fate from pluripotent stem cells provides an opportunity to understand the molecular underpinnings of germ cell development. Here, we established robust methods for induced pluripotent stem cell (iPSC) culture in the common marmoset (Callithrix jacchus [cj]), allowing stable propagation in an undifferentiated state. Notably, iPSCs cultured on a feeder layer in the presence of a WNT signaling inhibitor upregulated genes related to ubiquitin-dependent protein catabolic processes and enter a permissive state that enables differentiation into primordial germ cell-like cells (PGCLCs) bearing immunophenotypic and transcriptomic similarities to pre-migratory cjPGCs in vivo. Induction of cjPGCLCs is accompanied by transient upregulation of mesodermal genes, culminating in the establishment of a primate-specific germline transcriptional network. Moreover, cjPGCLCs can be expanded in monolayer while retaining the germline state. Upon co-culture with mouse testicular somatic cells, these cells acquire an early prospermatogonia-like phenotype. Our findings provide a framework for understanding and reconstituting marmoset germ cell development in vitro, thus providing a comparative tool and foundation for a preclinical modeling of human in vitro gametogenesis.


Assuntos
Células-Tronco Pluripotentes Induzidas , Células-Tronco Pluripotentes , Animais , Camundongos , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Callithrix , Diferenciação Celular , Células-Tronco Pluripotentes/metabolismo , Células Germinativas/metabolismo
11.
Development ; 149(22)2022 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-36250451

RESUMO

In mammalian testes, premeiotic spermatogonia respond to retinoic acid by completing an essential lengthy differentiation program before initiating meiosis. The molecular and cellular changes directing these developmental processes remain largely undefined. This wide gap in knowledge is due to two unresolved technical challenges: (1) lack of robust and reliable in vitro models to study differentiation and meiotic initiation; and (2) lack of methods to isolate large and pure populations of male germ cells at each stage of differentiation and at meiotic initiation. Here, we report a facile in vitro differentiation and meiotic initiation system that can be readily manipulated, including the use of chemical agents that cannot be safely administered to live animals. In addition, we present a transgenic mouse model enabling fluorescence-activated cell sorting-based isolation of millions of spermatogonia at specific developmental stages as well as meiotic spermatocytes.


Assuntos
Espermatogênese , Espermatogônias , Masculino , Camundongos , Animais , Espermatócitos , Testículo , Meiose , Diferenciação Celular , Camundongos Transgênicos , Mamíferos
12.
PLoS Genet ; 18(9): e1010416, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-36129965

RESUMO

Control over gene expression is exerted, in multiple stages of spermatogenesis, at the post-transcriptional level by RNA binding proteins (RBPs). We identify here an essential role in mammalian spermatogenesis and male fertility for 'RNA binding protein 46' (RBM46). A highly evolutionarily conserved gene, Rbm46 is also essential for fertility in both flies and fish. We found Rbm46 expression was restricted to the mouse germline, detectable in males in the cytoplasm of premeiotic spermatogonia and meiotic spermatocytes. To define its requirement for spermatogenesis, we generated Rbm46 knockout (KO, Rbm46-/-) mice; although male Rbm46-/- mice were viable and appeared grossly normal, they were infertile. Testes from adult Rbm46-/- mice were small, with seminiferous tubules containing only Sertoli cells and few undifferentiated spermatogonia. Using genome-wide unbiased high throughput assays RNA-seq and 'enhanced crosslinking immunoprecipitation' coupled with RNA-seq (eCLIP-seq), we discovered RBM46 could bind, via a U-rich conserved consensus sequence, to a cohort of mRNAs encoding proteins required for completion of differentiation and subsequent meiotic initiation. In summary, our studies support an essential role for RBM46 in regulating target mRNAs during spermatogonia differentiation prior to the commitment to meiosis in mice.


Assuntos
Proteínas de Ligação a RNA/metabolismo , Espermatogênese , Espermatogônias , Animais , Diferenciação Celular/genética , Masculino , Mamíferos/genética , Meiose/genética , Camundongos , Camundongos Knockout , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Proteínas de Ligação a RNA/genética , Espermatócitos/metabolismo , Espermatogênese/genética , Espermatogônias/metabolismo , Testículo
13.
JCI Insight ; 6(11)2021 06 08.
Artigo em Inglês | MEDLINE | ID: mdl-34100386

RESUMO

The omega-3 fatty acid docosahexaenoic acid (DHA) inversely relates to neurological impairments with aging; however, limited nondietary models manipulating brain DHA have hindered a direct linkage. We discovered that loss of long-chain acyl-CoA synthetase 6 in mice (Acsl6-/-) depletes brain membrane phospholipid DHA levels, independent of diet. Here, Acsl6-/- brains contained lower DHA compared with controls across the life span. The loss of DHA- and increased arachidonate-enriched phospholipids were visualized by MALDI imaging predominantly in neuron-rich regions where single-molecule RNA in situ hybridization localized Acsl6 to neurons. ACSL6 is also astrocytic; however, we found that astrocyte-specific ACSL6 depletion did not alter membrane DHA because astrocytes express a non-DHA-preferring ACSL6 variant. Across the life span, Acsl6-/- mice exhibited hyperlocomotion, impairments in working spatial memory, and increased cholesterol biosynthesis genes. Aging caused Acsl6-/- brains to decrease the expression of membrane, bioenergetic, ribosomal, and synaptic genes and increase the expression of immune response genes. With age, the Acsl6-/- cerebellum became inflamed and gliotic. Together, our findings suggest that ACSL6 promotes membrane DHA enrichment in neurons, but not in astrocytes, and is important for neuronal DHA levels across the life span. The loss of ACSL6 impacts motor function, memory, and age-related neuroinflammation, reflecting the importance of neuronal ACSL6-mediated lipid metabolism across the life span.


Assuntos
Envelhecimento/genética , Encéfalo/metabolismo , Coenzima A Ligases/genética , Ácidos Docosa-Hexaenoicos/metabolismo , Neuroproteção/genética , Envelhecimento/metabolismo , Envelhecimento/patologia , Envelhecimento/fisiologia , Animais , Encéfalo/patologia , Cerebelo/metabolismo , Cerebelo/patologia , Colesterol/biossíntese , Coenzima A Ligases/metabolismo , Expressão Gênica , Gliose/genética , Gliose/metabolismo , Gliose/patologia , Locomoção/fisiologia , Memória de Curto Prazo/fisiologia , Camundongos , Camundongos Knockout , Doenças Neuroinflamatórias/metabolismo , Memória Espacial/fisiologia , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz
14.
Reproduction ; 161(6): 645-655, 2021 05 05.
Artigo em Inglês | MEDLINE | ID: mdl-33835049

RESUMO

Initiation of spermatogonial differentiation in the mouse testis begins with the response to retinoic acid (RA) characterized by activation of KIT and STRA8 expression. In the adult, spermatogonial differentiation is spatiotemporally coordinated by a pulse of RA every 8.6 days that is localized to stages VII-VIII of the seminiferous epithelial cycle. Dogmatically, progenitor spermatogonia that express retinoic acid receptor gamma (RARG) at these stages will differentiate in response to RA, but this has yet to be tested functionally. Previous single-cell RNA-seq data identified phenotypically and functionally distinct subsets of spermatogonial stem cells (SSCs) and progenitor spermatogonia, where late progenitor spermatogonia were defined by expression of RARG and Dppa3. Here, we found late progenitor spermatogonia (RARGhigh KIT-) were further divisible into two subpopulations based on Dppa3 reporter expression (Dppa3-ECFP or Dppa3-EGFP) and were observed across all stages of the seminiferous epithelial cycle. However, nearly all Dppa3+ spermatogonia were differentiating (KIT+) late in the seminiferous epithelial cycle (stages X-XII), while Dppa3- late progenitors remained abundant, suggesting that Dppa3+ and Dppa3- late progenitors differentially responded to RA. Following acute RA treatment (2-4 h), significantly more Dppa3+ late progenitors induced KIT, including at the midpoint of the cycle (stages VI-IX), than Dppa3- late progenitors. Subsequently, single-cell analyses indicated a subset of Dppa3+ late progenitors expressed higher levels of Rxra, which we confirmed by RXRA whole-mount immunostaining. Together, these results indicate RARG alone is insufficient to initiate a spermatogonial response to RA in the adult mouse testis and suggest differential RXRA expression may discriminate responding cells.


Assuntos
Células-Tronco Germinativas Adultas/metabolismo , Proteínas Cromossômicas não Histona/metabolismo , Receptores do Ácido Retinoico/metabolismo , Receptor X Retinoide alfa/metabolismo , Espermatogênese , Espermatogônias/metabolismo , Tretinoína/farmacologia , Células-Tronco Germinativas Adultas/citologia , Células-Tronco Germinativas Adultas/efeitos dos fármacos , Animais , Antineoplásicos/farmacologia , Proteínas Cromossômicas não Histona/genética , Masculino , Camundongos , Receptores do Ácido Retinoico/genética , Receptor X Retinoide alfa/genética , Espermatogônias/citologia , Espermatogônias/efeitos dos fármacos , Receptor gama de Ácido Retinoico
15.
Cell Rep ; 34(7): 108752, 2021 02 16.
Artigo em Inglês | MEDLINE | ID: mdl-33596419

RESUMO

Spermatogonial stem cells (SSCs) sustain spermatogenesis by balancing self-renewal and initiation of differentiation to produce progenitor spermatogonia committed to forming sperm. To define the regulatory logic among SSCs and progenitors, we performed single-cell RNA velocity analyses and validated results in vivo. A predominant quiescent SSC population spawns a small subset of cell-cycle-activated SSCs via mitogen-activated protein kinase (MAPK)/AKT signaling. Activated SSCs form early progenitors and mTORC1 inhibition drives activated SSC accumulation consistent with blockade to progenitor formation. Mechanistically, mTORC1 inhibition suppresses transcription among spermatogonia and specifically alters expression of insulin growth factor (IGF) signaling in early progenitors. Tex14-/- testes lacking intercellular bridges do not accumulate activated SSCs following mTORC1 inhibition, indicating that steady-state mTORC1 signaling drives activated SSCs to produce progenitor clones. These results are consistent with a model of SSC self-renewal dependent on interconversion between activated and quiescent SSCs, and mTORC1-dependent initiation of differentiation from SSCs to progenitor clones.


Assuntos
Células-Tronco Germinativas Adultas/fisiologia , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , Espermatogônias/fisiologia , Células-Tronco Germinativas Adultas/citologia , Células-Tronco Germinativas Adultas/metabolismo , Animais , Diferenciação Celular/fisiologia , Células Cultivadas , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Transdução de Sinais , Espermatogônias/metabolismo
16.
Nat Commun ; 11(1): 5656, 2020 11 09.
Artigo em Inglês | MEDLINE | ID: mdl-33168808

RESUMO

Establishment of spermatogonia throughout the fetal and postnatal period is essential for production of spermatozoa and male fertility. Here, we establish a protocol for in vitro reconstitution of human prospermatogonial specification whereby human primordial germ cell (PGC)-like cells differentiated from human induced pluripotent stem cells are further induced into M-prospermatogonia-like cells and T1 prospermatogonia-like cells (T1LCs) using long-term cultured xenogeneic reconstituted testes. Single cell RNA-sequencing is used to delineate the lineage trajectory leading to T1LCs, which closely resemble human T1-prospermatogonia in vivo and exhibit gene expression related to spermatogenesis and diminished proliferation, a hallmark of quiescent T1 prospermatogonia. Notably, this system enables us to visualize the dynamic and stage-specific regulation of transposable elements during human prospermatogonial specification. Together, our findings pave the way for understanding and reconstructing human male germline development in vitro.


Assuntos
Células Germinativas Embrionárias/fisiologia , Células-Tronco Pluripotentes Induzidas/fisiologia , Espermatogênese/genética , Espermatogênese/fisiologia , Animais , Diferenciação Celular , Epigenômica , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Masculino , Camundongos , Camundongos Endogâmicos ICR , Elementos Reguladores de Transcrição , Análise de Sequência de RNA , Espermatogônias/citologia , Espermatozoides , Testículo/citologia , Transcriptoma
17.
iScience ; 23(10): 101596, 2020 Oct 23.
Artigo em Inglês | MEDLINE | ID: mdl-33083754

RESUMO

Spermatogonial stem cells (SSCs) both self-renew and give rise to progenitors that initiate spermatogenic differentiation in the mammalian testis. Questions remain regarding the extent to which the SSC and progenitor states are functionally distinct. Here we provide the first multiparametric integrative analysis of mammalian germ cell epigenomes comparable with that done for >100 somatic cell types by the ENCODE Project. Differentially expressed genes distinguishing SSC- and progenitor-enriched spermatogonia showed distinct histone modification patterns, particularly for H3K27ac and H3K27me3. Motif analysis predicted transcription factors that may regulate spermatogonial subtype-specific fate, and immunohistochemistry and gene-specific chromatin immunoprecipitation analyses confirmed subtype-specific differences in target gene binding of a subset of these factors. Taken together, these results show that SSCs and progenitors display distinct epigenetic profiling consistent with these spermatogonial subtypes being differentially programmed to either self-renew and maintain regenerative capacity as SSCs or lose regenerative capacity and initiate lineage commitment as progenitors.

18.
J Biol Chem ; 294(39): 14394-14405, 2019 09 27.
Artigo em Inglês | MEDLINE | ID: mdl-31399511

RESUMO

Docosahexaenoic acid (DHA) is an ω-3 dietary-derived polyunsaturated fatty acid of marine origin enriched in testes and necessary for normal fertility, yet the mechanisms regulating the enrichment of DHA in the testes remain unclear. Long-chain ACSL6 (acyl-CoA synthetase isoform 6) activates fatty acids for cellular anabolic and catabolic metabolism by ligating a CoA to a fatty acid, is highly expressed in testes, and has high preference for DHA. Here, we investigated the role of ACSL6 for DHA enrichment in the testes and its requirement for male fertility. Acsl6-/- males were severely subfertile with smaller testes, reduced cauda epididymal sperm counts, germ cell loss, and disorganization of the seminiferous epithelium. Total fatty acid profiling of Acsl6-/- testes revealed reduced DHA and increased ω-6 arachidonic acid, a fatty acid profile also reflected in phospholipid composition. Strikingly, lipid imaging demonstrated spatial redistribution of phospholipids in Acsl6-/- testes. Arachidonic acid-containing phospholipids were predominantly interstitial in control testes but diffusely localized across Acsl6-/- testes. In control testes, DHA-containing phospholipids were predominantly within seminiferous tubules, which contain Sertoli cells and spermatogenic cells but relocalized to the interstitium in Acsl6-/- testes. Taken together, these data demonstrate that ACSL6 is an initial driving force for germ cell DHA enrichment and is required for normal spermatogenesis and male fertility.


Assuntos
Coenzima A Ligases/genética , Ácidos Graxos Ômega-6/metabolismo , Infertilidade Masculina/genética , Túbulos Seminíferos/metabolismo , Animais , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Fosfolipídeos/metabolismo , Túbulos Seminíferos/citologia , Espermatogênese
19.
Biol Reprod ; 101(3): 617-634, 2019 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-31077285

RESUMO

Mammalian spermatogenesis is a complex developmental program that transforms mitotic testicular germ cells (spermatogonia) into mature male gametes (sperm) for production of offspring. For decades, it has been known that this several-weeks-long process involves a series of highly ordered and morphologically recognizable cellular changes as spermatogonia proliferate, spermatocytes undertake meiosis, and spermatids develop condensed nuclei, acrosomes, and flagella. Yet, much of the underlying molecular logic driving these processes has remained opaque because conventional characterization strategies often aggregated groups of cells to meet technical requirements or due to limited capability for cell selection. Recently, a cornucopia of single-cell transcriptome studies has begun to lift the veil on the full compendium of gene expression phenotypes and changes underlying spermatogenic development. These datasets have revealed the previously obscured molecular heterogeneity among and between varied spermatogenic cell types and are reinvigorating investigation of testicular biology. This review describes the extent of available single-cell RNA-seq profiles of spermatogenic and testicular somatic cells, how those data were produced and evaluated, their present value for advancing knowledge of spermatogenesis, and their potential future utility at both the benchtop and bedside.


Assuntos
Mamíferos/genética , RNA-Seq/métodos , Análise de Célula Única/métodos , Espermatogênese/genética , Animais , Humanos , Masculino , Transcriptoma/fisiologia , Pesquisa Translacional Biomédica/métodos , Pesquisa Translacional Biomédica/tendências
20.
Development ; 146(12)2019 05 13.
Artigo em Inglês | MEDLINE | ID: mdl-31023878

RESUMO

In the mammalian testis, sustained spermatogenesis relies on spermatogonial stem cells (SSCs); their progeny either remain as stem cells (self-renewal) or proliferate and differentiate to enter meiosis in response to retinoic acid (RA). Here, we sought to uncover elusive mechanisms regulating a key switch fundamental to spermatogonial fate: the capacity of spermatogonia to respond to RA. Using the developing mouse testis as a model, we found that spermatogonia and precursor prospermatogonia exhibit a heterogeneous capacity to respond to RA with at least two underlying causes. First, progenitor spermatogonia are prevented from responding to RA by catabolic activity of cytochrome P450 family 26 enzymes. Second, a smaller subset of undifferentiated spermatogonia enriched for SSCs exhibit catabolism-independent RA insensitivity. Moreover, for the first time, we observed that precursor prospermatogonia are heterogeneous and comprise subpopulations that exhibit the same differential RA responsiveness found in neonatal spermatogonia. We propose a novel model by which mammalian prospermatogonial and spermatogonial fates are regulated by their intrinsic capacity to respond (or not) to the differentiation signal provided by RA before, and concurrent with, the initiation of spermatogenesis.


Assuntos
Regulação da Expressão Gênica , Espermatogênese , Espermatogônias/citologia , Células-Tronco/citologia , Testículo/crescimento & desenvolvimento , Tretinoína/metabolismo , Animais , Diferenciação Celular , Linhagem da Célula , Família 26 do Citocromo P450/metabolismo , Genômica , Proteínas de Fluorescência Verde/metabolismo , Masculino , Meiose , Camundongos , Células de Sertoli/citologia , Transdução de Sinais , Testículo/embriologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA