Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
Dev Biol ; 509: 11-27, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38311163

RESUMEN

Undifferentiated spermatogonia are composed of a heterogeneous cell population including spermatogonial stem cells (SSCs). Molecular mechanisms underlying the regulation of various spermatogonial cohorts during their self-renewal and differentiation are largely unclear. Here we show that AKT1S1, an AKT substrate and inhibitor of mTORC1, regulates the homeostasis of undifferentiated spermatogonia. Although deletion of Akt1s1 in mouse appears not grossly affecting steady-state spermatogenesis and male mice are fertile, the subset of differentiation-primed OCT4+ spermatogonia decreased significantly, whereas self-renewing GFRα1+ and proliferating PLZF+ spermatogonia were sustained. Both neonatal prospermatogonia and the first wave spermatogenesis were greatly reduced in Akt1s1-/- mice. Further analyses suggest that OCT4+ spermatogonia in Akt1s1-/- mice possess altered PI3K/AKT-mTORC1 signaling, gene expression and carbohydrate metabolism, leading to their functionally compromised developmental potential. Collectively, these results revealed an important role of AKT1S1 in mediating the stage-specific signals that regulate the self-renewal and differentiation of spermatogonia during mouse spermatogenesis.


Asunto(s)
Proteínas Proto-Oncogénicas c-akt , Espermatogonias , Masculino , Animales , Ratones , Proteínas Proto-Oncogénicas c-akt/metabolismo , Testículo/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Espermatogénesis/genética , Diferenciación Celular/fisiología , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo
2.
BMC Genomics ; 25(1): 464, 2024 May 13.
Artículo en Inglés | MEDLINE | ID: mdl-38741085

RESUMEN

Gonad development includes sex determination and divergent maturation of the testes and ovaries. Recent advances in measuring gene expression in single cells are providing new insights into this complex process. However, the underlying epigenetic regulatory mechanisms remain unclear. Here, we profiled chromatin accessibility in mouse gonadal cells of both sexes from embryonic day 11.5 to 14.5 using single-cell assay for transposase accessible chromatin by sequencing (scATAC-seq). Our results showed that individual cell types can be inferred by the chromatin landscape, and that cells can be temporally ordered along developmental trajectories. Integrative analysis of transcriptomic and chromatin-accessibility maps identified multiple putative regulatory elements proximal to key gonadal genes Nr5a1, Sox9 and Wt1. We also uncover cell type-specific regulatory factors underlying cell type specification. Overall, our results provide a better understanding of the epigenetic landscape associated with the progressive restriction of cell fates in the gonad.


Asunto(s)
Linaje de la Célula , Cromatina , Gónadas , Factor de Transcripción SOX9 , Análisis de la Célula Individual , Animales , Cromatina/metabolismo , Cromatina/genética , Ratones , Linaje de la Célula/genética , Femenino , Masculino , Factor de Transcripción SOX9/genética , Factor de Transcripción SOX9/metabolismo , Gónadas/metabolismo , Gónadas/citología , Gónadas/embriología , Factor Esteroidogénico 1/genética , Factor Esteroidogénico 1/metabolismo , Proteínas WT1/genética , Proteínas WT1/metabolismo , Testículo/metabolismo , Testículo/citología , Epigénesis Genética , Regulación del Desarrollo de la Expresión Génica , Ovario/metabolismo , Ovario/citología
3.
Allergy ; 2024 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-38727640

RESUMEN

BACKGROUND: Meteorin-like protein (METRNL)/Interleukin-41 (IL-41) is a novel immune-secreted cytokine/myokine involved in several inflammatory diseases. However, how METRNL exerts its regulatory properties on skin inflammation remains elusive. This study aims to elucidate the functionality and regulatory mechanism of METRNL in atopic dermatitis (AD). METHODS: METRNL levels were determined in skin and serum samples from patients with AD and subsequently verified in the vitamin D3 analogue MC903-induced AD-like mice model. The cellular target of METRNL activity was identified by multiplex immunostaining, single-cell RNA-seq and RNA-seq. RESULTS: METRNL was significantly upregulated in lesions and serum of patients with dermatitis compared to healthy controls (p <.05). Following repeated MC903 exposure, AD model mice displayed elevated levels of METRNL in both ears and serum. Administration of recombinant murine METRNL protein (rmMETRNL) ameliorated allergic skin inflammation and hallmarks of AD in mice, whereas blocking of METRNL signaling led to the opposite. METRNL enhanced ß-Catenin activation, limited the expression of Th2-related molecules that attract the accumulation of Arginase-1 (Arg1)hi macrophages, dendritic cells, and activated mast cells. CONCLUSIONS: METRNL can bind to KIT receptor and subsequently alleviate the allergic inflammation of AD by inhibiting the expansion of immune cells, and downregulating inflammatory gene expression by regulating the level of active WNT pathway molecule ß-Catenin.

4.
PLoS Genet ; 17(7): e1009369, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34237055

RESUMEN

Spermatogonial stem cells (SSC), the foundation of spermatogenesis and male fertility, possess lifelong self-renewal activity. Aging leads to the decline in stem cell function and increased risk of paternal age-related genetic diseases. In the present study, we performed a comparative genomic analysis of mouse SSC-enriched undifferentiated spermatogonia (Oct4-GFP+/KIT-) and differentiating progenitors (Oct4-GFP+/KIT+) isolated from young and aged testes. Our transcriptome data revealed enormous complexity of expressed coding and non-coding RNAs and alternative splicing regulation during SSC differentiation. Further comparison between young and aged undifferentiated spermatogonia suggested these differentiation programs were affected by aging. We identified aberrant expression of genes associated with meiosis and TGF-ß signaling, alteration in alternative splicing regulation and differential expression of specific lncRNAs such as Fendrr. Epigenetic profiling revealed reduced H3K27me3 deposition at numerous pro-differentiation genes during SSC differentiation as well as aberrant H3K27me3 distribution at genes in Wnt and TGF-ß signaling upon aging. Finally, aged undifferentiated spermatogonia exhibited gene body hypomethylation, which is accompanied by an elevated 5hmC level. We believe this in-depth molecular analysis will serve as a reference for future analysis of SSC aging.


Asunto(s)
Células Madre Germinales Adultas/citología , Células Madre Germinales Adultas/fisiología , Envejecimiento/fisiología , Epigenoma , 5-Metilcitosina/metabolismo , Envejecimiento/genética , Empalme Alternativo , Animales , Diferenciación Celular , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Lisina/genética , Lisina/metabolismo , Masculino , Ratones Endogámicos C57BL , Ratones Transgénicos , ARN Largo no Codificante/genética , Testículo/citología
5.
Development ; 146(6)2019 03 27.
Artículo en Inglés | MEDLINE | ID: mdl-30824552

RESUMEN

Neonatal germ cell development provides the foundation of spermatogenesis. However, a systematic understanding of this process is still limited. To resolve cellular and molecular heterogeneity in this process, we profiled single cell transcriptomes of undifferentiated germ cells from neonatal mouse testes and employed unbiased clustering and pseudotime ordering analysis to assign cells to distinct cell states in the developmental continuum. We defined the unique transcriptional programs underlying migratory capacity, resting cellular states and apoptosis regulation in transitional gonocytes. We also identified a subpopulation of primitive spermatogonia marked by CD87 (plasminogen activator, urokinase receptor), which exhibited a higher level of self-renewal gene expression and migration potential. We further revealed a differentiation-primed state within the undifferentiated compartment, in which elevated Oct4 expression correlates with lower expression of self-renewal pathway factors, higher Rarg expression, and enhanced retinoic acid responsiveness. Lastly, a knockdown experiment revealed the role of Oct4 in the regulation of gene expression related to the MAPK pathway and cell adhesion, which may contribute to stem cell differentiation. Our study thus provides novel insights into cellular and molecular regulation during early germ cell development.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Análisis de Secuencia de ARN , Espermatogonias/citología , Animales , Animales Recién Nacidos , Apoptosis , Adhesión Celular , Diferenciación Celular , Perfilación de la Expresión Génica , Sistema de Señalización de MAP Quinasas , Masculino , Ratones , Microscopía Fluorescente , Factor 3 de Transcripción de Unión a Octámeros/fisiología , Receptores de Ácido Retinoico/fisiología , Receptores del Activador de Plasminógeno Tipo Uroquinasa/fisiología , Espermatogénesis/genética , Transcriptoma , Tretinoina/fisiología , Receptor de Ácido Retinoico gamma
6.
Exp Cell Res ; 373(1-2): 71-79, 2018 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-30266657

RESUMEN

During spermatogenesis, a group of undifferentiated spermatogonia undergoes an essential transition to a differentiating stage, which involves gain of Kit receptor. In the current study, we showed that a small non-coding RNA, miRNA-26b could induce transition from Kit- to Kit+ and inhibit proliferation of spermatogonia. A key transcriptional factor for undifferentiated spermatogonia, Plzf, was proven as a direct target of miR-26b. When undifferentiated spermatogonia were treated with Retinoic acid (RA), miR-26b was increased, further promoting RA-induced differentiation of spermatogonia. In addition, miR-26b could repress 5-hydroxymethylcytosine (5hmC) via repression of Tet3 in spermatogonia. These findings demonstrate that miR-26b might play a role in promoting the transition from Kit- to Kit+ SSCs.


Asunto(s)
MicroARNs/fisiología , Espermatogénesis , Espermatogonias/metabolismo , 5-Metilcitosina/análogos & derivados , 5-Metilcitosina/metabolismo , Animales , Apoptosis , Diferenciación Celular/efectos de los fármacos , Proliferación Celular , Células Cultivadas , Proteínas de Unión al ADN/metabolismo , Dioxigenasas , Masculino , Ratones , MicroARNs/metabolismo , Proteína de la Leucemia Promielocítica con Dedos de Zinc/genética , Proteína de la Leucemia Promielocítica con Dedos de Zinc/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-kit/análisis , Espermatogonias/citología , Espermatogonias/efectos de los fármacos , Tretinoina/farmacología
7.
Kidney Int ; 93(1): 173-187, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29042082

RESUMEN

Src activation has been associated with fibrogenesis after kidney injury. Macrophage-myofibroblast transition is a newly identified process to generate collagen-producing myofibroblasts locally in the kidney undergoing fibrosis in a TGF-ß/Smad3-dependent manner. The potential role of the macrophage-myofibroblast transition in Src-mediated renal fibrosis is unknown. In studying this by RNA sequencing at single-cell resolution, we uncovered a unique Src-centric regulatory gene network as a key underlying mechanism of macrophage-myofibroblast transition. A total of 501 differentially expressed genes associated with macrophage-myofibroblast transition were identified. However, Smad3-knockout largely reduced the transcriptome diversity. More importantly, inhibition of Src largely suppresses ureteral obstruction-induced macrophage-myofibroblast transition in the injured kidney in vivo along with transforming growth factor-ß1-induced elongated fibroblast-like morphology, α-smooth muscle actin expression and collagen production in bone marrow derived macrophages in vitro. Unexpectedly, we further uncovered that Src serves as a direct Smad3 target gene and also specifically up-regulated in macrophages during macrophage-myofibroblast transition. Thus, macrophage-myofibroblast transition contributes to Src-mediated tissue fibrosis. Hence, targeting Src may represent as a precision therapeutic strategy for macrophage-myofibroblast transition-driven fibrotic diseases.


Asunto(s)
Transdiferenciación Celular , Cicatriz/enzimología , Enfermedades Renales/enzimología , Riñón/enzimología , Macrófagos/enzimología , Miofibroblastos/enzimología , Familia-src Quinasas/metabolismo , Animales , Transdiferenciación Celular/efectos de los fármacos , Transdiferenciación Celular/genética , Células Cultivadas , Cicatriz/genética , Cicatriz/patología , Cicatriz/prevención & control , Modelos Animales de Enfermedad , Fibrosis , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Redes Reguladoras de Genes , Riñón/efectos de los fármacos , Riñón/patología , Enfermedades Renales/genética , Enfermedades Renales/patología , Enfermedades Renales/prevención & control , Macrófagos/efectos de los fármacos , Macrófagos/patología , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Miofibroblastos/efectos de los fármacos , Miofibroblastos/patología , Inhibidores de Proteínas Quinasas/farmacología , Análisis de Secuencia de ARN , Transducción de Señal , Análisis de la Célula Individual , Proteína smad3/genética , Proteína smad3/metabolismo , Obstrucción Ureteral/tratamiento farmacológico , Obstrucción Ureteral/enzimología , Obstrucción Ureteral/genética , Familia-src Quinasas/genética
8.
Nucleic Acids Res ; 43(16): 7805-22, 2015 Sep 18.
Artículo en Inglés | MEDLINE | ID: mdl-26130713

RESUMEN

Ten eleven translocation (Tet) family-mediated DNA oxidation on 5-methylcytosine (5mC) to 5-hydroxymethylcytosine (5hmC) represents a novel epigenetic modification that regulates dynamic gene expression during embryonic stem cells (ESCs) differentiation. Through the role of Tet on 5hmC regulation in stem cell development is relatively defined, how the Tet family is regulated and impacts on ESCs lineage development remains elusive. In this study, we show non-coding RNA regulation on Tet family may contribute to epigenetic regulation during ESCs differentiation, which is suggested by microRNA-29b (miR-29b) binding sites on the Tet1 3' untranslated region (3' UTR). We demonstrate miR-29b increases sharply after embyoid body (EB) formation, which causes Tet1 repression and reduction of cellular 5hmC level during ESCs differentiation. Importantly, we show this miR-29b/Tet1 regulatory axis promotes the mesendoderm lineage formation both in vitro and in vivo by inducing the Nodal signaling pathway and repressing the key target of the active demethylation pathway, Tdg. Taken together, our findings underscore the contribution of small non-coding RNA mediated regulation on DNA demethylation dynamics and the differential expressions of key mesendoderm regulators during ESCs lineage specification. MiR-29b could potentially be applied to enrich production of mesoderm and endoderm derivatives and be further differentiated into desired organ-specific cells.


Asunto(s)
Diferenciación Celular/genética , Proteínas de Unión al ADN/metabolismo , Epigénesis Genética , MicroARNs/metabolismo , Células Madre Embrionarias de Ratones/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , 5-Metilcitosina/análogos & derivados , Animales , Células Cultivadas , Citosina/análogos & derivados , Citosina/metabolismo , Proteínas de Unión al ADN/antagonistas & inhibidores , Proteínas de Unión al ADN/genética , Dioxigenasas , Ectodermo/citología , Cuerpos Embrioides/citología , Endodermo/citología , Células HEK293 , Humanos , Factores de Determinación Derecha-Izquierda/genética , Mesodermo/citología , Ratones , MicroARNs/biosíntesis , Células Madre Embrionarias de Ratones/citología , Proteínas Proto-Oncogénicas/antagonistas & inhibidores , Proteínas Proto-Oncogénicas/genética , Timina ADN Glicosilasa/metabolismo
9.
Mol Syst Biol ; 11(6): 814, 2015 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-26063786

RESUMEN

Coordination of cell division timing is crucial for proper cell fate specification and tissue growth. However, the differential regulation of cell division timing across or within cell types during metazoan development remains poorly understood. To elucidate the systems-level genetic architecture coordinating division timing, we performed a high-content screening for genes whose depletion produced a significant reduction in the asynchrony of division between sister cells (ADS) compared to that of wild-type during Caenorhabditis elegans embryogenesis. We quantified division timing using 3D time-lapse imaging followed by computer-aided lineage analysis. A total of 822 genes were selected for perturbation based on their conservation and known roles in development. Surprisingly, we find that cell fate determinants are not only essential for establishing fate asymmetry, but also are imperative for setting the ADS regardless of cellular context, indicating a common genetic architecture used by both cellular processes. The fate determinants demonstrate either coupled or separate regulation between the two processes. The temporal coordination appears to facilitate cell migration during fate specification or tissue growth. Our quantitative dataset with cellular resolution provides a resource for future analyses of the genetic control of spatial and temporal coordination during metazoan development.


Asunto(s)
Proteínas de Caenorhabditis elegans/biosíntesis , Diferenciación Celular/genética , División Celular/genética , Desarrollo Embrionario , Animales , Caenorhabditis elegans/embriología , Caenorhabditis elegans/genética , Linaje de la Célula/genética , Movimiento Celular , Regulación del Desarrollo de la Expresión Génica
10.
J Leukoc Biol ; 115(1): 19-35, 2024 01 05.
Artículo en Inglés | MEDLINE | ID: mdl-37675661

RESUMEN

Age-related immunosenescence is characterized by progressive dysfunction of adaptive immune response and increased autoimmunity. Nevertheless, the impact of aging on CD4+ regulatory T cells that are master regulators of the immune system remains largely unclear. Here, we report cellular and molecular hallmarks of regulatory T cells derived from murine lymphoid and adipose tissues at 3, 18, and 24 mo of age, respectively, by analyzing their heterogeneity that displays dynamic changes in transcriptomic effector signatures at a single-cell resolution. Although the proportion of regulatory T cells among total Cd4+ T cells, as well as their expression levels of Foxp3, did not show any global change with time, we have identified 6 transcriptomically distinct clusters of regulatory T cells with cross-tissue conserved hallmarks of aging, including increased numbers of proinflammatory regulatory T cells, reduced precursor cells, increased immature and mature T follicular regulatory cells potentially supported by a metabolic switch from oxidative phosphorylation to glycolysis, a gradual loss of CD150hi regulatory T cells that support hematopoiesis, and increased adipose tissue-specific regulatory T cells that are associated with metabolic disease. To dissect the impact of immunosenescence on humoral immunity, we propose some potential mechanisms underlying T follicular regulatory cell-mediated dysfunction by interactome analysis on T follicular regulatory cells, T follicular helper cells, and B cells during aging. Lastly, spatiotemporal analysis further revealed trajectories of regulatory T-cell aging that demonstrate the most significant changes in marrow and adipose tissues that might contribute to the development of age-related immunosenescence and type 2 diabetes. Taken together, our findings could provide a better understanding of age-associated regulatory T-cell heterogeneity in lymphoid and adipose tissues, as well as regulatory T-cell hallmarks during progressive adaptation to aging that could be therapeutically targeted for rejuvenating the aging immune system in the future.


Asunto(s)
Diabetes Mellitus Tipo 2 , Linfocitos T Reguladores , Ratones , Animales , Linfocitos T Colaboradores-Inductores , Diabetes Mellitus Tipo 2/metabolismo , Envejecimiento/genética , Perfilación de la Expresión Génica
11.
Elife ; 122023 04 25.
Artículo en Inglés | MEDLINE | ID: mdl-37096870

RESUMEN

Spermatogenesis depends on an orchestrated series of developing events in germ cells and full maturation of the somatic microenvironment. To date, the majority of efforts to study cellular heterogeneity in testis has been focused on single-cell gene expression rather than the chromatin landscape shaping gene expression. To advance our understanding of the regulatory programs underlying testicular cell types, we analyzed single-cell chromatin accessibility profiles in more than 25,000 cells from mouse developing testis. We showed that single-cell sequencing assay for transposase-accessible chromatin (scATAC-Seq) allowed us to deconvolve distinct cell populations and identify cis-regulatory elements (CREs) underlying cell-type specification. We identified sets of transcription factors associated with cell type-specific accessibility, revealing novel regulators of cell fate specification and maintenance. Pseudotime reconstruction revealed detailed regulatory dynamics coordinating the sequential developmental progressions of germ cells and somatic cells. This high-resolution dataset also unveiled previously unreported subpopulations within both the Sertoli and Leydig cell groups. Further, we defined candidate target cell types and genes of several genome-wide association study (GWAS) signals, including those associated with testosterone levels and coronary artery disease. Collectively, our data provide a blueprint of the 'regulon' of the mouse male germline and supporting somatic cells.


Asunto(s)
Cromatina , Testículo , Masculino , Embarazo , Femenino , Animales , Ratones , Cromatina/metabolismo , Testículo/metabolismo , Estudio de Asociación del Genoma Completo , Factores de Transcripción/metabolismo , Espermatogénesis/genética , Análisis de la Célula Individual
12.
Sci Rep ; 12(1): 21482, 2022 12 12.
Artículo en Inglés | MEDLINE | ID: mdl-36509798

RESUMEN

Spermatogonial stem cells are the most primitive spermatogonia in testis, which can self-renew to maintain the stem cell pool or differentiate to give rise to germ cells including haploid spermatids. All-trans-retinoic acid (RA), a bioactive metabolite of vitamin A, plays a fundamental role in initiating spermatogonial differentiation. In this study, single-cell ATAC-seq (scATAC-seq) was used to obtain genome-wide chromatin maps of cultured germline stem cells (GSCs) that were in control and RA-induced differentiation states. We showed that different subsets of GSCs can be distinguished based on chromatin accessibility of self-renewal and differentiation signature genes. Importantly, both progenitors and a subset of stem cells are able to respond to RA and give rise to differentiating cell subsets with distinct chromatin accessibility profiles. In this study, we identified regulatory regions that undergo chromatin remodeling and are associated with the retinoic signaling pathway. Moreover, we reconstructed the differentiation trajectory and identified novel transcription factor candidates enriched in different spermatogonia subsets. Collectively, our work provides a valuable resource for understanding the heterogeneity associated with differentiation and RA response in GSCs.


Asunto(s)
Espermatogénesis , Espermatogonias , Masculino , Humanos , Espermatogénesis/genética , Espermatogonias/metabolismo , Testículo/metabolismo , Diferenciación Celular , Tretinoina/farmacología , Tretinoina/metabolismo , Células Madre/metabolismo , Cromatina/genética , Cromatina/metabolismo
13.
Stem Cell Res Ther ; 13(1): 380, 2022 07 29.
Artículo en Inglés | MEDLINE | ID: mdl-35906661

RESUMEN

BACKGROUND: Tendon healing is clinically challenging largely due to its inferior regenerative capacity. We have previously prepared a soluble, DNA-free, urea-extracted bovine tendon-derived extracellular matrix (tECM) that exhibits strong pro-tenogenic bioactivity on human adipose-derived stem cells (hASCs). In this study, we aimed to elucidate the mechanism of tECM bioactivity via characterization of tECM protein composition and comparison of transcriptomic profiles of hASC cultures treated with tECM versus collagen type I (Col1) as a control ECM component. METHODS: The protein composition of tECM was characterized by SDS-PAGE, hydroxyproline assay, and proteomics analysis. To investigate tECM pro-tenogenic bioactivity and mechanism of action, differentiation of tECM-treated hASC cultures was compared to serum control medium or Col1-treated groups, as assessed via immunofluorescence for tenogenic markers and RNA Sequencing (RNA-Seq). RESULTS: Urea-extracted tECM yielded consistent protein composition, including collagens (20% w/w) and at least 17 non-collagenous proteins (< 100 kDa) based on MS analysis. Compared to current literature, tECM included key tendon ECM components that are functionally involved in tendon regeneration, as well as those that are involved in similar principal Gene Ontology (GO) functions (ECM-receptor interaction and collagen formation) and signaling pathways (ECM-receptor interaction and focal adhesion). When used as a cell culture supplement, tECM enhanced hASC proliferation and tenogenic differentiation compared to the Col1 and FBS treatment groups based on immunostaining of tenogenesis-associated markers. Furthermore, RNA-Seq analysis revealed a total of 584 genes differentially expressed among the three culture groups. Specifically, Col1-treated hASCs predominantly exhibited expression of genes and pathways related to ECM-associated processes, while tECM-treated hASCs expressed a mixture of ECM- and cell activity-associated processes, which may explain in part the enhanced proliferation and tenogenic differentiation of tECM-treated hASCs. CONCLUSIONS: Our findings showed that urea-extracted tECM contained 20% w/w collagens and is significantly enriched with other non-collagenous tendon ECM components. Compared to Col1 treatment, tECM supplementation enhanced hASC proliferation and tenogenic differentiation as well as induced distinct gene expression profiles. These findings provide insights into the potential mechanism of the pro-tenogenic bioactivity of tECM and support the development of future tECM-based approaches for tendon repair.


Asunto(s)
Células Madre , Transcriptoma , Animales , Bovinos , Diferenciación Celular , Colágeno/metabolismo , Matriz Extracelular/metabolismo , Humanos , Células Madre/metabolismo , Tendones/metabolismo , Ingeniería de Tejidos , Urea/metabolismo , Urea/farmacología
14.
Nat Commun ; 13(1): 2500, 2022 05 06.
Artículo en Inglés | MEDLINE | ID: mdl-35523793

RESUMEN

Maintenance of male fertility requires spermatogonial stem cells (SSCs) that self-renew and generate differentiating germ cells for production of spermatozoa. Germline cells are sensitive to genotoxic drugs and patients receiving chemotherapy can become infertile. SSCs surviving treatment mediate germline recovery but pathways driving SSC regenerative responses remain poorly understood. Using models of chemotherapy-induced germline damage and recovery, here we identify unique molecular features of regenerative SSCs and characterise changes in composition of the undifferentiated spermatogonial pool during germline recovery by single-cell analysis. Increased mitotic activity of SSCs mediating regeneration is accompanied by alterations in growth factor signalling including PI3K/AKT and mTORC1 pathways. While sustained mTORC1 signalling is detrimental for SSC maintenance, transient mTORC1 activation is critical for the regenerative response. Concerted inhibition of growth factor signalling disrupts core features of the regenerative state and limits germline recovery. We also demonstrate that the FOXM1 transcription factor is a target of growth factor signalling in undifferentiated spermatogonia and provide evidence for a role in regeneration. Our data confirm dynamic changes in SSC functional properties following damage and support an essential role for microenvironmental growth factors in promoting a regenerative state.


Asunto(s)
Fosfatidilinositol 3-Quinasas , Espermatogénesis , Diferenciación Celular/fisiología , Humanos , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Masculino , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Espermatogénesis/genética , Espermatogonias , Células Madre/metabolismo , Testículo/metabolismo
15.
Sci Adv ; 8(40): eabn5535, 2022 10 07.
Artículo en Inglés | MEDLINE | ID: mdl-36206343

RESUMEN

Tumor innervation is a common phenomenon with unknown mechanism. Here, we discovered a direct mechanism of tumor-associated macrophage (TAM) for promoting de novo neurogenesis via a subset showing neuronal phenotypes and pain receptor expression associated with cancer-driven nocifensive behaviors. This subset is rich in lung adenocarcinoma associated with poorer prognosis. By elucidating the transcriptome dynamics of TAM with single-cell resolution, we discovered a phenomenon "macrophage to neuron-like cell transition" (MNT) for directly promoting tumoral neurogenesis, evidenced by macrophage depletion and fate-mapping study in lung carcinoma models. Encouragingly, we detected neuronal phenotypes and activities of the bone marrow-derived MNT cells (MNTs) in vitro. Adoptive transfer of MNTs into NOD/SCID mice markedly enhanced their cancer-associated nocifensive behaviors. We identified macrophage-specific Smad3 as a pivotal regulator for promoting MNT at the genomic level; its disruption effectively blocked the tumor innervation and cancer-dependent nocifensive behaviors in vivo. Thus, MNT may represent a precision therapeutic target for cancer pain.


Asunto(s)
Dolor en Cáncer , Neoplasias Pulmonares , Animales , Dolor en Cáncer/metabolismo , Dolor en Cáncer/patología , Neoplasias Pulmonares/metabolismo , Macrófagos/metabolismo , Ratones , Ratones Endogámicos NOD , Ratones SCID , Neuronas , Análisis de Secuencia de ARN
16.
Cell Death Differ ; 26(11): 2194-2207, 2019 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-30760872

RESUMEN

Lethal (3) malignant brain tumor like 2 (L3MBTL2) is a member of the MBT-domain proteins, which are involved in transcriptional repression and implicated in chromatin compaction. Our previous study has shown that L3MBTL2 is highly expressed in the testis, but its role in spermatogenesis remains unclear. In the present study, we found that L3MBTL2 was most highly expressed in pachytene spermatocytes within the testis. Germ cell-specific ablation of L3mbtl2 in the testis led to increased abnormal spermatozoa, progressive decrease of sperm counts and premature testicular failure in mice. RNA-sequencing analysis on L3mbtl2 deficient testes confirmed that L3MBTL2 was a transcriptional repressor but failed to reveal any significant changes in spermatogenesis-associated genes. Interestingly, L3mbtl2 deficiency resulted in increased γH2AX deposition in the leptotene spermatocytes, subsequent inappropriate retention of γH2AX on autosomes, and defective crossing-over and synapsis during the pachytene stage of meiosis I, and more germ cell apoptosis and degeneration in aging mice. L3MBTL2 interacted with the histone ubiquitin ligase RNF8. Inhibition of L3MBTL2 reduced nuclear RNF8 and ubH2A levels in GC2 cells. L3mbtl2 deficiency led to decreases in the levels of the RNF8 and ubH2A pathway and in histone acetylation in elongating spermatids, and in protamine 1 deposition and chromatin condensation in sperm. These results suggest that L3MBTL2 plays important roles in chromatin remodeling during meiosis and spermiogenesis.


Asunto(s)
Ensamble y Desensamble de Cromatina/genética , Cromatina/metabolismo , Proteínas Nucleares/genética , Espermatocitos/metabolismo , Espermatogénesis/genética , Factores de Transcripción/genética , Acetilación , Animales , Apoptosis/genética , Ensamble y Desensamble de Cromatina/fisiología , Histonas/metabolismo , Masculino , Profase Meiótica I/fisiología , Ratones , Ratones Noqueados , Proteínas Nucleares/metabolismo , Fase Paquiteno/fisiología , Proteínas del Grupo Polycomb/metabolismo , Recuento de Espermatozoides , Testículo/metabolismo , Factores de Transcripción/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo
17.
Int J Biochem Cell Biol ; 107: 1-5, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30529019

RESUMEN

Single-cell parallel sequencing allows us to explore how genetic and epigenetic variations correlate of gene expression in the same cell. Beads-based approach and non-beads-based approach are the two present methods to separate DNA and RNA from the same cell. However, systematic difference between the two methods are lacking. In our study, we compared the performances of the two methods using transcriptome and methylome profiles generated simultaneously from single mouse oocytes. Our results showed that the beads-based approach could capture maximum quantity of mRNA but loss of DNA was inevitable, while the non-beads-based approach could obtain more DNA due to the undamaged nucleus obtained but at a cost of partial loss of mRNA. As the sequencing coverage of methylome sequencing in a single cell was relatively low, single-cell whole genome bisulfite sequencing (scWGBS) was preferable to generate the methylome map in single-cell parallel sequencing in comparison to single-cell reduced representation bisulfite sequencing (scRRBS). To the best of our knowledge, this is the first study to compare the two methods of single-cell parallel sequencing which offers a basic idea for deciding between the two methods and a direction of single-cell parallel sequencing development.


Asunto(s)
Secuenciación de Nucleótidos de Alto Rendimiento , Oocitos/metabolismo , Análisis de la Célula Individual , Animales , Femenino , Ratones , Ratones Endogámicos C57BL , Análisis de Secuencia de ARN
18.
Stem Cells Dev ; 28(20): 1365-1375, 2019 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-31580778

RESUMEN

Glaucoma is characterized by retinal ganglion cell (RGC) degeneration and is the second leading cause of blindness worldwide. However, current treatments such as eye drop or surgery have limitations and do not target the loss of RGC. Regenerative therapy using embryonic stem cells (ESCs) holds a promising option, but ethical concern hinders clinical applications on human subjects. In this study, we employed spermatogonial stem cells (SSCs) as an alternative source of ESCs for cell-based regenerative therapy in mouse glaucoma model. We generated functional RGCs from SSCs with a two-step protocol without applying viral transfection or chemical induction. SSCs were first dedifferentiated to embryonic stem-like cells (SSC-ESCs) that resemble ESCs in morphology, gene expression signatures, and stem cell properties. The SSC-ESCs then differentiated toward retinal lineages. We showed SSC-ESC-derived retinal cells expressed RGC-specific marker Brn3b and functioned as bona fide RGCs. To allow in vivo RGC tracing, Brn3b-EGFP reporter SSC-ESCs were generated and the derived RGCs were subsequently transplanted into the retina of glaucoma mouse models by intravitreal injection. We demonstrated that the transplanted RGCs could survive in host retina for at least 10 days after transplantation. SSC-ESC-derived RGCs can thus potentially be a novel alternative to replace the damaged RGCs in glaucomatous retina.


Asunto(s)
Células Madre Germinales Adultas/citología , Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Glaucoma/terapia , Células Ganglionares de la Retina/trasplante , Células Madre Germinales Adultas/metabolismo , Animales , Biomarcadores/metabolismo , Diferenciación Celular , Modelos Animales de Enfermedad , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Expresión Génica , Genes Reporteros , Glaucoma/inducido químicamente , Glaucoma/genética , Glaucoma/patología , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Inyecciones Intravítreas , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , N-Metilaspartato/administración & dosificación , Factor 3 de Transcripción de Unión a Octámeros/genética , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Cultivo Primario de Células , Retina/efectos de los fármacos , Retina/metabolismo , Retina/patología , Células Ganglionares de la Retina/citología , Células Ganglionares de la Retina/metabolismo , Testículo/citología , Testículo/metabolismo , Factor de Transcripción Brn-3B/genética , Factor de Transcripción Brn-3B/metabolismo
19.
Database (Oxford) ; 2015: bav044, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25982314

RESUMEN

Spermatogenic failure is a major cause of male infertility, which affects millions of couples worldwide. Recent discovery of long non-coding RNAs (lncRNAs) as critical regulators in normal and disease development provides new clues for delineating the molecular regulation in male germ cell development. However, few functional lncRNAs have been characterized to date. A major limitation in studying lncRNA in male germ cell development is the absence of germ cell-specific lncRNA annotation. Current lncRNA annotations are assembled by transcriptome data from heterogeneous tissue sources; specific germ cell transcript information of various developmental stages is therefore under-represented, which may lead to biased prediction or fail to identity important germ cell-specific lncRNAs. GermlncRNA provides the first comprehensive web-based and open-access lncRNA catalogue for three key male germ cell stages, including type A spermatogonia, pachytene spermatocytes and round spermatids. This information has been developed by integrating male germ transcriptome resources derived from RNA-Seq, tiling microarray and GermSAGE. Characterizations on lncRNA-associated regulatory features, potential coding gene and microRNA targets are also provided. Search results from GermlncRNA can be exported to Galaxy for downstream analysis or downloaded locally. Taken together, GermlncRNA offers a new avenue to better understand the role of lncRNAs and associated targets during spermatogenesis. Database URL: http://germlncrna.cbiit.cuhk.edu.hk/


Asunto(s)
Bases de Datos de Ácidos Nucleicos , Sistemas en Línea , ARN Largo no Codificante , Espermatozoides/metabolismo , Transcriptoma , Animales , Humanos , Masculino , ARN Largo no Codificante/biosíntesis , ARN Largo no Codificante/genética
20.
Int J Biochem Cell Biol ; 67: 115-20, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25979370

RESUMEN

For decades, DNA methylation at the 5 position of cytosine (5mC) catalyzed by DNA methyltransferases (DNMTs) is a well-known epigenetic modification in mammalian genome, where it modulates chromatin remodeling and transcriptional silencing. The discovery of Ten-eleven translocation (TET) enzymes that oxidize 5mC to 5-hydroxymethylcytosine (5hmC) prompts a new era of DNA demethylation research. It is now established that in DNA demethylation pathway 5mC is first converted to 5-hydroxymethylcytosine (5hmC), then 5-formylcytosine (5fC) and 5-carboxylcytosine (5caC) through TETs. Conversion to unmethylated cytosine (5C) is further facilitated by excision mechanism through thymine-DNA glycosylase (TDG) or base excision repair (BER) pathway. Our understanding of DNMTs and TETs on epigenetic dynamics of cytosine methylation has led to a completion of the methylation (Yin) - demethylation (Yang) cycle on epigenetic modifications on cytosine. However, the regulations on DNA demethylation pathway remain largely unknown. In this review, we provide the recent advances on epigenetic dynamics of DNA demethylation and its potential control from the prespective of small non-coding RNA-mediated regulation. Specifically, we will illustrate how microRNAs contribute to active DNA demethylation control in normal and disease development based on recent findings in stem cells and cancer. This article is part of a Directed Issue entitled: Epigenetics dynamics in development and disease.


Asunto(s)
Citosina/metabolismo , Metilación de ADN , Epigénesis Genética , MicroARNs/genética , Neoplasias/genética , Animales , Ensamble y Desensamble de Cromatina , ADN (Citosina-5-)-Metiltransferasas/genética , ADN (Citosina-5-)-Metiltransferasas/metabolismo , Reparación del ADN , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Humanos , MicroARNs/metabolismo , Oxigenasas de Función Mixta , Neoplasias/metabolismo , Neoplasias/patología , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Transducción de Señal , Timina ADN Glicosilasa/genética , Timina ADN Glicosilasa/metabolismo , Yin-Yang
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA