Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Br J Cancer ; 130(12): 1893-1903, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38649788

RESUMEN

BACKGROUND: Immune cell infiltration is heterogeneous but common in testicular germ cell tumors (TGCT) and pre-invasive germ cell neoplasia in situ (GCNIS). Tumor-infiltrating T cells including regulatory T (Treg) and follicular helper T (Tfh) cells are found in other cancer entities, but their contributions to TGCT are unknown. METHODS: Human testis specimens from independent patient cohorts were analyzed using immunohistochemistry, flow cytometry and single-cell RNA sequencing (scRNA-seq) with special emphasis on delineating T cell subtypes. RESULTS: Profound changes in immune cell composition within TGCT, shifting from macrophages in normal testes to T cells plus B and dendritic cells in TGCT, were documented. In most samples (96%), the CD4+ T cell frequency exceeded that of CD8+ cells, with decreasing numbers from central to peripheral tumor areas, and to tumor-free, contralateral testes. T cells including Treg and Tfh were most abundant in seminoma compared to mixed tumors and embryonal carcinoma. CONCLUSION: Despite considerable heterogeneity between patients, T cell subtypes form a key part of the TGCT microenvironment. The novel finding of rare Treg and Tfh cells in human testis suggests their involvement in TGCT pathobiology, with implications for understanding tumor progression, to assess patients' prognosis, and as putative targets for personalized immunotherapy.


Asunto(s)
Neoplasias de Células Germinales y Embrionarias , Linfocitos T Reguladores , Neoplasias Testiculares , Microambiente Tumoral , Humanos , Neoplasias Testiculares/inmunología , Neoplasias Testiculares/patología , Masculino , Neoplasias de Células Germinales y Embrionarias/patología , Neoplasias de Células Germinales y Embrionarias/inmunología , Linfocitos T Reguladores/inmunología , Microambiente Tumoral/inmunología , Linfocitos Infiltrantes de Tumor/inmunología , Linfocitos Infiltrantes de Tumor/patología , Linfocitos T CD8-positivos/inmunología , Análisis de la Célula Individual , Testículo/patología , Testículo/inmunología , Adulto
2.
Am J Hum Genet ; 108(10): 1924-1945, 2021 10 07.
Artículo en Inglés | MEDLINE | ID: mdl-34626582

RESUMEN

Klinefelter syndrome (KS), also known as 47, XXY, is characterized by a distinct set of physiological abnormalities, commonly including infertility. The molecular basis for Klinefelter-related infertility is still unclear, largely because of the cellular complexity of the testis and the intricate endocrine and paracrine signaling that regulates spermatogenesis. Here, we demonstrate an analysis framework for dissecting human testis pathology that uses comparative analysis of single-cell RNA-sequencing data from the biopsies of 12 human donors. By comparing donors from a range of ages and forms of infertility, we generate gene expression signatures that characterize normal testicular function and distinguish clinically distinct forms of male infertility. Unexpectedly, we identified a subpopulation of Sertoli cells within multiple individuals with KS that lack transcription from the XIST locus, and the consequence of this is increased X-linked gene expression compared to all other KS cell populations. By systematic assessment of known cell signaling pathways, we identify 72 pathways potentially active in testis, dozens of which appear upregulated in KS. Altogether our data support a model of pathogenic changes in interstitial cells cascading from loss of X inactivation in pubertal Sertoli cells and nominate dosage-sensitive factors secreted by Sertoli cells that may contribute to the process. Our findings demonstrate the value of comparative patient analysis in mapping genetic mechanisms of disease and identify an epigenetic phenomenon in KS Sertoli cells that may prove important for understanding causes of infertility and sex chromosome evolution.


Asunto(s)
Infertilidad Masculina/patología , Síndrome de Klinefelter/complicaciones , Células Intersticiales del Testículo/patología , Células de Sertoli/patología , Análisis de la Célula Individual/métodos , Testículo/patología , Transcriptoma , Humanos , Infertilidad Masculina/etiología , Infertilidad Masculina/metabolismo , Síndrome de Klinefelter/cirugía , Células Intersticiales del Testículo/metabolismo , Masculino , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Células de Sertoli/metabolismo , Espermatogénesis , Testículo/metabolismo , Inactivación del Cromosoma X
3.
Cancer Cell Int ; 19: 280, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31708690

RESUMEN

BACKGROUND: Glioma is the most common central nervous system (CNS) tumour. p62, an important autophagy adaptor, plays a crucial role in cancer. However, the role of p62 in the progression of glioma is poorly characterized. METHODS: We examined the expression of p62 in glioma tissues and cell lines. Then we investigated the function of p62 in vitro, and clarified the mechanism underlying the regulation of p62 expression. RESULTS: We revealed that p62 was upregulated at both the mRNA and protein levels in human glioma tissues irrelevant to isocitrate dehydrogenase (IDH) status. Then, we found that overexpression of p62 promoted glioma progression by promoting proliferation, migration, glycolysis, temozolomide (TMZ) resistance and nuclear factor κB (NF-κB) signalling pathway, and repressing autophagic flux and reactive oxygen species (ROS) in vitro. In accordance with p62 overexpression, knockdown of p62 exerted anti-tumour effects in glioma cells. Subsequently, we demonstrated that miR-124-3p directly targeted the 3'-UTR of p62 mRNA, leading to the downregulation of p62. Finally, we found that p62 function could be partially reversed by miR-124-3p overexpression. CONCLUSIONS: Our results demonstrate that p62 can be targeted by miR-124-3p and acts as an oncogene in glioma, suggesting the potential value of p62 as a novel therapeutic target for glioma.

4.
Int J Syst Evol Microbiol ; 67(5): 1235-1240, 2017 May.
Artículo en Inglés | MEDLINE | ID: mdl-28581924

RESUMEN

A Gram-stain-negative, aerobic, yellow-pigmented, non-spore-forming, non-motile, rod-shaped bacterium, designated strain DHOB07T, was isolated from a soil sample collected from the lower subtropical forest of the Dinghushan Biosphere Reserve, Guangdong Province, PR China (23° 10' N 112° 31' E). Strain DHOB07T grew at 10-37 °C, pH 4-7 and 0-0.5 % (w/v) NaCl, with an optimum at 28 °C, pH 5-5.5 and 0% (w/v) NaCl on R2A medium. Phylogenetic analyses based on 16S rRNA gene sequences showed that the strain formed a clade with Dyella jejuensis JP1T, Dyella nitratireducens DHG59T, Dyella koreensis BB4T, Dyella marensis CS5-B2Tand Dyellasoli JS12-10T, with sequence similarities of 98.9, 98.0, 97.9, 97.9 and 97.8 %, respectively. Multilocus sequence analysis based on the concatenated sequences of partial housekeeping genes gyrB, lepA and recA confirmed that strain DHOB07T belongs to thegenus Dyella but is distinct from all currently known species of the genus Dyella. The G+C content of the genomic DNA was 58.2 mol%. The DNA-DNA relatedness value between strain DHOB07T and D. jejuensis JP1T was 41.8 %. Iso-C16 : 0, iso-C15 : 0 and iso-C17 : 1ω9c were the major fatty acids, and ubiquinone-8 was the only respiratory quinone detected, all of which supported the affiliation of strain DHOB07T to the genus Dyella. On the basis of the polyphasic characterization results presented above, strain DHOB07T represents a novel species of the genus Dyella, for which the name Dyella lipolytica sp. nov. is proposed. The type strain is DHOB07T (=NBRC 111473T=KCTC 52132T).


Asunto(s)
Bosques , Filogenia , Microbiología del Suelo , Xanthomonadaceae/clasificación , Técnicas de Tipificación Bacteriana , Composición de Base , China , ADN Bacteriano/genética , Ácidos Grasos/química , Genes Bacterianos , ARN Ribosómico 16S/genética , Análisis de Secuencia de ADN , Ubiquinona/química , Xanthomonadaceae/genética , Xanthomonadaceae/aislamiento & purificación
5.
Dev Cell ; 58(20): 2097-2111.e3, 2023 10 23.
Artículo en Inglés | MEDLINE | ID: mdl-37582368

RESUMEN

Formation of either an ovary or a testis during human embryonic life is one of the most important sex-specific events leading to the emergence of secondary sexual characteristics and sex assignment of babies at birth. Our study focused on the sex-specific and sex-indifferent characteristics of the prenatal ovarian stromal cells, cortical cords, and germline, with the discovery that the ovarian mesenchymal cells of the stroma are transcriptionally indistinguishable from the mesenchymal cells of the testicular interstitium. We found that first-wave pre-granulosa cells emerge at week 7 from early supporting gonadal cells with stromal identity and are spatially defined by KRT19 levels. We also identified rare transient state f0 spermatogonia cells within the ovarian cords between weeks 10 and 16. Taken together, our work illustrates a unique plasticity of the embryonic ovary during human development.


Asunto(s)
Gónadas , Ovario , Masculino , Femenino , Recién Nacido , Humanos , Testículo , Células Germinativas , Análisis de la Célula Individual
6.
Nat Commun ; 14(1): 8462, 2023 Dec 20.
Artículo en Inglés | MEDLINE | ID: mdl-38123589

RESUMEN

Seminoma is the most common malignant solid tumor in 14 to 44 year-old men. However, its molecular features and tumor microenvironment (TME) is largely unexplored. Here, we perform a series of studies via genomics profiling (single cell multi-omics and spatial transcriptomics) and functional examination using seminoma samples and a seminoma cell line. We identify key gene expression programs share between seminoma and primordial germ cells, and further characterize the functions of TFAP2C in promoting tumor invasion and migration. We also identify 15 immune cell subtypes in TME, and find that subtypes with exhaustion features were located closer to the tumor region through combined spatial transcriptome analysis. Furthermore, we identify key pathways and genes that may facilitate seminoma disseminating beyond the seminiferous tubules. These findings advance our knowledge of seminoma tumorigenesis and produce a multi-omics atlas of in situ human seminoma microenvironment, which could help discover potential therapy targets for seminoma.


Asunto(s)
Neoplasias de Células Germinales y Embrionarias , Seminoma , Neoplasias Testiculares , Masculino , Humanos , Adolescente , Adulto Joven , Adulto , Seminoma/genética , Seminoma/metabolismo , Seminoma/patología , Multiómica , Neoplasias de Células Germinales y Embrionarias/genética , Neoplasias Testiculares/metabolismo , Microambiente Tumoral/genética
7.
Elife ; 112022 01 04.
Artículo en Inglés | MEDLINE | ID: mdl-34982026

RESUMEN

Vertebrate embryos achieve developmental competency during zygotic genome activation (ZGA) by establishing chromatin states that silence yet poise developmental genes for subsequent lineage-specific activation. Here, we reveal the order of chromatin states in establishing developmental gene poising in preZGA zebrafish embryos. Poising is established at promoters and enhancers that initially contain open/permissive chromatin with 'Placeholder' nucleosomes (bearing H2A.Z, H3K4me1, and H3K27ac), and DNA hypomethylation. Silencing is initiated by the recruitment of polycomb repressive complex 1 (PRC1), and H2Aub1 deposition by catalytic Rnf2 during preZGA and ZGA stages. During postZGA, H2Aub1 enables Aebp2-containing PRC2 recruitment and H3K27me3 deposition. Notably, preventing H2Aub1 (via Rnf2 inhibition) eliminates recruitment of Aebp2-PRC2 and H3K27me3, and elicits transcriptional upregulation of certain developmental genes during ZGA. However, upregulation is independent of H3K27me3 - establishing H2Aub1 as the critical silencing modification at ZGA. Taken together, we reveal the logic and mechanism for establishing poised/silent developmental genes in early vertebrate embryos.


Asunto(s)
Cromatina/genética , Regulación del Desarrollo de la Expresión Génica , Silenciador del Gen , Proteínas del Grupo Polycomb/metabolismo , Pez Cebra/genética , Cigoto/crecimiento & desarrollo , Animales , Línea Celular , Metilación de ADN , Drosophila , Proteínas de Drosophila/genética , Genes del Desarrollo , Genómica/métodos , Proteínas del Grupo Polycomb/genética , Cigoto/metabolismo
8.
Dev Cell ; 57(9): 1160-1176.e5, 2022 05 09.
Artículo en Inglés | MEDLINE | ID: mdl-35504286

RESUMEN

Aging men display reduced reproductive health; however, testis aging is poorly understood at the molecular and genomic levels. Here, we utilized single-cell RNA-seq to profile over 44,000 cells from both young and older men and examined age-related changes in germline development and in the testicular somatic cells. Age-related changes in spermatogonial stem cells appeared modest, whereas age-related dysregulation of spermatogenesis and somatic cells ranged from moderate to severe. Altered pathways included signaling and inflammation in multiple cell types, metabolic signaling in Sertoli cells, hedgehog signaling and testosterone production in Leydig cells, cell death and growth in testicular peritubular cells, and possible developmental regression in both Leydig and peritubular cells. Remarkably, the extent of dysregulation correlated with body mass index in older but not in younger men. Collectively, we reveal candidate molecular mechanisms underlying the complex testicular changes conferred by aging and their possible exacerbation by concurrent chronic conditions such as obesity.


Asunto(s)
Análisis de la Célula Individual , Testículo , Anciano , Envejecimiento , Índice de Masa Corporal , Proteínas Hedgehog/metabolismo , Humanos , Masculino , Células de Sertoli , Espermatogénesis/genética , Testículo/metabolismo
9.
Cell Rep ; 37(5): 109915, 2021 11 02.
Artículo en Inglés | MEDLINE | ID: mdl-34731600

RESUMEN

Single-cell RNA sequencing has revealed extensive molecular diversity in gene programs governing mammalian spermatogenesis but fails to delineate their dynamics in the native context of seminiferous tubules, the spatially confined functional units of spermatogenesis. Here, we use Slide-seq, a spatial transcriptomics technology, to generate an atlas that captures the spatial gene expression patterns at near-single-cell resolution in the mouse and human testis. Using Slide-seq data, we devise a computational framework that accurately localizes testicular cell types in individual seminiferous tubules. Unbiased analysis systematically identifies spatially patterned genes and gene programs. Combining Slide-seq with targeted in situ RNA sequencing, we demonstrate significant differences in the cellular compositions of spermatogonial microenvironment between mouse and human testes. Finally, a comparison of the spatial atlas generated from the wild-type and diabetic mouse testis reveals a disruption in the spatial cellular organization of seminiferous tubules as a potential mechanism of diabetes-induced male infertility.


Asunto(s)
Perfilación de la Expresión Génica , Espermatogénesis/genética , Espermatogonias/metabolismo , Testículo/metabolismo , Transcriptoma , Algoritmos , Animales , Microambiente Celular , Bases de Datos Genéticas , Diabetes Mellitus/genética , Diabetes Mellitus/metabolismo , Diabetes Mellitus/patología , Modelos Animales de Enfermedad , Regulación del Desarrollo de la Expresión Génica , Humanos , Infertilidad Masculina/genética , Infertilidad Masculina/metabolismo , Infertilidad Masculina/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Microscopía Confocal , RNA-Seq , Análisis de la Célula Individual , Especificidad de la Especie , Espermatogonias/patología , Testículo/patología , Factores de Tiempo
10.
Cell Stem Cell ; 28(4): 764-778.e4, 2021 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-33453151

RESUMEN

Human testis development in prenatal life involves complex changes in germline and somatic cell identity. To better understand, we profiled and analyzed ∼32,500 single-cell transcriptomes of testicular cells from embryonic, fetal, and infant stages. Our data show that at 6-7 weeks postfertilization, as the testicular cords are established, the Sertoli and interstitial cells originate from a common heterogeneous progenitor pool, which then resolves into fetal Sertoli cells (expressing tube-forming genes) or interstitial cells (including Leydig-lineage cells expressing steroidogenesis genes). Almost 10 weeks later, beginning at 14-16 weeks postfertilization, the male primordial germ cells exit mitosis, downregulate pluripotent transcription factors, and transition into cells that strongly resemble the state 0 spermatogonia originally defined in the infant and adult testes. Therefore, we called these fetal spermatogonia "state f0." Overall, we reveal multiple insights into the coordinated and temporal development of the embryonic, fetal, and postnatal male germline together with the somatic niche.


Asunto(s)
Células de Sertoli , Testículo , Adulto , Femenino , Humanos , Masculino , Embarazo , Análisis de la Célula Individual , Espermatogonias , Células Madre
11.
Cell Stem Cell ; 26(2): 262-276.e4, 2020 02 06.
Artículo en Inglés | MEDLINE | ID: mdl-31928944

RESUMEN

The human testis undergoes dramatic developmental and structural changes during puberty, including proliferation and maturation of somatic niche cells, and the onset of spermatogenesis. To characterize this understudied process, we profiled and analyzed single-cell transcriptomes of ∼10,000 testicular cells from four boys spanning puberty and compared them to those of infants and adults. During puberty, undifferentiated spermatogonia sequentially expand and differentiate prior to the initiation of gametogenesis. Notably, we identify a common pre-pubertal progenitor for Leydig and myoid cells and delineate candidate factors controlling pubertal differentiation. Furthermore, pre-pubertal Sertoli cells exhibit two distinct transcriptional states differing in metabolic profiles before converging to an alternative single mature population during puberty. Roles for testosterone in Sertoli cell maturation, antimicrobial peptide secretion, and spermatogonial differentiation are further highlighted through single-cell analysis of testosterone-suppressed transfemale testes. Taken together, our transcriptional atlas of the developing human testis provides multiple insights into developmental changes and key factors accompanying male puberty.


Asunto(s)
Espermatogonias , Testículo , Adulto , Humanos , Lactante , Masculino , Pubertad , Células de Sertoli , Espermatogénesis/genética
12.
Cell Rep ; 23(3): 838-851, 2018 Apr 17.
Artículo en Inglés | MEDLINE | ID: mdl-29669288

RESUMEN

p38 signaling is broadly involved in controlling inflammation and stress-induced cell death; however, the mechanisms controlling its activity have seldom been studied. Here, we report that TRIM9 short isoform (TRIM9s) potentiates p38 signaling by stabilizing MKK6. Mechanistic studies revealed that TRIM9s promotes the K63-linked ubiquitination of MKK6 at Lys82, thus inhibiting the degradative K48-linked ubiquitination of MKK6 at the same lysine. MKK6 could also stabilize TRIM9s by promoting the phosphorylation of TRIM9s at Ser76/80 via p38, thereby blocking the ubiquitin-proteasome pathway. Further functional analyses showed that p38 signaling plays a critical role in suppressing glioblastoma progression. Co-reduction of MKK6 and TRIM9s is significantly associated with overall poor survival of glioblastoma patients. We identify a positive feedback loop in p38 signaling generated by MKK6-TRIM9s, which suppresses glioblastoma progression, and we provide insights into the mechanisms by which TRIM9s and MKK6 potentiate p38 signaling through mutual stabilization.


Asunto(s)
Glioblastoma/patología , MAP Quinasa Quinasa 6/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Transducción de Señal , Proteínas de Motivos Tripartitos/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Animales , Línea Celular Tumoral , Glioblastoma/tratamiento farmacológico , Glioblastoma/mortalidad , Humanos , MAP Quinasa Quinasa 6/deficiencia , MAP Quinasa Quinasa 6/genética , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Proteínas del Tejido Nervioso/antagonistas & inhibidores , Proteínas del Tejido Nervioso/genética , Fosforilación , Isoformas de Proteínas/antagonistas & inhibidores , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , ARN Interferente Pequeño/uso terapéutico , Tasa de Supervivencia , Proteínas de Motivos Tripartitos/antagonistas & inhibidores , Proteínas de Motivos Tripartitos/genética , Ubiquitina/metabolismo , Ubiquitina-Proteína Ligasas/antagonistas & inhibidores , Ubiquitina-Proteína Ligasas/genética , Ubiquitinación , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
13.
Cell Res ; 28(12): 1141-1157, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30315278

RESUMEN

Human adult spermatogenesis balances spermatogonial stem cell (SSC) self-renewal and differentiation, alongside complex germ cell-niche interactions, to ensure long-term fertility and faithful genome propagation. Here, we performed single-cell RNA sequencing of ~6500 testicular cells from young adults. We found five niche/somatic cell types (Leydig, myoid, Sertoli, endothelial, macrophage), and observed germline-niche interactions and key human-mouse differences. Spermatogenesis, including meiosis, was reconstructed computationally, revealing sequential coding, non-coding, and repeat-element transcriptional signatures. Interestingly, we identified five discrete transcriptional/developmental spermatogonial states, including a novel early SSC state, termed State 0. Epigenetic features and nascent transcription analyses suggested developmental plasticity within spermatogonial States. To understand the origin of State 0, we profiled testicular cells from infants, and identified distinct similarities between adult State 0 and infant SSCs. Overall, our datasets describe key transcriptional and epigenetic signatures of the normal adult human testis, and provide new insights into germ cell developmental transitions and plasticity.


Asunto(s)
Espermatogénesis/genética , Espermatogonias/metabolismo , Testículo/citología , Testículo/metabolismo , Adolescente , Adulto , Animales , Atlas como Asunto , Secuencia de Bases , Ciclo Celular/genética , Plasticidad de la Célula/genética , Humanos , Lactante , Masculino , Ratones , Análisis de Secuencia de ARN/métodos , Análisis de la Célula Individual/métodos , Espermatogonias/citología , Espermatogonias/crecimiento & desarrollo , Transcriptoma
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA