Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 130
Filtrar
1.
Zhongguo Dang Dai Er Ke Za Zhi ; 25(8): 864-869, 2023 Aug 15.
Artigo em Chinês | MEDLINE | ID: mdl-37668036

RESUMO

Neonatal hypoxic-ischemic encephalopathy (HIE) remains one of the leading causes of death and long-term neurodevelopmental disorders in full-term neonates, and there is currently no curative treatment. Therapeutic hypothermia is now a standard therapy for HIE in the neonatal intensive care unit, but its safety and efficacy in remote areas remains unclear. Melatonin is an indole endocrine hormone mainly produced by the pineal gland and it has the ability to easily penetrate the blood-brain barrier. Through receptor and non-receptor mechanisms, melatonin exerts anti-oxidative and anti-inflammatory effects and participates in the regulation of organelle function and the inhibition of cell death. Melatonin is considered one of the most promising drugs for the treatment of HIE based on its reliable safety profile and clinical/preclinical results. This article reviews the recent research on the use of melatonin in combination with therapeutic hypothermia for the treatment of neonatal HIE.


Assuntos
Hipotermia Induzida , Hipóxia-Isquemia Encefálica , Melatonina , Recém-Nascido , Humanos , Melatonina/uso terapêutico , Hipóxia-Isquemia Encefálica/terapia , Unidades de Terapia Intensiva Neonatal
2.
Zhongguo Dang Dai Er Ke Za Zhi ; 25(7): 697-704, 2023 Jul 15.
Artigo em Chinês | MEDLINE | ID: mdl-37529951

RESUMO

OBJECTIVES: To investigate the risk factors for neonatal asphyxia in Hubei Enshi Tujia and Miao Autonomous Prefecture and establish a nomogram model for predicting the risk of neonatal asphyxia. METHODS: A retrospective study was conducted with 613 cases of neonatal asphyxia treated in 20 cooperative hospitals in Enshi Tujia and Miao Autonomous Prefecture from January to December 2019 as the asphyxia group, and 988 randomly selected non-asphyxia neonates born and admitted to the neonatology department of these hospitals during the same period as the control group. Univariate and multivariate analyses were used to identify risk factors for neonatal asphyxia. R software (4.2.2) was used to establish a nomogram model. Receiver operator characteristic curve, calibration curve, and decision curve analysis were used to assess the discrimination, calibration, and clinical usefulness of the model for predicting the risk of neonatal asphyxia, respectively. RESULTS: Multivariate logistic regression analysis showed that minority (Tujia), male sex, premature birth, congenital malformations, abnormal fetal position, intrauterine distress, maternal occupation as a farmer, education level below high school, fewer than 9 prenatal check-ups, threatened abortion, abnormal umbilical cord, abnormal amniotic fluid, placenta previa, abruptio placentae, emergency caesarean section, and assisted delivery were independent risk factors for neonatal asphyxia (P<0.05). The area under the curve of the model for predicting the risk of neonatal asphyxia based on these risk factors was 0.748 (95%CI: 0.723-0.772). The calibration curve indicated high accuracy of the model for predicting the risk of neonatal asphyxia. The decision curve analysis showed that the model could provide a higher net benefit for neonates at risk of asphyxia. CONCLUSIONS: The risk factors for neonatal asphyxia in Hubei Enshi Tujia and Miao Autonomous Prefecture are multifactorial, and the nomogram model based on these factors has good value in predicting the risk of neonatal asphyxia, which can help clinicians identify neonates at high risk of asphyxia early, and reduce the incidence of neonatal asphyxia.


Assuntos
Asfixia Neonatal , Nomogramas , Recém-Nascido , Humanos , Masculino , Gravidez , Feminino , Estudos Retrospectivos , Cesárea , Fatores de Risco , Asfixia Neonatal/epidemiologia , Asfixia Neonatal/etiologia
3.
Molecules ; 27(7)2022 Apr 06.
Artigo em Inglês | MEDLINE | ID: mdl-35408755

RESUMO

Heat shock proteins (HSPs) are highly conserved stress proteins known as molecular chaperones, which are considered to be cytoplasmic proteins with functions restricted to the intracellular compartment, such as the cytoplasm or cellular organelles. However, an increasing number of observations have shown that HSPs can also be released into the extracellular matrix and can play important roles in the modulation of inflammation and immune responses. Recent studies have demonstrated that extracellular HSPs (eHSPs) were involved in many human diseases, such as cancers, neurodegenerative diseases, and kidney diseases, which are all diseases that are closely linked to inflammation and immunity. In this review, we describe the types of eHSPs, discuss the mechanisms of eHSPs secretion, and then highlight their functions in the modulation of inflammation and immune responses. Finally, we take cancer as an example and discuss the possibility of targeting eHSPs for human disease therapy. A broader understanding of the function of eHSPs in development and progression of human disease is essential for developing new strategies to treat many human diseases that are critically related to inflammation and immunity.


Assuntos
Nefropatias , Neoplasias , Proteínas de Choque Térmico/metabolismo , Humanos , Inflamação/tratamento farmacológico , Nefropatias/tratamento farmacológico , Chaperonas Moleculares/fisiologia , Neoplasias/tratamento farmacológico , Neoplasias/metabolismo
4.
Zhonghua Nan Ke Xue ; 27(2): 140-144, 2021 Feb.
Artigo em Chinês | MEDLINE | ID: mdl-34914330

RESUMO

OBJECTIVE: To investigate the relationship of the prostate volume with the count of inflammatory cells in the peripheral blood and clarify the pathogenesis of BPH. METHODS: From 2015 to 2019, we enrolled 104 men pathologically diagnosed with BPH. Using univariate and multivariate linear regression analysis models, we analyzed the correlation of the prostate volume with the neutrophil count, platelet count, neutrophil-lymphocyte ratio (NLR), platelet-WBC ratio (PWR), and lymphocyte-monocyte ratio (LMR) in the peripheral blood of the patients. RESULTS: Both the platelet count (r = 0.401, P < 0.001) and PWR (r = 0.343, P < 0.001) in the peripheral blood were positively correlated with the prostate volume and serum PSA level, but not with IPSS. No evident relationship was found between the prostate volume and the systemic inflammatory markers NLR and LMR. CONCLUSIONS: The platelet count in the peripheral blood is an important predictor of BPH and may play an important role in the development and progression of BPH.


Assuntos
Próstata , Humanos , Masculino , Contagem de Plaquetas
5.
Mol Reprod Dev ; 87(2): 223-230, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-32011766

RESUMO

Sertoli cells (SCs) are presumed to be the center of testis differentiation because they provide both structural support and biological regulation for spermatogenesis. Previous studies suggest that SCs control germ cell (GC) count and Leydig cell (LC) development in mouse testes. However, the regulatory role of SCs on peritubular myoid (PTM) cell fate in fetal testis has not been clearly reported. Here, we employed Amh-Cre; diphtheria toxin fragment A (DTA) mouse model to selectively ablate SCs from embryonic day (E) 14.5. Results found that SC ablation in the fetal stage caused the disruption of testis cords and the massive loss of GCs. Furthermore, the number of α-smooth muscle actin-labeled PTM cells was gradually decreased from E14.5 and almost lost at E18.5 in SC ablation testis. Interestingly, some Ki67 and 3ß-HSD double-positive fetal LCs could be observed in Amh-Cre; DTA testes at E16.5 and E18.5. Consistent with this phenomenon, the messenger RNA levels of Hsd3b1, Cyp11a1, Lhr, Star and the protein levels of 3ß-HSD and P450Scc were significantly elevated by SC ablation. SC ablation appears to induce ectopic proliferation of fetal LCs although the total LC number appeared reduced. Together, these findings bring us a better understanding of SCs' central role in fetal testis development.


Assuntos
Diferenciação Celular/genética , Toxina Diftérica/genética , Maturidade dos Órgãos Fetais , Integrases/genética , Fragmentos de Peptídeos/genética , Túbulos Seminíferos/embriologia , Células de Sertoli/metabolismo , Animais , Proliferação de Células/genética , Toxina Diftérica/metabolismo , Células Germinativas/metabolismo , Integrases/metabolismo , Células Intersticiais do Testículo/metabolismo , Masculino , Camundongos , Modelos Animais , Fragmentos de Peptídeos/metabolismo , Ratos Transgênicos , Espermatogênese
6.
Cell Mol Life Sci ; 76(9): 1713-1727, 2019 May.
Artigo em Inglês | MEDLINE | ID: mdl-30671589

RESUMO

Testicular tumors are the most common tumors in adolescent and young men and germ cell tumors (TGCTs) account for most of all testicular cancers. Increasing incidence of TGCTs among males provides strong motivation to understand its biological and genetic basis. Gains of chromosome arm 12p and aneuploidy are nearly universal in TGCTs, but TGCTs have low point mutation rate. It is thought that TGCTs develop from premalignant intratubular germ cell neoplasia that is believed to arise from the failure of normal maturation of gonocytes during fetal or postnatal development. Progression toward invasive TGCTs (seminoma and nonseminoma) then occurs after puberty. Both inherited genetic factors and environmental risk factors emerge as important contributors to TGCT susceptibility. Genome-wide association studies have so far identified more than 30 risk loci for TGCTs, suggesting that a polygenic model fits better with the genetic landscape of the disease. Despite high cure rates because of its particular sensitivity to platinum-based chemotherapy, exploration of mechanisms underlying the occurrence, progression, metastasis, recurrence, chemotherapeutic resistance, early diagnosis and optional clinical therapeutics without long-term side effects are urgently needed to reduce the cancer burden in this underserved age group. Herein, we present an up-to-date review on clinical challenges, origin and progression, risk factors, TGCT mouse models, serum diagnostic markers, resistance mechanisms, miRNA regulation, and database resources of TGCTs. We appeal that more attention should be paid to the basic research and clinical diagnosis and treatment of TGCTs.


Assuntos
Cromossomos Humanos Par 12/genética , Predisposição Genética para Doença/genética , Neoplasias Embrionárias de Células Germinativas , Neoplasias Testiculares , Animais , Antígeno B7-H1/metabolismo , Progressão da Doença , Células Germinativas/patologia , Humanos , Masculino , Camundongos , MicroRNAs/genética , Neoplasias Embrionárias de Células Germinativas/diagnóstico , Neoplasias Embrionárias de Células Germinativas/tratamento farmacológico , Neoplasias Embrionárias de Células Germinativas/genética , Neoplasias Embrionárias de Células Germinativas/patologia , Receptor de Morte Celular Programada 1/antagonistas & inibidores , Fatores de Risco , Testes Sorológicos , Neoplasias Testiculares/diagnóstico , Neoplasias Testiculares/tratamento farmacológico , Neoplasias Testiculares/genética , Neoplasias Testiculares/patologia
7.
J Proteome Res ; 18(4): 1819-1826, 2019 04 05.
Artigo em Inglês | MEDLINE | ID: mdl-30835130

RESUMO

Seminoma and embryonal carcinoma (EC), two typical types of testicular germ cell tumors (TGCTs), present significant differences in growth behavior, expression characteristics, differentiation potential, clinical features, therapy, and prognosis. The purpose of this study was to compare the distinctive or preference metabolic pathways between seminoma and EC. The Cancer Genome Atlas revealed that many genes encoding metabolic enzymes could distinguish between seminoma and EC. Using well-characterized cell line models for seminoma (Tcam-2 cells) and EC (NT2 cells), we characterized their metabolite profiles using ultraperformance liquid chromatography coupled to Q-TOF mass spectrometry (UPLC/Q-TOF MS). In general, the integrated results from transcriptome and metabolite profiling revealed that seminoma and EC exhibited distinctive characteristics in the metabolisms of amino acids, glucose, fatty acids, sphingolipids, nucleotides, and drugs. Notably, an attenuation of citric acid cycle/mitochondrial oxidative phosphorylation and sphingolipid biosynthesis as well as an increase in arachidonic acid metabolism and (very) long-chain fatty acid abundance occurred in seminoma as compared with EC. Our study suggests histologic subtype-dependent metabolic reprogramming in TGCTs and will lead to a better understanding of the metabolic signatures and biology of TGCT subtypes.


Assuntos
Carcinoma Embrionário/metabolismo , Metaboloma/genética , Neoplasias Embrionárias de Células Germinativas/metabolismo , Seminoma/metabolismo , Neoplasias Testiculares/metabolismo , Transcriptoma/genética , Carcinoma Embrionário/genética , Linhagem Celular Tumoral , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica/genética , Humanos , Masculino , Espectrometria de Massas , Neoplasias Embrionárias de Células Germinativas/genética , Seminoma/genética , Neoplasias Testiculares/genética
8.
Mol Hum Reprod ; 25(9): 507-518, 2019 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-31328782

RESUMO

Spermatogonial stem cells (SSCs) self-renew and contribute genetic information to the next generation. Pig is wildly used as a model animal for understanding reproduction mechanisms of human being. Inducing directional differentiation of porcine SSCs may be an important strategy in exploring the mechanisms of spermatogenesis and developing better treatment methods for male infertility. Here, we established an in-vitro culture model for porcine small seminiferous tubule segments, to induce SSCs to differentiate into single-tail haploid spermatozoa. The culture model subsequently enabled spermatozoa to express the sperm-specific protein acrosin and oocytes to develop to blastocyst stage after round spermatid injection. The addition of retinoic acid (RA) to the differentiation media promoted the efficiency of haploid differentiation. RT-PCR analysis indicated that RA stimulated the expression of Stra8 but reduced the expression of NANOS2 in spermatogonia. Genes involved in post-meiotic development, transition protein 1 (Tnp1) and protamine 1 (Prm1) were upregulated in the presence of RA. The addition of an RA receptor (RAR) inhibitor, BMS439, showed that RA enhanced the expression of cAMP responsive-element binding protein through RAR and promoted the formation of round spermatids. We established an efficient culture system for in-vitro differentiation of pig SSCs. Our study represents a model for human testis disease and toxicology screening. Molecular regulators of SSC differentiation revealed in this study might provide a therapeutic strategy for male infertility.


Assuntos
Diferenciação Celular , Haploidia , Espermatogônias/fisiologia , Espermatozoides/fisiologia , Suínos , Animais , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/genética , Células Cultivadas , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Masculino , Cultura Primária de Células/métodos , Cultura Primária de Células/veterinária , Espermatogênese/efeitos dos fármacos , Espermatogênese/fisiologia , Espermatogônias/citologia , Espermatogônias/efeitos dos fármacos , Espermatozoides/citologia , Espermatozoides/efeitos dos fármacos , Espermatozoides/metabolismo , Tretinoína/farmacologia
9.
FASEB J ; 32(3): 1653-1664, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29183964

RESUMO

The assembly of the blood-testis barrier (BTB) during postnatal development is crucial to support meiosis. However, the role of germ cells in BTB assembly remains unclear. Herein, KitW/KitWV mice were used as a study model. These mice were infertile, failing to establish a functional BTB to support meiosis due to c-Kit mutation. Transplantation of undifferentiated spermatogonia derived from normal mice into the testis of KitW/KitWV mice triggered functional BTB assembly, displaying cyclic remodeling during the epithelial cycle. Also, transplanted germ cells were capable of inducing Leydig cell testosterone production, which could enhance the expression of integral membrane protein claudin 3 in Sertoli cells. Early spermatocytes were shown to play a vital role in directing BTB assembly by expressing claudin 3, which likely created a transient adhesion structure to mediate BTB and cytoskeleton assembly in adjacent Sertoli cells. In summary, the positive modulation of germ cells on somatic cell function provides useful information regarding somatic-germ cell interactions.-Li, X.-Y., Zhang, Y., Wang, X.-X., Jin, C., Wang, Y.-Q., Sun, T.-C., Li, J., Tang, J.-X., Batool, A., Deng, S.-L., Chen, S.-R., Cheng, C. Y., Liu, Y.-X. Regulation of blood-testis barrier assembly in vivo by germ cells.


Assuntos
Barreira Hematotesticular/metabolismo , Claudina-3/biossíntese , Células Intersticiais do Testículo/metabolismo , Células de Sertoli/metabolismo , Espermatogônias/metabolismo , Animais , Barreira Hematotesticular/citologia , Claudina-3/genética , Células Intersticiais do Testículo/citologia , Masculino , Camundongos , Camundongos Transgênicos , Células de Sertoli/citologia , Espermatogônias/citologia
10.
Semin Cell Dev Biol ; 59: 89-98, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-26968934

RESUMO

During fetal testis development, fetal Leydig cells (FLCs) are found to be originated from multiple progenitor cells. FLC specification and function are under tight regulation of specific genes and signaling proteins. Furthermore, Sertoli cells play a crucial role to regulate FLC differentiation during fetal testis development. FLC progenitor- and FLC-produced biomolecules are also involved in the differentiation and activity of rodent FLCs. The main function of FLCs is to produce androgens to masculinize XY embryos. However, FLCs are capable of producing androstenedione but not testosterone due to the lack of 17ß-HSD (17ß-hydroxysteroid dehydrogenase), but fetal Sertoli cells express 17ß-HSD which thus transforms androstenedione to testosterone in the fetal testis. On the other hand, FLCs produce activin A to regulate Sertoli cell proliferation, and Sertoli cells in turn modulate testis cord expansion. It is now generally accepted that adult Leydig cells (ALCs) gradually replace FLCs during postnatal development to produce testosterone to support spermatogenesis as FLCs undergo degeneration in neonatal and pre-pubertal testes. However, based on studies using genetic tracing mouse models, FLCs are found to persist in adult testes, making up ∼20% of total Leydig cells. In this review, we evaluate the latest findings regarding the development, function and fate of FLCs during fetal and adult testis development.


Assuntos
Linhagem da Célula , Feto/citologia , Células Intersticiais do Testículo/citologia , Animais , Diferenciação Celular , Humanos , Masculino , Modelos Biológicos
11.
Biochim Biophys Acta Biomembr ; 1860(1): 141-153, 2018 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-28450047

RESUMO

Signaling pathways that regulate blood-tissue barriers are important for studying the biology of various blood-tissue barriers. This information, if deciphered and better understood, will provide better therapeutic management of diseases particularly in organs that are sealed by the corresponding blood-tissue barriers from systemic circulation, such as the brain and the testis. These barriers block the access of antibiotics and/or chemotherapeutical agents across the corresponding barriers. Studies in the last decade using the blood-testis barrier (BTB) in rats have demonstrated the presence of several signaling pathways that are crucial to modulate BTB function. Herein, we critically evaluate these findings and provide hypothetical models regarding the underlying mechanisms by which these signaling molecules/pathways modulate BTB dynamics. This information should be carefully evaluated to examine their applicability in other tissue barriers which shall benefit future functional studies in the field. This article is part of a Special Issue entitled: Gap Junction Proteins edited by Jean Claude Herve.


Assuntos
Barreira Hematotesticular/metabolismo , Conexinas/metabolismo , Junções Comunicantes/metabolismo , Modelos Cardiovasculares , Transdução de Sinais/fisiologia , Animais , Humanos , Masculino
12.
BMC Biotechnol ; 18(1): 61, 2018 10 03.
Artigo em Inglês | MEDLINE | ID: mdl-30285700

RESUMO

BACKGROUND: Clustered Regularly Interspaced Short Palindromic Repeats/CRISPR associated protein 9 (CRISPR/Cas9) has been wildly used to generate gene knockout models through inducing indels causing frame-shift. However, there are few studies concerning the post-transcript effects caused by CRISPR-mediated genome editing. RESULTS: In the present study, we showed that gene knockdown model also could be generated using CRISPR-mediated gene editing by disrupting the boundary of exon and intron in mice (C57BL/6 J). CRISPR induced indel at the boundary of exon and intron (5' splice site) caused alternative splicing and produced multiple different mRNAs, most of these mRNAs introduced premature termination codon causing down expression of the gene. CONCLUSIONS: These results showed that alternative splicing mutants were able to generate through CRISPR-mediated genome editing by deleting the boundary of exon and intron causing disruption of 5' splice site. Although alternative splicing was an unexpected outcome, this finding could be developed as a technology to generate gene knockdown models or to investigate pre-mRNA splicing.


Assuntos
Sistemas CRISPR-Cas , Edição de Genes , Técnicas de Silenciamento de Genes/métodos , Camundongos/genética , Precursores de RNA/genética , Splicing de RNA , Animais , Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas , Éxons , Mutação INDEL , Íntrons , Camundongos Endogâmicos C57BL
13.
Reproduction ; 156(4): 343­351, 2018 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-30306767

RESUMO

Reduced contractility of the testicular peritubular myoid (PTM) cells may contribute to human male subfertility or infertility. Transcription factor GATA4 in Sertoli and Leydig cells is essential for murine spermatogenesis, but limited attention has been paid to the potential role of GATA4 in PTM cells. In primary cultures of mouse PTM cells, siRNA knockdown of GATA4 increased the contractile activity, while GATA4 overexpression significantly attenuated the contractility of PTM cells using a collagen gel contraction assay. Using RNA sequencing and qRT-PCR, we identified a set of genes that exhibited opposite expressional alternation between Gata4 siRNA vs nontargeting siRNA-treated PTM cells and Gata4 adenovirus vs control adenovirus-treated PTM cells. Notably, ion channels, smooth muscle function, cytokines and chemokines, cytoskeleton, adhesion and extracellular matrix were the top four enriched pathways, as revealed by cluster analysis. Natriuretic peptide type B (NPPB) content was significantly upregulated by GATA4 overexpression in both PTM cells and their culture supernatant. More importantly, the addition of 100 µM NPPB could abolish the promoting effect of Gata4 silencing on PTM cell contraction. Taken together, we suggest that the inhibitory action of GATA4 on PTM cell contraction is mediated at least partly by regulating genes belonging to smooth muscle contraction pathway (e.g. Nppb).


Assuntos
Fator de Transcrição GATA4/fisiologia , Testículo/fisiologia , Animais , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Canais Iônicos/genética , Canais Iônicos/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Contração Muscular , Peptídeo Natriurético Encefálico/metabolismo , Cultura Primária de Células , Espermatogênese , Testículo/citologia
14.
J Assist Reprod Genet ; 35(2): 229-236, 2018 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-29152689

RESUMO

PURPOSE: Spermatozoa maturation, a process required for spermatozoa to acquire progressive motility and the ability to fertilize ova, primarily occurs in the caput and corpus of the epididymis. Despite considerable efforts, the factor(s) promoting epididymal sperm maturation remains unclear. Recently, WNT signaling has been implicated in epididymal sperm maturation. METHODS: To further investigate WNT signaling function in epididymal sperm maturation, we generated Wntless conditional knockout mice (Wls cKO), Wls flox/flox ; Lcn5-Cre. RESULTS: In these mice, WNTLESS (WLS), a conserved membrane protein required for all WNT protein secretion, was specifically disrupted in the principal cells of the caput epididymidis. Immunoblot analysis showed that WLS was significantly reduced in the caput epididymidis of Wls cKO mice. In the caput epididymidis of Wls cKO mice, WNT 10A and WNT 2b, which are typically secreted by the principal cells of the caput epididymis, were not secreted. Interestingly, sperm motility analysis showed that the WLS deficiency in the caput epididymidis had no effect on sperm motility. Moreover, fertility tests showed that Wls cKO male mice had normal fertility. CONCLUSION: These results indicate that the disruption of WLS in principal cells of the caput epididymidis inhibits WNT protein secretion but has no effect on sperm motility and male fertility, suggesting that WNT signaling in the caput epididymidis may be dispensable for epididymal sperm maturation in mice.


Assuntos
Epididimo/citologia , Maturação do Esperma/fisiologia , Via de Sinalização Wnt/fisiologia , Animais , Epididimo/fisiologia , Feminino , Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Masculino , Camundongos Knockout , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Gravidez , Taxa de Gravidez , Transporte Proteico , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Motilidade dos Espermatozoides , Proteínas Wnt/genética , Proteínas Wnt/metabolismo
15.
Molecules ; 23(2)2018 Feb 17.
Artigo em Inglês | MEDLINE | ID: mdl-29462985

RESUMO

Melatonin is a ubiquitous molecule and exhibits different effects in long-day and short-day breeding animals. Testosterone, the main resource of androgens in the testis, is produced by Leydig cells but regulated mainly by cytokine secreted by Sertoli cells. Melatonin acts as a local modulator of the endocrine activity in Leydig cells. In Sertoli cells, melatonin influences cellular proliferation and energy metabolism and, consequently, can regulate steroidogenesis. These suggest melatonin as a key player in the regulation of steroidogenesis. However, the melatonin-induced regulation of steroid hormones may differ among species, and the literature data indicate that melatonin has important effects on steroidogenesis and male reproduction.


Assuntos
Hormônios Esteroides Gonadais/biossíntese , Melatonina/farmacologia , Reprodução/efeitos dos fármacos , Animais , Humanos , Células Intersticiais do Testículo/efeitos dos fármacos , Células Intersticiais do Testículo/metabolismo , Masculino , Células de Sertoli/efeitos dos fármacos , Células de Sertoli/metabolismo , Testosterona/metabolismo
16.
Reproduction ; 154(5): 615-625, 2017 11.
Artigo em Inglês | MEDLINE | ID: mdl-28982932

RESUMO

Spermatogenesis is crucial for male fertility and is therefore tightly controlled by a variety of epigenetic regulators. However, the function of enhancer of zeste homolog 2 (EZH2) in spermatogenesis and the molecular mechanisms underlying its activity remain poorly defined. Here, we demonstrate that deleting EZH2 promoted spermatogonial differentiation and apoptosis. EZH2 is expressed in spermatogonia, spermatocytes and round and elongated spermatids from stage 9 to 11 but not in leptotene and zygotene spermatocytes. Knocking down Ezh2 in vitro using a lentivirus impaired self-renewal in spermatogonial stem cells (SSCs), and the conditional knockout of Ezh2 in spermatogonial progenitors promoted precocious spermatogonial differentiation. EZH2 functions to balance self-renewal and differentiation in spermatogonia by suppressing NEUROG3 and KIT via a direct interaction that is independent of its histone methyltransferase activity. Moreover, deleting Ezh2 enhanced the activation of CASP3 in spermatids, resulting in reduced spermatozoa production. Collectively, these data demonstrate that EZH2 plays a nonclassical role in the regulation of spermatogonial differentiation and apoptosis in murine spermatogenesis.


Assuntos
Apoptose/genética , Diferenciação Celular/genética , Proteína Potenciadora do Homólogo 2 de Zeste/genética , Espermatogônias/fisiologia , Animais , Células Cultivadas , Feminino , Deleção de Genes , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Camundongos Knockout , Espermatogênese/genética
17.
Int J Mol Sci ; 18(7)2017 Jul 22.
Artigo em Inglês | MEDLINE | ID: mdl-28737671

RESUMO

Aneuploidy is a leading genetic cause of birth defects and lower implantation rates in humans. Most errors in chromosome number originate from oocytes. Aneuploidy in oocytes increases with advanced maternal age. Recent studies support the hypothesis that cohesion deterioration with advanced maternal age represents a leading cause of age-related aneuploidy. Cohesin generates cohesion, and is established only during the premeiotic S phase of fetal development without any replenishment throughout a female's period of fertility. Cohesion holds sister chromatids together until meiosis resumes at puberty, and then chromosome segregation requires the release of sister chromatid cohesion from chromosome arms and centromeres at anaphase I and anaphase II, respectively. The time of cohesion cleavage plays an important role in correct chromosome segregation. This review focuses specifically on the causes and effects of age-related cohesion deterioration in female meiosis.


Assuntos
Envelhecimento , Anáfase/genética , Aneuploidia , Cromátides , Segregação de Cromossomos/genética , Cromossomos Humanos , Envelhecimento/genética , Envelhecimento/metabolismo , Envelhecimento/patologia , Animais , Cromátides/genética , Cromátides/metabolismo , Cromossomos Humanos/genética , Cromossomos Humanos/metabolismo , Feminino , Humanos , Masculino , Oócitos/patologia
18.
Hum Mol Genet ; 23(2): 333-41, 2014 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-24009315

RESUMO

The Wt1 gene encodes a nuclear transcription factor that is specifically expressed in ovarian granulosa cells. However, the physiological significance of Wt1 in ovarian follicle development remains elusive. In this study, we found that Wt1(+/R394W) mice were grossly normal, however, the females displayed severe reproductive defects. Only ∼15% of the Wt1(+/R394W) females became pregnant after mating with wild-type males, compared with 88.2% of control females. Further study revealed that the subfertility of Wt1(+/R394W) females was caused by aberrant ovarian follicle development. Compared with control females, the ovary size and the number of developing follicles was significantly decreased in Wt1 mutant ovaries which was very similar to premature ovarian failure (POF) in human patients. The results of in vitro studies demonstrated that the expression of follicle stimulating hormone receptor (FSHR), 3ß-hydroxysteroid dehydrogenase and Aromatase was inhibited by Wt1 in granulosa cells, and mutation of Wt1 resulted in the upregulation of these genes and in the premature differentiation of granulosa cells. We also found that Wt1 was likely involved in granulosa cell development via the regulation of E-cadherin and Par6b expression. Mutation in Wt1 caused defects in polarity establishment in granulosa cells, which also likely contributed to the observed aberrant follicle development. The results of this study provide new mechanisms for understanding the regulation of ovarian follicle development and potential pathological cause of POF in human patients.


Assuntos
Células da Granulosa/citologia , Células da Granulosa/metabolismo , Folículo Ovariano/fisiologia , Proteínas WT1/fisiologia , 3-Hidroxiesteroide Desidrogenases/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Aromatase/metabolismo , Caderinas/metabolismo , Diferenciação Celular , Polaridade Celular , Feminino , Fertilidade , Regulação da Expressão Gênica , Células da Granulosa/patologia , Humanos , Masculino , Camundongos , Folículo Ovariano/patologia , Ovulação , Gravidez , Insuficiência Ovariana Primária/patologia , Receptores do FSH/metabolismo , Proteínas WT1/genética
19.
Biol Reprod ; 94(2): 42, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26792939

RESUMO

Testis cords, embryonic precursors of the seminiferous tubules, are fundamental for testis structure and function. Delay or disruption of testis cord formation could result in gonadal dysgenesis. Although mechanisms regulating testis cord formation during sex determination have been well-studied, the genes and signaling pathways involving in testis cord maintenance after the cords have formed are not well characterized. It is now clear that the maintenance of cord structure is an active process. In this review, we summarize the recent findings regarding the regulation of testis cord integrity by a series of Sertoli cell transcription factors, including the WT1-SOX8/SOX9-beta-CATENIN-DHH network, GPR56, STIM1, and NR0B1 (also known as DAX1). In particularly, we emphasize the underappreciated role of peritubular myoid cells in testis cord maintenance and their cooperation with Sertoli cells. The regulation of the size, shape, and number of testis cords by Sertoli cell proliferation (e.g., SMAD4, GATA4, and TGF-beta signaling), Leydig cell products (e.g., ACTIVIN A), vascular development (a lesson learned from PDGF signaling), and available gonad space (as observed in Ift144 mutant mice) is also addressed. Further efforts and new genetic models are needed to unveil the gene networks and underlying mechanisms regulating testis cord integrity and morphology after sex determination.


Assuntos
Desenvolvimento Embrionário/genética , Túbulos Seminíferos/embriologia , Transdução de Sinais/genética , Cordão Espermático/embriologia , Animais , Masculino , Camundongos , Proteínas Repressoras/metabolismo , Fatores de Transcrição SOX9/metabolismo , Túbulos Seminíferos/metabolismo , Células de Sertoli/metabolismo , Cordão Espermático/metabolismo , Proteínas WT1 , beta Catenina/metabolismo
20.
Mol Reprod Dev ; 83(7): 615-23, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27265621

RESUMO

SMAD4 is the central component of canonical signaling in the transforming growth factor beta (TGFß) superfamily. Loss of Smad4 in Sertoli cells affects the expansion of the fetal testis cords, whereas selective deletion of Smad4 in Leydig cells alone does not appreciably alter fetal or adult testis development. Loss of Smad4 in Sertoli and Leydig cells, on the other hand, leads to testicular dysgenesis, and tumor formation in mice. Within the murine testes, Smad4 is also expressed in germ cells of the seminiferous tubules. We therefore, crossed Ngn3-Cre or Stra8-Cre transgenic mice with Smad4-flox mice to generate conditional knockout animals in which Smad4 was specifically deleted in postnatal germ cells to further uncover cell type-specific requirement of Smad4. Unexpectedly, these germ-cell-knockout mice were fertile and did not exhibit any detectable abnormalities in spermatogenesis, indicating that Smad4 is not required for the production of sperm; instead, these data indicate a cell type-specific requirement of Smad4 primarily during testis development. Mol. Reprod. Dev. 83: 615-623, 2016. © 2016 Wiley Periodicals, Inc.


Assuntos
Fertilidade/fisiologia , Proteína Smad4/metabolismo , Espermatogênese/fisiologia , Testículo/crescimento & desenvolvimento , Animais , Deleção de Genes , Masculino , Camundongos , Camundongos Transgênicos , Proteína Smad4/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA