Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
1.
Cell Physiol Biochem ; 57(2): 137-156, 2023 Apr 30.
Artigo em Inglês | MEDLINE | ID: mdl-37125425

RESUMO

BACKGROUND/AIMS: Earlier studies have revealed the miRNAs and mRNAs involved in Polycystic Ovarian Syndrome (PCOS), but little is known about their regulatory networks. METHODS: To address this issue, we applied a comprehensive miRNA, mRNA profiling approach in peripheral blood of PCOS patients. We identified 30 differential miRNAs and 3310 differential transcripts. A robust computational framework was created to integrate matched miRNA and mRNA expression profiles in PCOS using feed-forward loops. RESULTS: The network consisted of differential miRNAs, transcription factors (TFs), and their common predicted target genes. The key network consisted of 14 non-orphan network clusters with 50 TF-gene pairs, 8 TF-TF pairs, 6 miRNA-TF pairs and 36 miRNA- gene pairs which were later dissected into 16 subclusters. Gene ontology annotations revealed that a host of signals (hormone, growth factors -EGF/ PDGF, thrombopoietin, oxidative stress and vitamin/nutrition) regulate MAPK signaling altering angiogenesis, JAK-STAT signaling, apoptosis, inflammatory and immune response and steroidogenesis in PCOS women. CONCLUSION: MAPK signaling is identified as the syndrome´s major dysregulated pathway. Our data imparts a robust foundation to expand the work and pave the way to focus efforts on p38MAPK targeted therapeutic strategies in PCOS.


Assuntos
MicroRNAs , Síndrome do Ovário Policístico , Humanos , Feminino , MicroRNAs/genética , MicroRNAs/metabolismo , Síndrome do Ovário Policístico/genética , Síndrome do Ovário Policístico/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Perfilação da Expressão Gênica , Fatores de Transcrição/genética , Redes Reguladoras de Genes
2.
Hum Reprod ; 37(8): 1835-1855, 2022 07 30.
Artigo em Inglês | MEDLINE | ID: mdl-35728080

RESUMO

STUDY QUESTION: Do circadian genes exhibit an altered profile in peripheral blood mononuclear cells (PBMCs) of polycystic ovary syndrome (PCOS) patients and do they have a potential role in androgen excess? SUMMARY ANSWER: Our findings revealed that an impaired circadian clock could hamper the regulation of peripheral steroid metabolism in PCOS women. WHAT IS KNOWN ALREADY: PCOS patients exhibit features of metabolic syndrome. Circadian rhythm disruption is involved in the development of metabolic diseases and subfertility. An association between shift work and the incidence of PCOS in females was recently reported. STUDY DESIGN, SIZE, DURATION: This is a retrospective case-referent study in which peripheral blood samples were obtained from 101 control and 101 PCOS subjects. PCOS diagnoses were based on Rotterdam Consensus criteria. PARTICIPANTS/MATERIALS, SETTING, METHODS: This study comprised 101 women with PCOS and 101 control volunteers, as well as Swiss albino mice treated with dehydroepiandrosterone (DHEA) to induce PCOS development. Gene expression analyses of circadian and steroidogenesis genes in human PBMC and mice ovaries and blood were executed by quantitative real-time PCR. MAIN RESULTS AND THE ROLE OF CHANCE: We observed aberrant expression of peripheral circadian clock genes in PCOS, with a significant reduction in the core clock genes, circadian locomotor output cycles kaput (CLOCK) (P ≤ 0.00001), brain and muscle ARNT-like 1 (BMAL1) (P ≤ 0.00001) and NPAS2 (P ≤ 0.001), and upregulation of their negative feedback loop genes, CRY1 (P ≤ 0.00003), CRY2 (P ≤ 0.00006), PER1 (P ≤ 0.003), PER2 (P ≤ 0.002), DEC1 (P ≤ 0.0001) and DEC2 (P ≤ 0.00005). Transcript levels of an additional feedback loop regulating BMAL1 showed varied expression, with reduced RORA (P ≤ 0.008) and increased NR1D1 (P ≤ 0.02) in PCOS patients in comparison with the control group. We also demonstrated the expression pattern of clock genes in PBMCs of PCOS women at three different time points. PCOS patients also exhibited increased mRNA levels of steroidogenic enzymes like StAR (P ≤ 0.0005), CYP17A1 (P ≤ 0.005), SRD5A1 (P ≤ 0.00006) and SRD5A2 (P ≤ 0.009). Knockdown of CLOCK/BMAL1 in PBMCs resulted in a significant reduction in estradiol production, by reducing CYP19A1 and a significant increase in dihydrotestosterone production, by upregulating SRD5A1 and SRD5A2 in PBMCs. Our data also showed that CYP17A1 as a direct CLOCK-BMAL1 target in PBMCs. Phenotypic classification of PCOS subgroups showed a higher variation in expression of clock genes and steroidogenesis genes with phenotype A of PCOS. In alignment with the above results, altered expression of ovarian core clock genes (Clock, Bmal1 and Per2) was found in DHEA-treated PCOS mice. The expression of peripheral blood core clock genes in DHEA-induced PCOS mice was less robust and showed a loss of periodicity in comparison with that of control mice. LARGE SCALE DATA: N/A. LIMITATIONS, REASONS FOR CAUTION: We could not evaluate the circadian oscillation of clock genes and clock-controlled genes over a 24-h period in the peripheral blood of control versus PCOS subjects. Additionally, circadian genes in the ovaries of PCOS women could not be evaluated due to limitations in sample availability, hence we employed the androgen excess mouse model of PCOS for ovarian circadian assessment. Clock genes were assessed in the whole ovary of the androgen excess mouse model of PCOS rather than in granulosa cells, which is another limitation of the present work. WIDER IMPLICATIONS OF THE FINDINGS: Our observations suggest that the biological clock is one of the contributing factors in androgen excess in PCOS, owing to its potential role in modulating peripheral androgen metabolism. Considering the increasing prevalence of PCOS and the rising frequency of delayed circadian rhythms and insufficient sleep among women, our study emphasizes the potential in modulating circadian rhythm as an important strategy in PCOS management, and further research on this aspect is highly warranted. STUDY FUNDING/COMPETING INTEREST(S): This work was supported by the RGCB-DBT Core Funds and a grant (#BT/PR29996/MED/97/472/2020) from the Department of Biotechnology (DBT), India, to M.L. B.S.J. was supported by a DST/INSPIRE Fellowship/2015/IF150361 and M.B.K. was supported by the Research Fellowship from Council of Scientific & Industrial Research (CSIR) (10.2(5)/2007(ii).E.U.II). The authors declare no competing interests. TRIAL REGISTRATION NUMBER: N/A.


Assuntos
Síndrome do Ovário Policístico , 3-Oxo-5-alfa-Esteroide 4-Desidrogenase , Fatores de Transcrição ARNTL , Androgênios , Animais , Desidroepiandrosterona , Feminino , Humanos , Leucócitos Mononucleares/metabolismo , Proteínas de Membrana , Camundongos , Síndrome do Ovário Policístico/genética , Síndrome do Ovário Policístico/metabolismo , Estudos Retrospectivos
3.
Cell Physiol Biochem ; 52(1): 141-155, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30790510

RESUMO

BACKGROUND/AIMS: Type 1 Diabetes (T1D) involves autoimmune attack due to reduced regulatory T cells as an effect of mutant Stat5b(C1462A) in non-obese diabetic (NOD) mice, a T1D model resulting in pancreatic ß-cell destruction. Although reactive oxygen species are considered to orchestrate the immune attack, the role of nitric oxide (·NO) still remains debatable. Since JAK-STAT pathway is known to induce Nos2, we investigated the role of STAT5B in nitric oxide generation and oxidative stress. METHODS: In this study, we have used chromatin immunoprecipitation with STAT5B antibody to explore whether STAT5B binds Nos2 promoter. Using Stat5b gene silencing and overexpression models in MIN6 mouse pancreatic ß-cell line we have assayed nitric oxide and its end products, superoxide levels, H2O2 levels, and expression of genes related to redox pathway by immunocytochemistry, biochemical assays, quantitative real time PCR and western blotting. RESULTS: Our results prove that STAT5B binds to the candidate gamma-interferon-activated (GAS) element in Nos2 promoter thereby inducing Nos2 mRNA transcription resulting in NOS2 protein expression in MIN6, a mouse pancreatic ß-cell line. Our findings are substantiated by reduced ·NO as well as nitric oxide end products (nitrate and nitrite), and increased superoxide production in Stat5b silenced MIN6 cells. Our results indicate that C1462A mutant STAT5B shows lack of ·NO generation ability. To detoxify excess superoxide as a consequence of lowered Nos2, an overexpressed SOD2 in Stat5b silenced cells results in increased H2O2 production. H2O2 metabolizing enzymes do not show upregulation upon Stat5b silencing, and thus oxidative stress is brought about by amassed H2O2. Stat5b silencing finally reduces AKT expression, a prosurvival signal. CONCLUSION: Our study enables us to conclude that ß-cell stress is aggravated by the incapability of STAT5B to induce Nos2 resulting in H2O2 accumulation and the ensuing oxidative stress enhances ß-cell damage.


Assuntos
Células Secretoras de Insulina/metabolismo , Óxido Nítrico Sintase Tipo II/metabolismo , Óxido Nítrico/metabolismo , Fator de Transcrição STAT5/metabolismo , Substituição de Aminoácidos , Animais , Linhagem Celular , Peróxido de Hidrogênio/farmacologia , Camundongos Endogâmicos NOD , Mutação de Sentido Incorreto , Óxido Nítrico/genética , Óxido Nítrico Sintase Tipo II/genética , Estresse Oxidativo/efeitos dos fármacos , Estresse Oxidativo/genética , Regiões Promotoras Genéticas , Ligação Proteica , Fator de Transcrição STAT5/genética
4.
Cell Physiol Biochem ; 43(5): 1880-1892, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29055959

RESUMO

BACKGROUND: Though oxidative stress is associated with Polycystic Ovary Syndrome (PCOS), the status of nitric oxide is still unclear. Nitric Oxide (NO) plays pivotal roles in many physiological functions which are compromised in PCOS. Our recent study reveals lowered T-regulatory cells (Tregs) in PCOS, and Treg generation is known to be regulated by NO levels. However concrete evidences are lacking on mechanisms modulating NO levels under PCOS. METHODS: This is a retrospective case-control cohort study, comprised of PCOS women (N=29) and normal menstruating women as controls (N=20). We analysed NOx (nitrite+nitrate) and hydrogen peroxide (H2O2) concentrations, transcript levels of endothelial nitric oxide synthase (eNOS)/inducible nitric oxide synthase (iNOS) and arginine modulators, hydrogen peroxide regulators in the cohort. RESULTS: PCOS women showed reduced plasma NOx(nitrate+nitrite) and H2O2 compared to controls. We report reduction in transcript levels of iNOS/NOS2 and eNOS/NOS3 in PCOS peripheral blood. The transcripts involved in arginine bioavailability: Argininosuccinate lyase (ASL), Solute Carrier Family1, member 7 (SLC7A1) and Arginase 1 (ARG1) and Asymmetric Dimethyl Arginine (ADMA) metabolism: Protein arginine methyltransferase 1 (PRMT1) and Dimethylarginine dimethylaminohydrolase 2 (DDAH2) also showed differential expression. H2O2 concentration in PCOS women was also found to be reduced. The reduction can be attributed to increase in catalase levels as a consequence of the body's effort to alleviate the oxidative burden in the system. CONCLUSION: Our study advocates that PCOS women have lowered NO due to reduced iNOS/eNOS expression, low H2O2, high ADMA synthesis and reduced arginine bioavailability. An in-depth analysis of redox biology of PCOS to open up potential therapeutic strategies is highly recommended.


Assuntos
Arginina/metabolismo , Óxido Nítrico/sangue , Síndrome do Ovário Policístico/sangue , Síndrome do Ovário Policístico/metabolismo , Amidoidrolases/genética , Arginina/análogos & derivados , Argininossuccinato Liase/genética , Transportador 1 de Aminoácidos Catiônicos/genética , Feminino , Humanos , Peróxido de Hidrogênio/sangue , Nitratos/sangue , Óxido Nítrico Sintase Tipo II/genética , Óxido Nítrico Sintase Tipo III/genética , Nitritos/sangue , Oxirredução , Proteína-Arginina N-Metiltransferases/genética , Proteínas Repressoras/genética
5.
Mol Cell Proteomics ; 14(12): 3185-95, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26432663

RESUMO

DYNLT1 is a member of a gene family identified within the t-complex of the mouse, which has been linked with male germ cell development and function in the mouse and the fly. Though defects in the expression of this gene are associated with male sterility in both these models, there has been no study examining its association with spermatogenic defects in human males. In this study, we evaluated the levels of DYNLT1 and its expression product in the germ cells of fertile human males and males suffering from spermatogenic defects. We screened fertile (n = 14), asthenozoospermic (n = 15), oligozoospermic (n = 20) and teratozoospermic (n = 23) males using PCR and Western blot analysis. Semiquantitative PCR indicated either undetectable or significantly lower levels of expression of DYNLT1 in the germ cells from several patients from across the three infertility syndrome groups, when compared with that of fertile controls. DYNLT1 was localized on head, mid-piece, and tail segments of spermatozoa from fertile males. Spermatozoa from infertile males presented either a total absence of DYNLT1 or its absence in the tail region. Majority of the infertile individuals showed negligible levels of localization of DYNLT1 on the spermatozoa. Overexpression of DYNLT1 in GC1-spg cell line resulted in the up-regulation of several cytoskeletal proteins and molecular chaperones involved in cell cycle regulation. Defective expression of DYNLT1 was associated with male factor infertility syndromes in our study population. Proteome level changes in GC1-spg cells overexpressing DYNLT1 were suggestive of its possible function in germ cell development. We have discussed the implications of these observations in the light of the known functions of DYNLT1, which included protein trafficking, membrane vesiculation, cell cycle regulation, and stem cell differentiation.


Assuntos
Dineínas/genética , Dineínas/metabolismo , Infertilidade Masculina/metabolismo , Espermatogênese , Espermatozoides/metabolismo , Animais , Astenozoospermia/metabolismo , Linhagem Celular , Proteínas do Citoesqueleto/metabolismo , Regulação da Expressão Gênica , Humanos , Infertilidade Masculina/genética , Masculino , Camundongos , Chaperonas Moleculares/metabolismo , Oligospermia/metabolismo
6.
J Cell Sci ; 127(Pt 8): 1738-50, 2014 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-24481815

RESUMO

Embryo implantation is effected by a myriad of signaling cascades acting on the embryo-endometrium axis. Here we show, by using MALDI TOF analysis, far-western analysis and colocalization and co-transfection studies, that STAT3 and MCL-1 are interacting partners during embryo implantation. We show in vitro that the interaction between the two endogenous proteins is strongly regulated by estrogen and progesterone. Implantation, pregnancy and embryogenesis are distinct from any other process in the body, with extensive, but controlled, proliferation, cell migration, apoptosis, cell invasion and differentiation. Cellular plasticity is vital during the early stages of development for morphogenesis and organ homeostasis, effecting the epithelial to mesenchymal transition (EMT) and, the reverse process, mesenchymal to epithelial transition (MET). STAT3 functionally associates with MCL-1 in the mammalian breast cancer cell line MCF7 that overexpresses STAT3 and MCL-1, which leads to an increased rate of apoptosis and decreased cellular invasion, disrupting the EMT. Association of MCL-1 with STAT3 modulates the normal, anti-apoptotic, activity of MCL-1, resulting in pro-apoptotic effects. Studying the impact of the association of STAT3 with MCL-1 on MET could lead to an enhanced understanding of pregnancy and infertility, and also metastatic tumors.


Assuntos
Transdiferenciação Celular , Proteína de Sequência 1 de Leucemia de Células Mieloides/metabolismo , Fator de Transcrição STAT3/metabolismo , Animais , Apoptose , Implantação do Embrião , Estrogênios/fisiologia , Feminino , Pontos de Checagem da Fase G2 do Ciclo Celular , Humanos , Células MCF-7 , Camundongos , Proteína de Sequência 1 de Leucemia de Células Mieloides/química , Fosfoproteínas/química , Fosfoproteínas/metabolismo , Gravidez , Progesterona/fisiologia , Regiões Promotoras Genéticas , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas , Mapeamento de Interação de Proteínas , Transporte Proteico , Fator de Transcrição STAT3/química , Ativação Transcricional , Útero/citologia , Útero/metabolismo
7.
Biol Reprod ; 89(4): 84, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23926286

RESUMO

Transforming growth factor-beta (TGF-B) plays an important role in embryo implantation; however, TGF-B requires liberation from its inactive latent forms (i.e., large latent TGF-B complex [LLC] and small latent TGF-B complex [SLC]) to its biologically active (i.e., monomer or dimer) forms in order to act on its receptors (TGF-BRs), which in turn activate SMAD2/3. Activation of TGF-B1 from its latent complexes in the uterus is not yet deciphered. We investigated uterine latent TGF-B1 complex and its biologically active form during implantation, decidualization, and delayed implantation. Our study, utilizing nonreducing SDS-PAGE followed by Western blotting and immunoblotting with TGF-B1, LTBP1, and latency-associated peptide, showed the presence of LLC and SLC in the uterine extracellular matrix and plasma membranous protein fraction during stages of the implantation period. A biologically active form of TGF-B1 (~17-kDa monomer) was highly elevated in the uterine plasma membranous compartment at the peri-implantation stage (implantation and nonimplantation sites). Administration of hydroxychloroquine (an inhibitor of pro-TGF-B processing) at the preimplantation stage was able to block the liberation of biologically active TGF-B1 from its latent complex at the postimplantation stage; as a consequence, the number of implantation sites was reduced at Day 5 (1000 h), as was the number of fetuses at Day 13. The inhibition of TGF-B1 showed reduced levels of phosphorylated SMAD3. Further, the delayed-implantation mouse model showed progesterone and estradiol coordination to release the active TGF-B1 form from its latent complex in the receptive endometrium. This study demonstrates the importance of liberation of biologically active TGF-B1 during the implantation period and its regulation by estradiol.


Assuntos
Implantação do Embrião , Endométrio/metabolismo , Estradiol/metabolismo , Processamento de Proteína Pós-Traducional , Fator de Crescimento Transformador beta1/agonistas , Animais , Decídua/metabolismo , Modelos Animais de Doenças , Implantação Tardia do Embrião , Feminino , Infertilidade Feminina/metabolismo , Proteínas de Ligação a TGF-beta Latente/metabolismo , Camundongos , Peptídeos/metabolismo , Fosforilação/efeitos dos fármacos , Placentação , Gravidez , Progesterona/metabolismo , Precursores de Proteínas/metabolismo , Sinais Direcionadores de Proteínas , Proteína Smad3/agonistas , Proteína Smad3/antagonistas & inibidores , Proteína Smad3/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Fator de Crescimento Transformador beta1/antagonistas & inibidores , Fator de Crescimento Transformador beta1/metabolismo
8.
J Mol Endocrinol ; 70(1)2023 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-36103132

RESUMO

Estrogen accounts for several biological processes in the body; embryo implantation and pregnancy being one of the vital events. This manuscript aims to unearth the nuclear role of Son of sevenless1 (SOS1), its interaction with estrogen receptor alpha (ERα), and signal transducer and activator of transcription 3 (STAT3) in the uterine nucleus during embryo implantation. SOS1, a critical cytoplasmic linker between receptor tyrosine kinase and rat sarcoma virus signaling, translocates into the nucleus via its bipartite nuclear localization signal (NLS) during the 'window of implantation' in pregnant mice. SOS1 associates with chromatin, interacts with histones, and shows intrinsic histone acetyltransferase (HAT) activity specifically acetylating lysine 16 (K16) residue of histone H4. SOS1 is a coactivator of STAT3 and a co-repressor of ERα. SOS1 creates a partial mesenchymal-epithelial transition by acting as a transcriptional modulator. Finally, our phylogenetic tree reveals that the two bipartite NLS surface in reptiles and the second acetyl coenzymeA (CoA) (RDNGPG) important for HAT activity emerges in mammals. Thus, SOS1 has evolved into a moonlighting protein, the special class of multi-tasking proteins, by virtue of its newly identified nuclear functions in addition to its previously known cytoplasmic function.


Assuntos
Implantação do Embrião , Receptor alfa de Estrogênio , Proteína SOS1 , Fator de Transcrição STAT3 , Animais , Camundongos , Receptor alfa de Estrogênio/genética , Filogenia , Fatores ras de Troca de Nucleotídeo Guanina , Fator de Transcrição STAT3/genética , Proteína SOS1/genética
9.
Am J Reprod Immunol ; 89(2): e13637, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-36305192

RESUMO

PROBLEM: Autoimmune polyendocrinopathy-candidiasis- ectodermal dystrophy (APECED) pathology due to autoimmune regulator (AIRE) gene mutations leads to loss of central tolerance triggering immune attack, a factor causing infertility. One of the targets of autoimmune attack is ovary and its repercussion results in polycystic ovarian syndrome (PCOS). Although reduced Tregs have been reported in PCOS, a lacunae exists on the status of AIRE gene expression and its role in treg insufficiency via HIF1A-FOXP3 axis in PCOS. METHOD OF STUDY: This is a case-control cohort study recruiting 40 normal and 40 PCOS volunteers for peripheral blood sample collection and PCOS diagnoses were based on Rotterdam Consensus criteria. AIRE and HIF1A expression status was analysed by qRT PCR and western blot. FACS analyses was conducted on AIRE silenced peripheral blood mononuclear cells (PBMCs) after Treg induction. RESULTS: Our results indicate a reduced AIRE (fold change log2 (RQ) = -2.6, P < .01) and increased HIF1A (fold change log2 (RQ) = 3.6, P < .02) in PBMCs of PCOS subjects compared to age-matched controls. Western blot of AIRE and HIF1A corroborates with qRT PCR data. Our CHIP data demonstrate AIRE mediated HIF1A promoter regulation. Silencing of AIRE in PBMCs contributes to the upregulation of HIF1A transcripts by two-fold (P < .0015) and downregulation in FOXP3 expression by three-fold (P < .0017). FACS analyses revealed that silencing of AIRE reduces Tcell to Treg conversion. CONCLUSIONS: Our consolidated results derive a new connection among AIRE-HIF1A-FOXP3 with AIRE reduction enabling increased HIF1A resulting in reduced FOXP3 in PBMCs of PCOS patients leading to Treg insufficiency.


Assuntos
Fatores de Transcrição Forkhead , Subunidade alfa do Fator 1 Induzível por Hipóxia , Síndrome do Ovário Policístico , Fatores de Transcrição , Feminino , Humanos , Estudos de Casos e Controles , Fatores de Transcrição Forkhead/genética , Fatores de Transcrição Forkhead/metabolismo , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Leucócitos Mononucleares/metabolismo , Síndrome do Ovário Policístico/genética , Poliendocrinopatias Autoimunes/genética , Fatores de Transcrição/metabolismo , Proteína AIRE
10.
ACS Omega ; 7(10): 8601-8612, 2022 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-35309488

RESUMO

A considerable section of males suffered from COVID-19, with many experiencing long-term repercussions. Recovered males have been documented to have compromised fertility, albeit the mechanisms remain unclear. We investigated the impact of COVID-19 on semen proteome following complete clinical recovery using mass spectrometry. A label-free quantitative proteomics study involved 10 healthy fertile subjects and 17 COVID-19-recovered men. With 1% false discovery rate and >1 unique peptide stringency, MaxQuant analysis found 1099 proteins and 8503 peptides. Of the 48 differentially expressed proteins between the healthy and COVID-19-recovered groups, 21 proteins were downregulated and 27 were upregulated in COVID-19-recovered males. The major pathways involved in reproductive functions, such as sperm-oocyte recognition, testosterone response, cell motility regulation, adhesion regulation, extracellular matrix adhesion, and endopeptidase activity, were downregulated in COVID-19-recovered patients according to bioinformatics analysis. Furthermore, the targeted approach revealed significant downregulation of semenogelin 1 and prosaposin, two proteins related to male fertility. Therefore, we demonstrate the alteration of semen proteome in response to COVID-19, thus disrupting the male reproductive function despite the patient's clinical remission. Hence, to understand fertility-related biological processes triggered by this infection, a protracted evaluation of the consequences of COVID-19 in recovered men is warranted.

11.
Biochem Cell Biol ; 89(4): 411-22, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21819345

RESUMO

The autoimmune regulator gene Aire shows predominant expression in thymus and other immunologically relevant tissues, and is assigned the major function of programming autoreactive T-cell deletion. However, the expression of this gene in tissues outside the immune system raises a question about its possible function beyond the T-cell deletion dogma. We detected Aire in mouse testis, and the expression of AIRE protein was remarkably high in postmeiotic germ cells. Sequencing results indicate that testis expressed Aire variant 1a. AIRE could be detected in spermatozoa, with heavy localization on the principal acrosomal domains. Mouse oocytes stained negatively for AIRE before fertilization, but stained positively for AIRE 30 min after fertilization. In the zygote, the levels of AIRE correlated negatively with cyclin B2 levels. Goat testicular lysates spiked with recombinant human AIRE exhibited augmented cyclin B2 degradation in the presence of protease inhibitors, which was inhibited by MG-132, indicating the operation of proteasomal pathways. Thus, this study identifies a correlation between the presence of AIRE and proteasomal breakdown of cyclin B2, which leads us to speculate that cyclin B2 could be a target of AIRE's E3-ubiquitin ligase activity.


Assuntos
Extratos Celulares/química , Ciclina B2/metabolismo , Isoformas de Proteínas/genética , Testículo/metabolismo , Fatores de Transcrição/genética , Aglutinação , Animais , Especificidade de Anticorpos , Sequência de Bases , Ciclina B2/química , Feminino , Expressão Gênica , Cabras , Humanos , Soros Imunes/química , Imunoprecipitação , Masculino , Camundongos , Dados de Sequência Molecular , Gravidez , Isoformas de Proteínas/química , Isoformas de Proteínas/metabolismo , Coelhos , Alinhamento de Sequência , Espermatozoides/química , Espermatozoides/crescimento & desenvolvimento , Espermatozoides/metabolismo , Testículo/citologia , Fatores de Transcrição/química , Fatores de Transcrição/metabolismo , Ubiquitina/metabolismo , Zona Pelúcida/metabolismo , Zigoto/citologia , Zigoto/metabolismo , Proteína AIRE
12.
Cytokine Growth Factor Rev ; 54: 32-42, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32747157

RESUMO

The seventh human coronavirus SARS-CoV2 belongs to the cluster of extremely pathogenic coronaviruses including SARS-CoV and MERS-CoV, which can cause fatal lower respiratory tract infection. Likewise, SARS-CoV2 infection can be fatal as the disease advances to pneumonia, followed by acute respiratory distress syndrome (ARDS). The development of lethal clinical symptons is associated with an exaggerated production of inflammatory cytokines, referred to as the cytokine storm, is a consequence of a hyperactivated immune response aginst the infection. In this article, we discuss the pathogenic consequences of the cytokine storm and its relationship with COVID-19 associated risk factors. The increased pro-inflammatory immune status in patients with risk factors (diabetes, hypertension, cardiovascular disease, COPD) exacerbates the Cytokine-storm of COVID-19 into a 'Cytokine Super Cyclone'. We also evaluate the antiviral immune responses provided by BCG vaccination and the potential role of 'trained immunity' in early protection against SARS-CoV2.


Assuntos
Vacina BCG/uso terapêutico , Infecções por Coronavirus/prevenção & controle , Síndrome da Liberação de Citocina/prevenção & controle , Citocinas/sangue , Pandemias/prevenção & controle , Pneumonia Viral/prevenção & controle , Antivirais/uso terapêutico , Betacoronavirus/imunologia , COVID-19 , Doenças Cardiovasculares/patologia , Infecções por Coronavirus/tratamento farmacológico , Infecções por Coronavirus/imunologia , Síndrome da Liberação de Citocina/imunologia , Síndrome da Liberação de Citocina/patologia , Diabetes Mellitus/patologia , Humanos , Hipertensão/patologia , Coronavírus da Síndrome Respiratória do Oriente Médio/imunologia , Mycobacterium bovis/imunologia , Pneumonia Viral/tratamento farmacológico , Pneumonia Viral/imunologia , Doença Pulmonar Obstrutiva Crônica/patologia , Fatores de Risco , SARS-CoV-2 , Síndrome Respiratória Aguda Grave/imunologia , Vacinação
13.
Free Radic Biol Med ; 161: 198-211, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-33065180

RESUMO

The redox-active transition metals such as copper, iron, chromium, vanadium, and silica are known for its ROS generation via mechanisms such as Haber-Weiss and Fenton-type reactions. Nanoparticles of these metals induce oxidative stress due to acellular factors owing to their small size and more reactive surface area, leading to various cellular responses. The intrinsic enzyme-like activity of nano vanadium has fascinated the scientific community. However, information concerning their cellular uptake and time-dependent induced effects on their cellular organelles and biological activity is lacking. This comprehensive study focuses on understanding the precise molecular interactions of vanadium pentoxide nanoparticles (VnNp) and evaluate their specific "nano" induced effects on MDA-MB-231 cancer cells. Understanding the mechanism behind NP-induced ROS generation could help design a model for selective NP induced toxicity, useful for cancer management. The study demonstrated the intracellular persistence of VnNp and insights into its molecular interactions with various organelles and its overall effects at the cellular level. Where triple-negative breast cancer MDA-MB-231 cells resulted in 59.6% cell death towards 48 h of treatment and the normal fibroblast cells showed only 15.4% cell death, indicating an inherent anticancer property of VnNp. It acts as an initial reactive oxygen species quencher, by serving itself as an antioxidant, while; it was also found to alter the cellular antioxidant system with prolonged incubation. The VnNp accumulated explicitly in the lysosomes and mitochondria and modulated various cellular processes including impaired lysosomal function, mitochondrial damage, and autophagy. At more extended time points, VnNp influenced cell cycle arrest, inhibited cell migration, and potentiated the onset of apoptosis. Results are indicative of the fact that VnNp selectively induced breast cancer cell death and hence could be developed as a future drug molecule for breast cancer management. This could override the most crucial challenge of chemo-resistance that still remain as the main hurdle to cancer therapy.


Assuntos
Autofagia , Nanopartículas , Apoptose , Humanos , Oxirredução , Espécies Reativas de Oxigênio , Compostos de Vanádio
14.
Am J Reprod Immunol ; 80(1): e12844, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29516628

RESUMO

PROBLEM: Dedicator of cytokinesis (DOCK 180) involved in cytoskeletal reorganization is primarily a cytosolic molecule. It is recently shown to be nuclear in HeLa cells but its nuclear function is not known. METHOD OF STUDY: The spatiotemporal distribution of DOCK180 in uterus was studied in uterine cytoplasmic and nuclear compartments during the "window of implantation." The functional significance of nuclear DOCK180 was explored by homology modeling, co-immunoprecipitation assays, and mass spectrometric analysis. Dock180's role in early pregnancy was ascertained by Dock 180 silencing and subsequent quantitative real-time PCR and Western blotting analysis. RESULTS: Our study shows a nuclear DOCK180 in the uterus during "window of implantation." Estrogen and progesterone mediate expression and nuclear translocation of DOCK180. The nuclear function of DOCK180 is attributed to its ability to import autoimmune regulator (AIRE) into the nucleus. Silencing of Dock180 inhibited AIRE nuclear shuttling which influenced its downstream targets, thereby affecting decidualization with AIRE and HOXA-10 as the major players as well as lack of implantation site formation due to impact on angiogenesis-associated genes. CONCLUSION: DOCK180 has an indispensable role in pregnancy establishment as knocking down Dock180 abrogates pregnancy by a consolidated impact on decidualization and angiogenesis by regulating AIRE nuclear entry.


Assuntos
Núcleo Celular/genética , Citocinese/efeitos dos fármacos , Inativação Gênica/efeitos dos fármacos , Fatores de Transcrição/genética , Proteínas rac de Ligação ao GTP/genética , Indutores da Angiogênese/metabolismo , Autoimunidade/efeitos dos fármacos , Autoimunidade/genética , Linhagem Celular Tumoral , Núcleo Celular/efeitos dos fármacos , Núcleo Celular/metabolismo , Citocinese/genética , Implantação do Embrião/efeitos dos fármacos , Implantação do Embrião/genética , Estrogênios/genética , Feminino , Células HeLa , Humanos , Gravidez , Progesterona/genética , Proteína AIRE
15.
Diabetes ; 55(3): 734-41, 2006 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-16505237

RESUMO

A point mutation in the Stat5b DNA binding domain in the nonobese diabetic (NOD) mouse was shown to have weaker DNA binding compared with the B6 Stat5b. Here, we assessed the binding ability of the mutant Stat5b in the B6 genetic background (B6.NOD-c11) and the wild-type Stat5b in the NOD background (NOD.Lc11). To our surprise, the binding ability of Stat5b is inconsistent with the presence or absence of the Stat5b mutation in these congenic mice but is correlated with the expression levels of the Crkl protein, which was coprecipitated by an anti-Stat5b antibody. Both the expression of Crkl and the Stat5b binding ability are the highest in B6.NOD-c11 and the lowest in NOD while intermediate in B6 and NOD.Lc11 mice. We demonstrated that the adapter molecule Crkl can bind Stat5b and that the Crkl protein is a Stat5b binding cofactor. More importantly, profection of Crkl recombinant protein significantly increased Stat5b binding ability and rescued the binding defect of the NOD mutant Stat5b, suggesting that Crkl is a key regulatory molecule for Stat5b binding. Therefore, the defective Crkl expression may contribute to the development of diabetes in the NOD mice by exacerbating the defective Stat5b binding ability.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/fisiologia , DNA/metabolismo , Diabetes Mellitus Tipo 1/metabolismo , Proteínas Nucleares/fisiologia , Fator de Transcrição STAT5/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/química , Animais , Sequência de Bases , Western Blotting , Feminino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos NOD , Dados de Sequência Molecular , Proteínas Nucleares/química , Receptores de Antígenos de Linfócitos T/fisiologia , Linfócitos T/fisiologia , Domínios de Homologia de src
16.
FEBS Lett ; 580(24): 5653-60, 2006 Oct 16.
Artigo em Inglês | MEDLINE | ID: mdl-17011554

RESUMO

Establishment of early pregnancy is promoted by a complex network of signalling molecules that mediate cell-to-cell and cell-to-extracellular matrix communications between the receptive endometrium and the invasive trophectoderm. In this study, we have attempted to evaluate the expression profiles of cadherin and catenin during embryo implantation in the mouse. Western blotting studies along with immunocytochemical analysis revealed that E-cadherin is expressed rather ubiquitously in the uterine epithelial cells, distinct enrichment is observed on the apical membrane in the endometrium of peri-implantation uterus specifically at the implantation sites and not at the inter-implanation sites. beta-Catenin also is upregulated and is specifically restricted to apical membrane of epithelial cells of implantation sites. Progesterone induced expression of E-cadherin and 17beta-estradiol regulated the expression of catenin in implantation-delayed uteri. Interestingly, estradiol imparted negative modulation on cadherin expression when co-administered with progesterone. On the contrary, trophoblast exhibits a striking down regulation of cadherin, catenin and Ca(2+) at peri implanting stage. These observations suggest that the trophoblasts exhibited an invasive phenotype while the endometrial epithelium displayed an adhesive phenotype during the window of implantation. Thus, embryo implantation presents an instance where two interacting surfaces showed mutually complementing interaction phenotypes.


Assuntos
Caderinas/metabolismo , Cálcio/metabolismo , Implantação do Embrião , Embrião de Mamíferos/fisiologia , Útero/fisiologia , beta Catenina/metabolismo , Animais , Embrião de Mamíferos/metabolismo , Feminino , Camundongos , Fatores de Tempo , Útero/metabolismo
17.
Photochem Photobiol ; 82(5): 1358-64, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16842022

RESUMO

The photoreceptive extreme tip of the wheat coleoptile exhibits intense green-yellow fluorescence under UV light, suggesting the presence of UV-absorbing materials. Fluorescence spectra of the intact coleoptile tip and tip homogenate showed the presence of the known photoreceptor pigments flavin and carotene, and a preponderance of phenolic compounds. Absorption spectra and fluorescence spectra of various phenolic compounds showed close overlap with the absorption and fluorescence spectra of the wheat coleoptile tip homogenate. Fluorescence spectra of several phenolic compounds showed close overlap with the absorption bands of flavin, carotene and pterine, suggesting possible energy transduction from phenols to these photoreceptors. Excitation of gentisic acid and ferulic acid with 340 nm light in the presence of flavin showed enhancement of flavin fluorescence in a concentration- and viscosity-dependent fashion, indicating fluorescence resonance energy transfer between them and riboflavin. Furthermore, several phenolic compounds tested generated superoxide anion on excitation at 340 nm, suggesting that superoxide-dependent signal cascades could operate in a polyphenol-mediated pathway. Phenolic compounds thus may act as accessory photoreceptors bringing about excitation energy transfer to the reactive photoreceptor molecules, or they may take over the function of the normal photoreceptor in genetic mutations lacking the system, or both processes may occur. The responses of plants to UV-B and UV-A light in mutants may be explained in terms of various phenolics acting as energy transducers in photoreceptor functioning.


Assuntos
Flavonoides/química , Luz , Fenóis/química , Cotilédone/química , Cotilédone/fisiologia , Cotilédone/efeitos da radiação , Transferência Ressonante de Energia de Fluorescência/métodos , Glicerol , Complexo de Proteínas do Centro de Reação Fotossintética/fisiologia , Complexo de Proteínas do Centro de Reação Fotossintética/efeitos da radiação , Polifenóis , Triticum/química , Triticum/fisiologia , Triticum/efeitos da radiação
18.
Am J Reprod Immunol ; 75(3): 256-62, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26662181

RESUMO

Failed implantation is the major restraining factor in assisted reproduction and is defined as the 'black-box of assisted reproduction'. Although work on understanding the complex process of implantation has substantially advanced, it has been limited to studies on mechanism of steroid hormone-mediated signaling during embryo implantation and knocking out single molecules and assessing their impact on embryo implantation. It is important to realize that most proteins exert their function via interaction with other proteins in order to relay downstream signals and/or regulate gene expression via interactions within promoter complexes. Such networks of biomolecular interactions constitute the basis for life as protein interactions are obligatory for cellular functioning. Thus, this review will focus on highlighting protein interactions during the complex process of embryo implantation as they attain a larger significance as pregnancy is fundamental to childbirth and the continuity of life per se.


Assuntos
Implantação do Embrião/imunologia , Embrião de Mamíferos/imunologia , Estrogênios/imunologia , Regulação da Expressão Gênica no Desenvolvimento/imunologia , Gravidez/imunologia , Transdução de Sinais/imunologia , Animais , Embrião de Mamíferos/citologia , Feminino , Humanos
19.
Am J Reprod Immunol ; 76(3): 224-34, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27432359

RESUMO

PROBLEM: Autoimmune polyendocrinopathy, candidiasis, and ectodermal dystrophy (APECED, APS-1) patients characterized by Aire (autoimmune regulator) mutations and Aire homozygous knockouts (Aire(-/-) ) exhibit infertility. It is not clear as to what contributes to infertility in the above. METHOD OF STUDY: This study investigates the expression of "AIRE in the uterus" and its contribution to early pregnancy of mice by using quantitative real-time PCR analysis, immunohistochemistry, Western blotting, and in vivo Aire silencing experiments. RESULTS: Aire (Isoform 1a) is expressed in the uterus during the "window of implantation" and decidualization. In vivo Aire silencing interfered with formation of implantation sites and stromal cell transformation by regulating bone morphogenetic protein-2,4 (Bmp2, Bmp4), homeobox A10 (Hoxa10), and insulin-like growth factor-binding protein 1(Igfbp1) leading to pregnancy failure. CONCLUSION: Our consolidated results on extrathymic uterine expression of AIRE during early pregnancy and decidualization and impaired fertility on in vivo silencing are suggestive of its importance in pregnancy via a role beyond immune tolerance.


Assuntos
Regulação da Expressão Gênica/imunologia , Prenhez/imunologia , Fatores de Transcrição/imunologia , Útero/imunologia , Animais , Feminino , Camundongos , Camundongos Knockout , Poliendocrinopatias Autoimunes/genética , Poliendocrinopatias Autoimunes/imunologia , Poliendocrinopatias Autoimunes/metabolismo , Gravidez , Prenhez/genética , Prenhez/metabolismo , Isoformas de Proteínas/biossíntese , Isoformas de Proteínas/genética , Isoformas de Proteínas/imunologia , Fatores de Transcrição/biossíntese , Fatores de Transcrição/genética , Útero/metabolismo , Proteína AIRE
20.
J Clin Endocrinol Metab ; 100(1): 282-92, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25303485

RESUMO

CONTEXT: The immunesupressive action of CD4(+)CD25(+) CD127(-/low) T regulatory cells (Tregs) is vital for an efficient reproductive function. However no data exists on their number or functionality in polycystic ovary syndrome (PCOS). OBJECTIVE: The study aimed to analyze the frequency of circulating Tregs and key factors modulating them in women with PCOS. DESIGN, SETTING, AND PARTICIPANTS: This is a retrospective, case-control cohort study conducted in women with PCOS recruited from Samad IVF hospitals and Women and Children Hospital, Thiruvananthapuram, India. Women with PCOS (N = 20) were diagnosed according to Rotterdam Consensus and normal menstruating women were taken as controls (N = 2331). MAIN OUTCOME MEASURES: We analyzed the proportion of CD4(+)CD25(+) CD127(-/low) Tregs in women with PCOS by fluorescent activated cell sorting. RESULTS: The study discovered that the women with PCOS have reduced numbers of Tregs (2.626 ± 0.62) compared with controls (4.253 ± 0.87) (t = 6.963, P < .0001, mean difference = -1.627; 95% confidence interval = -2.099--1.155). We documented a decrease in the follicular phase Treg expansion in women with PCOS. Our results revealed a reduced STAT5A (fold change [FC] = 7.642, P < .0004)/STAT5B (FC = 3.824, P < .0001), FOXP3 (FC = 4.1343, P = .0004)/CTLA4 (FC = 2.569, P = .0001) and elevated AKT (FC = 7.39, P = .05)/PIK3 (FC = 5.326, P = .0002) expression in women with PCOS. Recombinant interleukin 2 (rIL2) treatment failed to improve FOXP3/CTLA4 levels but caused a reduction of AKT/PIK3 arm, possibly due to an elevated PTEN in women with PCOS. CONCLUSION: The study suggests that women with PCOS have reduced Tregs due to an inherent hyporesponsiveness to IL2, which is unable to activate STAT5B and reduce FOXP3 expression. IL2-based therapeutic strategies can ameliorate complications in PCOS by suppressing the AKT/PIK3 arm.


Assuntos
Interleucina-2/metabolismo , Síndrome do Ovário Policístico/metabolismo , Transdução de Sinais/imunologia , Linfócitos T Reguladores/imunologia , Adulto , Estudos de Casos e Controles , Separação Celular , Feminino , Citometria de Fluxo , Humanos , Síndrome do Ovário Policístico/sangue , Síndrome do Ovário Policístico/imunologia , Estudos Retrospectivos , Linfócitos T Reguladores/metabolismo
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa