Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Cell Death Dis ; 15(2): 136, 2024 Feb 12.
Artigo em Inglês | MEDLINE | ID: mdl-38346941

RESUMO

Histone methylation plays a crucial role in various cellular processes. We previously reported the in vitro function of histone lysine demethylase 7 A (KDM7A) in osteoblast and adipocyte differentiation. The current study was undertaken to investigate the physiological role of KDM7A in bone homeostasis and elucidate the underlying mechanisms. A conditional strategy was employed to delete the Kdm7a gene specifically in osterix-expressing osteoprogenitor cells in mice. The resulting mutant mice exhibited a significant increase in cancellous bone mass, accompanied by an increase in osteoblasts and bone formation, as well as a reduction in osteoclasts, marrow adipocytes and bone resorption. The bone marrow stromal cells (BMSCs) and calvarial pre-osteoblastic cells derived from the mutant mice exhibited enhanced osteogenic differentiation and suppressed adipogenic differentiation. Additionally, osteoclastic precursor cells from the mutant mice exhibited impaired osteoclast differentiation. Co-culturing BMSCs from the mutant mice with wild-type osteoclast precursor cells resulted in the inhibition of osteoclast differentiation. Mechanistic investigation revealed that KDM7A was able to upregulate the expression of fibroblast activation protein α (FAP) and receptor activator of nuclear factor κB ligand (RANKL) in BMSCs through removing repressive di-methylation marks of H3K9 and H3K27 from Fap and Rankl promoters. Moreover, recombinant FAP attenuated the dysregulation of osteoblast and adipocyte differentiation in BMSCs from Kdm7a deficient mice. Finally, Kdm7a deficiency prevented ovariectomy-induced bone loss in mice. This study establish the role of KDM7A in bone homeostasis through its epigenetic regulation of osteoblast and osteoclast differentiation. Consequently, inhibiting KDM7A may prove beneficial in ameliorating osteoporosis. KDM7A suppresses osteoblast differentiation and bone formation through. upregulating FAP expression and inactivating canonical Wnt signaling, and conversely promotes osteoclast differentiation and bone resorption through upregulating RANKL expression. These are based on its epigenetic removal of the repressive H3K9me2 and H3K27me2 marks from Fap and Rankl promoters. As a result, the expression of KDM7A in osteoprogenitor cells tends to negatively modulate bone mass.


Assuntos
Reabsorção Óssea , Histona Desmetilases com o Domínio Jumonji , Osteoclastos , Animais , Feminino , Camundongos , Reabsorção Óssea/genética , Reabsorção Óssea/metabolismo , Diferenciação Celular , Epigênese Genética , Histona Desmetilases/genética , Histona Desmetilases/metabolismo , Homeostase , Osteoblastos/metabolismo , Osteoclastos/metabolismo , Osteogênese/genética , Ligante RANK/genética , Ligante RANK/metabolismo , Histona Desmetilases com o Domínio Jumonji/genética , Histona Desmetilases com o Domínio Jumonji/metabolismo
2.
NPJ Precis Oncol ; 7(1): 121, 2023 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-37968457

RESUMO

In recent years, lung adenocarcinoma (LUAD) has become a focus of attention due to its low response to treatment, poor prognosis, and lack of reliable indicators to predict the progression or therapeutic effect of LUAD. Different cell death patterns play a crucial role in tumor development and are promising for predicting LUAD prognosis. From the TCGA and GEO databases, we obtained bulk transcriptomes, single-cell transcriptomes, and clinical information. Genes in 15 types of cell death were analyzed for cell death index (CDI) signature establishment. The CDI signature using necroptosis + immunologic cell death-related genes was established in the TCGA cohort with the 1-, 2-, 3-, 4- and 5-year AUC values were 0.772, 0.736, 0.723, 0.795, and 0.743, respectively. The prognosis was significantly better in the low CDI group than in the high CDI group. We also investigated the relationship between the CDI signature and clinical variables, published prognosis biomarkers, immune cell infiltration, functional enrichment pathways, and immunity biomarkers. In vitro assay showed that HNRNPF and FGF2 promoted lung cancer cell proliferation and migration and were also involved in cell death. Therefore, as a robust prognosis biomarker, CDI signatures can screen for patients who might benefit from immunotherapy and improve diagnostic accuracy and LUAD patient outcomes.

3.
Cell Mol Life Sci ; 80(9): 247, 2023 Aug 14.
Artigo em Inglês | MEDLINE | ID: mdl-37578641

RESUMO

Controlled mRNA storage and stability is essential for oocyte meiosis and early embryonic development. However, how to regulate mRNA storage and stability in mammalian oogenesis remains elusive. Here we showed that LSM14B, a component of membraneless compartments including P-body-like granules and mitochondria-associated ribonucleoprotein domain (MARDO) in germ cell, is indispensable for female fertility. To reveal loss of LSM14B disrupted primordial follicle assembly and caused mRNA reduction in non-growing oocytes, which was concomitant with the impaired assembly of P-body-like granules. 10× Genomics single-cell RNA-sequencing and immunostaining were performed. Meanwhile, we conducted RNA-seq analysis of GV-stage oocytes and found that Lsm14b deficiency not only impaired the maternal mRNA accumulation but also disrupted the translation in fully grown oocytes, which was closely associated with dissolution of MARDO components. Moreover, Lsm14b-deficient oocytes reassembled a pronucleus containing decondensed chromatin after extrusion of the first polar body, through compromising the activation of maturation promoting factor, while the defects were restored via WEE1/2 inhibitor. Together, our findings reveal that Lsm14b plays a pivotal role in mammalian oogenesis by specifically controlling of oocyte mRNA storage and stability.


Assuntos
Oócitos , Oogênese , Animais , Feminino , RNA Mensageiro/genética , Oogênese/genética , Folículo Ovariano , Meiose/genética , Fertilidade/genética , Mamíferos
4.
BMC Biol ; 20(1): 121, 2022 05 24.
Artigo em Inglês | MEDLINE | ID: mdl-35606800

RESUMO

BACKGROUND: Men with prediabetes often exhibit concomitant low-quality sperm production or even infertility, problems which urgently require improved therapeutic options. In this study, we have established a sheep model of diet-induced prediabetes that is associated with spermatogenic defects and have explored the possible underlying metabolic causes. RESULTS: We compared male sheep fed a normal diet with those in which prediabetes was induced by a rich diet and with a third group in which the rich diet was supplemented by melatonin. Only the rich diet group had symptoms of prediabetes, and in these sheep, we found impaired spermatogenesis characterized by a block in the development of round spermatids and an increased quantity of testicular apoptotic cells. Comparing the gut microbiomes and intestinal digest metabolomes of the three groups revealed a distinctive difference in the taxonomic composition of the microbiota in prediabetic sheep, and an altered metabolome, whose most significant feature was altered sphingosine metabolism; elevated sphingosine was also found in blood and testes. Administration of melatonin alleviated the symptoms of prediabetes, including those of impaired spermatogenesis, while restoring a more normal microbiota and metabolic levels of sphingosine. Fecal microbiota transplantation from prediabetic sheep induced elevated sphingosine levels and impaired spermatogenesis in recipient mice, indicating a causal role of gut microbiota in these phenotypes. CONCLUSIONS: Our results point to a key role of sphingosine in the disruption of spermatogenesis in prediabetic sheep and suggest it could be a useful disease marker; furthermore, melatonin represents a potential prebiotic agent for the treatment of male infertility caused by prediabetes.


Assuntos
Microbioma Gastrointestinal , Melatonina , Estado Pré-Diabético , Animais , Apoptose , Humanos , Masculino , Camundongos , Estado Pré-Diabético/complicações , Ovinos , Esfingosina , Testículo
5.
FASEB J ; 36(6): e22348, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35583907

RESUMO

Spermatogenesis is a highly coordinated and complex process, and is pivotal for transmitting genetic information between mammalian generations. In this study, we investigated the conservation, differences, and biological functions of homologous genes during spermatogenesis in Mongolia sheep, humans, cynomolgus monkey, and mice using single-cell RNA sequencing technology. We compared X chromosome meiotic inactivation events in Mongolia sheep, humans, cynomolgus monkey, and mice to uncover the concerted activity of X chromosome genes. Subsequently, we focused on the dynamics of gene expression, key biological functions, and signaling pathways at various stages of spermatogenesis in Mongolia sheep and humans. Additionally, the ligand-receptor networks of Mongolia sheep and humans in testicular somatic and germ cells at different developmental stages were mapped to reveal conserved germ cell-soma communication using single-cell resolution. These datasets provided novel information and insights to unravel the molecular regulatory mechanisms of Mongolia sheep spermatogenesis and highlight conservation in gene expression during spermatogenesis between Mongolia sheep and humans, providing a foundation for the establishment of a large mammalian disease model of male infertility.


Assuntos
Testículo , Transcriptoma , Animais , Macaca fascicularis/genética , Masculino , Mamíferos/genética , Camundongos , Mongólia , Análise de Sequência de RNA , Ovinos/genética , Espermatogênese/genética , Testículo/metabolismo
6.
Stem Cell Res Ther ; 13(1): 53, 2022 02 04.
Artigo em Inglês | MEDLINE | ID: mdl-35120575

RESUMO

BACKGROUND: N-myc downstream regulated gene 1 (NDRG1) plays a role in a variety of biological processes including differentiation of osteoclasts. However, it is not known if and how NDRG1 regulates osteogenic differentiation of marrow stromal progenitor cells. METHODS: Gene expression profiling analysis was performed to study the expression level of Ndrg1 during osteogenic and adipogenic differentiation. Gain-of-function and/or loss-of function experiments were carried out to study the role of NDRG1 in the proliferation and differentiation of marrow stromal progenitor cells and the mechanism underlying the function was investigated. Finally, in vivo transfection of Ndrg1 siRNA was done and its effect on osteogenic and adipogenic differentiation in mice was explored. RESULTS: Gene expression profiling analysis revealed that NDRG1 level was regulated during osteogenic and adipogenic differentiation of progenitor cells. The functional experiments demonstrated that NDRG1 negatively regulated the cell growth, and reciprocally modulated the osteogenic and adipogenic commitment of marrow stromal progenitor cells, driving the cells to differentiate toward adipocytes at the expense of osteoblast differentiation. Moreover, NDRG1 interacted with low-density lipoprotein receptor-related protein 6 (LRP6) in the stromal progenitor cells and inactivated the canonical Wnt/ß-catenin signaling cascade. Furthermore, the impaired differentiation of progenitor cells induced by Ndrg1 siRNA could be attenuated when ß-catenin was simultaneously silenced. Finally, in vivo transfection of Ndrg1 siRNA to the marrow of mice prevented the inactivation of canonical Wnt signaling in the BMSCs of ovariectomized mice, and ameliorated the reduction of osteoblasts on the trabeculae and increase of fat accumulation in the marrow observed in the ovariectomized mice. CONCLUSION: This study has provided evidences that NDRG1 plays a role in reciprocally modulating osteogenic and adipogenic commitment of marrow stromal progenitor cells through inactivating canonical Wnt signaling.


Assuntos
Osteogênese , Via de Sinalização Wnt , Animais , Proteínas de Ciclo Celular , Diferenciação Celular/fisiologia , Peptídeos e Proteínas de Sinalização Intracelular , Camundongos , Osteoblastos/metabolismo , Osteogênese/genética , Via de Sinalização Wnt/genética , beta Catenina/genética , beta Catenina/metabolismo
7.
Cell Mol Life Sci ; 79(2): 107, 2022 Jan 30.
Artigo em Inglês | MEDLINE | ID: mdl-35094173

RESUMO

Metastasis suppressor 1 (MTSS1) plays an inhibitory role in tumorigenesis and metastasis of a variety of cancers. To date, the function of MTSS1 in the differentiation of marrow stromal progenitor cells remains to be explored. In the current study, we investigated whether and how MTSS1 has a role in osteoblast differentiation and bone homeostasis. Our data showed that MTSS1 mRNA was upregulated during osteoblast differentiation and downregulated in the osteoblastic lineage cells of ovariectomized and aged mice. Functional studies revealed that MTSS1 promoted the osteogenic differentiation from marrow stromal progenitor cells. Mechanistic explorations uncovered that the inactivation of Src and afterward activation of canonical Wnt signaling were involved in osteoblast differentiation induced by MTSS1. The enhanced osteogenic differentiation induced by MTSS1 overexpression was attenuated when Src was simultaneously overexpressed, and conversely, the inhibition of osteogenic differentiation by MTSS1 siRNA was rescued when the Src inhibitor was supplemented to the culture. Finally, the in vivo transfection of MTSS1 siRNA to the marrow of mice significantly reduced the trabecular bone mass, along with the reduction of trabecular osteoblasts, the accumulation of marrow adipocytes, and the increase of phospho-Src-positive cells on the trabeculae. No change in the number of osteoclasts was observed. This study has unraveled that MTSS1 contributes to osteoblast differentiation and bone homeostasis through regulating Src-Wnt/ß-catenin signaling. It also suggests the potential of MTSS1 as a new target for the treatment of osteoporosis.


Assuntos
Osso e Ossos/metabolismo , Diferenciação Celular/genética , Proteínas dos Microfilamentos/genética , Proteínas de Neoplasias/genética , Osteoblastos/metabolismo , Via de Sinalização Wnt/genética , beta Catenina/genética , Quinases da Família src/genética , Animais , Western Blotting , Células Cultivadas , Feminino , Regulação da Expressão Gênica , Homeostase/genética , Humanos , Camundongos Endogâmicos C57BL , Proteínas dos Microfilamentos/metabolismo , Proteínas de Neoplasias/metabolismo , Osteoblastos/citologia , Osteogênese/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , beta Catenina/metabolismo , Quinases da Família src/metabolismo
8.
Gut ; 71(1): 78-87, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-33504491

RESUMO

OBJECTIVE: Effects of the diet-induced gut microbiota dysbiosis reach far beyond the gut. We aim to uncover the direct evidence involving the gut-testis axis in the aetiology of impaired spermatogenesis. DESIGN: An excessive-energy diet-induced metabolic syndrome (MetS) sheep model was established. The testicular samples, host metabolomes and gut microbiome were analysed. Faecal microbiota transplantation (FMT) confirmed the linkage between gut microbiota and spermatogenesis. RESULTS: We demonstrated that the number of arrested spermatogonia was markedly elevated by using 10× single-cell RNA-seq in the MetS model. Furthermore, through using metabolomics profiling and 16S rDNA-seq, we discovered that the absorption of vitamin A in the gut was abolished due to a notable reduction of bile acid levels, which was significantly associated with reduced abundance of Ruminococcaceae_NK4A214_group. Notably, the abnormal metabolic effects of vitamin A were transferable to the testicular cells through the circulating blood, which contributed to abnormal spermatogenesis, as confirmed by FMT. CONCLUSION: These findings define a starting point for linking the testicular function and regulation of gut microbiota via host metabolomes and will be of potential value for the treatment of male infertility in MetS.


Assuntos
Microbioma Gastrointestinal/fisiologia , Síndrome Metabólica/fisiopatologia , Espermatogênese/fisiologia , Testículo/fisiologia , Vitamina A/metabolismo , Animais , Ácidos e Sais Biliares/metabolismo , Modelos Animais de Doenças , Disbiose/fisiopatologia , Masculino , Metaboloma , Ovinos
9.
Int J Biol Sci ; 17(14): 3795-3817, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34671200

RESUMO

Background: SARS-CoV-2, the cause of the worldwide COVID-19 pandemic, utilizes the mechanism of binding to ACE2 (a crucial component of the renin-angiotensin system [RAS]), subsequently mediating a secondary imbalance of the RAS family and leading to severe injury to the host. However, very few studies have been conducted to reveal the mechanism behind the effect of SARS-CoV-2 on tumors. Methods: Demographic data extracted from 33 cancer types and over 10,000 samples were employed to determine the comprehensive landscape of the RAS. Expression distribution, pretranscriptional and posttranscriptional regulation and posttranslational modifications (PTMs) as well as genomic alterations, DNA methylation and m6A modification were analyzed in both tissue and cell lines. The clinical phenotype, prognostic value and significance of the RAS during immune infiltration were identified. Results: Low expression of AGTR1 was common in tumors compared to normal tissues, while very low expression of AGTR2 and MAS1 was detected in both tissues and cell lines. Differential expression patterns of ACE in ovarian serous cystadenocarcinoma (OV) and kidney renal clear cell carcinoma (KIRC) were correlated with ubiquitin modification involving E3 ligases. Genomic alterations of the RAS family were infrequent across TCGA pan-cancer program, and ACE had the highest alteration frequency compared with other members. Low expression of AGTR1 may result from hypermethylation in the promoter. Downregulation of RAS family was linked to higher clinical stage and worse survival (as measured by disease-specific survival [DSS], overall survival [OS] or progression-free interval [PFI]), especially for ACE2 and AGTR1 in KIRC. ACE-AGTR1, a classical axis of the RAS family related to immune infiltration, was positively correlated with M2-type macrophages, cancer-associated fibroblasts (CAFs) and immune checkpoint genes in most cancers. Conclusion: ACE, ACE2, AGT and AGTR1 were differentially expressed in 33 types of cancers. PTM of RAS family was found to rely on ubiquitination. ACE2 and AGTR1 might serve as independent prognostic factors for LGG and KIRC. SARS-CoV-2 might modify the tumor microenvironment by regulating the RAS family, thus affecting the biological processes of cancer.


Assuntos
Neoplasias/metabolismo , Sistema Renina-Angiotensina , SARS-CoV-2/metabolismo , COVID-19/complicações , COVID-19/metabolismo , Metilação de DNA , Regulação Neoplásica da Expressão Gênica , Humanos , Imunoterapia , Neoplasias/etiologia , Neoplasias/mortalidade , Neoplasias/terapia , Processamento de Proteína Pós-Traducional , Proto-Oncogene Mas
10.
Stem Cells ; 39(10): 1349-1361, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34028135

RESUMO

It was previously reported that the loss of the transcription factor nuclear factor I/X (NFIX) gene in mice impaired endochondral ossification and mineralization in bone. However, the cellular and molecular basis for the defect remains unexplored. In this study, we investigated if and how NFIX regulates osteoblast differentiation. Nfix mRNA was induced during osteogenic and adipogenic differentiation of progenitor cells. Loss-of-function and gain-of-function studies revealed that NFIX induced osteoblast differentiation and impaired adipocyte formation from progenitor cells. RNA-seq and promoter analysis revealed that NFIX transcriptionally stimulated the expression of high-mobility group AT-Hook 1 (HMGA1). We then demonstrated that HMGA1 stimulated osteogenic differentiation of progenitor cells at the expense of adipogenic differentiation. The effect of Nfix siRNA on the differentiation of progenitor cells could be attenuated when HMGA1 was simultaneously overexpressed. Further investigations revealed the stimulatory effect of NFIX and HMGA1 on canonical wingless-type MMTV integration site family (Wnt) signaling. HMGA1 transcriptionally activates the expression of low-density lipoprotein receptor-related protein 5. Finally, in vivo transfection of Nfix siRNA to the marrow of mice reduced osteoblasts and increased fat accumulation in the marrow, and inactivated HMGA1/ß-catenin signaling in bone marrow mesenchymal stem cells. This study suggests that HMGA1 plays a role in osteoblast commitment and mediates the function of NFIX through transcriptionally activating canonical Wnt signaling.


Assuntos
Proteína HMGA1a , Fatores de Transcrição NFI , Osteogênese , Via de Sinalização Wnt , Animais , Diferenciação Celular , Proteína HMGA1a/genética , Proteína HMGA1a/metabolismo , Camundongos , Fatores de Transcrição NFI/metabolismo , Osteoblastos/metabolismo , Osteogênese/genética , RNA Interferente Pequeno/metabolismo , beta Catenina/metabolismo
11.
Front Cell Infect Microbiol ; 10: 593674, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33520734

RESUMO

Symptomatic hepatitis E virus (HEV) infection is sporadic, and usually occurs in a limited number of infected patients, which hinders the investigation of risk factors for clinical outcomes in patients with acute HEV infection. A retrospective cohort study enrolling 1913 patients with symptomatic acute hepatitis E in Beijing 302 Hospital from January 1, 2001 to December 31, 2018 was conducted. The baseline characteristics, clinical features and laboratory data of these HEV infection cases were analyzed. Albumin (ALB), platelet (PLT), alanine aminotransferase (ALT), total bilirubin (T-BiL), international normalized ratio (INR) and serum creatinine (SCR) levels, along with the model for end-stage liver disease (MELD) score, hospitalization days, co-morbidity number and mortality were taken as major parameters for comparing the clinical manifestations in our study. We found that not all pre-existing chronic liver diseases exacerbate clinical manifestations of acute hepatitis E. Alcoholic hepatitis, fatty liver hepatitis, hepatic cyst, drug-induced hepatitis and hepatocellular carcinoma were not significantly associated with mortality of HEV patients. Among all of the comorbidities, end-stage liver diseases (ESLDs, including ascites, cirrhosis, hepatic coma and hepatorenal syndrome), respiratory tract infection and chronic kidney diseases (CKDs, including renal insufficiency and renal failure) were found to remarkably increase the mortality of patients with symptomatic HEV infection. Furthermore, the severity evaluation indexes (SEI), such as MELD score, duration of hospital stay, and co-morbidity number in HEV patients with underlying comorbidities were much worse than that of their counterparts without relevant comorbidities.


Assuntos
Doença Hepática Terminal , Hepatite E , Insuficiência Renal Crônica , Infecções Respiratórias , Hepatite E/complicações , Hepatite E/epidemiologia , Humanos , Prognóstico , Estudos Retrospectivos , Índice de Gravidade de Doença
13.
Zhonghua Kou Qiang Yi Xue Za Zhi ; 41(8): 494-7, 2006 Aug.
Artigo em Chinês | MEDLINE | ID: mdl-17074191

RESUMO

OBJECTIVE: To investigate the expression of the T cell-specific transcription factors T-bet/GATA-3 in recurrent aphthous ulcerations (RAU). METHODS: Peripheral blood mononuclear cells (PBMC) were isolated by density gradient centrifugation from RAU patients and healthy controls. Expressions of T-bet and GATA-3 mRNA and protein in the PBMCs were detected by reverse transcription-polymerase chain reaction (RT-PCR) and Western blot respectively. The concentrations of gamma-interferon (gamma-IFN) and interleukin-4 (IL-4) in the serum were determined by enzyme-linked immunosorbent assay (ELISA). RESULTS: The expressions of T-bet mRNA were 0.73 +/- 0.14 in the control group and 0.12 +/- 0.04 in the recurrent aphthous ulcerations group, the levels of T-bet protein expressions were 0.73 +/- 0.18 in the control group and 0.24 +/- 0.09 in the recurrent aphthous ulcerations group, there was a significant difference between the two groups (P < 0.01). The levels of GATA-3 mRNA expressions were 0.89 +/- 0.11 in the control group and 1.30 +/- 0.13 in the recurrent aphthous ulcerations group; GATA-3 protein expressions were 1.09 +/- 0.16 in the control group and 1.80 +/- 0.16 in the recurrent aphthous ulcerations group, there was a significant difference between the two groups (P < 0.01). In the control group, the concentration of gamma-interferon in the serum was (22.15 +/- 1.52) ng/L, which was significantly elevated compared with that in the recurrent aphthous ulcerations group (16.32 +/- 0.22) ng/L (P < 0.01), in the control group, the concentration of IL-4 in the serum was (25.58 +/- 2.59) ng/L, which was significantly lower than that of the recurrent aphthous ulcerations group (43.60 +/- 3.15) ng/L (P < 0.01). The ratio of gamma-IFN and IL-4 was positively correlated with the ratio of T-bet and GATA-3 mRNA and protein expression (r = 0.96, 0.94, P < 0.01). CONCLUSIONS: Imbalance of transcription factors T-bet and GATA-3 may be one of the key factors in immune dysregulation of recurrent aphthous ulcerations.


Assuntos
Fator de Transcrição GATA3/metabolismo , Estomatite Aftosa/metabolismo , Proteínas com Domínio T/metabolismo , Adolescente , Adulto , Feminino , Fator de Transcrição GATA3/genética , Regulação da Expressão Gênica , Humanos , Masculino , Pessoa de Meia-Idade , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Proteínas com Domínio T/genética , Linfócitos T/metabolismo , Adulto Jovem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA