Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 118
Filtrar
1.
Front Immunol ; 15: 1386260, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38975349

RESUMO

Introduction: Lrba is a cytoplasmic protein involved in vesicular trafficking. Lrba-deficient (Lrba-/-) mice exhibit substantially higher levels of IgA in both serum and feces than wild-type (WT) mice. Transforming growth factor ß1 (TGFß1) and its receptors (TGFßR I and II) is essential for differentiating IgA+ B cells. Furthermore, increased IgA production suggests a potential connection between Lrba and the TGFßR signaling pathway in IgA production. However, the specific function of Lrba in B cell biology remains unknown. Aim: Given the increased IgA levels in Lrba-/- mice, the goal in this work was to explore the lymph organs where the switch to IgA occurs, and if TGFßR function is affected. Methods: Non-immunized Lrba-/- mice were compared with Lrba+/+ mice. IgA levels in the serum and feces, as well as during peripheral B cell development, were determined. IgA+ B cells and plasma cells were assessed in the small intestine and secondary lymphoid organs, such as the spleen, mesenteric lymph nodes, and Peyer's patches. The TGFßR signaling pathway was evaluated by determining the expression of TGFßR on B cells. Additionally, SMAD2 phosphorylation was measured under basal conditions and in response to recombinant TGFß. Finally, confocal microscopy was performed to investigate a possible interaction between Lrba and TGFßR in B cells. Results: Lrba-/- mice exhibited significantly higher levels of circulating IgA, IgA+ B, and plasma cells than in peripheral lymphoid organs those in WT mice. TGFßR expression on the membrane of B cells was similar in both Lrba-/- and Lrba+/+ mice. However, intracellular TGFßR expression was reduced in Lrba-/- mice. SMAD2 phosphorylation showed increased levels under basal conditions; stimulation with recombinant TGFß elicited a poorer response than in that in Lrba+/+ B cells. Finally, we found that Lrba colocalizes with TGFßR in B cells. Conclusion: Lrba is essential in controlling TGFßR signaling, subsequently regulating SMAD2 phosphorylation on B cells. This mechanism may explain the increased differentiation of IgA+ B cells and production of IgA-producing plasma cells.


Assuntos
Linfócitos B , Diferenciação Celular , Imunoglobulina A , Transdução de Sinais , Animais , Camundongos , Linfócitos B/imunologia , Linfócitos B/metabolismo , Diferenciação Celular/imunologia , Imunoglobulina A/imunologia , Camundongos Endogâmicos C57BL , Camundongos Knockout , Nódulos Linfáticos Agregados/imunologia , Nódulos Linfáticos Agregados/metabolismo , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Receptores de Fatores de Crescimento Transformadores beta/genética , Proteína Smad2/metabolismo , Proteínas de Transporte Vesicular/genética , Proteínas de Transporte Vesicular/metabolismo
2.
Biomol Biomed ; 24(2): 302-314, 2024 Mar 11.
Artigo em Inglês | MEDLINE | ID: mdl-37688493

RESUMO

Marfan syndrome (MFS) is a multisystem genetic disorder with over 3000 mutations described in the fibrillin 1 (FBN1) gene. Like MFS, other connective tissue disorders also require a deeper understanding of the phenotype-genotype relationship due to the complexity of the clinical presentation, where diagnostic criteria often overlap. Our objective was to identify mutations in patients with connective tissue disorders using a genetic multipanel and to analyze the genotype-phenotype associations in a cohort of Mexican patients. We recruited 136 patients with MFS and related syndromes from the National Institute of Cardiology. Mutations were identified using next-generation sequencing (NGS). To examine the correlation between mutation severity and severe cardiovascular conditions, we focused on patients who had undergone Bentall-de Bono surgery or aortic valve repair. The genetic data obtained allowed us to reclassify the initial clinical diagnosis across various types of connective tissue disorders. The transforming growth factor beta receptor 2 (TGFBR2) rs79375991 mutation was found in 10 out of 16 (63%) Loeys-Dietz patients. We observed a high prevalence (65%) of more severe mutations, such as frameshift indels and stop codons, among patients requiring invasive treatments like aortic valve-sparing surgery, Bentall and de Bono procedures, or aortic valve replacement due to severe cardiovascular injury. Although our study did not achieve precise phenotype-genotype correlations, it underscores the importance of a multigenetic panel evaluation. This could pave the way for a more comprehensive diagnostic approach and inform medical and surgical treatment decision-making.


Assuntos
Doenças Cardiovasculares , Doenças do Tecido Conjuntivo , Síndrome de Marfan , Humanos , Síndrome de Marfan/diagnóstico , Receptores de Fatores de Crescimento Transformadores beta/genética , Proteínas Serina-Treonina Quinases/genética , Fibrilina-1/genética , Tecido Conjuntivo
3.
Cancer Gene Ther ; 31(1): 174-185, 2024 01.
Artigo em Inglês | MEDLINE | ID: mdl-37993543

RESUMO

The TGF signaling pathway is a key regulator of cancer progression. In this work, we report for the first time the antitumor activity of TßRII-SE/Fc, a novel peptibody whose targeting domain is comprised of the soluble endogenous isoform of the human TGF-ß type II receptor (TßRII-SE). Overexpression of TßRIISE/Fc reduces in vitro cell proliferation and migration while inducing cell cycle arrest and apoptosis in human colorectal cancer-derived cell lines. Moreover, TßRII-SE/Fc overexpression reduces tumorigenicity in BALB/c nude athymic mice. Our results revealed that TRII-SE/Fc-expressing tumors were significantly reduced in size or were even incapable of developing. We also demonstrated that the novel peptibody has the ability to inhibit the canonical TGF-ß and BMP signaling pathways while identifying SMAD-dependent and independent proteins involved in tumor progression that are modulated by TßRII-SE/Fc. These findings provide insights into the underlying mechanism responsible for the antitumor activity of TßRII-SE/Fc. Although more studies are required to demonstrate the effectiveness and safety of the novel peptibody as a new therapeutic for the treatment of cancer, our initial in vitro and in vivo results in human colorectal tumor-derived cell lines are highly encouraging. Our results may serve as the foundation for further research and development of a novel biopharmaceutical for oncology.


Assuntos
Neoplasias , Receptores de Fatores de Crescimento Transformadores beta , Camundongos , Animais , Humanos , Receptor do Fator de Crescimento Transformador beta Tipo II/genética , Receptores de Fatores de Crescimento Transformadores beta/genética , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Xenoenxertos , Lentivirus/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Linhagem Celular
4.
Mol Ecol ; 32(6): 1398-1410, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-35403749

RESUMO

The evolution of sex chromosomes and their differentiation from autosomes is a major event during genome evolution that happened many times in several lineages. The repeated evolution and lability of sex-determination mechanisms in fishes makes this a well-suited system to test for general patterns in evolution. According to current theory, differentiation is triggered by the suppression of recombination following the evolution of a new master sex-determining gene. However, the molecular mechanisms that establish recombination suppression are known from few examples, owing to the intrinsic difficulties of assembling sex-determining regions (SDRs). The development of forward-genetics and long-read sequencing have generated a wealth of data questioning central aspects of the current theory. Here, we demonstrate that sex in Midas cichlids is determined by an XY system, and identify and assemble the SDR by combining forward-genetics, long-read sequencing and optical mapping. We show how long-reads aid in the detection of artefacts in genotype-phenotype mapping that arise from incomplete genome assemblies. The male-specific region is restricted to a 100-kb segment on chromosome 4 that harbours transposable elements and a Y-specific duplicate of the anti-Mullerian receptor 2 gene, which has evolved master sex-determining functions repeatedly. Our data suggest that amhr2Y originated by an interchromosomal translocation from chromosome 20 to 4 pre-dating the split of Midas and Flier cichlids. In the latter, it is pseudogenized and translocated to another chromosome. Duplication of anti-Mullerian genes is a common route to establishing new sex determiners, highlighting the role of molecular parallelism in the evolution of sex determination.


Assuntos
Ciclídeos , Masculino , Animais , Ciclídeos/genética , Receptores de Fatores de Crescimento Transformadores beta , Elementos de DNA Transponíveis
5.
Ann Hepatol ; 28(1): 100775, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36280014

RESUMO

INTRODUCTION AND OBJECTIVES: Liver fibrosis is a common pathological change in many chronic liver diseases. Activation of hepatic stellate cells (HSCs) is the core event in liver fibrosis. This study aimed to investigate the role of testicular orphan receptor 4 (TR4) in the activation of HSCs. MATERIALS AND METHODS: In vivo, bile duct ligation (BDL)-induced rat liver fibrosis model was established, and the expressions of TR4 and α-smooth muscle actin (α-SMA) in liver tissues were detected. In vitro, TR4 knockdown and overexpression in JS-1 cells using lentiviral vectors were constructed, and the expressions of TR4, α-SMA, Col-I, and TGF-ß1/smads and retinoid X receptor (RXR) pathway-related genes were detected. RESULTS: TR4 was highly expressed in BDL-induced fibrotic liver, accompanied by increased expression of α-SMA. Knockdown of TR4 significantly inhibited the expressions of α-SMA, Col-I, p-TßRI, and p-Smad2/3, and up-regulated the expression of RXRα in HSCs in vitro. In contrast, TR4 overexpression significantly increased the expressions of α-SMA, Col-I, p-TßRI, and p-Smad2/3, and inhibited the expression of RXRα. CONCLUSIONS: TR4 may promote the activation of HSCs by up-regulating TßR I/Smad2/3 signaling pathway and down-regulating RXRα signaling, thereby promoting the progression of liver fibrosis. Our findings may provide a new therapeutic target against hepatic fibrosis.


Assuntos
Células Estreladas do Fígado , Fator de Crescimento Transformador beta1 , Ratos , Animais , Células Estreladas do Fígado/metabolismo , Fator de Crescimento Transformador beta1/metabolismo , Cirrose Hepática/metabolismo , Transdução de Sinais , Fígado/patologia , Receptores de Fatores de Crescimento Transformadores beta/metabolismo
6.
Int J Mol Sci ; 23(22)2022 Nov 18.
Artigo em Inglês | MEDLINE | ID: mdl-36430763

RESUMO

Genes associated with growth factors were previously analyzed in a radiation- and estrogen-induced experimental breast cancer model. Such in vitro experimental breast cancer model was developed by exposure of the immortalized human breast epithelial cell line, MCF-10F, to low doses of high linear energy transfer (LET) α particle radiation (150 keV/µm) and subsequent growth in the presence or absence of 17ß-estradiol. The MCF-10F cell line was analyzed in different stages of transformation after being irradiated with either a single 60 cGy dose or 60/60 cGy doses of alpha particles. In the present report, the profiling of differentially expressed genes associated with growth factors was analyzed in their relationship with clinical parameters. Thus, the results indicated that Fibroblast growth factor2 gene expression levels were higher in cells transformed by radiation or in the presence of ionizing radiation; whereas the fibroblast growth factor-binding protein 1gene expression was higher in the tumor cell line derived from this model. Such expressions were coincident with higher values in normal than malignant tissues and with estrogen receptor (ER) negative samples for both gene types. The results also showed that transforming growth factor alpha gene expression was higher in the tumor cell line than the tumorigenic A5 and the transformed A3 cell line, whereas the transforming growth factor beta receptor 3 gene expression was higher in A3 and A5 than in Tumor2 cell lines and the untreated controls and the E cell lines. Such gene expression was accompanied by results indicating negative and positive receptors for transforming growth factor alpha and the transforming growth factor beta receptor 3, respectively. Such expressions were low in malignant tissues when compared with benign ones. Furthermore, Fibroblast growth factor2, the fibroblast growth factor-binding protein 1, transforming growth factor alpha, the transforming growth factor beta receptor 3, and the insulin growth factor receptor gene expressions were found to be present in all BRCA patients that are BRCA-Basal, BRCA-LumA, and BRCA-LumB, except in BRCA-Her2 patients. The results also indicated that the insulin growth factor receptor gene expression was higher in the tumor cell line Tumor2 than in Alpha3 cells transformed by ionizing radiation only; then, the insulin growth factor receptor was higher in the A5 than E cell line. The insulin growth factor receptor gene expression was higher in breast cancer than in normal tissues in breast cancer patients. Furthermore, Fibroblast growth factor2, the fibroblast growth factor-binding protein 1, transforming growth factor alpha, the transforming growth factor beta receptor 3, and the insulin growth factor receptor gene expression levels were in stages 3 and 4 of breast cancer patients. It can be concluded that, by using gene technology and molecular information, it is possible to improve therapy and reduce the side effects of therapeutic radiation use. Knowing the different genes involved in breast cancer will make possible the improvement of clinical chemotherapy.


Assuntos
Neoplasias da Mama , Fator de Crescimento Transformador alfa , Humanos , Feminino , Neoplasias da Mama/genética , Neoplasias da Mama/radioterapia , Estrogênios , Radiação Ionizante , Insulina Regular Humana , Linhagem Celular Tumoral , Insulina , Receptores de Fatores de Crescimento Transformadores beta , Fatores de Crescimento de Fibroblastos
7.
Gynecol Obstet Invest ; 87(3-4): 248-255, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35853432

RESUMO

BACKGROUND: Inhibins and their co-receptor betaglycan are members of the transforming growth factor ß superfamily, a group of signaling molecules that control the differentiation of human endometrium in the secretory phase of the menstrual cycle. OBJECTIVE: Since endometriosis is associated with endometrial dysfunction and infertility, this study aimed at evaluating the expression of α-inhibin and betaglycan mRNA and proteins in endometrial samples of infertile women with and without endometriosis. DESIGN: This was a cross-sectional study. Participants/Materials: Endometrial samples of women with (n = 17) and without (n = 22) endometriosis were subdivided according to the menstrual cycle phase into proliferative and secretory. SETTING: University hospital. METHODS: We used real-time RT-PCR to quantify mRNA levels and immunohistochemistry to localize the proteins. RESULTS: α-inhibin mRNA levels were significantly increased in the secretory phase (p < 0.01 vs. proliferative phase) only among women with endometriosis. Conversely, betaglycan mRNA levels were downregulated in the secretory endometrium of controls (p < 0.01 vs. proliferative) but failed to change between cycle phases of patients with endometriosis. Both proteins were present in the glandular epithelium and stroma in the endometrium of women with and without endometriosis. Immunostaining analysis showed that while α-inhibin protein expression did not vary significantly, the intensity of betaglycan immunostaining decreased in the secretory phase in the control group (p = 0.038 vs. proliferative phase) but not in the endometriosis group. LIMITATIONS: We cannot determine whether endometriosis causes the abnormal expression of α-inhibin and betaglycan in the eutopic endometrium or if this alteration already existed before the establishment of endometriotic lesions. CONCLUSION: Our findings suggest an abnormally increased expression of α-inhibin mRNA (not protein) and betaglycan (mRNA and protein) in the secretory-phase endometrium of women with endometriosis.


Assuntos
Endometriose , Infertilidade Feminina , Estudos Transversais , Endometriose/complicações , Endometriose/genética , Endométrio/patologia , Feminino , Humanos , Infertilidade Feminina/complicações , Infertilidade Feminina/genética , Inibinas/metabolismo , Proteoglicanas/metabolismo , RNA Mensageiro/metabolismo , Receptores de Fatores de Crescimento Transformadores beta , Fator de Crescimento Transformador beta/metabolismo
8.
Dev Dyn ; 251(1): 213-225, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34228380

RESUMO

BACKGROUND: The Transforming Growth Factor ß (TGFß) family is a group of related proteins that signal through a type I and type II receptors. Betaglycan, also known as the type III receptor (Tgfbr3), is a coreceptor for various ligands of the TGFß family that participates in heart, liver and kidney development as revealed by the tgfbr3-null mouse, as well as in angiogenesis as revealed by Tgfbr3 downregulation in morphant zebrafish. RESULTS: Here, we present CRISPR/Cas9-derived zebrafish Tgfbr3-null mutants, which exhibited unaltered embryonic angiogenesis and developed into fertile adults. One reproducible phenotype displayed by these Tgfbr3-null mutants is delayed chordacentra mineralization, which nonetheless does not result in vertebral abnormalities in the adult fishes. We also report that the canonical TGFß signaling pathway is needed for proper chordacentra mineralization and that Tgfbr3 absence decreases this signal in the notochordal cells responsible for this process. CONCLUSION: Betaglycan's "ligand presentation" function contributes to the optimal TGFß signaling required for zebrafish chordacentra mineralization.


Assuntos
Receptores de Fatores de Crescimento Transformadores beta , Peixe-Zebra , Animais , Camundongos , Proteoglicanas/genética , Receptores de Fatores de Crescimento Transformadores beta/genética , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Transdução de Sinais/genética , Fator de Crescimento Transformador beta/genética , Fator de Crescimento Transformador beta/metabolismo , Peixe-Zebra/genética , Peixe-Zebra/metabolismo
9.
Sex Dev ; 16(2-3): 138-146, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-34515230

RESUMO

Anti-müllerian hormone (AMH) is 1 of the 2 testicular hormones involved in male development of the genitalia during fetal life. When the testes differentiate, AMH is secreted by Sertoli cells and binds to its specific receptor type II (AMHR2) on the müllerian ducts, inducing their regression. In the female fetus, the lack of AMH allows the müllerian ducts to form the fallopian tubes, the uterus, and the upper part of the vagina. The human AMH gene maps to 19p13.3 and consists of 5 exons and 4 introns spanning 2,764 bp. The AMHR2 gene maps to 12q13.13, consists of 11 exons, and is 7,817 bp long. Defects in the AMH pathway are the underlying etiology of a subgroup of disorders of sex development (DSD) in 46,XY patients. The condition is known as the persistent müllerian duct syndrome (PMDS), characterized by the existence of a uterus and fallopian tubes in a boy with normally virilized external genitalia. Approximately 200 cases of patients with PMDS have been reported to date with clinical, biochemical, and molecular genetic characterization. An updated review is provided in this paper. With highly sensitive techniques, AMH and AMHR2 expression has also been detected in other tissues, and massive sequencing technologies have unveiled variants in AMH and AMHR2 genes in hitherto unsuspected conditions.


Assuntos
Hormônio Antimülleriano , Transtorno 46,XY do Desenvolvimento Sexual , Transtornos do Desenvolvimento Sexual , Receptores de Peptídeos , Receptores de Fatores de Crescimento Transformadores beta , Feminino , Humanos , Masculino , Hormônio Antimülleriano/genética , Transtorno 46,XY do Desenvolvimento Sexual/genética , Transtornos do Desenvolvimento Sexual/genética , Ductos Paramesonéfricos , Desenvolvimento Sexual , Receptores de Peptídeos/genética , Receptores de Fatores de Crescimento Transformadores beta/genética
10.
BMC Cancer ; 21(1): 209, 2021 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-33648463

RESUMO

BACKGROUND: There is evidence to consider that the tumor microenvironment (TME) composition associates with antitumor immune response, and may predict the outcome of various non-Hodgkin lymphoma subtypes. However, in the case of mantle cell lymphoma (MCL), a rare and aggressive disease, there is lacking a detailed study of the TME components, as well as an integrative approach among them in patients' samples. Also, from the genetic point of view, it is known that single nucleotide variants (SNVs) in immune-response genes are among important regulators of immunity. At present, it is uncertain whether SNVs in candidate immune-response genes and the TME composition are able to alter the prognosis in MCL. METHODS: We assessed a detailed TME composition in 88 MCL biopsies using immunohistochemistry, which was automatically analyzed by pixel counting (Aperio system). We also genotyped SNVs located in candidate immune-response genes (IL12A, IL2, IL10, TGFB1, TGFBR1, TGFBR2, IL17A, IL17F) in 95 MCL patients. We tested whether the SNVs could modulate the respective protein expression and TME composition in the tumor compartment. Finally, we proposed survival models in rituximab-treated patients, considering immunohistochemical and SNV models. RESULTS: High FOXP3/CD3 ratios (p = 0.001), high IL17A levels (p = 0.003) and low IL2 levels (p = 0.03) were individual immunohistochemical predictors of poorer survival. A principal component, comprising high quantities of macrophages and high Ki-67 index, also worsened outcome (p = 0.02). In the SNV model, the CC haplotype of IL10 (p < 0.01), the GG genotype of IL2 rs2069762 (p = 0.02) and the AA+AG genotypes of TGFBR2 rs3087465 (p < 0.01) were independent predictors of outcome. Finally, the GG genotype of TGFB1 rs6957 associated with lower tumor TGFß levels (p = 0.03) and less CD163+ macrophages (p = 0.01), but did not modulate patients' survival. CONCLUSIONS: Our results indicate that the TME composition has relevant biological roles in MCL. In this setting, immunohistochemical detection of T-reg cells, IL17A and IL2, coupled with SNV genotyping in IL10, TGFBR2 and IL2, may represent novel prognostic factors in this disease, following future validations.


Assuntos
Imunidade/genética , Linfoma de Célula do Manto/genética , Polimorfismo de Nucleotídeo Único , Microambiente Tumoral , Adulto , Idoso , Idoso de 80 Anos ou mais , Antineoplásicos/uso terapêutico , Terapia Combinada , Progressão da Doença , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Estudos de Associação Genética , Genótipo , Transplante de Células-Tronco Hematopoéticas , Humanos , Imunossupressores/uso terapêutico , Interleucinas/genética , Estimativa de Kaplan-Meier , Linfoma de Célula do Manto/imunologia , Linfoma de Célula do Manto/patologia , Linfoma de Célula do Manto/terapia , Masculino , Pessoa de Meia-Idade , Proteínas de Neoplasias/genética , Análise de Componente Principal , Prognóstico , Modelos de Riscos Proporcionais , Receptores de Fatores de Crescimento Transformadores beta/genética , Rituximab/uso terapêutico , Fatores de Transcrição SOXC/análise , Fator de Crescimento Transformador beta1/genética
11.
Acta Obstet Gynecol Scand ; 100(5): 868-875, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-32524577

RESUMO

INTRODUCTION: Oocyte competence and quality depend on communication between the oocyte and the cumulus and theca cells. In the preantral phase, the members of the transforming growth factor ß (TGF-ß) superfamily are responsible for this communication and play an important role in folliculogenesis. Members of the TGF-ß superfamily are related to endometriosis (overexpression in the ectopic endometrium); however, few studies have explored these proteins as influencing fertility in endometriosis. Considering endometriosis-related infertility and to better understand the role of the TGF-ß superfamily members in the antral phase in women with endometriosis, this research investigated the gene expression of the genes for ligands AMH, BMP-6, GDF-9, INHA, INHBB, and TGFß3; receptors AMHR2, BMPR2, and TGFßR3; and intracellular signalling: SMAD3 and SMAD4. MATERIAL AND METHODS: The gene expression of AMH, BMP-6, GDF-9, INHA, INHBB, TGFß3, AMHR2, BMPR2, TGFßR3, SMAD3, and SMAD4 in cumulus cells was investigated through quantitative real-time PCR in a case-control study including infertile women with and without peritoneal endometriosis undergoing in vitro fertilization. RESULTS: Age and outcomes of assisted reproduction were similar between the groups (P > .05). However, women with endometriosis showed reduced expression of BMP-6 and SMAD4 (P < .05) in cumulus cells compared with the control group, other genes did not present altered gene expression in women with endometriosis (P > .05). CONCLUSIONS: The reduced expression of BMP-6 and SMAD4 in women with peritoneal endometriosis compared with the control group indicates that granulosa (cumulus) cell function could be altered in these women.


Assuntos
Proteína Morfogenética Óssea 6/genética , Células do Cúmulo/metabolismo , Endometriose/complicações , Expressão Gênica , Infertilidade Feminina/complicações , Proteína Smad4/genética , Adulto , Hormônio Antimülleriano , Receptores de Proteínas Morfogenéticas Ósseas Tipo II , Estudos de Casos e Controles , Feminino , Fator 9 de Diferenciação de Crescimento , Humanos , Subunidades beta de Inibinas , Inibinas , Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Ligantes , Proteoglicanas , Reação em Cadeia da Polimerase em Tempo Real , Receptores de Superfície Celular/genética , Receptores de Superfície Celular/metabolismo , Receptores de Peptídeos , Receptores de Fatores de Crescimento Transformadores beta , Proteína Smad3 , Fator de Crescimento Transformador beta/genética , Fator de Crescimento Transformador beta/metabolismo , Fator de Crescimento Transformador beta3
12.
Mediators Inflamm ; 2020: 3185015, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33149723

RESUMO

Individuals with sickle cell disease (SCD) present both chronic and acute inflammatory events. The TGF-ß pathway is known to play a role in immune response, angiogenesis, inflammation, hematopoiesis, vascular inflammation, and cell proliferation. Polymorphisms in the transforming growth factor-beta receptor 3 (TGFBR3) gene have been linked to several inflammatory diseases. This study investigated associations between two TGFBR3 haplotypes and classical laboratory parameters, as well as clinical manifestations, in SCD. We found that individuals with the GG haplotype presented higher levels of total cholesterol (TC), low-density lipoprotein cholesterol (LDL-C), triglycerides, non-HDL cholesterol, total proteins, and globulin than individuals with non-GG haplotypes. In addition, the GG haplotype was associated with a previous history of pneumonia. Individuals with the CGG haplotype presented increased plateletcrit, TC, LDL-C levels, and non-HDL cholesterol. The CCG haplotype was also associated with a previous history of pneumonia. Our findings suggest that individuals with the GG and CGG haplotypes of TGFBR3 present important alterations in lipid profile.


Assuntos
Anemia Falciforme/sangue , Anemia Falciforme/genética , Haplótipos , Hemoglobinas/metabolismo , Lipídeos/química , Polimorfismo de Nucleotídeo Único , Proteoglicanas/genética , Receptores de Fatores de Crescimento Transformadores beta/genética , Adolescente , Biomarcadores/metabolismo , Proliferação de Células , Criança , Colesterol/metabolismo , LDL-Colesterol , Feminino , Genótipo , Humanos , Inflamação , Desequilíbrio de Ligação , Masculino , Pneumonia/metabolismo , Prognóstico , Proteoglicanas/sangue , Receptores de Fatores de Crescimento Transformadores beta/sangue , Adulto Jovem
13.
Dis Markers ; 2020: 8867986, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33062074

RESUMO

Individuals with sickle cell anemia (SCA) present chronic anemia, hemolysis, an exacerbated inflammatory response, and heterogeneous clinical complications, which may be modulated by the transforming growth factor beta (TGF-ß) pathway. Thus, we aimed to investigate polymorphisms (rs1805110 and rs7526590) of the transforming growth factor beta receptor III gene (TGFBR3) with regard to laboratory biomarkers and clinical manifestations in individuals with SCA. Hematological, biochemical, immunological, and genetic analyses were carried out, as well as serum endothelin-1 measurements. The minor allele (A) of the TGFBR3 rs1805110 polymorphism was associated with increased hemoglobin, hematocrit, reticulocyte counts, total cholesterol, low-density lipoprotein, uric acid, and endothelin levels, as well as decreased platelet distribution width (PDW) and the occurrence of bone alterations. The minor allele (T) of TGFBR3 rs7526590 was associated with increased red cell distribution width, PDW, alkaline phosphatase, aspartate aminotransferase, total and indirect bilirubin, and lactate dehydrogenase levels, as well as lower ferritin levels and the occurrence of leg ulcers. Our data suggest that the minor allele (A) of TGFBR3 rs1805110 is associated with inflammation and bone alterations, while the minor allele (T) of TGFBR3 rs7526590 is related to hemolysis and the occurrence of leg ulcers.


Assuntos
Anemia Falciforme/patologia , Biomarcadores/sangue , Índices de Eritrócitos , Polimorfismo de Nucleotídeo Único , Proteoglicanas/genética , Receptores de Fatores de Crescimento Transformadores beta/genética , Adolescente , Anemia Falciforme/sangue , Anemia Falciforme/genética , Criança , Estudos Transversais , Feminino , Seguimentos , Hemólise , Humanos , Masculino , Prognóstico
14.
Hum Mol Genet ; 28(19): 3211-3218, 2019 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-31238341

RESUMO

The persistent Müllerian duct syndrome (PMDS) is a 46,XY disorder of sexual development characterized by the persistence of Müllerian duct derivatives, uterus and tubes, in otherwise normally masculinized males. The condition, transmitted as a recessive autosomal trait, is usually due to mutations in either the anti-Müllerian hormone (AMH) gene or its main receptor. Many variants of these genes have been described, all targeting the coding sequences. We report the first case of PMDS due to a regulatory mutation. The AMH promoter contains two binding sites for steroidogenic factor 1 (SF1), one at -102 and the other at -228. Our patient carries a single base deletion at -225, significantly decreasing its capacity for binding SF1, as measured by the electrophoresis mobility shift assay. Furthermore, by linking the AMH promoter to the luciferase gene, we show that the transactivation capacity of the promoter is significantly decreased by the mutation, in contrast to the disruption of the -102 binding site. To explain the difference in impact we hypothesize that SF1 could partially overcome the lack of binding to the -102 binding site by interacting with a GATA4 molecule linked to a nearby response element. We show that disruption of both the -102 SF1 and the -84 GATA response elements significantly decreases the transactivation capacity of the promoter. In conclusion, we suggest that the distance between mutated SF1 sites and potentially rescuing GATA binding motifs might play a role in the development of PMDS.


Assuntos
Hormônio Antimülleriano/química , Hormônio Antimülleriano/metabolismo , Transtorno 46,XY do Desenvolvimento Sexual/genética , Mutação , Fatores de Processamento de RNA/metabolismo , Receptores de Peptídeos/genética , Receptores de Fatores de Crescimento Transformadores beta/genética , Hormônio Antimülleriano/genética , Sítios de Ligação/genética , Linhagem Celular , Criança , Pré-Escolar , Feminino , Humanos , Recém-Nascido , Masculino , Linhagem , Regiões Promotoras Genéticas , Ligação Proteica
15.
Reprod Domest Anim ; 54(1): 46-54, 2019 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-30120850

RESUMO

The objective of this study was to examine the expression of transforming growth factor beta receptor (TGFBR)1, TGFBR2, TGFBR3, activin receptor (ACVR)1B and ACVR2B in ovaries of cows with cystic ovarian disease (COD). The expression of the selected receptors was determined by immunohistochemistry in sections of ovaries from cows with ACTH-induced and spontaneous COD. Expression of TGFBR1 and TGFBR3 was higher in granulosa cells of cysts from cows with spontaneous COD than in tertiary follicles from the control group. Additionally, TGFBR3 expression was higher in granulosa cells of cysts from cows with ACTH-induced COD than in those from the control group and lower in theca cells of spontaneous and ACTH-induced cysts than in tertiary control follicles. There were no changes in the expression of TGFBR2. ACVR1B expression was higher in granulosa cells of tertiary follicles of cows with spontaneous COD than in the control group, whereas ACVR2B expression was higher in cysts of the spontaneous COD group than in tertiary follicles from the control group. The alterations here detected, together with the altered expression of the ligands previously reported, indicate alterations in the response of the ligands in the target cells, modifying their actions at cellular level.


Assuntos
Receptores de Ativinas/metabolismo , Doenças dos Bovinos/metabolismo , Cistos Ovarianos/veterinária , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Hormônio Adrenocorticotrópico/administração & dosagem , Animais , Bovinos , Feminino , Células da Granulosa/metabolismo , Imuno-Histoquímica , Cistos Ovarianos/induzido quimicamente , Cistos Ovarianos/metabolismo , Ovário/metabolismo , Células Tecais/metabolismo
16.
Zygote ; 26(3): 232-241, 2018 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-29880072

RESUMO

SummaryDuring preimplantation development, embryos are exposed and have the capacity to respond to different growth factors present in the maternal environment. Among these factors, transforming growth factor ß1 (TGF-ß1) is a well known modulator of embryonic growth and development. However, its action during the first stages of development, when the embryo transits through the oviduct, has not been yet elucidated. The objective of the present study was to examine the effect of early exposure to exogenous TGF-ß1 on embryo development and expression of pluripotency (OCT4, NANOG) and DNA methylation (DNMT1, DNMT3A, DNMT3B) genes in bovine embryos produced in vitro. First, gene expression analysis of TGF-ß receptors confirmed a stage-specific expression pattern, showing greater mRNA abundance of TGFBR1 and TGFBR2 from the 2- to the 8-cell stage, before embryonic genome activation. Second, embryo culture for the first 48 h in serum-free CR1aa medium supplemented with 50 or 100 ng/ml recombinant TGF-ß1 did not affect the cleavage and blastocyst rate (days 7 and 8). However, RT-qPCR analysis showed a significant increase in the relative abundance of NANOG and DNMT3A in the 8-cell stage embryos and expanded blastocysts (day 8) derived from TGF-ß1 treated embryos. These results suggest an early action of exogenous TGF-ß1 on the bovine embryo, highlighting the importance to provide a more comprehensive understanding of the role of TGF-ß signalling during early embryogenesis.


Assuntos
Desenvolvimento Embrionário/efeitos dos fármacos , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Fator de Crescimento Transformador beta1/farmacologia , Animais , Blastocisto/efeitos dos fármacos , Blastocisto/fisiologia , Bovinos , DNA (Citosina-5-)-Metiltransferase 1/genética , DNA (Citosina-5-)-Metiltransferases/genética , Metilação de DNA/efeitos dos fármacos , Metilação de DNA/genética , Técnicas de Cultura Embrionária/métodos , Técnicas de Cultura Embrionária/veterinária , Desenvolvimento Embrionário/genética , Feminino , Fertilização in vitro , Masculino , Proteína Homeobox Nanog/genética , Fator 3 de Transcrição de Octâmero/genética , Receptores de Fatores de Crescimento Transformadores beta/genética
17.
J Oral Pathol Med ; 47(1): 78-85, 2018 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-29032608

RESUMO

BACKGROUND: Cemento-ossifying fibroma (COF) is a benign fibro-osseous neoplasm of uncertain pathogenesis, and its treatment results in morbidity. MicroRNAs (miRNA) are small non-coding RNAs that regulate gene expression and may represent therapeutic targets. The purpose of the study was to generate a comprehensive miRNA profile of COF compared to normal bone. Additionally, the most relevant pathways and target genes of differentially expressed miRNA were investigated by in silico analysis. METHODS: Nine COF and ten normal bone samples were included in the study. miRNA profiling was carried out by using TaqMan® OpenArray® Human microRNA panel containing 754 validated human miRNAs. We identified the most relevant miRNAs target genes through the leader gene approach, using STRING and Cytoscape software. Pathways enrichment analysis was performed using DIANA-miRPath. RESULTS: Eleven miRNAs were downregulated (hsa-miR-95-3p, hsa-miR-141-3p, hsa-miR-205-5p, hsa-miR-223-3p, hsa-miR-31-5p, hsa-miR-944, hsa-miR-200b-3p, hsa-miR-135b-5p, hsa-miR-31-3p, hsa-miR-223-5p and hsa-miR-200c-3p), and five were upregulated (hsa-miR-181a-5p, hsa-miR-181c-5p, hsa-miR-149-5p, hsa-miR-138-5p and hsa-miR-199a-3p) in COF compared to normal bone. Eighteen common target genes were predicted, and the leader genes approach identified the following genes involved in human COF: EZH2, XIAP, MET and TGFBR1. According to the biology of bone and COF, the most relevant KEGG pathways revealed by enrichment analysis were proteoglycans in cancer, miRNAs in cancer, pathways in cancer, p53-, PI3K-Akt-, FoxO- and TGF-beta signalling pathways, which were previously found to be differentially regulated in bone neoplasms, odontogenic tumours and osteogenesis. CONCLUSION: miRNA dysregulation occurs in COF, and EZH2, XIAP, MET and TGFBR1 are potential targets for functional analysis validation.


Assuntos
Neoplasias Ósseas/genética , Neoplasias Ósseas/metabolismo , Fibroma Ossificante/genética , Fibroma Ossificante/metabolismo , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Redes Reguladoras de Genes , MicroRNAs/genética , Adolescente , Adulto , Biologia Computacional , Regulação para Baixo , Proteína Potenciadora do Homólogo 2 de Zeste/genética , Feminino , Fatores de Transcrição Forkhead/metabolismo , Estudos de Associação Genética , Humanos , Masculino , MicroRNAs/classificação , Pessoa de Meia-Idade , Proteínas de Neoplasias/metabolismo , Tumores Odontogênicos , Osteogênese , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Serina-Treonina Quinases/genética , Proteínas Proto-Oncogênicas c-met/genética , RNA não Traduzido , Receptor do Fator de Crescimento Transformador beta Tipo I , Receptores de Fatores de Crescimento Transformadores beta/genética , Proteína Supressora de Tumor p53/metabolismo , Regulação para Cima , Proteínas Inibidoras de Apoptose Ligadas ao Cromossomo X/genética , Adulto Jovem
18.
Exp Cell Res ; 362(2): 260-267, 2018 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-29208458

RESUMO

Chagasic cardiomyopathy (CC) is the main manifestation of Chagas Disease (CD). CC is a progressive dysfunctional illness, in which transforming growth factor beta (TGF-ß) plays a central role in fibrogenesis and hypertrophy. In the present study, we tested in a three-dimensional (3D) model of cardiac cells culture (named cardiac spheroids), capable of mimicking the aspects of fibrosis and hypertrophy observed in CC, the role of TGF-ß pathway inhibition in restoring extracellular matrix (ECM) balance disrupted by T. cruzi infection. Treatment of T. cruzi-infected cardiac spheroids with SB 431542, a selective inhibitor of TGF-ß type I receptor, resulted in a reduction in the size of spheroids, which was accompanied by a decrease in parasite load and in fibronectin expression. The inhibition of TGF-ß pathway also promoted an increase in the activity of matrix metalloproteinase (MMP)-2 and a decrease in tissue inhibitor of matrix metalloproteinase (TIMP)-1 expression, which may be one of the mechanisms regulating extracellular matrix remodeling. Therefore, our study provides new insights into the molecular mechanisms by which inhibition of TGF-ß signaling reverts fibrosis and hypertrophy generated by T. cruzi during CC and also highlights the use of cardiac spheroids as a valuable tool for the study of fibrogenesis and anti-fibrotic compounds.


Assuntos
Cardiomiopatias/tratamento farmacológico , Doença de Chagas/tratamento farmacológico , Coração/fisiopatologia , Proteínas Serina-Treonina Quinases/genética , Receptores de Fatores de Crescimento Transformadores beta/genética , Benzamidas/farmacologia , Cardiomiopatias/genética , Cardiomiopatias/parasitologia , Cardiomiopatias/fisiopatologia , Doença de Chagas/genética , Doença de Chagas/parasitologia , Doença de Chagas/fisiopatologia , Dioxóis/farmacologia , Matriz Extracelular/genética , Fibronectinas/genética , Regulação da Expressão Gênica/efeitos dos fármacos , Coração/parasitologia , Humanos , Metaloproteinase 2 da Matriz/genética , Receptor do Fator de Crescimento Transformador beta Tipo I , Esferoides Celulares/efeitos dos fármacos , Esferoides Celulares/patologia , Inibidor Tecidual de Metaloproteinase-1/genética , Fator de Crescimento Transformador beta/genética , Trypanosoma cruzi/efeitos dos fármacos , Trypanosoma cruzi/patogenicidade
19.
Cell Biol Int ; 42(4): 443-456, 2018 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-29227005

RESUMO

Diabetes is associated with metabolic and functional alterations in the gut. Using an experimental model of streptozotocin (STZ)-induced diabetes in rodents, we analyzed the extracellular matrix (ECM) and TGF-ß/Smad signaling in the colon mucosa. Male rats were divided into normal control, diabetic and insulin treated diabetic groups during 4 and 9 weeks. Sirius red staining showed marked increase in the extracellular matrix deposition in diabetic mucosa. High levels of fibrillar collagen (I and III) and fibronectin mRNAs were also detected with an imbalance between MMPs/TIMPs activities. Moreover, an increased mesenchymal cell proliferation together with an enhanced expression of myofibroblasts markers vimentin and α-SMA were observed. TGF-ß/Smad signaling-related genes were determined using RT-PCR, Western blotting, and immunohistochemistry. Diabetic rats showed a significant up-regulation of TGF-ß1, TGF-ß receptors and the effectors p-Smad2/3 in the mucosa compared with control rats. Insulin treatment attenuated the stimulating effect of diabetes on colon ECM deposition and TGF-ß/Smad signaling. In conclusion, the overall results showed a deregulation of the TGFß1 pathway associated with the appearance of myofibroblasts and the accumulation of ECM in the mucosa of diabetic colon. These data provide the first in vivo evidence that TGF-ß1/Smad is a key component of intestinal tissue remodeling in diabetes.


Assuntos
Colo/metabolismo , Diabetes Mellitus Experimental/metabolismo , Matriz Extracelular/metabolismo , Mucosa Intestinal/metabolismo , Proteínas Smad/metabolismo , Fator de Crescimento Transformador beta1/metabolismo , Animais , Diabetes Mellitus Experimental/patologia , Colágenos Fibrilares/efeitos dos fármacos , Fibronectinas/metabolismo , Masculino , Miofibroblastos/metabolismo , Ratos , Ratos Wistar , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Transdução de Sinais/efeitos dos fármacos
20.
Biochem Biophys Res Commun ; 494(1-2): 82-87, 2017 12 09.
Artigo em Inglês | MEDLINE | ID: mdl-29050936

RESUMO

TGF-ß type III receptor (TßRIII) is a co-receptor for TGFß family members required for high-affinity binding of these ligands to their receptors, potentiating their cellular functions. TGF-ßs, Bone Morphogenetic Proteins (BMP2/4) and Inhibins/Activins regulate different checkpoints during T cell differentiation. We have previously reported that TßRIII modulates T cell development by protecting developing thymocytes from apoptosis, however the role of this co-receptor in peripheral lymphocytes still remains elusive. Here we describe a detailed characterization of TßRIII expression in murine and human lymphocyte subpopulations demonstrating that this co-receptor is significantly expressed in T but not B lymphocytes and among them, preferentially expressed on naïve and central memory T cells. TßRIII was upregulated after TCR stimulation, in parallel to other early activation markers. In contrast, natural and induced Tregs downregulated TßRIII in association with FoxP3 upregulation. Finally, anti-TßRIII blocking experiments demonstrated that TßRIII promotes TGFß-dependent iTreg conversion in vitro, and suggest that this co-receptor may be involved in modulating peripheral T cell tolerance and could be considered as a potential target to boost T cell immune responses.


Assuntos
Proteoglicanas/metabolismo , Receptores de Antígenos de Linfócitos T/metabolismo , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Linfócitos T Reguladores/imunologia , Animais , Regulação para Baixo , Fatores de Transcrição Forkhead/metabolismo , Humanos , Memória Imunológica , Técnicas In Vitro , Ativação Linfocitária , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteoglicanas/antagonistas & inibidores , Proteoglicanas/genética , Receptores de Fatores de Crescimento Transformadores beta/antagonistas & inibidores , Receptores de Fatores de Crescimento Transformadores beta/genética , Transdução de Sinais , Linfócitos T Reguladores/classificação , Linfócitos T Reguladores/metabolismo , Regulação para Cima
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA