Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 767
Filtrar
1.
Injury ; 53(12): 4165-4168, 2022 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-36261312

RESUMO

The repair of the tendon-bone interface, which is composed of tendon, fibrocartilage, and bony attachment, remains a clinical challenge. The application of mesenchymal stem cells (MSCs), collagen-rich extracellular matrix (ECMs), as well as growth factors, has the potential to regenerate this special multiple-tissue structure through the so-called biological augmentation. We present here an in vitro tendon regeneration model with C3H10T1/2 cells cultured on Collagen I matrix and evaluated the lineage determination effects of Growth Differentiation Factor 7 (GDF-7). We found that besides tenogenic effect, GDF-7 also stimulates the expression of osteoblastic as well as adipocytic genes. Our results indicate that GDF-7 might be a promising growth factor for regeneration of the tendon-bone interface due to its multiple-lineage stimulating effects. However, the side effect on adipogenic differentiation should be of concern, as it is a known risk factor for repair failures.


Assuntos
Proteínas Morfogenéticas Ósseas , Diferenciação Celular , Células-Tronco Mesenquimais , Proteínas Morfogenéticas Ósseas/fisiologia , Colágeno/metabolismo , Animais , Camundongos , Linhagem Celular , Tendões , Regeneração
2.
Reprod Biol Endocrinol ; 20(1): 34, 2022 Feb 19.
Artigo em Inglês | MEDLINE | ID: mdl-35183204

RESUMO

BACKGROUND: Growth differentiation factor-11 (GDF-11) belongs to the transforming growth factor-ß (TGF-ß) superfamily. To date, the expression of GDF-11 in the ovary and its role in regulating ovarian function are completely unknown. Ovarian granulosa cell-mediated steroidogenesis plays a pivotal role in maintaining normal female reproductive function. GDF-11 and GDF-8 share high sequence similarity and exhibit many similar features and functions. Steroidogenic acute regulatory protein (StAR) regulates the rate-limiting step in steroidogenesis and its expression can be downregulated by GDF-8. Polycystic ovary syndrome (PCOS) is the most common cause of female infertility. The expression levels of GDF-8 are upregulated in the human follicular fluid and granulosa-lutein (hGL) cells of PCOS patients. However, whether similar results can be observed for the GDF-11 needs to be determined. METHODS: The effect of GDF-11 on StAR expression and the underlying molecular mechanisms were explored by a series of in vitro experiments in a primary culture of hGL cells obtained from patients undergoing in vitro fertilization (IVF) treatment. Human follicular fluid samples were obtained from 36 non-PCOS patients and 36 PCOS patients. GDF-11 levels in follicular fluid were measured by ELISA. RESULTS: GDF-11 downregulates StAR expression, whereas the expression levels of the P450 side-chain cleavage enzyme (P450scc) and 3ß-hydroxysteroid dehydrogenase (3ß-HSD) are not affected by GDF-11 in hGL cells. Using pharmacological inhibitors and a siRNA-mediated approach, we reveal that ALK5 but not ALK4 mediates the suppressive effect of GDF-11 on StAR expression. Although GDF-11 activates both SMAD2 and SMAD3 signaling pathways, only SMAD3 is involved in the GDF-11-induced downregulation of StAR expression. In addition, we show that SMAD1/5/8, ERK1/2, and PI3K/AKT signaling pathways are not activated by GDF-11 in hGL cells. RT-qPCR and ELISA detect GDF-11 mRNA expression in hGL cells and GDF-11 protein expression in human follicular fluid, respectively. Interestingly, unlike GDF-8, the expression levels of GDF-11 are not varied in hGL cells and follicular fluid between non-PCOS and PCOS patients. CONCLUSIONS: This study increases the understanding of the biological function of GDF-11 and provides important insights into the regulation of ovarian steroidogenesis.


Assuntos
Proteínas Morfogenéticas Ósseas/fisiologia , Fatores de Diferenciação de Crescimento/fisiologia , Células Lúteas/metabolismo , Fosfoproteínas/genética , Adulto , Células Cultivadas , Regulação para Baixo/genética , Feminino , Líquido Folicular/metabolismo , Células da Granulosa/metabolismo , Humanos , Infertilidade Feminina/genética , Infertilidade Feminina/metabolismo , Fosfoproteínas/metabolismo , Síndrome do Ovário Policístico/genética , Síndrome do Ovário Policístico/metabolismo , Receptor do Fator de Crescimento Transformador beta Tipo I/metabolismo , Transdução de Sinais/fisiologia , Proteína Smad3/metabolismo
3.
Int J Mol Sci ; 22(9)2021 Apr 29.
Artigo em Inglês | MEDLINE | ID: mdl-33946990

RESUMO

Ubiquitination and deubiquitination are protein post-translational modification processes that have been recognized as crucial mediators of many complex cellular networks, including maintaining ubiquitin homeostasis, controlling protein stability, and regulating several signaling pathways. Therefore, some of the enzymes involved in ubiquitination and deubiquitination, particularly E3 ligases and deubiquitinases, have attracted attention for drug discovery. Here, we review recent findings on USP15, one of the deubiquitinases, which regulates diverse signaling pathways by deubiquitinating vital target proteins. Even though several basic previous studies have uncovered the versatile roles of USP15 in different signaling networks, those have not yet been systematically and specifically reviewed, which can provide important information about possible disease markers and clinical applications. This review will provide a comprehensive overview of our current understanding of the regulatory mechanisms of USP15 on different signaling pathways for which dynamic reverse ubiquitination is a key regulator.


Assuntos
Processamento de Proteína Pós-Traducional/fisiologia , Transdução de Sinais/fisiologia , Proteases Específicas de Ubiquitina/fisiologia , Animais , Receptores de Proteínas Morfogenéticas Ósseas Tipo I/metabolismo , Proteínas Morfogenéticas Ósseas/fisiologia , Complexo do Signalossomo COP9/fisiologia , Humanos , Imunidade Inata , Masculino , Camundongos , NF-kappa B/metabolismo , Neoplasias da Próstata/metabolismo , Domínios Proteicos , Isoformas de Proteínas , Proteínas Proto-Oncogênicas c-mdm2/metabolismo , Precursores de RNA/metabolismo , Receptor do Fator de Crescimento Transformador beta Tipo I/metabolismo , Proteínas Smad/metabolismo , Fator de Crescimento Transformador beta/fisiologia , Proteína Supressora de Tumor p53/metabolismo , Proteases Específicas de Ubiquitina/química , Proteases Específicas de Ubiquitina/genética , Ubiquitinação , Via de Sinalização Wnt/fisiologia , Proteínas de Xenopus/metabolismo
4.
Theranostics ; 11(13): 6573-6591, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33995677

RESUMO

Mesenchymal stem cells (MSCs) have been identified in many adult tissues. MSCs can regenerate through cell division or differentiate into adipocytes, osteoblasts and chondrocytes. As a result, MSCs have become an important source of cells in tissue engineering and regenerative medicine for bone tissue and cartilage. Several epigenetic factors are believed to play a role in MSCs differentiation. Among these, microRNA (miRNA) regulation is involved in the fine modulation of gene expression during osteogenic/chondrogenic differentiation. It has been reported that miRNAs are involved in bone homeostasis by modulating osteoblast gene expression. In addition, countless evidence has demonstrated that miRNAs dysregulation is involved in the development of osteoporosis and bone fractures. The deregulation of miRNAs expression has also been associated with several malignancies including bone cancer. In this context, bone-associated circulating miRNAs may be useful biomarkers for determining the predisposition, onset and development of osteoporosis, as well as in clinical applications to improve the diagnosis, follow-up and treatment of cancer and metastases. Overall, this review will provide an overview of how miRNAs activities participate in osteogenic/chondrogenic differentiation, while addressing the role of miRNA regulatory effects on target genes. Finally, the role of miRNAs in pathologies and therapies will be presented.


Assuntos
Doenças Ósseas/genética , Condrogênese/genética , Células-Tronco Mesenquimais/citologia , MicroRNAs/genética , Osteogênese/genética , Proteínas Morfogenéticas Ósseas/fisiologia , Subunidade alfa 1 de Fator de Ligação ao Core/fisiologia , Sistemas de Liberação de Medicamentos , Fraturas Ósseas/metabolismo , Histona Desacetilases/fisiologia , Humanos , Metaloproteinase 13 da Matriz/fisiologia , Proteínas Repressoras/fisiologia , Transdução de Sinais , Proteínas Smad/fisiologia , Fator de Transcrição Sp7/fisiologia , Fator de Crescimento Transformador beta/fisiologia , Fator A de Crescimento do Endotélio Vascular/fisiologia
5.
Curr Treat Options Oncol ; 22(6): 48, 2021 04 17.
Artigo em Inglês | MEDLINE | ID: mdl-33866453

RESUMO

OPINION STATEMENT: Malignant melanoma is a deadly form of skin cancer caused by neoplastic transformation of melanocytic cells. Despite recent progress in melanoma therapy, by inhibition of activated oncogenes or immunotherapy, survival rate for metastatic melanoma patients remains low. The remarkable phenotypic plasticity of melanoma cells allows for rapid development of invasive properties and metastatic tumors, the main cause of mortality in melanoma patients. Phenotypic and molecular analyses of developing tumors revealed that epithelial-mesenchymal transition (EMT), a cellular and molecular mechanism, controls transition from mature melanocyte to less differentiated melanocyte lineage progenitor cells forming melanoma tumors. This transition is facilitated by persistence of transcriptional regulatory circuit characteristic of embryonic stage in mature melanocytes. Switching of the developmental program of mature melanocyte to EMT is induced by accumulated mutations, especially targeting BRAF, N-RAS, or MEK1/2 signaling pathways, and further promoted by dynamic stimuli from local environment including hypoxia, interactions with extracellular matrix and growth factors or cytokines. Recent reports demonstrate that signaling mediated by transforming growth factor-ß (TGF-ß) and bone morphogenic proteins (BMPs) play critical roles in inducing EMT by controlling expression of critical transcription factors. BMPs are essential modulators of differentiation, proliferation, apoptosis, invasiveness, and metastases in developing melanoma tumors. They control transcription and epigenetic landscape of melanoma cells. Better understanding of the role of BMPs may lead to new strategies to control EMT processes in melanocyte cell lineage and to achieve clinical benefits for the patients.


Assuntos
Proteínas Morfogenéticas Ósseas/fisiologia , Melanoma/etiologia , Neoplasias Cutâneas/etiologia , Transformação Celular Neoplásica , Transição Epitelial-Mesenquimal , Humanos , Melanoma/patologia , Transdução de Sinais/fisiologia , Neoplasias Cutâneas/patologia
6.
Surg Today ; 51(3): 340-349, 2021 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-32754843

RESUMO

Organ liver transplantation and hepatocyte transplantation are not performed to their full potential because of donor shortage, which could be resolved by identifying new donor sources for the development of hepatocyte-like cells (HLCs). HLCs have been differentiated from some stem cell sources as alternative primary hepatocytes throughout the world; however, the currently available techniques cannot differentiate HLCs to the level of normal adult primary hepatocytes. The outstanding questions are as follows: which stem cells are the best cell sources? which protocol is the best way to differentiate them into HLCs? what is the definition of differentiated HLCs? how can we enforce the function of HLCs? what is the difference between HLCs and primary hepatocytes? what are the problems with HLC transplantation? This review summarizes the current status of HLCs, focusing on stem cell sources, the differentiation protocol for HLCs, the general characterization of HLCs, the generation of more functional HLCs, comparison with primary hepatocytes, and HLCs in cell-transplantation-based liver regeneration.


Assuntos
Diferenciação Celular , Terapia Baseada em Transplante de Células e Tecidos/métodos , Técnicas Citológicas/métodos , Hepatócitos/transplante , Hepatopatias/terapia , Células-Tronco/fisiologia , Proteínas Morfogenéticas Ósseas/fisiologia , Diferenciação Celular/genética , Células Cultivadas , Técnicas de Cocultura , Fatores de Crescimento de Fibroblastos/fisiologia , Fator 4 Nuclear de Hepatócito/fisiologia , Proteínas de Homeodomínio/fisiologia , Humanos , Regeneração Hepática/fisiologia , Fatores de Transcrição SOXF/fisiologia , Fatores de Transcrição/fisiologia
7.
Int J Mol Sci ; 21(18)2020 Sep 05.
Artigo em Inglês | MEDLINE | ID: mdl-32899497

RESUMO

Activins transduce the TGF-ß pathway through a heteromeric signaling complex consisting of type I and type II receptors, and activins also inhibit bone morphogenetic protein (BMP) signaling mediated by type I receptor ALK2. Recent studies indicated that activin A cross-activates the BMP pathway through ALK2R206H, a mutation associated with Fibrodysplasia Ossificans Progressiva (FOP). How activin A inhibits ALK2WT-mediated BMP signaling but activates ALK2R206H-mediated BMP signaling is not well understood, and here we offer some insights into its molecular mechanism. We first demonstrated that among four BMP type I receptors, ALK2 is the only subtype able to mediate the activin A-induced BMP signaling upon the dissociation of FKBP12. We further showed that BMP4 does not cross-signal TGF-ß pathway upon FKBP12 inhibition. In addition, although the roles of type II receptors in the ligand-independent BMP signaling activated by FOP-associated mutant ALK2 have been reported, their roles in activin A-induced BMP signaling remains unclear. We demonstrated in this study that the known type II BMP receptors contribute to activin A-induced BMP signaling through their kinase activity. Together, the current study provided important mechanistic insights at the molecular level into further understanding physiological and pathophysiological BMP signaling.


Assuntos
Receptores de Ativinas Tipo I/metabolismo , Ativinas/metabolismo , Proteínas Morfogenéticas Ósseas/metabolismo , Receptores de Ativinas Tipo I/genética , Receptores de Activinas Tipo II/genética , Receptores de Activinas Tipo II/metabolismo , Ativinas/fisiologia , Animais , Receptores de Proteínas Morfogenéticas Ósseas Tipo I/genética , Receptores de Proteínas Morfogenéticas Ósseas Tipo II/genética , Proteínas Morfogenéticas Ósseas/fisiologia , Diferenciação Celular/fisiologia , Linhagem Celular , Regulação da Expressão Gênica/genética , Células HEK293 , Células Hep G2 , Humanos , Camundongos , Ossificação Heterotópica/genética , Fosforilação , Transdução de Sinais/genética , Transdução de Sinais/fisiologia , Fator de Crescimento Transformador beta/metabolismo
8.
Med Sci Monit ; 26: e920943, 2020 Apr 05.
Artigo em Inglês | MEDLINE | ID: mdl-32248202

RESUMO

BACKGROUND Bone morphogenetic proteins (BMPs) are widely involved in cancer development. However, a wealth of conflicting data raises the question of whether BMPs serve as oncogenes or as cancer suppressors. MATERIAL AND METHODS By integrating multi-omics data across cancers, we comprehensively analyzed the genomic and pharmacogenomic landscape of BMP genes across cancers. RESULTS Surprisingly, our data indicate that BMPs are globally downregulated in cancers. Further genetics and epigenetics analyses show that this abnormal expression is driven by copy number variations, especially heterozygous amplification. We next assessed the BMP-associated pathways and demonstrated that they suppress cell cycle and estrogen hormone pathways. Bone morphogenetic protein interacts with 58 compounds, and their dysfunction can induce drug sensitivity. CONCLUSIONS Our results define the landscape of the BMP family at a systems level and open potential therapeutic opportunities for cancer patients.


Assuntos
Proteínas Morfogenéticas Ósseas , Neoplasias/metabolismo , Animais , Proteínas Morfogenéticas Ósseas/genética , Proteínas Morfogenéticas Ósseas/metabolismo , Proteínas Morfogenéticas Ósseas/fisiologia , Variações do Número de Cópias de DNA , Humanos , Fator de Crescimento Transformador beta/genética , Fator de Crescimento Transformador beta/metabolismo , Fator de Crescimento Transformador beta/fisiologia
9.
Sci Rep ; 10(1): 1136, 2020 01 24.
Artigo em Inglês | MEDLINE | ID: mdl-31980720

RESUMO

One current concept suggests that unchecked proliferation of clonally selected precursors of endothelial cells (ECs) contribute to severe pulmonary arterial hypertension (PAH). We hypothesized that clonally selected ECs expressing the progenitor marker CD117 promote severe occlusive pulmonary hypertension (PH). The remodelled pulmonary arteries of PAH patients harboured CD117+ ECs. Rat lung CD117+ ECs underwent four generations of clonal expansion to enrich hyperproliferative ECs. The resulting clonally enriched ECs behaved like ECs, as measured by in vitro and in vivo angiogenesis assays. The same primitive ECs showed a limited ability for mesenchymal lineage differentiation. Endothelial differentiation and function were enhanced by blocking TGF-ß signalling, promoting bone morphogenic protein (BMP) signalling. The transplantation of the EC clones caused arterio-occlusive PH in rats exposed to chronic hypoxia. These EC clones engrafted in the pulmonary arteries. Yet cessation of chronic hypoxia promoted lung cell apoptosis and resolution of vascular lesions. In conclusion, this is to the best of our knowledge, the first report that clonally enriched primitive ECs promote occlusive pulmonary arteriopathy and severe PH. These primitive EC clones further give rise to cells of endothelial and mesenchymal lineage as directed by BMP and TGF-ß signaling.


Assuntos
Arteriopatias Oclusivas/etiologia , Células Endoteliais/patologia , Hipertensão Pulmonar/etiologia , Hipóxia/patologia , Artéria Pulmonar/patologia , Animais , Apoptose , Arteriopatias Oclusivas/patologia , Proteínas Morfogenéticas Ósseas/fisiologia , Linhagem da Célula , Separação Celular , Células Cultivadas , Doença Crônica , Células Clonais , Células Endoteliais/química , Células Endoteliais/transplante , Citometria de Fluxo , Humanos , Hipertensão Pulmonar/genética , Hipertensão Pulmonar/patologia , Hipóxia/complicações , Masculino , Mesoderma/citologia , Proteínas Proto-Oncogênicas c-kit/análise , Ratos , Ratos Sprague-Dawley , Receptor do Fator de Crescimento Transformador beta Tipo I/antagonistas & inibidores , Transdução de Sinais , Transcriptoma , Fator de Crescimento Transformador beta/fisiologia
10.
J Orthop Surg Res ; 14(1): 462, 2019 Dec 23.
Artigo em Inglês | MEDLINE | ID: mdl-31870454

RESUMO

BACKGROUND: This study was conducted with the aim of exploring the effect of the BMP signaling pathway on osteoblastic differentiation in rat bone marrow mesenchymal stem cells (rBMSCs) in rats with osteoporosis (OP). METHODS: The bilateral ovaries of female SD rats were resected for the establishment of a rat OP model. The osteoblastic differentiation of isolated rBMSCs was identified through osteogenic induction. Adipogenetic induction and flow cytometry (FCM) were used to detect adipogenic differentiation and the expression of rBMSC surface markers. The rBMSCs were grouped into the blank group, NC group, si-BMP2 group, and oe-BMP2 group. The expression levels of key factors and osteogenesis-related factors were determined by Western blot and quantitative real-time polymerase chain reaction (qRT-PCR). The formation of calcified nodules was observed by alizarin red staining. ALP activity was measured by alkaline phosphatase staining. RESULTS: The rats with OP had greater weight but decreased bone mineral density (BMD) than normal rats (all P < 0.01). The rBMSCs from rats with OP were capable of osteoblastic differentiation and adipogenic differentiation and showed high expression of CD44 (91.3 ± 2.9%) and CD105 (94.8 ± 2.1%). Compared with the blank group, the oe-BMP2 group had elevated BMP-2 and Smad1 levels and an increase in calcified nodules and ALP-positive staining areas (all P < 0.05). Moreover, the expression levels of Runx2, OC, and OPN in the oe-BMP2 group were relatively higher than those in the blank group (all P < 0.05). The findings in the si-BMP2 group were opposite to those in the oe-BMP2 group. CONCLUSION: BMP signaling pathways activated by BMP-2 can promote the osteoblastic differentiation of rBMSCs from rats with OP.


Assuntos
Proteínas Morfogenéticas Ósseas/fisiologia , Diferenciação Celular , Células-Tronco Mesenquimais/citologia , Osteoblastos/citologia , Osteoporose/patologia , Transdução de Sinais , Animais , Feminino , Ratos , Ratos Sprague-Dawley
11.
Toxicol Appl Pharmacol ; 385: 114779, 2019 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-31697996

RESUMO

Osteoporosis is a common bone disease resulting from imbalance between bone formation and bone resorption. Currently, anti-resorptive agents that inhibit bone resorption are the most available drugs on the market. Biosphosphonates, anti-resorptive drugs most commonly used to treat osteoporosis, are limited by their side effects for long-term continuous treatment. It is important to develop appropriate therapeutic stragegies capable of promoting bone formation to counteract osteoporotic bone loss. Thus, anabolic agents that stimulate bone formation are undoubtedly of interest. Here, we purified and identified two novel osteogenic peptides AWLNH and PHDL from ark shell protein hydrolysates. AWLNH and PHDL stimulated osteoblast differentiation via mitogen-activated protein kinase (MAPK) and bone morphogenetic protein-2 (BMP-2) pathways. The activation of BMP-2 pathway stimulated by AWLNH and PHDL was abolished by treating noggin, BMP antagonist, in bone marrow-derived mesenchymal stem cells (BMMSCs), but not the phosphorylation of JNK1/2, ERK1/2, and p38 MAPK. However, treatment with MAPK inhibitors in BMMSCs downregulated the expression of BMP-2 and p-Smad1/5 and inhibited alkaline phosphatase activity. The dominant inhibitory effects by JNK inhibitor and ERK inhibitor are observed. In ovariectomized (OVX) mice, a reduction of femoral bone mineral density (BMD) was significantly observed, however, AWLNH and PHDL (0.2 mg/kg/per day) injection restored BMD as well as the osteoporotic conditions in OVX mice. Moreover, the increased serum osteocalcin and alkaline phosphatase activity in OVX mice were significantly reduced in AWLNH and PHDL injected-OVX mice. These results suggest that two novel osteogenic peptides AWLNH and PHDL could be attractive therapeutic agents for osteoporosis treatment.


Assuntos
Arcidae/química , Osteoblastos/efeitos dos fármacos , Osteoporose/prevenção & controle , Peptídeos/farmacologia , Fosfatase Alcalina/sangue , Animais , Proteínas Morfogenéticas Ósseas/fisiologia , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Feminino , Humanos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Ovariectomia , Hidrolisados de Proteína/análise , Hidrolisados de Proteína/farmacologia
12.
Clin Sci (Lond) ; 133(20): 2069-2084, 2019 10 30.
Artigo em Inglês | MEDLINE | ID: mdl-31654062

RESUMO

Growth differentiation factor 11 (GDF11) is a member of the transforming growth factor (TGF)-ß superfamily. The rejuvenative effect of GDF11 has been called into question recently, and its role in liver regeneration is unclear. Here, we investigated the pathophysiologic role of GDF11, as well as its plausible signaling mechanisms in a mouse model of partial hepatectomy (PH). We demonstrated that both serum and hepatic GDF11 protein expression increased following PH. Treatment with adeno-associated viruses-GDF11 and recombinant GDF11 protein severely impaired liver regeneration, whereas inhibition of GDF11 activity with neutralizing antibodies significantly improved liver regeneration after PH. In vitro, GDF11 treatment significantly delayed cell proliferation and induced cell-cycle arrest in α mouse liver 12 (AML12) cells. Moreover, GDF11 activated TGF-ß-SMAD2/3 signaling pathway. Inhibition of GDF11-induced SMAD2/3 activity significantly blocked GDF11-mediated reduction in cell proliferation both in vivo and in vitro. In the clinical setting, GDF11 levels were significantly elevated in patients after hepatectomy. Collectively, these results indicate that rather than a 'rejuvenating' agent, GDF11 impairs liver regeneration after PH. Suppression of cell-cycle progression via TGF-ß-SMAD2/3 signaling pathway may be a key mechanism by which GDF11 inhibits liver regeneration.


Assuntos
Proteínas Morfogenéticas Ósseas/fisiologia , Fatores de Diferenciação de Crescimento/fisiologia , Regeneração Hepática/fisiologia , Animais , Proteínas Morfogenéticas Ósseas/antagonistas & inibidores , Proteínas Morfogenéticas Ósseas/sangue , Proteínas Morfogenéticas Ósseas/metabolismo , Proteínas Morfogenéticas Ósseas/farmacologia , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Modelos Animais de Doenças , Fatores de Diferenciação de Crescimento/antagonistas & inibidores , Fatores de Diferenciação de Crescimento/sangue , Fatores de Diferenciação de Crescimento/metabolismo , Fatores de Diferenciação de Crescimento/farmacologia , Hepatectomia , Hepatócitos/efeitos dos fármacos , Hepatócitos/patologia , Humanos , Fígado/metabolismo , Fígado/patologia , Regeneração Hepática/efeitos dos fármacos , Masculino , Camundongos Endogâmicos C57BL , Período Pós-Operatório , Proteínas Recombinantes/farmacologia , Transdução de Sinais/fisiologia , Proteína Smad2/metabolismo , Proteína Smad3/metabolismo
13.
Cells ; 8(10)2019 09 23.
Artigo em Inglês | MEDLINE | ID: mdl-31547567

RESUMO

The Transforming Growth Factor beta (TGFß) and Bone Morphogenic Protein (BMP) pathways intersect at multiple signaling hubs and cooperatively or counteractively participate to bring about cellular processes which are critical not only for tissue morphogenesis and organogenesis during development, but also for adult tissue homeostasis. The proper functioning of the TGFß/BMP pathway depends on its communication with other signaling pathways and any deregulation leads to developmental defects or diseases, including fibrosis and cancer. In this review we explore the cellular and physio-pathological contexts in which the synergism or antagonism between the TGFß and BMP pathways are crucial determinants for the normal developmental processes, as well as the progression of fibrosis and malignancies.


Assuntos
Proteínas Morfogenéticas Ósseas/fisiologia , Neoplasias/genética , Organogênese/genética , Fator de Crescimento Transformador beta/fisiologia , Animais , Proteínas Morfogenéticas Ósseas/genética , Progressão da Doença , Fibrose/genética , Fibrose/patologia , Redes Reguladoras de Genes/fisiologia , Humanos , Neoplasias/patologia , Transdução de Sinais/genética , Fator de Crescimento Transformador beta/genética
14.
Am J Hematol ; 94(11): 1227-1235, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31400017

RESUMO

The erythroferrone (ERFE) is the erythroid regulator of hepatic iron metabolism by suppressing the expression of hepcidin. Congenital dyserythropoietic anemia type II (CDAII) is an inherited hyporegenerative anemia due to biallelic mutations in the SEC23B gene. Patients with CDAII exhibit marked clinical variability, even among individuals sharing the same pathogenic variants. The ERFE expression in CDAII is increased and related to abnormal erythropoiesis. We identified a recurrent low-frequency variant, A260S, in the ERFE gene in 12.5% of CDAII patients with a severe phenotype. We demonstrated that the ERFE-A260S variant leads to increased levels of ERFE, with subsequently marked impairment of iron regulation pathways at the hepatic level. Functional characterization of ERFE-A260S in the hepatic cell system demonstrated its modifier role in iron overload by impairing the BMP/SMAD pathway. We herein described for the first time an ERFE polymorphism as a genetic modifier variant. This was with a mild effect on disease expression, under a multifactorial-like model, in a condition of iron-loading anemia due to ineffective erythropoiesis.


Assuntos
Anemia Diseritropoética Congênita/genética , Proteínas Morfogenéticas Ósseas/fisiologia , Sobrecarga de Ferro/etiologia , Fígado/metabolismo , Hormônios Peptídicos/genética , Transdução de Sinais/genética , Proteínas Smad/fisiologia , Adolescente , Adulto , Anemia Diseritropoética Congênita/complicações , Anemia Diseritropoética Congênita/metabolismo , Transfusão de Sangue , Proteína Morfogenética Óssea 6/farmacologia , Linhagem Celular , Criança , Eritropoese/genética , Feminino , Estudos de Associação Genética , Hepcidinas/biossíntese , Hepcidinas/sangue , Hepcidinas/genética , Humanos , Masculino , Hormônios Peptídicos/sangue , Hormônios Peptídicos/farmacologia , Hormônios Peptídicos/fisiologia , Proteínas Recombinantes/farmacologia , Índice de Gravidade de Doença , Proteínas Smad/biossíntese , Proteínas Smad/genética , Adulto Jovem
15.
J Dent Res ; 98(8): 904-911, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-31136721

RESUMO

Bone morphogenetic protein (BMP) signaling performs multiple essential functions during craniofacial development. In this study, we used the adult mouse incisor as a model to uncover how BMP signaling maintains tissue homeostasis and regulates mesenchymal stem cell (MSC) fate by mediating WNT and FGF signaling. We observed a severe defect in the proximal region of the adult mouse incisor after loss of BMP signaling in the Gli1+ cell lineage, indicating that BMP signaling is required for cell proliferation and odontoblast differentiation. Our study demonstrates that BMP signaling serves as a key regulator that antagonizes WNT and FGF signaling to regulate MSC lineage commitment. In addition, BMP signaling in the Gli1+ cell lineage is also required for the maintenance of quiescent MSCs, suggesting that BMP signaling not only is important for odontoblast differentiation but also plays a crucial role in providing feedback to the MSC population. This study highlights multiple important roles of BMP signaling in regulating tissue homeostasis.


Assuntos
Proteínas Morfogenéticas Ósseas/fisiologia , Incisivo/crescimento & desenvolvimento , Células-Tronco Mesenquimais/citologia , Transdução de Sinais , Animais , Diferenciação Celular , Fatores de Crescimento de Fibroblastos/metabolismo , Homeostase , Camundongos , Odontoblastos/citologia , Via de Sinalização Wnt
16.
Cells ; 8(5)2019 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-31096638

RESUMO

Non-alcoholic fatty liver disease (NAFLD) is considered to be the hepatic manifestation of the metabolic syndrome. The bone morphogenetic protein-8B (BMP8B) has been shown to be expressed in brown adipose tissues and the hypothalamus and to affect thermogenesis and susceptibility to diet-induced obesity. Here, we aimed to analyze BMP8B expression in NAFLD and to gain insight into BMP8B effects on pathophysiological steps of NAFLD progression. BMP8B mRNA and protein expression were dose-dependently induced in primary human hepatocytes in vitro upon incubation with fatty acids. Furthermore, hepatic BMP8B expression was significantly increased in a murine NAFLD model and in NAFLD patients compared with controls. Incubation with recombinant BMP8B further enhanced the fatty acid-induced cellular lipid accumulation as well as NFκB activation and pro-inflammatory gene expression in hepatocytes, while siRNA-mediated BMP8B depletion ameliorated these fatty acid-induced effects. Analysis of the expression of key factors of hepatocellular lipid transport and metabolisms indicated that BMP8B effects on fatty acid uptake as well as de novo lipogenesis contributed to hepatocellular accumulation of fatty acids leading to increased storage in the form of triglycerides and enhanced combustion by beta oxidation. In conclusion, our data indicate that BMP8B enhances different pathophysiological steps of NAFLD progression and suggest BMP8B as a promising prognostic marker and therapeutic target for NAFLD and, potentially, also for other chronic liver diseases.


Assuntos
Proteínas Morfogenéticas Ósseas/fisiologia , Ácidos Graxos/metabolismo , Células Estreladas do Fígado/metabolismo , Inflamação/metabolismo , Hepatopatia Gordurosa não Alcoólica/metabolismo , Animais , Biomarcadores/metabolismo , Modelos Animais de Doenças , Feminino , Células Hep G2 , Células Estreladas do Fígado/citologia , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Triglicerídeos/metabolismo
17.
J Craniofac Surg ; 30(3): 703-708, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30839467

RESUMO

Grafts and prosthetic materials used for the repair of bone defects are often accompanied by comorbidity and rejection. Therefore, there is an immense need for novel approaches to combating the issues surrounding such defects. Because of their accessibility, substantial proportion, and osteogenic differentiation potential, adipose-derived stem cells (ASCs) make for an ideal source of bone tissue in regenerative medicine. However, efficient induction of ASCs toward an osteoblastic lineage in vivo is met with challenges, and many signaling pathways must come together to secure osteoblastogenesis. Among them are bone morphogenic protein, wingless-related integration site protein, Notch, Hedgehog, fibroblast growth factor, vascular endothelial growth factor, and extracellular regulated-signal kinase. The goal of this literature review is to conglomerate the present research on these pathways to formulate a better understanding of how ASCs are most effectively transformed into bone in the context of tissue engineering.


Assuntos
Osteogênese/fisiologia , Células-Tronco/citologia , Adipócitos/metabolismo , Tecido Adiposo/citologia , Proteínas Morfogenéticas Ósseas/metabolismo , Proteínas Morfogenéticas Ósseas/fisiologia , Diferenciação Celular/fisiologia , Células Cultivadas , Regulação para Baixo , Fatores de Crescimento de Fibroblastos/fisiologia , Proteínas Hedgehog/fisiologia , Humanos , Sistema de Sinalização das MAP Quinases/fisiologia , Osteoblastos/citologia , Transdução de Sinais , Fator A de Crescimento do Endotélio Vascular/metabolismo , Via de Sinalização Wnt/fisiologia
18.
Geroscience ; 41(1): 1-11, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30729414

RESUMO

Growth differentiation factor 11 (GDF11) is a transforming growth factor ß (TGFß) protein that regulates aspects of central nervous system (CNS) formation and health throughout the lifespan. During development, GDF11 influences CNS patterning and the genesis, differentiation, maturation, and activity of new cells, which may be primarily dependent on local production and action. In the aged brain, exogenous, peripherally delivered GDF11 may enhance neurogenesis and angiogenesis, as well as improve neuropathological outcomes. This is in contrast to a predominantly negative influence on neurogenesis in the developing CNS. Seemingly antithetical effects may correspond to the cell types and mechanisms activated by local versus circulating concentrations of GDF11. Yet undefined, distinct mechanisms of action in young and aged brains may also play a role, which could include differential receptor and binding partner interactions. Exogenously increasing circulating GDF11 concentrations may be a viable approach for improving deleterious aspects of brain aging and neuropathology. Caution is warranted, however, since GDF11 appears to negatively influence muscle health and body composition. Nevertheless, an expanding understanding of GDF11 biology suggests that it is an important regulator of CNS formation and fate, and its manipulation may improve aspects of brain health in older organisms.


Assuntos
Proteínas Morfogenéticas Ósseas/fisiologia , Encéfalo/crescimento & desenvolvimento , Fatores de Diferenciação de Crescimento/fisiologia , Longevidade/fisiologia , Neurogênese , Fator de Crescimento Transformador beta/fisiologia , Animais , Composição Corporal/efeitos dos fármacos , Padronização Corporal/efeitos dos fármacos , Padronização Corporal/fisiologia , Proteínas Morfogenéticas Ósseas/genética , Proteínas Morfogenéticas Ósseas/farmacologia , Encéfalo/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Fatores de Diferenciação de Crescimento/genética , Fatores de Diferenciação de Crescimento/farmacologia , Humanos , Desenvolvimento Muscular/efeitos dos fármacos , Desenvolvimento Muscular/fisiologia , Miostatina/genética , Miostatina/metabolismo , Neovascularização Fisiológica/efeitos dos fármacos , Neovascularização Fisiológica/fisiologia , Neurogênese/efeitos dos fármacos , Neurogênese/fisiologia , Homologia de Sequência de Aminoácidos , Fator de Crescimento Transformador beta/genética , Fator de Crescimento Transformador beta/farmacologia
19.
Hua Xi Kou Qiang Yi Xue Za Zhi ; 36(4): 378-383, 2018 Aug 01.
Artigo em Chinês | MEDLINE | ID: mdl-30182564

RESUMO

OBJECTIVE: This study aimed to investigate the role and mechanism of neuropeptide substance P (SP) in ST2 cell (bone mesenchymal stem cells of mice) osteogenic differentiation to provide a basis for the treatment of temporomandibular joint osteoarthritis. METHODS: Third-generation ST2 cells were cultured with different concentrations of SP (0, 10⁻¹°, 10⁻8, 10⁻6, and 10⁻5 mol·L⁻¹). After 24, 48, and 72 h, cell proliferation was detected by CCK-8. The ST2 cells were cultured with 10⁻6 mol·L⁻¹ SP for 1, 3, 5, and 7 days. Subsequently, the expression of alkaline phosphatase (ALP), collagen typeⅠ(CollaⅠ), and osteocalcin (OCN) in the culture supernatant was tested by enzyme-linked immunosorbent assay (ELISA). ALP activity was detected by immunofluorescence staining. The ST2 cells were cultured with SP, Noggin (inhibitor of the bone morphogenetic protein signaling pathway), SP+Noggin, and 2% fetal bovine serum, respectively. Finally, the expression of ALP, CollaⅠ, and OCN in the culture supernatant was tested by ELISA. RESULTS: CCK-8 showed that the effect of cell proliferation was most obvious when the SP concentration was 10⁻6 mol·L⁻¹ (P<0.01). The ELISA results demonstrated that ALP expression significantly increased at day 5 compared with that in the control group (P<0.01), whereas the expression of CollaⅠand OCN significantly increased at day 7 (P<0.05). Immunofluorescence results showed that ALP activity was strongest at day 5. The expression of ALP, CollaⅠ, and OCN decreased after Noggin addition (P<0.05). CONCLUSIONS: SP can promote the proliferation and osteogenic differentiation of ST2 cells, and the bone morphogenetic protein signaling pathway may be involved in this process.


Assuntos
Proteínas Morfogenéticas Ósseas , Diferenciação Celular , Células-Tronco Mesenquimais , Substância P , Fosfatase Alcalina , Animais , Proteínas Morfogenéticas Ósseas/fisiologia , Células Cultivadas , Camundongos , Neuropeptídeos , Osteogênese , Substância P/fisiologia
20.
Surgery ; 164(2): 262-273, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29731246

RESUMO

BACKGROUND: GDF11 modulates embryonic patterning and kidney organogenesis. Herein, we sought to define GDF11 function in the adult kidney and in renal diseases. METHODS: In vitro renal cell lines, genetic, and murine in vivo renal injury models were examined. RESULTS: Among tissues tested, Gdf11 was highest in normal adult mouse kidney. Expression was increased acutely after 5/6 nephrectomy, ischemia-reperfusion injury, kanamycin toxicity, or unilateral ureteric obstruction. Systemic, high-dose GDF11 administration in adult mice led to renal failure, with accompanying kidney atrophy, interstitial fibrosis, epithelial-to-mesenchymal transition of renal tubular cells, and eventually death. These effects were associated with phosphorylation of SMAD2 and could be blocked by follistatin. In contrast, Gdf11 heterozygous mice showed reduced renal Gdf11 expression, renal fibrosis, and expression of fibrosis-associated genes both at baseline and after unilateral ureteric obstruction compared with wild-type littermates. The kidney-specific consequences of GDF11 dose modulation are direct effects on kidney cells. GDF11 induced proliferation and activation of NRK49f renal fibroblasts and also promoted epithelial-to-mesenchymal transition of IMCD-3 tubular epithelial cells in a SMAD3-dependent manner. CONCLUSION: Taken together, these data suggest that GDF11 and its downstream signals are critical in vivo mediators of renal injury. These effects are through direct actions of GDF11 on renal tubular cells and fibroblasts. Thus, regulation of GDF11 presents a therapeutic target for diseases involving renal fibrosis and impaired tubular function.


Assuntos
Proteínas Morfogenéticas Ósseas/fisiologia , Transição Epitelial-Mesenquimal , Fatores de Diferenciação de Crescimento/fisiologia , Nefroesclerose/etiologia , Insuficiência Renal/etiologia , Animais , Linhagem Celular , Feminino , Folistatina , Rim/patologia , Masculino , Camundongos Endogâmicos C57BL , Camundongos Nus , Insuficiência Renal/patologia , Proteína Smad2/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA