Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 225
Filtrar
1.
J Transl Med ; 22(1): 347, 2024 Apr 11.
Artigo em Inglês | MEDLINE | ID: mdl-38605354

RESUMO

BACKGROUND: THOC7-AS1 and FSTL1 expression are frequently upregulated in cutaneous squamous cell carcinoma (cSCC). However, their molecular biological mechanisms remain elusive and their potential as therapeutic targets needs urgent exploration. METHODS: Human tissue samples were used to evaluate clinical parameters. In vitro and in vivo experiments assessed biological functions. Quantitative PCR, western blot, immunohistochemistry, immunocytochemistry, immunoprecipitation, RNA fluorescence in situ hybridization, RNA pull-down, RNA immunoprecipitation, silver staining, chromatin immunoprecipitation, dual luciferase reporter assays etc. were utilized to explore the molecular biological mechanisms. RESULTS: We found FSTL1 is an oncogene in cSCC, with high expression in tumor tissues and cells. Its elevated expression closely associates with tumor size and local tissue infiltration. In vitro and in vivo, high FSTL1 expression promotes cSCC proliferation, migration and invasion, facilitating malignant behaviors. Mechanistically, FSTL1 interacts with ZEB1 to promote epithelial-to-mesenchymal transition (EMT) in cSCC cells. Exploring upstream regulation, we found THOC7-AS1 can interact with OCT1, which binds the FSTL1 promoter region and promotes FSTL1 expression, facilitating cSCC progression. Finally, treating tumors with THOC7-AS1 antisense oligonucleotides inhibited cSCC proliferative and migratory abilities, delaying tumor progression. CONCLUSIONS: The THOC7-AS1/OCT1/FSTL1 axis regulates EMT and promotes tumor progression in cSCC. This study provides clues and ideas for cSCC targeted therapy.


Assuntos
Carcinoma de Células Escamosas , Proteínas Relacionadas à Folistatina , Neoplasias Cutâneas , Humanos , Carcinoma de Células Escamosas/patologia , Linhagem Celular Tumoral , Movimento Celular/genética , Proliferação de Células/genética , Proteínas Relacionadas à Folistatina/genética , Proteínas Relacionadas à Folistatina/metabolismo , Regulação Neoplásica da Expressão Gênica , Hibridização in Situ Fluorescente , RNA , RNA Longo não Codificante/genética , Neoplasias Cutâneas/genética , Neoplasias Cutâneas/patologia
2.
Gene ; 906: 148263, 2024 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-38346455

RESUMO

Flolistatin-related protein 1 (FSTL1), a secreted glycoprotein that is involved in many physiological functions, has attracted much interest and has been implicated in a wide range of diseases, including heart diseases and inflammatory diseases. In recent years, the involvement of FSTL1 in cancer progression has been implicated and researched. FSTL1 plays a contradictory role in cancer, depending on the cancer type as well as the contents of the tumor microenvironment. As reviewed here, the structure and distribution of FSTL1 are first introduced. Subsequently, the expression and clinical significance of FSTL1 in various types of cancer as a tumor enhancer or inhibitor are addressed. Furthermore, we discuss the functional role of FSTL1 in various processes that involve tumor cell proliferation, metastasis, immune responses, stemness, cell apoptosis, and resistance to chemotherapy. FSTL1 expression is tightly controlled in cancer, and a multitude of cancer-related signaling cascades like TGF-ß/BMP/Smad signaling, AKT, NF-κB, and Wnt-ß-catenin signaling pathways are modulated by FSTL1. Finally, FSTL1 as a therapeutic target using monoclonal antibodies is stated. Herein, we review recent findings showing the double-edged characteristics and mechanisms of FSTL1 in cancer and elaborate on the current understanding of therapeutic approaches targeting FSTL1.


Assuntos
Proteínas Relacionadas à Folistatina , Neoplasias , Humanos , Proteínas Relacionadas à Folistatina/genética , Proteínas Relacionadas à Folistatina/metabolismo , Neoplasias/genética , Neoplasias/metabolismo , NF-kappa B/metabolismo , Microambiente Tumoral , Via de Sinalização Wnt , Animais
3.
Sci China Life Sci ; 67(3): 475-487, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-37219765

RESUMO

Cardiopulmonary bypass has been speculated to elicit systemic inflammation to initiate acute lung injury (ALI), including acute respiratory distress syndrome (ARDS), in patients after cardiac surgery. We previously found that post-operative patients showed an increase in endothelial cell-derived extracellular vesicles (eEVs) with components of coagulation and acute inflammatory responses. However, the mechanism underlying the onset of ALI owing to the release of eEVs after cardiopulmonary bypass, remains unclear. Plasma plasminogen-activated inhibitor-1 (PAI-1) and eEV levels were measured in patients with cardiopulmonary bypass. Endothelial cells and mice (C57BL/6, Toll-like receptor 4 knockout (TLR4-/-) and inducible nitric oxide synthase knockout (iNOS-/-)) were challenged with eEVs isolated from PAI-1-stimulated endothelial cells. Plasma PAI-1 and eEVs were remarkably enhanced after cardiopulmonary bypass. Plasma PAI-1 elevation was positively correlated with the increase in eEVs. The increase in plasma PAI-1 and eEV levels was associated with post-operative ARDS. The eEVs derived from PAI-1-stimulated endothelial cells could recognize TLR4 to stimulate a downstream signaling cascade identified as the Janus kinase 2/3 (JAK2/3)-signal transducer and activator of transcription 3 (STAT3)-interferon regulatory factor 1 (IRF-1) pathway, along with iNOS induction, and cytokine/chemokine production in vascular endothelial cells and C57BL/6 mice, ultimately contributing to ALI. ALI could be attenuated by JAK2/3 or STAT3 inhibitors (AG490 or S3I-201, respectively), and was relieved in TLR4-/- and iNOS-/- mice. eEVs activate the TLR4/JAK3/STAT3/IRF-1 signaling pathway to induce ALI/ARDS by delivering follistatin-like protein 1 (FSTL1), and FSTL1 knockdown in eEVs alleviates eEV-induced ALI/ARDS. Our data thus demonstrate that cardiopulmonary bypass may increase plasma PAI-1 levels to induce FSTL1-enriched eEVs, which target the TLR4-mediated JAK2/3/STAT3/IRF-1 signaling cascade and form a positive feedback loop, leading to ALI/ARDS after cardiac surgery. Our findings provide new insight into the molecular mechanisms and therapeutic targets for ALI/ARDS after cardiac surgery.


Assuntos
Lesão Pulmonar Aguda , Vesículas Extracelulares , Proteínas Relacionadas à Folistatina , Síndrome do Desconforto Respiratório , Animais , Humanos , Camundongos , Lesão Pulmonar Aguda/etiologia , Lesão Pulmonar Aguda/tratamento farmacológico , Lesão Pulmonar Aguda/metabolismo , Células Endoteliais/metabolismo , Vesículas Extracelulares/metabolismo , Proteínas Relacionadas à Folistatina/metabolismo , Proteínas Relacionadas à Folistatina/uso terapêutico , Inflamação/metabolismo , Lipopolissacarídeos/farmacologia , Pulmão/metabolismo , Camundongos Endogâmicos C57BL , Inibidor 1 de Ativador de Plasminogênio/metabolismo , Inibidor 1 de Ativador de Plasminogênio/uso terapêutico , Síndrome do Desconforto Respiratório/etiologia , Receptor 4 Toll-Like/metabolismo , Receptor 4 Toll-Like/uso terapêutico
4.
Obesity (Silver Spring) ; 32(2): 352-362, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38018497

RESUMO

OBJECTIVE: The aim of this study was to investigate the role of the follistatin-like 1 (Fstl1) and disco-interacting protein 2 homolog A (DIP2a) axis in relation to lipid metabolism during and after endurance exercise and to elucidate the mechanisms underlying the metabolic effects of Fstl1 on adipocytes, considering its regulation by exercise and muscle mass and its link to obesity. METHODS: Twenty-nine sedentary males participated in endurance exercise, and blood samples were collected during and after the exercise. Body composition, Fstl1, glycerol, epinephrine, growth hormone, and atrial natriuretic peptide were measured. 3T3-L1 adipocytes, with or without DIP2a knockdown, were treated with Fstl1 to assess glycerol release, cyclic AMP/cyclic GMP production, and hormone sensitive lipase phosphorylation. The association between DIP2a gene expression levels in human adipose tissues and exercise-induced lipolysis was examined. RESULTS: Fstl1 levels significantly increased during endurance exercise and following recovery, correlating with lean body mass and lipolysis. In 3T3-L1 adipocytes, Fstl1 increased glycerol release, cyclic GMP production, and hormone sensitive lipase activation, but these effects were attenuated by DIP2a knockdown. DIP2a gene expression in human adipose tissues correlated with serum glycerol concentrations during endurance exercise. CONCLUSIONS: Fstl1 is a myokine facilitating lipid mobilization during and after endurance exercise through DIP2a-mediated lipolytic effects in adipocytes.


Assuntos
Proteínas Relacionadas à Folistatina , Folistatina , Humanos , Masculino , GMP Cíclico/metabolismo , Folistatina/metabolismo , Proteínas Relacionadas à Folistatina/genética , Proteínas Relacionadas à Folistatina/metabolismo , Glicerol/metabolismo , Mobilização Lipídica , Lipólise/fisiologia , Miocinas , Esterol Esterase/metabolismo
5.
Clin Exp Hypertens ; 45(1): 2277654, 2023 Dec 31.
Artigo em Inglês | MEDLINE | ID: mdl-37963199

RESUMO

OBJECTIVE: Endothelial dysfunction is a critical initiating factor in the development of hypertension and related complications. Follistatin-like 1 (FSTL1) can promote endothelial cell function and stimulates revascularization in response to ischemic insult. However, it is unclear whether FSTL1 has an effect on ameliorating endothelial dysfunction in spontaneously hypertensive rats (SHRs). METHODS: Wistar Kyoto (WKY) and SHRs were treated with a tail vein injection of vehicle (1 mL/day) or recombinant FSTL1 (100 µg/kg body weight/day) for 4 weeks. Blood pressure was measured by tail-cuff plethysmograph, and vascular reactivity in mesenteric arteries was measured using wire myography. RESULTS: We found that treatment with FSTL1 reversed impaired endothelium-dependent relaxation (EDR) in mesenteric arteries and lowered blood pressure of SHRs. Decreased AMP-activated protein kinase (AMPK) phosphorylation, elevated endoplasmic reticulum (ER) stress markers, increased reactive oxygen species (ROS), and reduction of nitric oxide (NO) production in mesenteric arteries of SHRs were also reversed by FSTL1 treatment. Ex vivo treatment with FSTL1 improved the impaired EDR in mesenteric arteries from SHRs and reversed tunicamycin (ER stress inducer)-induced ER stress and the impairment of EDR in mesenteric arteries from WKY rats. The effects of FSTL1 were abolished by cotreatment of compound C (AMPK inhibitor). CONCLUSIONS: These results suggest that FSTL1 prevents endothelial dysfunction in mesenteric arteries of SHRs through inhibiting ER stress and ROS and increasing NO production via activation of AMPK signaling.


Assuntos
Proteínas Relacionadas à Folistatina , Hipertensão , Ratos , Animais , Ratos Endogâmicos SHR , Proteínas Quinases Ativadas por AMP/metabolismo , Folistatina/metabolismo , Folistatina/farmacologia , Ratos Endogâmicos WKY , Espécies Reativas de Oxigênio/metabolismo , Proteínas Relacionadas à Folistatina/metabolismo , Proteínas Relacionadas à Folistatina/farmacologia , Endotélio Vascular , Artérias Mesentéricas , Estresse do Retículo Endoplasmático
6.
Oncol Rep ; 50(6)2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37888756

RESUMO

The abnormal expression of follistatin­like protein 1 (FSTL1) in various tumors is a crucial regulator of the biological process of tumorigenesis. Nonetheless, the regulatory role of FSTL1 in cervical cancer is yet to be elucidated. Hence, the present study aimed to explore the expression, function, and molecular mechanism of FSTL1 in cervical cancer. The expression of FSTL1 in normal and cervical cancer tissues was examined using quantitative reverse transcription­polymerase chain reaction and immunohistochemistry assays. The effects of abnormal expression of FSTL1 on cervical cancer cells were assessed using colony formation, MTT, wound­healing, Transwell, apoptosis, and nude mouse tumorigenicity assays. FSTL1­related molecular mechanisms were screened using gene chip analysis. Western blotting analysis was used to verify the regulatory mechanisms of FSTL1 in cervical cancer. The results indicated that the expression of FSTL1 was downregulated in cervical cancer tissues and that its downregulation was associated with tumor differentiation, pathologic type, and infiltration depth. Moreover, FSTL1 inhibited the proliferation, migration, and invasion of cervical cancer cells as well as xenograft tumor growth and promoted cell apoptosis. In addition, the findings of gene chip analysis suggested that the differentially expressed genes of FSTL1 were predominantly enriched in multiple signaling pathways, of which the insulin­like growth factor (IGF)­1 signaling pathway was significantly activated. Western blotting suggested the involvement of FSTL1 in the regulation of the IGF­1R/PI3K/AKT/BCL­2 signaling pathway. These data establish the downregulation of FSTL1 in cervical cancer tissues. FSTL1 inhibited the proliferation, migration, and invasion of cervical cancer cells and promoted their apoptosis. Furthermore, xenograft tumor growth in nude mice was inhibited. FSTL1 may be involved in the regulation of the IGF­1R/PI3K/AKT/BCL­2 signaling pathway in cervical cancer. Therefore, FSTL1 may be employed as a novel biomarker to determine the extent of disease progression in patients with cervical cancer.


Assuntos
Proteínas Relacionadas à Folistatina , Neoplasias do Colo do Útero , Animais , Feminino , Camundongos , Humanos , Proteínas Proto-Oncogênicas c-akt/metabolismo , Neoplasias do Colo do Útero/patologia , Proteínas Relacionadas à Folistatina/genética , Proteínas Relacionadas à Folistatina/metabolismo , Proliferação de Células/genética , Camundongos Nus , Fosfatidilinositol 3-Quinases/genética , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Linhagem Celular Tumoral , Regulação Neoplásica da Expressão Gênica , Movimento Celular/genética
7.
Redox Biol ; 67: 102923, 2023 11.
Artigo em Inglês | MEDLINE | ID: mdl-37832398

RESUMO

As the predominant immunosuppressive component within the tumor microenvironment (TME), cancer-associated fibroblasts (CAFs) inhibit Natural Killer cell (NK cell) activity to promote tumor progression and immune escape; however, the mechanisms of cross-talk between CAFs and NK cells in gastric cancer (GC) remain poorly understood. In this study, we demonstrate that NK cell levels are inversely correlated with CAFs abundance in human GC. CAFs impair the anti-tumor capacity of NK cells by inducing ferroptosis, a cell death process characterized by the accumulation of iron-dependent lipid peroxides. CAFs induce ferroptosis in NK cells by promoting iron overload; conversely, decreased intracellular iron levels protect NK cells against CAF-induced ferroptosis. Mechanistically, CAFs increase the labile iron pool within NK cells via iron export into the TME, which is mediated by the upregulated expression of iron regulatory genes ferroportin1 and hephaestin in CAFs. Moreover, CAF-derived follistatin like protein 1(FSTL1) upregulates NCOA4 expression in NK cells via the DIP2A-P38 pathway, and NCOA4-mediated ferritinophagy is required for CAF-induced NK cell ferroptosis. In a human patient-derived organoid model, functional targeting of CAFs using a combination of deferoxamine and FSTL1-neutralizing antibody significantly alleviate CAF-induced NK cell ferroptosis and boost the cytotoxicity of NK cells against GC. This study demonstrates a novel mechanism of suppression of NK cell activity by CAFs in the TME and presents a potential therapeutic approach to augment the immune response against GC mediated by NK cells.


Assuntos
Antineoplásicos , Fibroblastos Associados a Câncer , Ferroptose , Proteínas Relacionadas à Folistatina , Neoplasias Gástricas , Humanos , Fibroblastos Associados a Câncer/metabolismo , Fibroblastos Associados a Câncer/patologia , Proteínas Relacionadas à Folistatina/metabolismo , Neoplasias Gástricas/metabolismo , Ferro/metabolismo , Células Matadoras Naturais/metabolismo , Células Matadoras Naturais/patologia , Antineoplásicos/farmacologia , Microambiente Tumoral
8.
Biomed Pharmacother ; 168: 115771, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37897975

RESUMO

Disco Interacting Protein 2 Homolog A (DIP2A) is expressed throughout the body and abundantly expressed in the brain tissue. It is activated by Follistatin-like 1 (FSTL1). Activated DIP2A interacts with several pathways, such as AMPK/mTOR and AKT pathways, to contribute to many biological processes, such as oxidative stress, transcriptional regulation, and apoptosis. Dysregulated DIP2A activation has been implicated in numerous processes in the brain. If the upstream pathways of DIP2A remain globally unexplored, many proteins, including cortactin, AMPK, and AKT, have been identified as its downstream targets in the literature. Recent studies have linked DIP2A to a variety of mechanisms in many types of brain disorders, suggesting that regulation of DIP2A could provide novel diagnostic and therapeutic approaches for brain disorders. In this review, we comprehensively summarized and discussed the current research on DIP2A in various brain disorders, such as stroke, autism spectrum disorders (ASD), Alzheimer's disease (AD), dyslexia, and glioma.


Assuntos
Encefalopatias , Proteínas Relacionadas à Folistatina , Humanos , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Quinases Ativadas por AMP/metabolismo , Proteínas Nucleares/genética , Regulação da Expressão Gênica , Proteínas Relacionadas à Folistatina/metabolismo
9.
Nat Commun ; 14(1): 6047, 2023 09 28.
Artigo em Inglês | MEDLINE | ID: mdl-37770480

RESUMO

Inter-organ crosstalk has gained increasing attention in recent times; however, the underlying mechanisms remain unclear. In this study, we elucidate an endocrine pathway that is regulated by skeletal muscle interferon regulatory factor (IRF) 4, which manipulates liver pathology. Skeletal muscle specific IRF4 knockout (F4MKO) mice exhibited ameliorated hepatic steatosis, inflammation, and fibrosis, without changes in body weight, when put on a nonalcoholic steatohepatitis (NASH) diet. Proteomics analysis results suggested that follistatin-like protein 1 (FSTL1) may constitute a link between muscles and the liver. Dual luciferase assays showed that IRF4 can transcriptionally regulate FSTL1. Further, inducing FSTL1 expression in the muscles of F4MKO mice is sufficient to restore liver pathology. In addition, co-culture experiments confirmed that FSTL1 plays a distinct role in various liver cell types via different receptors. Finally, we observed that the serum FSTL1 level is positively correlated with NASH progression in humans. These data indicate a signaling pathway involving IRF4-FSTL1-DIP2A/CD14, that links skeletal muscle cells to the liver in the pathogenesis of NASH.


Assuntos
Proteínas Relacionadas à Folistatina , Hepatopatia Gordurosa não Alcoólica , Camundongos , Humanos , Animais , Hepatopatia Gordurosa não Alcoólica/metabolismo , Proteínas Relacionadas à Folistatina/genética , Proteínas Relacionadas à Folistatina/metabolismo , Fígado/metabolismo , Transdução de Sinais/fisiologia , Músculo Esquelético/metabolismo , Cirrose Hepática/patologia , Camundongos Endogâmicos C57BL
10.
Genomics ; 115(5): 110677, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37406975

RESUMO

The matricellular protein, follistatin-like 1 (FSTL1), regulates lung development and saccular formation. Here, we employed single-cell RNA sequencing (scRNA-seq) to construct a transcriptomic atlas of 22,774 individual cells from wild-type (WT) and Fstl1-/- lung (E18.5) samples and identified 27 cell subtypes. We observed abnormal population sizes and gene expression profiles in diverse cell subtypes in Fstl1-/- lung samples. We identified Pdgfra and Tgfbi as genetic markers specifically expressed in postnatal myofibroblasts (MyoFBs). Fstl1 deletion decreased the number of MyoFB cells and downregulated their roles in ECM organization and muscle tissue/vasculature development, partly through the TGF-ß1/BMP4 signaling pathway. Our data provide a single-cell view of the cellular heterogeneity and the molecular mechanisms underlying abnormal saccular formation and atelectatic lungs in Fstl1-/- mice.


Assuntos
Proteínas Relacionadas à Folistatina , Pulmão , Miofibroblastos , Animais , Camundongos , Proteínas Relacionadas à Folistatina/genética , Proteínas Relacionadas à Folistatina/metabolismo , Pulmão/metabolismo , Miofibroblastos/metabolismo , Transdução de Sinais , Análise da Expressão Gênica de Célula Única
11.
FASEB J ; 37(8): e23064, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37440271

RESUMO

Off target damage to vital organ systems is an unfortunate side effect of cancer chemotherapy and remains a major limitation to the use of these essential drugs in the clinic. Despite decades of research, the mechanisms conferring susceptibility to chemotherapy driven cardiotoxicity and hepatotoxicity remain unclear. In the livers of patients with a history of chemotherapy, we observed a twofold increase in expression of G protein regulator RGS7 and a corresponding decrease in fellow R7 family member RGS11. Knockdown of RGS7 via introduction of RGS7 shRNA via tail vein injection decreased doxorubicin-induced hepatic collagen and lipid deposition, glycogen accumulation, and elevations in ALT, AST, and triglycerides by approximately 50%. Surprisingly, a similar result could be achieved via introduction of RGS7 shRNA directly to the myocardium without impacting RGS7 levels in the liver directly. Indeed, doxorubicin-treated cardiomyocytes secrete the endocrine factors transforming growth factor ß1 (TGFß1) and TGFß superfamily binding protein follistatin-related protein 1 (FSTL1). Importantly, RGS7 overexpression in the heart was sufficient to recapitulate the impacts of doxorubicin on the liver and inhibition of TGFß1 signaling with the receptor blocker GW788388 ameliorated the effect of cardiac RGS7 overexpression on hepatic fibrosis, steatosis, oxidative stress, and cell death as well as the resultant elevation in liver enzymes. Together these data demonstrate that RGS7 controls both the release of TGFß1 from the heart and the profibrotic and pro-oxidant actions of TGFß1 in the liver and emphasize the functional significance of endocrine cardiokine signaling in the pathogenesis of chemotherapy drive multiorgan damage.


Assuntos
Proteínas Relacionadas à Folistatina , Proteínas RGS , Humanos , Proteínas RGS/genética , Proteínas RGS/metabolismo , Fator de Crescimento Transformador beta1/genética , Fator de Crescimento Transformador beta1/metabolismo , Transdução de Sinais/fisiologia , Proteínas de Transporte/metabolismo , Fígado/metabolismo , Doxorrubicina/efeitos adversos , Proteínas Relacionadas à Folistatina/metabolismo
12.
Wilderness Environ Med ; 34(3): 341-345, 2023 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-37301628

RESUMO

INTRODUCTION: We have previously described negative energy balance (ie, -9.7±3.4 MJ/d) and weight loss (Δ-1.5 ± 0.7 kg) influenced by high levels of energy expenditure (ie, 17.4±2.6 MJ/d) during remote expeditionary hunting in Alaska. Despite negative energy balance, participants retained skeletal muscle. The purpose of this pilot study was to measure skeletal muscle protein synthesis and examine molecular markers of skeletal muscle protein metabolism under similar conditions of physical and nutrient stress. METHODS: The "virtual biopsy method" was used to evaluate integrated fractional synthetic rates (FSRs) of muscle protein from blood samples in 4 participants. Muscle biopsies were taken to measure molecular markers of muscle protein kinetics (ie, FSTL1, MEF2, MYOD1, B2M, and miR-1-3p, -206, -208b, 23a, and 499a) using real-time polymerase chain reaction. RESULTS: Our findings in 4 participants (2 females [28 and 62 y of age; 66.2 and 71.8 kg body weight; 25.5 and 26.7 kg/m2 body mass index] and 2 males [47 and 56 y of age; 87.5 and 91.4 kg body weight; 26.1 and 28.3 kg/m2 body mass index]) describe mean muscle FSRs of serum carbonic anhydrase (2.4%) and creatine kinase M-type (4.0%) and positive increments in molecular regulation. CONCLUSIONS: Preservation of skeletal muscle under conditions of physical and nutrient stress seems to be supported by positive inflection of skeletal muscle FSR and molecular activation.


Assuntos
Proteínas Relacionadas à Folistatina , Proteínas Musculares , Masculino , Feminino , Humanos , Proteínas Musculares/metabolismo , Alaska , Caça , Projetos Piloto , Músculo Esquelético , Peso Corporal , Metabolismo Energético , Proteínas Relacionadas à Folistatina/metabolismo
13.
Cell Biol Int ; 47(10): 1716-1727, 2023 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-37369969

RESUMO

Alveolar epithelial cell (AEC) senescence-induced changes of lung mesenchymal cells are key to starting the progress of pulmonary fibrosis. Follistatin-like 1 (FSTL1) plays a central regulatory role in the complex process of senescence and pulmonary fibrosis by enhancing transforming growth factor-ß1 (TGF-ß1) signal pathway activity. Activation of Smad4 and Ras relies on SUMO-specific peptidase 1 (SENP1)-mediated deSUMOylation during TGF-ß signaling pathway activation. We hypothesized that SENP1-mediated deSUMOylation may be a potential therapeutic target by modulating FSTL1-regulated cellular senescence in pulmonary fibrosis. In verifying this hypothesis, we found that FSTL1 expression was upregulated in the lung tissues of patients with idiopathic pulmonary fibrosis and that SENP1 was overexpressed in senescent AECs. TGF-ß1-induced FSTL1 not only promoted AEC senescence but also upregulated SENP1 expression. Interfering with SENP1 expression inhibited FSTL1-dependent promotion of AEC senescence and improved pulmonary fibrosis in mouse lungs. FSTL1 enhancement of TGF-ß1 signaling pathway activation was dependent on SENP1 in senescent AEC. Our work identifies a novel mechanism by which FSTL1 is involved in AEC senescence. Inhibition of SENP1 in epithelial cells alleviated pulmonary fibrosis by blocking FSTL1-enhanced TGF signaling.


Assuntos
Proteínas Relacionadas à Folistatina , Fibrose Pulmonar Idiopática , Animais , Camundongos , Envelhecimento , Células Epiteliais Alveolares , Proteínas Relacionadas à Folistatina/metabolismo , Fibrose Pulmonar Idiopática/metabolismo , Peptídeo Hidrolases/metabolismo , Fator de Crescimento Transformador beta1/metabolismo
14.
Int J Obes (Lond) ; 47(6): 520-527, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-36997723

RESUMO

BACKGROUND/OBJECTIVE: Compelling evidence indicates that myokines act in an autocrine, paracrine and endocrine manner to alter metabolic homeostasis. The mechanisms underlying exercise-induced changes in myokine secretion remain to be elucidated. Since exercise acutely decreases oxygen partial pressure (pO2) in skeletal muscle (SM), the present study was designed to test the hypothesis that (1) hypoxia exposure impacts myokine secretion in primary human myotubes and (2) exposure to mild hypoxia in vivo alters fasting and postprandial plasma myokine concentrations in humans. METHODS: Differentiated primary human myotubes were exposed to different physiological pO2 levels for 24 h, and cell culture medium was harvested to determine myokine secretion. Furthermore, we performed a randomized single-blind crossover trial to investigate the impact of mild intermittent hypoxia exposure (MIH: 7-day exposure to 15% O2, 3x2h/day vs. normoxia: 21% O2) on in vivo SM pO2 and plasma myokine concentrations in 12 individuals with overweight and obesity (body-mass index ≥ 28 kg/m2). RESULTS: Hypoxia exposure (1% O2) increased secreted protein acidic and rich in cysteine (SPARC, p = 0.043) and follistatin like 1 (FSTL1, p = 0.021), and reduced leukemia inhibitory factor (LIF) secretion (p = 0.009) compared to 3% O2 in primary human myotubes. In addition, 1% O2 exposure increased interleukin-6 (IL-6, p = 0.004) and SPARC secretion (p = 0.021), whilst reducing fatty acid binding protein 3 (FABP3) secretion (p = 0.021) compared to 21% O2. MIH exposure in vivo markedly decreased SM pO2 (≈40%, p = 0.002) but did not alter plasma myokine concentrations. CONCLUSIONS: Hypoxia exposure altered the secretion of several myokines in primary human myotubes, revealing hypoxia as a novel modulator of myokine secretion. However, both acute and 7-day MIH exposure did not induce alterations in plasma myokine concentrations in individuals with overweight and obesity. CLINICAL TRIALS IDENTIFIER: This study is registered at the Netherlands Trial Register (NL7120/NTR7325).


Assuntos
Proteínas Relacionadas à Folistatina , Osteonectina , Humanos , Osteonectina/metabolismo , Sobrepeso/metabolismo , Método Simples-Cego , Músculo Esquelético/metabolismo , Interleucina-6/metabolismo , Obesidade/metabolismo , Hipóxia/metabolismo , Proteínas Relacionadas à Folistatina/metabolismo
15.
J Cell Mol Med ; 27(5): 672-686, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36807490

RESUMO

Follistatin-like (FSTL) family members are associated with cancer progression. However, differences between FSTL members with identical cancer types have not been systematically investigated. Among the most malignant tumours worldwide, colorectal cancer (CRC) has high metastatic potential and chemoresistance, which makes it challenging to treat. A systematic examination of the relationship between the expression of FSTL family members in CRC will provide valuable information for prognosis and therapeutic development. Based on large cohort survival analyses, we determined that FSTL3 was associated with a significantly worse prognosis in CRC at the RNA and protein levels. Immunohistochemistry staining of CRC specimens revealed that FSTL3 expression levels in the cytosol were significantly associated with a poor prognosis in terms of overall and disease-free survival. Molecular simulation analysis showed that FSTL3 participated in multiple cell motility signalling pathways via the TGF-ß1/TWIST1 axis to control CRC metastasis. The findings provide evidence of the significance of FSTL3 in the oncogenesis and metastasis of CRC. FSTL3 may be useful as a diagnostic or prognostic biomarker, and as a potential therapeutic target.


Assuntos
Neoplasias Colorretais , Proteínas Relacionadas à Folistatina , Humanos , Citosol/metabolismo , Transformação Celular Neoplásica , Transdução de Sinais , Neoplasias Colorretais/genética , Neoplasias Colorretais/patologia , Estudos de Coortes , Biomarcadores Tumorais/genética , Proteínas Relacionadas à Folistatina/genética , Proteínas Relacionadas à Folistatina/metabolismo
16.
Obesity (Silver Spring) ; 31(1): 171-183, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36502285

RESUMO

OBJECTIVE: This study aimed to investigate the expression of follistatin-like 3 (FSTL3) in adipose tissue in individuals with overweight or obesity and to explore the role of FSTL3 in human adipocytes, as well as the relationship between serum FSTL3 levels and fat distribution and inflammation. METHODS: This study enrolled 236 individuals (171 with overweight or obesity; aged 18-67 years). Bulk transcriptome sequencing was performed on subcutaneous and visceral adipose tissue. The function of FSTL3 was studied in human adipocytes. Serum FSTL3 levels were measured using enzyme-linked immunosorbent assay. RESULTS: Adipose FTSL3 expression was higher in individuals with overweight or obesity than in individuals with normal weight. FSTL3 was mainly expressed in mature adipocytes and stimulated by tumor necrosis factor alpha (TNFα). FSTL3 suppressed inflammatory responses in human adipocytes, whereas FSTL3 knockdown promoted inflammatory responses. Serum FSTL3 levels were correlated with adipose FTSL3 expression and obesity-related indicators (all p < 0.05). Multiple linear regression analysis showed that serum FSTL3 levels were independently associated with the visceral fat area and serum TNFα levels (both p < 0.05). CONCLUSIONS: FSTL3 was highly expressed in adipose tissue in individuals with overweight or obesity and could suppress adipocyte inflammation. Serum FSTL3 levels might be considered as a biomarker of visceral obesity and inflammation.


Assuntos
Proteínas Relacionadas à Folistatina , Obesidade , Sobrepeso , Humanos , Tecido Adiposo/metabolismo , Proteínas Relacionadas à Folistatina/genética , Proteínas Relacionadas à Folistatina/metabolismo , Inflamação/metabolismo , Obesidade/genética , Obesidade/metabolismo , Sobrepeso/genética , Sobrepeso/metabolismo , Fator de Necrose Tumoral alfa/metabolismo
17.
Exp Biol Med (Maywood) ; 248(3): 193-200, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-36533576

RESUMO

Liver diseases, including viral hepatitis, fatty liver, metabolic-associated fatty liver disease, liver cirrhosis, alcoholic liver disease, and liver neoplasms, are major global health challenges. Despite the continued development of new drugs and technologies, the prognosis of end-stage liver diseases, including advanced liver cirrhosis and liver neoplasms, remains poor. Follistatin-like protein 1 (FSTL1), an extracellular glycoprotein, is secreted by various cell types. It is a glycoprotein that belongs to the family of secreted proteins acidic and rich in cysteine (SPARC). It is also known as transforming growth factor-beta inducible TSC-36 and follistatin-related protein (FRP). FSTL1 plays a key role in cell survival, proliferation, differentiation, and migration, as well as the regulation of inflammation and immunity. Studies have demonstrated that FSTL1 significantly affects the occurrence and development of liver diseases. This article reviews the role and mechanism of FSLT1 in liver diseases.


Assuntos
Proteínas Relacionadas à Folistatina , Cirrose Hepática , Neoplasias Hepáticas , Humanos , Diferenciação Celular , Proteínas Relacionadas à Folistatina/metabolismo , Cirrose Hepática/metabolismo , Cirrose Hepática/patologia , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patologia , Transdução de Sinais , Fator de Crescimento Transformador beta/metabolismo
18.
Eur J Sport Sci ; 23(4): 625-636, 2023 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-35152857

RESUMO

ABSTRACTIn rodents, exercise alters the plasma concentration of exerkines that regulate white adipose tissue (WAT) browning or brown adipose tissue (BAT) metabolism. This study aims to analyse the acute and chronic effect of exercise on the circulating concentrations of 16 of these exerkines in humans. Ten young sedentary adults (6 female) performed a maximum walking effort test and a resistance exercise session. The plasma concentration of 16 exerkines was assessed before, and 3, 30, 60, and 120 min after exercise. Those exerkines modified by exercise were additionally measured in another 28 subjects (22 women). We also measured the plasma concentrations of the exerkines before and after a 24-week exercise programme (endurance + resistance; 3-groups: control, moderate-intensity and vigorous-intensity) in 110 subjects (75 women). Endurance exercise acutely increased the plasma concentration of lactate, norepinephrine, brain-derived neurotrophic factor, interleukin 6, and follistatin-like protein 1 (3 min after exercise), and musclin and fibroblast growth factor 21 (30 and 60 min after exercise), decreasing the plasma concentration of leptin (30 min after exercise). Adiponectin, atrial natriuretic peptide (ANP), ß-aminoisobutyric acid, meteorin-like, follistatin, pro-ANP, irisin and myostatin were not modified or not detectable. The resistance exercise session increased the plasma concentration of lactate 3 min after exercise. Chronic exercise did not alter the plasma concentration of these exerkines. In sedentary young adults, acute endurance exercise releases to the bloodstream exerkines that regulate BAT metabolism and WAT browning. In contrast, neither a low-volume resistance exercise session nor a 24-week training programme modified plasma levels of these molecules.HighlightsAcute endurance exercise increases the plasma concentration of lactate, norepinephrine, brain-derived neurotrophic factor, interleukin 6, follistatin-like protein 1, musclin, and fibroblast growth factor 21, and decrease the plasma concentration of leptin.The exercise-induced change in lactate plasma concentration is positively associated with brown adipose tissue volume, glucose uptake and radiodensity.Neither acute resistance exercise nor chronic exercise significantly alter the plasma concentration of these exerkines.Trial registration: ClinicalTrials.gov identifier: NCT02365129.


Assuntos
Proteínas Relacionadas à Folistatina , Leptina , Adulto Jovem , Humanos , Feminino , Fator Neurotrófico Derivado do Encéfalo , Tecido Adiposo Marrom/metabolismo , Interleucina-6 , Proteínas Relacionadas à Folistatina/metabolismo , Lactatos/metabolismo
19.
J Biosci ; 472022.
Artigo em Inglês | MEDLINE | ID: mdl-36210730

RESUMO

Tuberculosis (TB) is a common disease caused by Mycobacterium tuberculosis (M.tb) infection. Our study was to explore the function and mechanism of circular RNA WD repeat domain 27 (circ-WDR27) in TB progression. Cell viability and apoptosis were detected by 3-(4, 5-dimethylthiazol-2-y1)-2, 5-diphenyl tetrazolium bromide assay and flow cytometry. Protein quantification was performed by Western blot. Inflammatory cytokines were examined using enzyme-linked immunosorbent assay. RNA levels were assayed via quantitative reverse-transcription polymerase chain reaction. M.tb survival was assessed using colony-forming unit assay. Target binding was analyzed via dual-luciferase reporter assay and RNA immunoprecipitation assay. Cell damages were induced by M.tb infection, and inflammatory cytokines were secreted in human macrophages. Circ-WDR27 was downregulated in TB patients and M.tb-infected macrophages. Circ-WDR27 overexpression reduced M.tb survival and released inflammatory cytokines in macrophages. Circ-WDR27 acted as a sponge for miR-370-3p. Circ-WDR27-mediated inhibition of TB progression was partly achieved by sponging miR-370-3p. miR-370-3p directly targeted Follistatin-like protein 1 (FSTL1). FSTL1 suppressed M.tb-induced cell damages, and reversed the protective role of miR-370-3p inhibition in TB progression. Circ- WDR27 regulated FSTL1 expression by targeting miR-370-3p. These results showed that circ-WDR27 repressed M.tb vitality and stimulated pro-inflammatory cytokines in M.tb-infected macrophages by affecting the miR-370-3p/FSTL1 axis.


Assuntos
Proteínas Relacionadas à Folistatina , MicroRNAs , Mycobacterium tuberculosis , Tuberculose , Apoptose/genética , Brometos/metabolismo , Proliferação de Células , Citocinas/genética , Citocinas/metabolismo , Proteínas Relacionadas à Folistatina/genética , Proteínas Relacionadas à Folistatina/metabolismo , Humanos , Macrófagos/metabolismo , MicroRNAs/genética , MicroRNAs/metabolismo , Mycobacterium tuberculosis/metabolismo , RNA Circular/genética , Tuberculose/genética , Tuberculose/metabolismo
20.
Int J Mol Sci ; 23(16)2022 Aug 14.
Artigo em Inglês | MEDLINE | ID: mdl-36012380

RESUMO

Arteriovenous malformation (AVM) is characterized by high-flow blood vessels connecting arteries and veins without capillaries. This disease shows increased angiogenesis and a pathophysiological hypoxic environment in proximal tissues. Here, we analyzed the effects of hypoxia on angiogenesis in the endothelial cells (ECs) of AVM and normal tissues. ECs from human normal and AVM tissues were evaluated using immunocytochemistry with CD31. In vitro tube formation under hypoxia was tested in both ECs using Matrigel. The relative expression of angiogenesis-related genes was measured using real-time PCR. Under normoxia, CD31 was significantly higher in AVM ECs (79.23 ± 0.65%) than in normal ECs (74.15 ± 0.70%). Similar results were observed under hypoxia in AVM ECs (63.85 ± 1.84%) and normal ECs (60.52 ± 0.51%). In the tube formation test under normoxic and hypoxic conditions, the junction count and total vessel length were significantly greater in AVM ECs than normal ECs. Under both normoxia and hypoxia, the angiogenesis-related gene FSTL1 showed a significantly higher expression in AVM ECs than in normal ECs. Under hypoxia, CSPG4 expression was significantly lower in AVM ECs than in normal ECs. Accordingly, the angiogenic effect was increased in AVM ECs compared with that in normal ECs. These results provide a basic knowledge for an AVM treatment strategy.


Assuntos
Proteínas Relacionadas à Folistatina , Malformações Arteriovenosas Intracranianas , Indutores da Angiogênese/metabolismo , Células Endoteliais/metabolismo , Proteínas Relacionadas à Folistatina/metabolismo , Humanos , Hipóxia/genética , Hipóxia/metabolismo , Malformações Arteriovenosas Intracranianas/genética , Malformações Arteriovenosas Intracranianas/metabolismo , Neovascularização Patológica/genética , Neovascularização Patológica/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA