Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
Cell Mol Neurobiol ; 43(1): 251-264, 2023 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-34853925

RESUMEN

Hypoxia-ischemia (HI) during crucial periods of brain formation can lead to changes in brain morphology, propagation of neuronal stimuli, and permanent neurodevelopmental impairment, which can have profound effects on cognitive function later in life. FAM3A, a subgroup of family with sequence similarity 3 (FAM3) gene family, is ubiquitously expressed in almost all cells. Overexpression of FAM3A has been evidenced to reduce hyperglycemia via the PI3K/Akt signaling pathway and protect mitochondrial function in neuronal HT22 cells. This study aims to evaluate the protective role of FAM3A in HI-induced brain impairment. Experimentally, maternal rats underwent uterine artery bilateral ligation to induce neonatal HI on day 14 of gestation. At 6 weeks of age, cognitive development assessments including NSS, wire grip, and water maze were carried out. The animals were then sacrificed to assess cerebral mitochondrial function as well as levels of FAM3A, TNF-α and IFN-γ. Results suggest that HI significantly reduced FAM3A expression in rat brain tissues, and that overexpression of FAM3A through lentiviral transduction effectively improved cognitive and motor functions in HI rats as reflected by improved NSS evaluation, cerebral water content, limb strength, as well as spatial learning and memory. At the molecular level, overexpression of FAM3A was able to promote ATP production, balance mitochondrial membrane potential, and reduce levels of pro-inflammatory cytokines TNF-α and IFN-γ. We conclude that FAM3A overexpression may have a protective effect on neuron morphology, cerebral mitochondrial as well as cognitive function. Created with Biorender.com.


Asunto(s)
Hipoxia-Isquemia Encefálica , Proteínas Proto-Oncogénicas c-akt , Animales , Ratas , Animales Recién Nacidos , Encéfalo/metabolismo , Isquemia , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
2.
FASEB J ; 34(3): 3915-3931, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31944392

RESUMEN

So far, the mechanism that links mitochondrial dysfunction to PDX1 inhibition in the pathogenesis of pancreatic ß cell dysfunction under diabetic condition remains largely unclear. This study determined the role of mitochondrial protein FAM3A in regulating PDX1 expression in pancreatic ß cells using gain- and loss-of function methods in vitro and in vivo. Within pancreas, FAM3A is highly expressed in ß, α, δ, and pp cells of islets. Islet FAM3A expression was correlated with insulin expression under physiological and diabetic conditions. Mice with specific knockout of FAM3A in islet ß cells exhibited markedly blunted insulin secretion and glucose intolerance. FAM3A-deficient islets showed significant decrease in PDX1 expression, and insulin expression and secretion. FAM3A overexpression upregulated PDX1 and insulin expressions, and augmented insulin secretion in cultured islets and ß cells. Mechanistically, FAM3A enhanced ATP production to elevate cellular Ca2+ level and promote insulin secretion. Furthermore, FAM3A-induced ATP release activated CaM to function as a co-activator of FOXA2, stimulating PDX1 gene transcription. In conclusion, FAM3A plays crucial roles in controlling PDX1 and insulin expressions in pancreatic ß cells. Inhibition of FAM3A will trigger mitochondrial dysfunction to repress PDX1 and insulin expressions.


Asunto(s)
Citocinas/metabolismo , Proteínas de Homeodominio/metabolismo , Insulina/metabolismo , Islotes Pancreáticos/metabolismo , Transactivadores/metabolismo , Adenosina Trifosfato/metabolismo , Animales , Calcio/metabolismo , Línea Celular , Células Cultivadas , Citocinas/genética , Glucosa/metabolismo , Factor Nuclear 3-beta del Hepatocito , Proteínas de Homeodominio/genética , Humanos , Immunoblotting , Inmunoprecipitación , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Transactivadores/genética
3.
Biochem Biophys Res Commun ; 516(1): 209-214, 2019 08 13.
Artículo en Inglés | MEDLINE | ID: mdl-31208715

RESUMEN

Chondrocyte death due to apoptosis is central for osteoarthritis (OA) pathogenesis. The family with sequence similarity 3A (FAM3A) is a mitochondrial protein that plays an important role for cellular adaptation to stress and cell survival. Yet, whether FAM3A is associated with chondrocyte apoptosis and OA pathogenesis remains uncharacterized. In this study, we found that FAM3A expression was downregulated in cartilage tissue from an experimental OA mouse model. Besides, FAM3A expression was also reduced in chondrocytes treated with interleukin-1ß (IL-1ß), an inflammatory cytokine that promotes cartilage degradation. Moreover, we discovered that FAM3A attenuated chondrocyte apoptosis induced by IL-1ß treatment in vitro, suggesting a protective effect of FAM3A against chondrocyte apoptosis. Moreover, mechanistically, FAM3A activated PI3K/Akt/mTOR pathway in IL-1ß-treated chondrocytes, and blockade of PI3K/Akt/mTOR pathway with specific inhibitors, wortmannin and LY294002, diminished FAM3A effect on IL-1ß-induced chondrocyte apoptosis, hence demonstrating that FAM3A attenuates IL-1ß-induced chondrocyte apoptosis through activating the pro-survival PI3K/Akt/mTOR pathway. In conclusion, our study may identify FAM3A as a potential regulator of chondrocyte apoptosis involved in OA pathogenesis.


Asunto(s)
Condrocitos/patología , Citocinas/inmunología , Interleucina-1beta/inmunología , Osteoartritis/inmunología , Animales , Apoptosis , Condrocitos/inmunología , Ratones , Ratones Endogámicos C57BL , Osteoartritis/patología , Fosfatidilinositol 3-Quinasas/inmunología , Proteínas Proto-Oncogénicas c-akt/inmunología , Transducción de Señal , Serina-Treonina Quinasas TOR/inmunología
4.
Int J Mol Sci ; 19(5)2018 Apr 26.
Artículo en Inglés | MEDLINE | ID: mdl-29701719

RESUMEN

Liver ischemia-reperfusion injury (IRI) is a major complication of hemorrhagic shock, liver transplantation, and other liver surgeries. It is one of the leading causes for post-surgery hepatic dysfunction, always leading to morbidity and mortality. Several strategies, such as low-temperature reperfusion and ischemic preconditioning, are useful for ameliorating liver IRI in animal models. However, these methods are difficult to perform in clinical surgeries. It has been reported that the activation of peroxisome proliferator activated receptor gamma (PPARγ) protects the liver against IRI, but with unidentified direct target gene(s) and unclear mechanism(s). Recently, FAM3A, a direct target gene of PPARγ, had been shown to mediate PPARγ’s protective effects in liver IRI. Moreover, noncoding RNAs, including LncRNAs and miRNAs, had also been reported to play important roles in the process of hepatic IRI. This review briefly discussed the roles and mechanisms of several classes of important molecules, including PPARγ, FAM3A, miRNAs, and LncRNAs, in liver IRI. In particular, oral administration of PPARγ agonists before liver surgery or liver transplantation to activate hepatic FAM3A pathways holds great promise for attenuating human liver IRI.


Asunto(s)
Hígado/metabolismo , Daño por Reperfusión/metabolismo , Animales , Citocinas/genética , Citocinas/metabolismo , Humanos , Hígado/irrigación sanguínea , Hígado/patología , MicroARNs/genética , MicroARNs/metabolismo , Terapia Molecular Dirigida/métodos , PPAR gamma/agonistas , PPAR gamma/genética , PPAR gamma/metabolismo , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Daño por Reperfusión/tratamiento farmacológico
5.
Cell Physiol Biochem ; 44(5): 2029-2041, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29241198

RESUMEN

BACKGROUND/AIMS: Stroke is the leading cause of adult disability, and glutamate-induced dysregulation of intracellular Ca2+ homeostasis is a key mechanism. FAM3A is the first member of the family with sequence similarity 3 (FAM3) gene family, and its biological function remains largely unknown. We have recently reported that FAM3A exerts protective effects against oxidative stress and mitochondrial dysfunction in HT22 cells. METHODS: Here, we investigated the protective effects of FAM3A using a glutamate-induced neuronal injury model in nerve growth factor (NGF)-differentiated PC12 cells. The protective effects were determined by measuring lactate dehydrogenase (LDH) release, apoptosis and mitochondrial oxidative stress. Ca2+ imaging was performed to detect changes in intracellular Ca2+ concentration in PC12 cells. The related molecular mechanisms were investigated by fluorescence staining, coimmunoprecipitation (Co-IP) and western blotting. RESULTS: Upregulation of FAM3A by lentivirus transfection markedly decreased LDH release, inhibited apoptosis and reduced mitochondrial oxidative stress, which were accompanied by alleviated intracellular Ca2+ levels as measured by calcium imaging. The results of western blotting showed that FAM3A significantly decreased the surface expression of metabotropic glutamate receptor 1/5 (mGluR1/5), with no effect on the expression of N-methyl-d-aspartic acid receptor (NMDAR) or α-amino-3-hydroxy-5-methyl-4-isoxazole-propionic acid receptor (AMPAR) subunits. FAM3A overexpression also inhibited the intracellular Ca2+ release mediated by mGluR1/5 and inositol 1,4,5-trisphosphate receptor (IP3R), but not the ryanodine receptor (RyR). In addition, FAM3A significantly attenuated the store-operated calcium entry (SOCE) induced by thapsigargin (Tg), but the expression of SOCE-related proteins was not altered. The results of coimmunoprecipitation (Co-IP) showed that FAM3A disrupted the interaction of stromal interaction molecule 1 (STIM1) with Orai1 triggered by glutamate. CONCLUSION: These results suggest that the upregulation of FAM3A protects against glutamate-induced dysfunction of Ca2+ homeostasis not only by inhibiting mGluR1/5-dependent endoplasmic reticulum (ER) Ca2+ release, but also by attenuating SOCE mediated by the STIM1-Orai1 interaction.


Asunto(s)
Calcio/metabolismo , Diferenciación Celular/efectos de los fármacos , Citocinas/metabolismo , Ácido Glutámico/toxicidad , Sustancias Protectoras/farmacología , Animales , Apoptosis , Citocinas/genética , Vectores Genéticos/genética , Vectores Genéticos/metabolismo , Receptores de Inositol 1,4,5-Trifosfato/metabolismo , L-Lactato Deshidrogenasa/metabolismo , Factor de Crecimiento Nervioso/farmacología , Proteína ORAI1/genética , Proteína ORAI1/metabolismo , Estrés Oxidativo , Células PC12 , Ratas , Receptor del Glutamato Metabotropico 5/metabolismo , Receptores de Glutamato Metabotrópico/metabolismo , Molécula de Interacción Estromal 1/genética , Molécula de Interacción Estromal 1/metabolismo , Molécula de Interacción Estromal 2/genética , Molécula de Interacción Estromal 2/metabolismo , Tapsigargina/farmacología
6.
J Mol Cell Cardiol ; 74: 173-82, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24857820

RESUMEN

The biological function of FAM3A, the first member of family with sequence similarity 3 (FAM3) gene family, remains largely unknown. This study aimed to determine its role in the proliferation and migration of vascular smooth muscle cells (VSMCs). Immunohistochemical staining revealed that FAM3A protein is expressed in the tunica media of rodent arteries, and its expression is reduced with an increase in prostaglandin E receptor 2 (EP2) expression after injury. In vitro, FAM3A overexpression promotes proliferation and migration of VSMCs, whereas FAM3A silencing inhibits these processes. In vivo, FAM3A overexpression results in exaggerated neointima formation of rat carotid artery after balloon injury. FAM3A activates Akt in a PI3K-dependent manner. In contrast, FAM3A induces ERK1/2 activation independent of PI3K. FAM3A protein is subcellularly located in mitochondria, where it affects ATP production and release. Activation of EP2 represses FAM3A expression, leading to impaired ATP production and release in VSMCs. FAM3A-induced activation of Akt and ERK1/2 pathways, proliferation and migration of VSMCs are inhibited by P2 receptor antagonist suramin. Furthermore, inhibition or knockdown of P2Y1 receptor inihibits FAM3A-induced proliferation and migration of VSMCs. In conclusion, FAM3A promotes proliferation and migration of VSMCs via P2Y1 receptor-mediated activation of Akt and ERK1/2 pathways. In injured vessels, FAM3A was repressed by upregulated EP2 expression, leading to the attenuation of ATP-P2Y1 receptor signaling, which is beneficial for preventing excessive proliferation and migration of VSMCs.


Asunto(s)
Arterias Carótidas/metabolismo , Traumatismos de las Arterias Carótidas/genética , Citocinas/genética , Neointima/genética , Neointima/metabolismo , Receptores Purinérgicos P2Y1/genética , Animales , Oclusión con Balón , Arterias Carótidas/patología , Traumatismos de las Arterias Carótidas/metabolismo , Traumatismos de las Arterias Carótidas/patología , Movimiento Celular , Proliferación Celular , Citocinas/metabolismo , Regulación de la Expresión Génica , Masculino , Proteína Quinasa 1 Activada por Mitógenos/genética , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/genética , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/patología , Miocitos del Músculo Liso/metabolismo , Miocitos del Músculo Liso/patología , Neointima/patología , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas , Ratas Sprague-Dawley , Subtipo EP2 de Receptores de Prostaglandina E/genética , Subtipo EP2 de Receptores de Prostaglandina E/metabolismo , Receptores Purinérgicos P2Y1/metabolismo , Transducción de Señal
7.
Redox Biol ; 74: 103225, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38875957

RESUMEN

Acute kidney injury (AKI) is in high prevalence worldwide but with no therapeutic strategies. Programmed cell death in tubular epithelial cells has been reported to accelerate a variety of AKI, but the major pathways and underlying mechanisms are not defined. Herein, we identified that pyroptosis was responsible for AKI progression and related to ATP depletion in renal tubular cells. We found that FAM3A, a mitochondrial protein that assists ATP synthesis, was decreased and negatively correlated with tubular cell injury and pyroptosis in both mice and patients with AKI. Knockout of FAM3A worsened kidney function decline, increased macrophage and neutrophil cell infiltration, and facilitated tubular cell pyroptosis in ischemia/reperfusion injury model. Conversely, FAM3A overexpression alleviated tubular cell pyroptosis, and inhibited kidney injury in ischemic AKI. Mechanistically, FAM3A promoted PI3K/AKT/NRF2 signaling, thus blocking mitochondrial reactive oxygen species (mt-ROS) accumulation. NLRP3 inflammasome sensed the overload of mt-ROS and then activated Caspase-1, which cleaved GSDMD, pro-IL-1ß, and pro-IL-18 into their mature forms to mediate pyroptosis. Of interest, NRF2 activator alleviated the pro-pyroptotic effects of FAM3A depletion, whereas the deletion of NRF2 blocked the anti-pyroptotic function of FAM3A. Thus, our study provides new mechanisms for AKI progression and demonstrates that FAM3A is a potential therapeutic target for treating AKI.


Asunto(s)
Lesión Renal Aguda , Túbulos Renales , Piroptosis , Especies Reactivas de Oxígeno , Animales , Humanos , Masculino , Ratones , Lesión Renal Aguda/metabolismo , Lesión Renal Aguda/patología , Lesión Renal Aguda/genética , Citocinas , Modelos Animales de Enfermedad , Inflamasomas/metabolismo , Túbulos Renales/metabolismo , Túbulos Renales/patología , Ratones Noqueados , Mitocondrias/metabolismo , Proteínas Mitocondriales/metabolismo , Proteínas Mitocondriales/genética , Factor 2 Relacionado con NF-E2/metabolismo , Factor 2 Relacionado con NF-E2/genética , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal
8.
J Cardiovasc Transl Res ; 17(1): 104-120, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-37014466

RESUMEN

Mitochondrial protein sequence similarity 3 gene family member A (FAM3A) plays important roles in the electron transfer chain, while its functions in the heart are still unknown. This study aims to explore the roles and mechanisms of FAM3A after myocardial infarction (MI). FAM3A-deficient (Fam3a-/-) mice were implemented with MI injury and showed lower survival rates at 4 weeks as well as decreased cardiac systolic function. Isolated cardiomyocytes of Fam3a-/- mice showed reduced basal, ATP-linked respiration and respiratory reserve compared to that of wild-type mice. Transmission electron microscopy studies showed Fam3a-/- mice had a larger size and elevated density of mitochondria. FAM3A deficiency also induced elevated mitochondrial Ca2+, higher opening level of mPTP, lower mitochondrial membrane potential and elevated apoptotic rates. Further analyses demonstrated that mitochondrial dynamics protein Opa1 contributed to the effects of FAM3A in cardiomyocytes. Our study discloses the important roles of mitochondrial protein FAM3A in the heart.


Asunto(s)
Insuficiencia Cardíaca , Enfermedades Mitocondriales , Infarto del Miocardio , Ratones , Animales , Insuficiencia Cardíaca/genética , Insuficiencia Cardíaca/metabolismo , Infarto del Miocardio/genética , Infarto del Miocardio/metabolismo , Mitocondrias/metabolismo , Proteínas Mitocondriales/metabolismo , Enfermedades Mitocondriales/metabolismo , Miocitos Cardíacos/metabolismo , Citocinas/metabolismo
9.
Mil Med Res ; 11(1): 41, 2024 Jun 27.
Artículo en Inglés | MEDLINE | ID: mdl-38937853

RESUMEN

BACKGROUND: Extracellular adenosine triphosphate (ATP) is an important signal molecule. In previous studies, intensive research had revealed the crucial roles of family with sequence similarity 3 member A (FAM3A) in controlling hepatic glucolipid metabolism, islet ß cell function, adipocyte differentiation, blood pressure, and other biological and pathophysiological processes. Although mitochondrial protein FAM3A plays crucial roles in the regulation of glucolipid metabolism via stimulating ATP release to activate P2 receptor pathways, its mechanism in promoting ATP release in hepatocytes remains unrevealed. METHODS: db/db, high-fat diet (HFD)-fed, and global pannexin 1 (PANX1) knockout mice, as well as liver sections of individuals, were used in this study. Adenoviruses and adeno-associated viruses were utilized for in vivo gene overexpression or inhibition. To evaluate the metabolic status in mice, oral glucose tolerance test (OGTT), pyruvate tolerance test (PTT), insulin tolerance test (ITT), and magnetic resonance imaging (MRI) were conducted. Protein-protein interactions were determined by coimmunoprecipitation with mass spectrometry (MS) assays. RESULTS: In livers of individuals and mice with steatosis, the expression of ATP-permeable channel PANX1 was increased (P < 0.01). Hepatic PANX1 overexpression ameliorated the dysregulated glucolipid metabolism in obese mice. Mice with hepatic PANX1 knockdown or global PANX1 knockout exhibited disturbed glucolipid metabolism. Restoration of hepatic PANX1 rescued the metabolic disorders of PANX1-deficient mice (P < 0.05). Mechanistically, ATP release is mediated by the PANX1-activated protein kinase B-forkhead box protein O1 (Akt-FOXO1) pathway to inhibit gluconeogenesis via P2Y receptors in hepatocytes. PANX1-mediated ATP release also activated calmodulin (CaM) (P < 0.01), which interacted with c-Jun N-terminal kinase (JNK) to inhibit its activity, thereby deactivating the transcription factor activator protein-1 (AP1) and repressing fatty acid synthase (FAS) expression and lipid synthesis (P < 0.05). FAM3A stimulated the expression of PANX1 via heat shock factor 1 (HSF1) in hepatocytes (P < 0.05). Notably, FAM3A overexpression failed to promote ATP release, inhibit the expression of gluconeogenic and lipogenic genes, and suppress gluconeogenesis and lipid deposition in PANX1-deficient hepatocytes and livers. CONCLUSIONS: PANX1-mediated release of ATP plays a crucial role in maintaining hepatic glucolipid homeostasis, and it confers FAM3A's suppressive effects on hepatic gluconeogenesis and lipogenesis.


Asunto(s)
Adenosina Trifosfato , Conexinas , Gluconeogénesis , Lipogénesis , Hígado , Proteínas del Tejido Nervioso , Animales , Conexinas/metabolismo , Ratones , Gluconeogénesis/fisiología , Proteínas del Tejido Nervioso/metabolismo , Proteínas del Tejido Nervioso/genética , Adenosina Trifosfato/metabolismo , Lipogénesis/fisiología , Hígado/metabolismo , Ratones Noqueados , Masculino , Humanos , Dieta Alta en Grasa/efectos adversos , Citocinas
10.
Metabolism ; 139: 155372, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36470472

RESUMEN

Reduced mitochondrial ATP synthase (ATPS) capacity plays crucial roles in the pathogenesis of metabolic disorders. However, there is currently no effective strategy for synchronously stimulating the expressions of ATPS key subunits to restore its assembly. This study determined the roles of mitochondrial protein FAM3A in regulating the activity and assembly of ATPS in hepatocytes. FAM3A is localized in mitochondrial matrix, where it interacts with F1-ATPS to initially activate ATP synthesis and release, and released ATP further activates P2 receptor-Akt-CREB pathway to induce FOXD3 expression. FOXD3 synchronously stimulates the transcriptions of ATPS key subunits and assembly genes to increase its assembly and capacity, augmenting ATP synthesis and inhibiting ROS production. FAM3A, FOXD3 and ATPS expressions were reduced in livers of diabetic mice and NAFLD patients. FOXD3 expression, ATPS capacity and ATP content were reduced in various tissues of FAM3A-deficient mice with dysregulated glucose and lipid metabolism. Hepatic FOXD3 activation increased ATPS assembly to ameliorate dysregulated glucose and lipid metabolism in obese mice. Hepatic FOXD3 inhibition or knockout reduced ATPS capacity to aggravate HFD-induced hyperglycemia and steatosis. In conclusion, FAM3A is an active ATPS component, and regulates its activity and assembly by activating FOXD3. Activating FAM3A-FOXD3 axis represents a viable strategy for restoring ATPS assembly to treat metabolic disorders.


Asunto(s)
Diabetes Mellitus Experimental , Enfermedad del Hígado Graso no Alcohólico , Ratones , Animales , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Glucosa , Homeostasis , Adenosina Trifosfato/metabolismo , Citocinas/metabolismo
11.
In Vitro Cell Dev Biol Anim ; 59(6): 431-442, 2023 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-37474885

RESUMEN

Family with sequence similarity 3 member A (FAM3A) is a multifunctional protein that is related to the pathological process of various disorders. FAM3A is reportedly able to affect the phenotypic change of vascular smooth muscle cells under a hypertensive state. Whether FAM3A mediates the phenotypic switch of vascular smooth muscle cells under an atherosclerotic state remains unaddressed. This work investigated the roles and mechanisms of FAM3A in mediating the phenotypic switch of human aortic smooth muscle cells (HASMCs) stimulated with oxidised low-density lipoprotein (ox-LDL) in vitro. FAM3A expression was elevated in HASMCs following ox-LDL treatment. FAM3A silencing led to a suppressive effect on ox-LDL-provoked proliferation, migration and inflammation of HASMCs, whereas FAM3A overexpression had an opposite effect. Ox-LDL elicited a change in HASMCs from a contractile phenotype to a synthetic phenotype, which was inhibited by FAM3A silencing or enhanced by FAM3A overexpression. Further investigation elucidated that FAM3A silencing repressed and FAM3A overexpression promoted ox-LDL-induced activation of the PI3K-AKT pathway in HASMCs. Reactivation of AKT reversed the suppressive effect of FAM3A silencing on the ox-LDL-induced phenotypic switch of HASMCs. Restraining AKT blocked the promoting effect of FAM3A overexpression on the ox-LDL-induced phenotypic switch of HASMCs. In summary, this work elucidates that FAM3A mediates the ox-LDL-induced phenotypic switch of HASMCs by influencing the PI3K-AKT pathway, indicating a potential role for FAM3A in atherosclerosis.


Asunto(s)
Aterosclerosis , Proteínas Proto-Oncogénicas c-akt , Humanos , Aterosclerosis/genética , Movimiento Celular , Proliferación Celular/genética , Células Cultivadas , Lipoproteínas LDL/farmacología , Lipoproteínas LDL/metabolismo , Miocitos del Músculo Liso , Fenotipo , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo
12.
Metabolism ; 136: 155292, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-35995281

RESUMEN

Mitochondrial FAM3A has been revealed to be a viable target for treating diabetes and nonalcoholic fatty liver disease (NAFLD). However, its distinct mechanism in ameliorating hepatic steatosis remained unrevealed. High-throughput RNA sequencing revealed that carnitine palmityl transferase 2 (CPT2), one of the key enzymes for lipid oxidation, is the downstream molecule of FAM3A signaling pathway in hepatocytes. Intensive study demonstrated that FAM3A-induced ATP release activated P2 receptor to promote the translocation of calmodulin (CaM) from cytoplasm into nucleus, where it functioned as a co-activator of forkhead box protein A2 (FOXA2) to promote the transcription of CPT2, increasing free fatty acid oxidation and reducing lipid deposition in hepatocytes. Furthermore, antidepressant imipramine activated FAM3A-ATP-P2 receptor-CaM-FOXA2-CPT2 pathway to reduce lipid deposition in hepatocytes. In FAM3A-deficient hepatocytes, imipramine failed to activate CaM-FOXA2-CPT2 axis to increase lipid oxidation. Imipramine administration significantly ameliorated hepatic steatosis, hyperglycemia and obesity of obese mice mainly by activating FAM3A-ATP-CaM-FOXA2-CPT2 pathway in liver and thermogenesis in brown adipose tissue (BAT). In FAM3A-deficient mice fed on high-fat-diet, imipramine treatment failed to correct the dysregulated lipid and glucose metabolism, and activate thermogenesis in BAT. In conclusion, imipramine activates FAM3A-ATP-CaM-FOXA2-CPT2 pathway to ameliorate steatosis. For depressive patients complicated with metabolic disorders, imipramine may be recommended in priority as antidepressive drug.


Asunto(s)
Imipramina , Enfermedad del Hígado Graso no Alcohólico , Adenosina Trifosfato/metabolismo , Animales , Calmodulina/metabolismo , Carnitina O-Palmitoiltransferasa/metabolismo , Citocinas/metabolismo , Dieta Alta en Grasa , Ácidos Grasos no Esterificados/metabolismo , Glucosa/metabolismo , Factor Nuclear 3-beta del Hepatocito/metabolismo , Imipramina/farmacología , Imipramina/uso terapéutico , Metabolismo de los Lípidos , Hígado/metabolismo , Ratones , Enfermedad del Hígado Graso no Alcohólico/tratamiento farmacológico , Enfermedad del Hígado Graso no Alcohólico/metabolismo
13.
Endocrinol Metab (Seoul) ; 37(1): 74-83, 2022 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-35144334

RESUMEN

BACKGROUND: Dulaglutide, a long-acting glucagon-like peptide-1 receptor agonist (GLP-1RA), has been shown to reduce body weight and liver fat content in patients with type 2 diabetes. Family with sequence similarity 3 member A (FAM3A) plays a vital role in regulating glucose and lipid metabolism. The aim of this study was to determine the mechanisms by which dulaglutide protects against hepatic steatosis in HepG2 cells treated with palmitic acid (PA). METHODS: HepG2 cells were pretreated with 400 µM PA for 24 hours, followed by treatment with or without 100 nM dulaglutide for 24 hours. Hepatic lipid accumulation was determined using Oil red O staining and triglyceride (TG) assay, and the expression of lipid metabolism-associated factor was analyzed using quantitative real time polymerase chain reaction and Western blotting. RESULTS: Dulaglutide significantly decreased hepatic lipid accumulation and reduced the expression of genes associated with lipid droplet binding proteins, de novo lipogenesis, and TG synthesis in PA-treated HepG2 cells. Dulaglutide also increased the expression of proteins associated with lipolysis and fatty acid oxidation and FAM3A in PA-treated cells. However, exendin-(9-39), a GLP-1R antagonist, reversed the expression of FAM3A, and fatty acid oxidation-associated factors increased due to dulaglutide. In addition, inhibition of FAM3A by siRNA attenuated the reducing effect of dulaglutide on TG content and its increasing effect on regulation of fatty acid oxidation. CONCLUSION: These results suggest that dulaglutide could be used therapeutically for improving nonalcoholic fatty liver disease, and its effect could be mediated in part via upregulation of FAM3A expression through a GLP-1R-dependent pathway.


Asunto(s)
Diabetes Mellitus Tipo 2 , Enfermedad del Hígado Graso no Alcohólico , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Péptidos Similares al Glucagón/análogos & derivados , Humanos , Fragmentos Fc de Inmunoglobulinas , Enfermedad del Hígado Graso no Alcohólico/inducido químicamente , Enfermedad del Hígado Graso no Alcohólico/tratamiento farmacológico , Enfermedad del Hígado Graso no Alcohólico/prevención & control , Ácido Palmítico/toxicidad , Proteínas Recombinantes de Fusión , Transducción de Señal
14.
EBioMedicine ; 43: 32-42, 2019 May.
Artículo en Inglés | MEDLINE | ID: mdl-31000420

RESUMEN

BACKGROUND: Angiogenesis improves reperfusion to the ischaemic tissue after vascular obstruction. The underlying molecular mechanisms of post-ischaemic angiogenesis are not clear. FAM3A belongs to the family with sequence similarity 3 (FAM3) genes, but its biological function in endothelial cells in regards to vascular diseases is not well understood. METHODS: Gain- and loss-of-function methods by adenovirus or associated-adenovirus (AAV) in different models were applied to investigate the effects of FAM3A on endothelial angiogenesis. Endothelial angiogenesis was analysed by tube formation, migration and proliferation in vitro, and the blood flow and capillary density in a hind limb ischaemic model in vivo. FINDINGS: Endothelial FAM3A expression is downregulated under hypoxic conditions. Overexpression of FAM3A promotes, but depletion of FAM3A suppresses, endothelial tube formation, proliferation and migration. Utilizing the mouse hind limb ischaemia model, we also observe that FAM3A overexpression can improve blood perfusion and increase capillary density, whereas FAM3A knockdown has the opposite effects. Mechanistically, mitochondrial FAM3A increases adenosine triphosphate (ATP) production and secretion; ATP binds to P2 receptors and then upregulates cytosolic free Ca2+ levels. Increased intracellular Ca2+ levels enhance phosphorylation of the transcriptional factor cAMP response element binding protein (CREB) and its recruitment to the VEGFA promoter, thus activating VEGFA transcription and the final endothelial angiogenesis. INTERPRETATION: In summary, our data demonstrate that FAM3A positively regulates angiogenesis through activation of VEGFA transcription, suggesting that FAM3A may constitute a novel molecular therapeutic target for ischaemic vascular disease.


Asunto(s)
Citocinas/genética , Células Endoteliales/metabolismo , Regulación de la Expresión Génica , Isquemia/metabolismo , Neovascularización Fisiológica/genética , Adenosina Trifosfato/metabolismo , Animales , Biomarcadores , Calcio/metabolismo , Señalización del Calcio , Línea Celular , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Citocinas/metabolismo , Modelos Animales de Enfermedad , Técnicas de Silenciamiento del Gen , Humanos , Inmunohistoquímica , Masculino , Ratones , Mitocondrias/metabolismo , Estrés Oxidativo , Consumo de Oxígeno , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo
15.
Life Sci ; 207: 23-29, 2018 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-29802941

RESUMEN

AIMS: Little is known regarding whether the NFE2/miR-423-5p and FAM3A-ATP-Akt pathway in liver mediates exercise allured alleviation of insulin resistance connected with diet-induced obesity. This research inquired the influence of exercise on liver insulin sensitivity and whole body insulin resistance in high-fat diet fed rats. MATERIALS AND METHODS: Forty male Sprague-Dawley rats at seven-week-old were assigned to four groups at random: standard diet as normal control group (NC, n = 10), high-fat diet group (HFD, n = 10), high-fat diet with chronic exercise intervention group (HFD-CE, n = 10) and high-fat diet with acute exercise intervention group (HFD-AE, n = 10). KEY FINDINGS: Compared with rats fed with a standard diet, eight-week high-fat diet feeding lead to elevated body weight, visceral fat content and serum FFAs, and decreased insulin sensitivity index. Moreover, high-fat diet enhanced NFE2 protein expression and miR-423-5p level, decreased FAM3A mRNA and protein expression, ATP level and Akt phosphorylation in liver. In contrast, physical exercise, both chronic and acute exercise alleviated whole body insulin resistance, reduced hepatic NFE2 and miR-423-5p expression, and serum FFAs level, meanwhile enhanced FAM3A mRNA and protein expression, ATP level and Akt phosphorylation in liver. The current findings indicated that exercise in diet-induced obesity, both chronic and acute, induce a momentous regulation in NFE2/miR-423-5p and FAM3A-ATP-Akt pathway in liver, and improve hepatic insulin sensitivity and whole body insulin resistance. SIGNIFICANCE: All these results supply crucial evidence in our comprehending of the molecular mechanism that connected exercise to an alleviation of insulin resistance.


Asunto(s)
Citocinas/metabolismo , Resistencia a la Insulina , Insulina/metabolismo , MicroARNs/metabolismo , Factor de Transcripción NF-E2/metabolismo , Condicionamiento Físico Animal , Adenosina Trifosfato/metabolismo , Animales , Área Bajo la Curva , Dieta , Dieta Alta en Grasa , Prueba de Tolerancia a la Glucosa , Hígado/metabolismo , Masculino , Obesidad/metabolismo , Fosforilación , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas , Ratas Sprague-Dawley
16.
Oncotarget ; 8(28): 45862-45873, 2017 Jul 11.
Artículo en Inglés | MEDLINE | ID: mdl-28515350

RESUMEN

FAM3A plays important roles in regulating hepatic glucose/lipid metabolism and the proliferation of VSMCs. This study determined the role and mechanism of FAM3A in the adipogenesis of 3T3-L1 preadipocytes. During the adipogenesis of 3T3-L1 preadipocytes, FAM3A expression was significantly increased. FAM3A overexpression enhanced 3T3-L1 preadipocyte adipogenesis with increased phosphorylated Akt (pAkt) level, whereas FAM3A silencing inhibited 3T3-L1 preadipocyte adipogenesis with reduced pAkt level. Moreover, FAM3A silencing reduced the expression and secretion of adipokines in 3T3-L1 cells. FAM3A protein is mainly located in mitochondrial fraction of 3T3-L1 cells and mouse adipose tissue. FAM3A overexpression increased, whereas FAM3A silencing decreased ATP production in 3T3-L1 preadipocytes. FAM3A-induced adipogenesis of 3T3-L1 preadipocytes was blunted by inhibitor of P2 receptor. In white adipose tissues of db/db and HFD-fed obese mice, FAM3A expression was reduced. One-month rosiglitazone administration upregulated FAM3A expression, and increased cellular ATP content and pAkt level in white adipose tissues of normal and obese mice. In conclusion, FAM3A enhances the adipogenesis of preadipocytes by activating ATP-P2 receptor-Akt pathway. Under obese condition, a decrease in FAM3A expression in adipose tissues plays important roles in the development of adipose dysfunction and type 2 diabetes.


Asunto(s)
Adenosina Trifosfato/metabolismo , Adipocitos/citología , Adipocitos/metabolismo , Adipogénesis , Citocinas/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptores Purinérgicos P2/metabolismo , Transducción de Señal , Células 3T3-L1 , Tejido Adiposo Blanco/citología , Tejido Adiposo Blanco/metabolismo , Animales , Diferenciación Celular/genética , Regulación de la Expresión Génica/efectos de los fármacos , Silenciador del Gen , Masculino , Ratones , PPAR gamma/metabolismo , Rosiglitazona , Tiazolidinedionas/farmacología
17.
Oncotarget ; 8(30): 49882-49896, 2017 Jul 25.
Artículo en Inglés | MEDLINE | ID: mdl-28562339

RESUMEN

FAM3A is a novel mitochondrial protein, and its biological function remains largely unknown. This study determined the role and mechanism of FAM3A in liver ischemia-reperfusion injury (IRI). In mouse liver after IRI, FAM3A expression was increased. FAM3A-deficient mice exhibited exaggerated liver damage with increased serum levels of AST, ALT, MPO, MDA and oxidative stress when compared with WT mice after liver IRI. FAM3A-deficient mouse livers had a decrease in ATP content, Akt activity and anti-apoptotic protein expression with an increase in apoptotic protein expression, inflammation and oxidative stress when compared WT mouse livers after IRI. Rosiglitazone pretreatment protected against liver IRI in wild type mice but not in FAM3A-deficient mice. In cultured hepatocytes, FAM3A overexpression protected against, whereas FAM3A deficiency exaggerated oxidative stress-induced cell death. FAM3A upregulation or FAM3A overexpression inhibited hypoxia/reoxygenation-induced activation of apoptotic gene and hepatocyte death in P2 receptor-dependent manner. FAM3A deficiency blunted rosiglitazone's beneficial effects on Akt activation and cell survival in cultured hepatocytes. Collectively, FAM3A protects against liver IRI by activating Akt survival pathways, repressing inflammation and attenuating oxidative stress. Moreover, the protective effects of PPARγ agonist(s) on liver IRI are dependent on FAM3A-ATP-Akt pathway.


Asunto(s)
Citocinas/metabolismo , Hepatopatías/metabolismo , Estrés Oxidativo , PPAR gamma/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Daño por Reperfusión/metabolismo , Transducción de Señal , Animales , Apoptosis , Supervivencia Celular , Citocinas/genética , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Expresión Génica , Hepatocitos/metabolismo , Hepatopatías/patología , Pruebas de Función Hepática , Masculino , Ratones , Ratones Noqueados , Estrés Oxidativo/efectos de los fármacos , Transporte de Proteínas/efectos de los fármacos , Daño por Reperfusión/patología , Rosiglitazona , Transducción de Señal/efectos de los fármacos , Tiazolidinedionas/farmacología
18.
Gene ; 595(1): 92-98, 2016 Dec 20.
Artículo en Inglés | MEDLINE | ID: mdl-27688071

RESUMEN

FAM3A (family with sequence similarity 3, member A) is regulated by PPARG and participates in the metabolism of lipid in liver. However, the transcriptional regulation analysis of FAM3A is very little and biological function of FAM3A still unclear. In this study, the core promoter region and transcription factor binding sites of FAM3A gene were identified and characterized using dual luciferase report experiments and electrophoretic mobility shift assays (EMSA). The promoter activity of FAM3A was dramatically decreased after the mutation of C/EBPß binding sites, suggesting that C/EBPß is a transcriptional activator of FAM3A. Overexpression of FAM3A significantly inhibited the efficiency of preadipocytes to differentiate into adipocytes as indicated by Western Blot and Oil Red O staining assay. These results suggest that C/EBPß plays an important role in regulating FAM3A promoter activity and FAM3A inhibits adipocyte differentiation.


Asunto(s)
Adipocitos/metabolismo , Diferenciación Celular/fisiología , Citocinas/biosíntesis , Elementos de Respuesta/fisiología , Transcripción Genética/fisiología , Células 3T3-L1 , Animales , Proteína beta Potenciadora de Unión a CCAAT/genética , Proteína beta Potenciadora de Unión a CCAAT/metabolismo , Células CHO , Cricetinae , Cricetulus , Citocinas/genética , Humanos , Ratones , Mutación
19.
Neurochem Int ; 94: 82-9, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26939760

RESUMEN

Endoplasmic reticulum (ER) stress is linked to several neurological disorders, and neuronal injury cascades initiated by excessive ER stress are mediated, in part, via mitochondrial dysfunction. In the present study, we identified FAM3A as an important regulator of ER stress-induced cell death in neuronal HT22 cells. The ER stress inductor tunicamycin (TM) significantly decreased the expression of FAM3A at both mRNA and protein levels, which was shown to be dependent on the induction of reactive oxygen species (ROS). Overexpression of FAM3A attenuated TM-induced apoptosis and activation of ER stress factors, but had no effect on ER calcium metabolism in HT22 cells. We also found decreased mitochondrial ROS generation, inhibited cytochrome c release and preserved mitochondrial membrane potential (MMP) in FAM3A overexpressed cells. In addition, the experiments using isolated mitochondria showed that overexpression of FAM3A attenuated mitochondrial swelling and loss of mitochondrial Ca(2+) buffering capacity after TM exposure. By using specific targeted small interfering RNA (siRNA) to knockdown the expression of the C/EBP homologous protein (CHOP), we found that FAM3A-induced protection and inhibition of ER stress was mediated by inverting TM-induced decrease of Wnt through the CHOP pathway. Our study demonstrates a pivotal role of FAM3A in protecting against TM-induced cytotoxicity via regulating CHOP-Wnt pathway, and suggests the therapeutic values of FAM3A overexpression against ER stress-associated neuronal injury.


Asunto(s)
Apoptosis/fisiología , Citocinas/biosíntesis , Estrés del Retículo Endoplásmico/fisiología , Mitocondrias/metabolismo , Factor de Transcripción CHOP/biosíntesis , Vía de Señalización Wnt/fisiología , Células 3T3 , Animales , Apoptosis/efectos de los fármacos , Estrés del Retículo Endoplásmico/efectos de los fármacos , Ratones , Mitocondrias/efectos de los fármacos , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Tunicamicina/toxicidad , Vía de Señalización Wnt/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA