Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
1.
Circ Res ; 126(12): 1746-1759, 2020 06 05.
Artículo en Inglés | MEDLINE | ID: mdl-32279581

RESUMEN

RATIONALE: Dysregulated purinergic signaling transduction plays important roles in the pathogenesis of cardiovascular diseases. However, the role and mechanism of vascular smooth muscle cell (VSMC)-released ATP in the regulation of blood pressure, and the pathogenesis of hypertension remain unknown. FAM3A (family with sequence similarity 3 member A) is a new mitochondrial protein that enhances ATP production and release. High expression of FAM3A in VSMC suggests it may play a role in regulating vascular constriction and blood pressure. OBJECTIVE: To determine the role and mechanism of FAM3A-ATP signaling pathway in VSMCs in the regulation of blood pressure and the pathogenesis of hypertension. METHODS AND RESULTS: In the media layer of hypertensive rat and mouse arteries, and the internal mammary artery of hypertensive patients, FAM3A expression was increased. VSMC-specific deletion of FAM3A reduced vessel contractility and blood pressure levels in mice. Moreover, deletion of FAM3A in VSMC attenuated Ang II (angiotensin II)-induced vascular constriction and remodeling, hypertension, and cardiac hypertrophy in mice. In cultured VSMCs, Ang II activated HSF1 (heat shock factor 1) to stimulate FAM3A expression, activating ATP-P2 receptor pathway to promote the change of VSMCs from contractile phenotype to proliferative phenotype. In the VSMC layer of spontaneously hypertensive rat arteries, Ang II-induced hypertensive mouse arteries and the internal mammary artery of hypertensive patients, HSF1 expression was increased. Treatment with HSF1 inhibitor reduced artery contractility and ameliorated hypertension of spontaneously hypertensive rats. CONCLUSIONS: FAM3A is an important regulator of vascular constriction and blood pressure. Overactivation of HSF1-FAM3A-ATP signaling cascade in VSMCs plays important roles in Ang II-induced hypertension and cardiovascular diseases. Inhibitors of HSF1 could be potentially used to treat hypertension.


Asunto(s)
Cardiomegalia/metabolismo , Citocinas/metabolismo , Hipertensión/metabolismo , Músculo Liso Vascular/metabolismo , Adenosina Trifosfato/metabolismo , Angiotensina II/farmacología , Animales , Arterias/efectos de los fármacos , Arterias/metabolismo , Arterias/fisiopatología , Presión Sanguínea , Cardiomegalia/fisiopatología , Células Cultivadas , Citocinas/genética , Femenino , Eliminación de Gen , Factores de Transcripción del Choque Térmico/metabolismo , Humanos , Hipertensión/fisiopatología , Masculino , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Músculo Liso Vascular/efectos de los fármacos , Músculo Liso Vascular/fisiopatología , Miocitos del Músculo Liso/metabolismo , Ratas , Receptores Purinérgicos P2/metabolismo , Vasoconstricción , Vasoconstrictores/farmacología
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.
J Cell Mol Med ; 24(14): 8138-8150, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32529766

RESUMEN

Recently, the first-line anti-diabetic drug metformin shows versatile protective effects against several diseases and is potentially prescribed to healthy individual for prophylactic use against ageing or other pathophysiological processes. However, for healthy individuals, it remains unclear what effects metformin treatment will induce on their bodies. A systematic profiling of the molecular landscape of metformin treatment is expected to provide crucial implications for this issue. Here, we delineated the first transcriptomic landscape induced by metformin in 10 tissues (aorta, brown adipose, brain, eye, heart, liver, kidney, skeletal muscle, stomach and testis) of healthy mice by using RNA-sequencing technique. A comprehensive computational analysis was performed. The overrepresentation of cardiovascular disease-related gene sets, positive correlation with hypertension-related transcriptomic signatures and the associations of drugs with hypertensive side effect together indicate that although metformin does exert various beneficial effects, it would also increase the risk of hypertension in healthy mice. This prediction was experimentally validated by an independent animal experiments. Together, this study provided important resource necessary for investigating metformin's beneficial/deleterious effects on various healthy tissues, when it is potentially prescribed to healthy individual for prophylactic use.


Asunto(s)
Perfilación de la Expresión Génica , Regulación de la Expresión Génica/efectos de los fármacos , Hipoglucemiantes/farmacología , Metformina/farmacología , Transcriptoma , Animales , Presión Sanguínea/efectos de los fármacos , Presión Sanguínea/genética , Biología Computacional/métodos , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Femenino , Hipertensión/etiología , Hipoglucemiantes/efectos adversos , Masculino , Metformina/efectos adversos , Ratones , Anotación de Secuencia Molecular , Especificidad de Órganos/efectos de los fármacos , Especificidad de Órganos/genética , Factores de Tiempo
4.
J Cell Mol Med ; 23(5): 3464-3475, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30887707

RESUMEN

Family with sequence similarity three member C (FAM3C) (interleukin-like EMT inducer [ILEI]), heat shock factor 1 (HSF1) and Ying-Yang 1 (YY1) have been independently reported to be involved in the pathogenesis of various cancers. However, whether they are coordinated to trigger the development of cancer remains unknown. This study determined the role and mechanism of YY1 and HSF1 in FAM3C-induced proliferation and migration of breast cancer cells. In human MDA-MB-231 breast cancer cell line, transforming growth factor-ß (TGFß) up-regulated FAM3C, HSF1 and YY1 expressions. FAM3C overexpression promoted the proliferation and migration of MDA-MB-231 cells with YY1 and HSF1 up-regulation, whereas FAM3C silencing exerted the opposite effects. FAM3C inhibition repressed TGFß-induced HSF1 activation, and proliferation and migration of breast cancer cells. YY1 was shown to directly activate HSF1 transcription to promote the proliferation and migration of breast cancer cells. YY1 silencing blunted FAM3C- and TGFß-triggered activation of HSF1-Akt-Cyclin D1 pathway, and proliferation and migration of breast cancer cells. Inhibition of HSF1 blocked TGFß-, FAM3C- and YY1-induced proliferation and migration of breast cancer cells. YY1 and HSF1 had little effect on FAM3C expression. Similarly, inhibition of HSF1 also blunted FAM3C- and TGFß-promoted proliferation and migration of human breast cancer BT-549 cells. In human breast cancer tissues, FAM3C, YY1 and HSF1 protein expressions were increased. In conclusion, FAM3C activated YY1-HSF1 signalling axis to promote the proliferation and migration of breast cancer cells. Furthermore, novel FAM3C-YY1-HSF1 pathway plays an important role in TGFß-triggered proliferation and migration of human breast cancer MDA-MB-231 cells.


Asunto(s)
Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Movimiento Celular , Citocinas/metabolismo , Factores de Transcripción del Choque Térmico/metabolismo , Proteínas de Neoplasias/metabolismo , Transducción de Señal , Factor de Crecimiento Transformador beta/farmacología , Factor de Transcripción YY1/metabolismo , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Femenino , Silenciador del Gen , Humanos , Transducción de Señal/efectos de los fármacos , Regulación hacia Arriba/efectos de los fármacos
5.
J Cell Mol Med ; 23(3): 1746-1758, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30488666

RESUMEN

FAM3B, also known as PANcreatic DERived factor (PANDER), promotes gluconeogenesis and lipogenesis in hepatocytes. However, the underlying mechanism(s) still remains largely unclear. This study determined the mechanism of PANDER-induced FOXO1 activation in hepatocytes. In mouse livers and cultured hepatocytes, PANDER protein is located in both the cytoplasm and nucleus. Nuclear PANDER distribution was increased in the livers of obese mice. In cultured mouse and human hepatocytes, PANDER was co-localized with FOXO1 in the nucleus. PANDER directly interacted with FOXO1 in mouse and human hepatocytes. PANDER overexpression enhanced PANDER-FOXO1 interaction, and detained FOXO1 in the nucleus upon insulin stimulation in hepatocytes. With the increase in PANDER-FOXO1 interaction, PANDER overexpression upregulated the expression of gluconeogenic genes and promoted gluconeogenesis in both human and mouse hepatocytes. Luciferase reporter assays further revealed that PANDER augmented the transcriptional activity of FOXO1 on gluconeogenic genes. Moreover, PANDER overexpression also interfered the binding of AS1842856, a specific FOXO1 inhibitor, with FOXO1, and impaired its inhibitory effects on gluconeogenic gene expression and gluconeogenesis in hepatocytes. siRNA mediated-silencing of FOXO1 inhibited PANDER-promoted gluconeogenic gene expression and glucose production in hepatocytes. In conclusion, PANDER protein is abundantly present in the nucleus, where it functions as a new co-activator of FOXO1 to induce gluconeogenic gene expression in hepatocytes.


Asunto(s)
Citocinas/metabolismo , Proteína Forkhead Box O1/metabolismo , Gluconeogénesis , Glucosa/metabolismo , Hepatocitos/metabolismo , Proteínas de Neoplasias/metabolismo , Animales , Células Cultivadas , Citocinas/genética , Proteína Forkhead Box O1/genética , Regulación de la Expresión Génica , Hepatocitos/citología , Humanos , Resistencia a la Insulina , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Obesos , Proteínas de Neoplasias/genética
6.
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
7.
Zhonghua Nei Ke Za Zhi ; 52(5): 403-6, 2013 May.
Artículo en Zh | MEDLINE | ID: mdl-23945307

RESUMEN

OBJECTIVE: To improve the diagnostic ability of leukoencephalopathy with cerebral calcifications and cysts (LCC), a rare central nervous system disease. METHODS: The clinical manifestations, neuroimages and neuropathological features of a 19-year-old male patient were analyzed. A total of 20 cases from 14 literatures were reviewed. RESULT: The patient was admitted with right limb weakness, cognitive decline, headache and blurred eyesight. Head CT scan showed multiple calcifications, cysts formation and leukoencephalopathy. Brain MRI showed several cysts in bilateral hemisphere, basal ganglia, thalamus and paraventricular areas. A mural nodule was noted inside one of the cyst, which was enhanced on the contrasted MRI. The wall of the cysts was partially enhanced, but not with the fluid inside the cysts. The corresponding CT calcifications foci showed on T1 and T2 with either both hyperintensity or both hypointensity, which was also partial enhanced. Extensive leukoencephalopathy was formed around the cysts and the ventricles. But neither Cho nor NAA changed a lot on MRS. Amplitude diagram of SWI series exhibited multiple round small dark signals all over the affected areas with mixed signals showed in the phase diagram, which indicated both calcifications and microbleeding at the lesions. Neuropathological examinations found no tumor cells in the operated cyst, and showed angiomatous small blood cells were dominant in the cyst wall. Hyaline degenerations, microcalcifications and hemosiderin deposition were observed. No obvious demyelination was discovered, while gliosis, numerous Rosenthal fibers and fibrinoid vascular necrosis were found around the lesions. The clinical, neuroimaging and pathological features of this patient were in accordance with the cases reported in the literatures. CONCLUSIONS: Neuroimaging is the most important method for the diagnosis of LCC. As small vessel lesions are probably closely related to the pathophysiology of LCC, SWI could be recommended to further reveal the etiology of LCC.


Asunto(s)
Leucoencefalopatías , Calcinosis/patología , Quistes/patología , Humanos , Leucoencefalopatías/diagnóstico , Leucoencefalopatías/patología , Masculino , Adulto Joven
8.
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
9.
Exp Clin Endocrinol Diabetes ; 130(8): 498-508, 2022 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-34592773

RESUMEN

FAM3A is a recently identified mitochondrial protein that stimulates pancreatic-duodenal homeobox 1 (PDX1) and insulin expressions by promoting ATP release in islet ß cells. In this study, the role of intracellular ATP in FAM3A-induced PDX1 expression in pancreatic ß cells was further examined. Acute FAM3A inhibition using siRNA transfection in mouse pancreatic islets significantly reduced PDX1 expression, impaired insulin secretion, and caused glucose intolerance in normal mice. In vitro, FAM3A overexpression elevated both intracellular and extracellular ATP contents and promoted PDX1 expression and insulin secretion. FAM3A-induced increase in cellular calcium (Ca2+) levels, PDX1 expression, and insulin secretion, while these were significantly repressed by inhibitors of P2 receptors or the L-type Ca2+ channels. FAM3A-induced PDX1 expression was abolished by a calmodulin inhibitor. Likewise, FAM3A-induced ß-cell proliferation was also inhibited by a P2 receptor inhibitor and an L-type Ca2+ channels inhibitor. Both intracellular and extracellular ATP contributed to FAM3A-induced PDX1 expression, insulin secretion, and proliferation of pancreatic ß cells.


Asunto(s)
Adenosina Trifosfato/metabolismo , Proteínas de Homeodominio/metabolismo , Células Secretoras de Insulina , Transducción de Señal , Transactivadores/metabolismo , Animales , Citocinas/metabolismo , Glucosa/metabolismo , Proteínas de Homeodominio/genética , Insulina/metabolismo , Células Secretoras de Insulina/metabolismo , Ratones , Regulación hacia Arriba
10.
J Pathol ; 218(3): 327-36, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19291713

RESUMEN

We report a new in situ tissue slide-based SELEX strategy targeting neoplastic tissues from breast cancer patients. The methodology, using the molecular differences between clinical specimens, can evolve aptamers to all fractions of tissue. The aptamers may be used as new molecular probes for pathological diagnosis and tumour imaging, and also to reveal the molecular differences that are responsible for the diseases. The specific aptamers were enriched by unequal length strand PCR employing a structured (-) strand primer. After 12 rounds of selection, using the paraffin tissue sections from infiltrating ductal carcinomas as targets, and using the adjacent normal tissue from the same case as controls, one of the enriched ssDNA aptamers, BC15, was selected from a nucleic acid library and characterized as recognizing breast cancer cells either within the tissue sections or from the culture medium, but only weakly binding to adjacent normal cells or immortalized breast cell line MCF10A. The calculated equilibrium dissociation constants (K(d)) of BC15 bound to MCF7 cells was 111.0 +/- 36.9 nM. Through streptavidin magnetic beads mediated affinity purification assay followed by mass spectrometry identification and western blot confirmation, the target of BC15 was characterized to be hnRNP A1, which was further verified to be specifically and highly expressed in cancerous tissues of breast by hnRNP A1 antibody immunostaining as well as western blot. BC15 aptamer was also used to probe cancer cells in tissues from other pathological types of breast cancers including lobular carcinoma, ductal carcinoma complicated with lobular carcinoma, comedo carcinoma, and lymph node metastasis of breast ductal carcinoma origin or breast lobular carcinoma origin. Therefore, tissue slide-based SELEX holds promise in identifying tumour markers and developing specific molecular probes for cancer diagnosis.


Asunto(s)
Aptámeros de Nucleótidos/genética , Biomarcadores de Tumor/genética , Neoplasias de la Mama/genética , Carcinoma Ductal de Mama/genética , Ribonucleoproteína Heterogénea-Nuclear Grupo A-B/genética , Secuencia de Aminoácidos , Biomarcadores de Tumor/metabolismo , Biopsia , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Carcinoma Ductal de Mama/metabolismo , Carcinoma Ductal de Mama/patología , ADN de Neoplasias/genética , Femenino , Biblioteca de Genes , Ribonucleoproteína Nuclear Heterogénea A1 , Ribonucleoproteína Heterogénea-Nuclear Grupo A-B/metabolismo , Humanos , Microscopía Confocal , Datos de Secuencia Molecular , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Técnica SELEX de Producción de Aptámeros/métodos , Células Tumorales Cultivadas
11.
Zhonghua Bing Li Xue Za Zhi ; 39(2): 100-5, 2010 Feb.
Artículo en Zh | MEDLINE | ID: mdl-20388375

RESUMEN

OBJECTIVES: To investigate molecular mechanisms of PAR-1 regulation on intracellular Ca²(+) mobilization in lung giant cell carcinoma cells in vitro and its involvement in tumor metastasis. METHODS: Free intracellular Ca²(+) ([Ca²(+)]i) was measured in lung giant cell carcinoma PLA801C and PLA801D cells by confocal microscopy. Sense and anti-sense PAR-1 expression vectors were transfected into PLA801C (C+)and PLA801D(D-) cells, respectively. The effects of PAR-1 expression were investigated by thrombin and TRAP-induced mobilization of [Ca²(+)]i in the C+ and D-cells. RESULTS: There were significant differences of the mean values of [Ca²(+)]i between PLA801D (59.55) and PLA801C cells (35.46, P < 0.01). The mean [Ca²(+)]i of C+ cells (45.77) was significantly higher than that of its control CV cells (35.46, P < 0.05), and the mean [Ca²(+)]i of D-cells (48.42) was significantly lower than that of its control DV cells (59.55, P < 0.05). The peaks of [Ca²(+)]i of C+ and CV cells were 48.19 ± 9.84 and 45.64 ± 9.87 (P < 0.05) respectively at 80 s and 100 s after thrombin treatment, but were 111.31 ± 25.00 and 52.93 ± 11.21 (P < 0.05) respectively at 60 s after TRAP treatment. The peaks of [Ca²(+)]i of D- and DV cells were 40.71 ± 5.89 and 61.07 ± 21.36 (P < 0.05) respectively at 60 s after thrombin treatment, but were 84.98 ± 11.23 and 102.58 ± 21.48 (P < 0.05) respectively at 40 s after TRAP treatment. CONCLUSIONS: The high metastatic potential of PLA801D and PLA801C may be related to [Ca²(+)]i of the tumor cells. PAR-1 may play an important role in the metastasis of lung giant cell carcinoma cells by up-regulating the intracellular Ca²(+).


Asunto(s)
Calcio/metabolismo , Carcinoma de Células Gigantes/metabolismo , Neoplasias Pulmonares/metabolismo , Receptor PAR-1/metabolismo , Señalización del Calcio/efectos de los fármacos , Carcinoma de Células Gigantes/patología , Línea Celular Tumoral , ADN sin Sentido/genética , Humanos , Neoplasias Pulmonares/patología , ARN Mensajero/metabolismo , Receptor PAR-1/genética , Receptor PAR-1/fisiología , Receptores de Trombina/metabolismo , Trombina/farmacología , Transfección , Regulación hacia Arriba
12.
Diabetes ; 69(6): 1126-1139, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32312868

RESUMEN

Mitochondrial protein FAM3A suppresses hepatic gluconeogenesis and lipogenesis. This study aimed to screen drug(s) that activates FAM3A expression and evaluate its effect(s) on hyperglycemia and steatosis. Drug-repurposing methodology predicted that antidepressive drug doxepin was among the drugs that potentially activated FAM3A expression. Doxepin was further validated to stimulate the translocation of transcription factor HNF4α from the cytoplasm into the nucleus, where it promoted FAM3A transcription to enhance ATP synthesis, suppress gluconeogenesis, and reduce lipid deposition in hepatocytes. HNF4α antagonism or FAM3A deficiency blunted doxepin-induced suppression on gluconeogenesis and lipid deposition in hepatocytes. Doxepin administration attenuated hyperglycemia, steatosis, and obesity in obese diabetic mice with upregulated FAM3A expression in liver and brown adipose tissues (BAT). Notably, doxepin failed to correct dysregulated glucose and lipid metabolism in FAM3A-deficient mice fed on high-fat diet. Doxepin's effects on ATP production, Akt activation, gluconeogenesis, and lipogenesis repression were also blunted in FAM3A-deficient mouse livers. In conclusion, FAM3A is a therapeutic target for diabetes and steatosis. Antidepressive drug doxepin activates FAM3A signaling pathways in liver and BAT to improve hyperglycemia and steatosis of obese diabetic mice. Doxepin might be preferentially recommended as an antidepressive drug in potential treatment of patients with diabetes complicated with depression.


Asunto(s)
Citocinas/metabolismo , Doxepina/uso terapéutico , Reposicionamiento de Medicamentos , Hígado Graso/tratamiento farmacológico , Regulación de la Expresión Génica/efectos de los fármacos , Hiperglucemia/tratamiento farmacológico , Animales , Antidepresivos Tricíclicos/uso terapéutico , Citocinas/genética , Bases de Datos Farmacéuticas , Proteína Forkhead Box O1/metabolismo , Células Hep G2 , Factor Nuclear 4 del Hepatocito/genética , Factor Nuclear 4 del Hepatocito/metabolismo , Hepatocitos/metabolismo , Humanos , Masculino , Ratones , Ratones Noqueados
13.
Oncol Rep ; 21(3): 697-706, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19212629

RESUMEN

Tumor metastasis, the important characteristic of malignant tumors, is closely associated with a series of changes in the expressions of genes and proteins. A novel gene MAG-2, which may have close correlation with lung cancer metastasis, was identified in our laboratory through an approach of suppressed subtractive hybridization using lung cancer cell strains with the same origin but different metastatic potential as models. The relations between MAG-2 gene and aspects of cancer metastasis including invasion, mobility, anchorage-independent growth capability and adherence to ECM, were investigated in our experiment models. MAG-2 gene was proved to be genuine and have mRNA and deduced proteins from itself by methods of expression profile analysis and fluorescence staining. Cytological experiments had demonstrated that hyper- or hypo-expressing of MAG-2 by gene transfection or RNAi leads to significant increase or decrease in the metastatic ability of cancer cells. In addition, CD44, MMP-2 and free calcium ion concentration intracellularly, were proved to be metastasis promoting factors, and found to be regulated by MAG-2 in lung cancer cells, this might be the mechanism of the metastasis promoting function of MAG-2 gene. The positive rate of MAG-2 mRNA was found to be significantly higher in tumor tissue from patients with metastatic lung cancer than tissues from patients with non-metastatic lung cancer. These data suggest that MAG-2 may be a novel causal gene for lung cancer invasion and metastasis.


Asunto(s)
Calcio/metabolismo , Adhesión Celular/genética , Receptores de Hialuranos/metabolismo , Neoplasias Pulmonares/genética , Metaloproteinasa 2 de la Matriz/metabolismo , Metástasis de la Neoplasia/genética , Secuencia de Aminoácidos , Western Blotting , Línea Celular Tumoral , Movimiento Celular/genética , Técnica del Anticuerpo Fluorescente , Perfilación de la Expresión Génica , Humanos , Immunoblotting , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Microscopía Confocal , Datos de Secuencia Molecular , Invasividad Neoplásica/genética , Interferencia de ARN , ARN Mensajero/análisis , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
14.
Front Microbiol ; 9: 1336, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29988362

RESUMEN

In recent years, the first-line anti-diabetic drug metformin has been shown to be also useful for the treatment of other diseases like cancer. To date, few reports were about the impact of metformin on gut microbiota. To fully understand the mechanism of action of metformin in treating diseases other than diabetes, it is especially important to investigate the impact of long-term metformin treatment on the gut microbiome in non-diabetic status. In this study, we treated healthy mice with metformin for 30 days, and observed 46 significantly changed gut microbes by using the 16S rRNA-based microbiome profiling technique. We found that microbes from the Verrucomicrobiaceae and Prevotellaceae classes were enriched, while those from Lachnospiraceae and Rhodobacteraceae were depleted. We further compared the altered microbiome profile with the profiles under various disease conditions using our recently developed comparative microbiome tool known as MicroPattern. Interestingly, the treatment of diabetes patients with metformin positively correlates with colon cancer and type 1 diabetes, indicating a confounding effect on the gut microbiome in patients with diabetes. However, the treatment of healthy mice with metformin exhibits a negative correlation with multiple inflammatory diseases, indicating a protective anti-inflammatory role of metformin in non-diabetes status. This result underscores the potential effect of metformin on gut microbiome homeostasis, which may contribute to the treatment of non-diabetic diseases.

15.
Front Genet ; 9: 380, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30356729

RESUMEN

Measuring the essentiality of genes is critically important in biology and medicine. Here we proposed a computational method, GIC (Gene Importance Calculator), which can efficiently predict the essentiality of both protein-coding genes and long noncoding RNAs (lncRNAs) based on only sequence information. For identifying the essentiality of protein-coding genes, GIC outperformed well-established computational scores. In an independent mouse lncRNA dataset, GIC also achieved an exciting performance (AUC = 0.918). In contrast, the traditional computational methods are not applicable to lncRNAs. Moreover, we explored several potential applications of GIC score. Firstly, we revealed a correlation between gene GIC score and research hotspots of genes. Moreover, GIC score can be used to evaluate whether a gene in mouse is representative for its homolog in human by dissecting its cross-species difference. This is critical for basic medicine because many basic medical studies are performed in animal models. Finally, we showed that GIC score can be used to identify candidate genes from a transcriptomics study. GIC is freely available at http://www.cuilab.cn/gic/.

16.
Metabolism ; 81: 71-82, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29221790

RESUMEN

Non-alcoholic fatty liver disease (NAFLD) and diabetes are severe public health issues worldwide. The Family with sequence similarity 3 (FAM3) gene family consists of four members designated as FAM3A, FAM3B, FAM3C and FAM3D, respectively. Recently, there had been increasing evidence that FAM3A, FAM3B and FAM3C are important regulators of glucose and lipid metabolism. FAM3A expression is reduced in the livers of diabetic rodents and NAFLD patients. Hepatic FAM3A restoration activates ATP-P2 receptor-Akt and AMPK pathways to attenuate steatosis and hyperglycemia in obese diabetic mice. FAM3C expression is also reduced in the liver under diabetic condition. FAM3C is a new hepatokine that activates HSF1-CaM-Akt pathway and represses mTOR-SREBP1-FAS pathway to suppress hepatic gluconeogenesis and lipogenesis. In contrast, hepatic expression of FAM3B, also called PANDER, is increased under obese state. FAM3B promotes hepatic lipogenesis and gluconeogenesis by repressing Akt and AMPK activities, and activating lipogenic pathway. Under obese state, the imbalance among hepatic FAM3A, FAM3B and FAM3C signaling networks plays important roles in the pathogenesis of NAFLD and type 2 diabetes. This review briefly discussed the latest research progress on the roles and mechanisms of FAM3A, FAM3B and FAM3C in the regulation of hepatic glucose and lipid metabolism.


Asunto(s)
Citocinas/fisiología , Diabetes Mellitus Tipo 2/metabolismo , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Animales , Citocinas/sangre , Diabetes Mellitus Tipo 2/etiología , Glucosa/metabolismo , Humanos , Resistencia a la Insulina , Metabolismo de los Lípidos , Hígado/metabolismo , Proteínas de Neoplasias/sangre , Enfermedad del Hígado Graso no Alcohólico/etiología , PPAR gamma/fisiología , Transducción de Señal
17.
Medicine (Baltimore) ; 97(26): e11136, 2018 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-29952957

RESUMEN

To investigate the relationship of the expression of vascular endothelial growth factor (VEGF)/vascular endothelial growth factor receptor-2 (VEGFR-2) and imaging features with the therapeutic efficacy of Phosphorus-32 colloid interstitial radiotherapy in recurrent craniopharyngioma.Thirty-two patients with recurrent craniopharyngioma underwent phosphorus-32 colloid interstitial radiotherapy. The tumor imaging features were classified into 4 types according to the thickness of the cyst wall and signals of the cyst contents as shown by computed tomography (CT) and magnetic resonance imaging (MRI) images. Protein expressions of VEGF and VEGFR-2 in craniopharyngioma tissues were evaluated with immunohistochemistry before radiotherapy. The tumor radiosensitivity was determined at 12 months after the interstitial radiotherapy.VEGF mainly expressed in the tumor cytoplasm, and VEGFR-2 expressed either in vascular endothelial cells or in tumor endothelial cells. VEGF/VEGFR-2 expressions varied significantly in cases sensitive or insensitive to the radiotherapy (VEGF: P = .028; VEGFR-2: P = .017). Tumor imaging features were associated with the therapeutic efficacy of interstitial radiotherapy (P = .000). VEGF expression had no association with the imaging features of tumors (P = .226), but VEGFR-2 expression was associated with the imaging features of tumors (P = .008).Our results confirmed the association among imaging features, VEGFR-2 expressions, and tumor radiosensitivity in craniopharyngiomas. Imaging features and VEGFR-2 expressions may add useful data to the radiosensitive assessment of craniopharyngiomas.


Asunto(s)
Braquiterapia/métodos , Craneofaringioma/radioterapia , Recurrencia Local de Neoplasia/radioterapia , Radioisótopos de Fósforo/uso terapéutico , Neoplasias Hipofisarias/radioterapia , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo , Factores de Crecimiento Endotelial Vascular/metabolismo , Adolescente , Adulto , Anciano , Niño , Preescolar , Craneofaringioma/clasificación , Craneofaringioma/diagnóstico por imagen , Craneofaringioma/metabolismo , Femenino , Humanos , Imagen por Resonancia Magnética/métodos , Masculino , Persona de Mediana Edad , Recurrencia Local de Neoplasia/diagnóstico por imagen , Recurrencia Local de Neoplasia/metabolismo , Radioisótopos de Fósforo/administración & dosificación , Neoplasias Hipofisarias/clasificación , Neoplasias Hipofisarias/diagnóstico por imagen , Neoplasias Hipofisarias/metabolismo , Tolerancia a Radiación/efectos de la radiación , Estudios Retrospectivos , Tomografía Computarizada por Rayos X/métodos , Resultado del Tratamiento , Adulto Joven
18.
Diabetes ; 67(4): 581-593, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29382663

RESUMEN

Mammalian genomes encode a huge number of long noncoding RNAs (lncRNAs) with unknown functions. This study determined the role and mechanism of a new lncRNA, lncRNA suppressor of hepatic gluconeogenesis and lipogenesis (lncSHGL), in regulating hepatic glucose/lipid metabolism. In the livers of obese mice and patients with nonalcoholic fatty liver disease, the expression levels of mouse lncSHGL and its human homologous lncRNA B4GALT1-AS1 were reduced. Hepatic lncSHGL restoration improved hyperglycemia, insulin resistance, and steatosis in obese diabetic mice, whereas hepatic lncSHGL inhibition promoted fasting hyperglycemia and lipid deposition in normal mice. lncSHGL overexpression increased Akt phosphorylation and repressed gluconeogenic and lipogenic gene expression in obese mouse livers, whereas lncSHGL inhibition exerted the opposite effects in normal mouse livers. Mechanistically, lncSHGL recruited heterogeneous nuclear ribonucleoprotein A1 (hnRNPA1) to enhance the translation efficiency of CALM mRNAs to increase calmodulin (CaM) protein level without affecting their transcription, leading to the activation of the phosphatidyl inositol 3-kinase (PI3K)/Akt pathway and repression of the mTOR/SREBP-1C pathway independent of insulin and calcium in hepatocytes. Hepatic hnRNPA1 overexpression also activated the CaM/Akt pathway and repressed the mTOR/SREBP-1C pathway to ameliorate hyperglycemia and steatosis in obese mice. In conclusion, lncSHGL is a novel insulin-independent suppressor of hepatic gluconeogenesis and lipogenesis. Activating the lncSHGL/hnRNPA1 axis represents a potential strategy for the treatment of type 2 diabetes and steatosis.


Asunto(s)
Diabetes Mellitus Tipo 2/genética , Galactosiltransferasas/metabolismo , Gluconeogénesis/genética , Ribonucleoproteína Nuclear Heterogénea A1/genética , Lipogénesis/genética , Hígado/metabolismo , Enfermedad del Hígado Graso no Alcohólico/metabolismo , ARN Largo no Codificante/genética , Animales , Glucemia/metabolismo , Calcio/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Modelos Animales de Enfermedad , Técnicas de Silenciamiento del Gen , Glucosa/metabolismo , Hepatocitos/metabolismo , Humanos , Insulina/metabolismo , Metabolismo de los Lípidos , Ratones , Ratones Obesos , Proteínas de Ensamble de Clatrina Monoméricas/genética , Proteínas de Ensamble de Clatrina Monoméricas/metabolismo , Fosfatidilinositol 3-Quinasa/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , ARN Mensajero/metabolismo , Transducción de Señal , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/metabolismo , Serina-Treonina Quinasas TOR/metabolismo
19.
Zhonghua Bing Li Xue Za Zhi ; 36(1): 19-23, 2007 Jan.
Artículo en Zh | MEDLINE | ID: mdl-17374233

RESUMEN

OBJECTIVE: To study the histopathologic features, differential diagnosis and prognosis of epithelioid angiomyolipoma (EAML) of kidney. METHODS: Two cases of EAML (including one case with recurrence) of kidney were retrieved from the archival files of Departments of Pathology, Navy General Hospital of PLA and Health Science Center of Peking University. The clinicopathologic features, immunohistochemistry, ultrastructural findings and follow-up data were studied and literature reviewed. RESULTS: Histologically, the tumors were predominantly composed of epithelioid cells with marked cellular pleomorphism. Focal perivascular arrangement was seen. Hemorrhage and necrosis were present and tumor emboli were found in the venous structures. The renal hilar lymph nodes were also involved by tumor cells. Immunohistochemical study showed that the tumor cells (including those in the hilar lymph nodes) were strongly and diffusely positive for HMB45, smooth muscle actin, neuron-specific enolase and vimentin. They were focally positive for S-100 protein, melan-pan and CD68. The staining for epithelial membrane antigen, AE1/3, CK7, CD117, muscle-specific actin, desmin, leukocyte common antigen, CD20, CD45RO, CD30, CD15, chromogranin A, synaptophysin, bcl-2, estrogen receptor, progesterone receptor and p53 were negative. Ultrastructural examination revealed the presence of melanosome-like dense granules, myofilaments and dense bodies in the tumor cell cytoplasm. Discontinuous and focally thickened basal lamina was seen surrounding the tumor cells. On follow up, both patients remained well and disease-free 10 months after operation. CONCLUSIONS: EAML is predominantly or almost entirely composed of epithelioid cells. Perivascular cellular arrangement, focal features of otherwise classic angiomyolipoma, as well as coexpression of HMB-45 and smooth muscle actin are clues to the correct diagnosis. The degree of cytologic atypia, presence of hemorrhage and necrosis and high mitotic activity may indicate the malignant potential of this tumor. On the other hand, the presence of lymph node involvement and even tumor emboli in renal veins may represent multifocaltumorigenicity rather than true malignancy. Definitive evidence of malignancy requires demonstration of distant metastasis.


Asunto(s)
Angiomiolipoma/patología , Neoplasias Renales/patología , Adulto , Angiomiolipoma/metabolismo , Angiomiolipoma/cirugía , Antígenos de Neoplasias/metabolismo , Diagnóstico Diferencial , Células Epitelioides/patología , Células Epitelioides/ultraestructura , Humanos , Inmunohistoquímica , Riñón/patología , Riñón/ultraestructura , Neoplasias Renales/metabolismo , Neoplasias Renales/cirugía , Masculino , Antígenos Específicos del Melanoma , Microscopía Electrónica , Proteínas de Neoplasias/metabolismo
20.
Zhonghua Bing Li Xue Za Zhi ; 36(5): 313-7, 2007 May.
Artículo en Zh | MEDLINE | ID: mdl-17706138

RESUMEN

OBJECTIVE: To study the functional aspects of protease-activated receptor 1 (PAR-1) gene involved in tumor metastasis. METHODS: Two human lung giant cell carcinoma cell lines PLA801C (low metastasis potential) and PLA801D (high metastasis potential) were chosen as in-vitro human cancer model systems. Sense and anti-sense expression constructs of PAR-1 gene (pC/PAR1s and pC/PAR1as) were transfected into PLA-801C and PLA-801D cells by lipofection. PAR-1 expression was determined by RT-PCR and western blot analysis. MTT growth, flow cytometry analysis, fibronectin adhesion, and matrigel invasion assays were used to study the effect of PAR-1 expression on the proliferation, adhesion, and invasion of the transfected cells. RESULTS: Appropriate up-regulation or down-regulation of protein expression of PAR-1 was observed in both transfected cell lines (PLA801C and PLA801D) to express PAR-1s or PAR-1as, respectively. Expression of the sense PAR-1 markedly increased cellular proliferation, adhesion and invasion of PLA-801C cells. In contrast, anti-sense PAR-1 significantly inhibited cell growth, adhesion and invasion capabilities, along with cell arrest at G0/G1 phase of the PLA-801D cells. CONCLUSIONS: Successful up- and down- regulation of expression of PAR-1 can be achieved by in-vitro transfection of sense and antisense PAR-1 constructs. PAR-1 may enhance metastasis of lung cancer through its regulation of cellular proliferation, adhesion and invasion. Down-regulation of expression of PAR-1 may provide a new therapeutic strategy against lung carcinoma.


Asunto(s)
Carcinoma de Células Gigantes/patología , Adhesión Celular , Proliferación Celular , Neoplasias Pulmonares/patología , Receptor PAR-1/genética , Carcinoma de Células Gigantes/metabolismo , Ciclo Celular , Línea Celular Tumoral , ADN sin Sentido , Regulación hacia Abajo , Regulación Neoplásica de la Expresión Génica , Humanos , Neoplasias Pulmonares/metabolismo , Invasividad Neoplásica , ARN Mensajero/metabolismo , Receptor PAR-1/metabolismo , Transfección
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA