Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Neural Regen Res ; 18(12): 2727-2732, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37449637

RESUMO

Fidgetin, a microtubule-severing enzyme, regulates neurite outgrowth, axonal regeneration, and cell migration by trimming off the labile domain of microtubule polymers. Because maintenance of the microtubule labile domain is essential for axon initiation, elongation, and navigation, it is of interest to determine whether augmenting the microtubule labile domain via depletion of fidgetin serves as a therapeutic approach to promote axonal regrowth in spinal cord injury. In this study, we constructed rat models of spinal cord injury and sciatic nerve injury. Compared with spinal cord injury, we found that expression level of tyrosinated microtubules in the labile portion of microtubules continuously increased, whereas fidgetin decreased after peripheral nerve injury. Depletion of fidgetin enhanced axon regeneration after spinal cord injury, whereas expression level of end binding protein 3 (EB3) markedly increased. Next, we performed RNA interference to knockdown EB3 or fidgetin. We found that deletion of EB3 did not change fidgetin expression. Conversely, deletion of fidgetin markedly increased expression of tyrosinated microtubules and EB3. Deletion of fidgetin increased the amount of EB3 at the end of neurites and thereby increased the level of tyrosinated microtubules. Finally, we deleted EB3 and overexpressed fidgetin. We found that fidgetin trimmed tyrosinated tubulins by interacting with EB3. When fidgetin was deleted, the labile portion of microtubules was elongated, and as a result the length of axons and number of axon branches were increased. These findings suggest that fidgetin can be used as a novel therapeutic target to promote axonal regeneration after spinal cord injury. Furthermore, they reveal an innovative mechanism by which fidgetin preferentially severs labile microtubules.

2.
Metab Brain Dis ; 38(7): 2369-2381, 2023 10.
Artigo em Inglês | MEDLINE | ID: mdl-37256467

RESUMO

Neuropsychiatric disorders have a high incidence worldwide. Kinesins, a family of microtubule-based molecular motor proteins, play essential roles in intracellular and axonal transport. Variants of kinesins have been found to be related to many diseases, including neurodevelopmental/neurodegenerative disorders. Kinesin-12 (also known as Kif15) was previously found to affect the frequency of both directional microtubule transports. However, whether Kif15 deficiency impacts mood in mice is yet to be investigated. In this study, we used the CRISPR/Cas9 method to obtain Kif15-/- mice. In behavioral tests, Kif15-/- female mice exhibited prominent depressive characteristics. Further studies showed that the expression of BDNF was significantly decreased in the frontal cortex, corpus callosum, and hippocampus of Kif15-/- mice, along with the upregulation of Interleukin-6 and Interleukin-1ß in the corpus callosum. In addition, the expression patterns of AnkG were notably changed in the developing brain of Kif15-/- mice. Based on our previous studies, we suggested that this appearance of altered AnkG was due to the maladjustment of the microtubule patterns induced by Kif15 deficiency. The distribution of PSD95 in neurites notably decreased after cultured neurons treated with the Kif15 inhibitor, but total PSD95 protein level was not impacted, which revealed that Kif15 may contribute to PSD95 transportation. This study suggested that Kif15 may serve as a potential target for future depression studies.


Assuntos
Depressão , Cinesinas , Animais , Feminino , Camundongos , Depressão/genética , Cinesinas/genética , Cinesinas/metabolismo , Microtúbulos/metabolismo , Neurônios/metabolismo
3.
Biochem Biophys Res Commun ; 655: 110-117, 2023 05 07.
Artigo em Inglês | MEDLINE | ID: mdl-36934586

RESUMO

Kif15, also name kinesin-12, is a microtubule (MT) associate protein, which functions as a regulator of MT-dependent transport or spindle organization. Previous studies reported Kif15 increases in many tumors, however the effect of host Kif15 gene lack on tumor growth is not investigated. In this study, CRISPR/Cas9 mediated Kif15 gene knockout (Kif15-/-) mice were established and HE (Hematoxylin-Eosin) assay revealed no significant differences of morphology in most adult tissues (heart, liver, lung, kidney, and brain) except a retarded development of spleen in adult Kif15-/- mice. RNA sequence analysis of adult spleen tissues of Kif15-/- and Kif15+/+ mice was performed, and the results revealed that a total of 438 mRNAs were significantly differentially expressed in Kif15 knockout spleen, showing the top biological process was immune system process. FCM (Flow Cytometry) assay showed the percentage of CD8+ T lymphocytes notably increased in spleens of 9 w and 12 w old Kif15-/- mice. The CD8+ T lymphocytes are cytotoxic effector cells fighting against tumor. We thus detected the tumor growth in Kif15-/- mice using the melanoma cells inoculated subcutaneously. The tumor size significantly reduced in Kif15-/- mice. We finally detected whether Kif15 dysfunction affects the phagocytic function of macrophages on tumor cells, and the result showed Kif15 inhibitor treated macrophages significantly promoted the phagocytosis in vitro. In summary, this study revealed that the tumor-bearing mice of Kif15 gene deficiency notably inhibited tumor growth due to innate immune activation, which was the first report of the relation of Kif15 on the immunoreactivity.


Assuntos
Neoplasias , Linfócitos T , Animais , Camundongos , Linfócitos T CD8-Positivos/metabolismo , Cinesinas/genética , Cinesinas/metabolismo , Microtúbulos/metabolismo , Neoplasias/metabolismo , Linfócitos T/metabolismo
4.
Exp Neurol ; 361: 114315, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36586551

RESUMO

Neurons require a constant increase in protein synthesis during axonal growth and regeneration. AKT-mTOR is a central pathway for mammalian cell survival and regeneration. Fidgetin (Fign) is an ATP-dependent microtubule (MT)-severing enzyme whose functions are associated with neurite outgrowth, axon regeneration and cell migration. Although most previous studies have indicated that depletion of Fign is involved in those biological activities by increasing labile MT mass, it remains unknown whether mTOR activation contributes to this process. Here, we showed that depletion of Fign enhanced p-mTOR/p-S6K activation, and the mTOR inhibitor Rapamycin inhibited axon outgrowth and p-rpS6 activation. We then investigated the effects of neuronal-specific Fign deletion in a rat spinal cord hemisection model by injecting syn-GFP Fign shRNA virus. BBB values revealed an improvement in functional recovery. The p-mTOR was activated along with neuronal Fign depletion. The syn-mCherry virus showed more sprouting neurites entering the injury region, which was confirmed by immunostaining GAP43 protein. Further, we showed that Fign siRNA treatment promoted axon outgrowth and branching, whose underlying mechanism was firstly attributed to local activation of the mTOR pathway, and increased MT dynamicity. Finally, considering L-leucine, promotes axonal growth and neuronal survival, we applied L-leucine with Fign depletion after spinal cord injury or in chondroitin sulfate proteoglycan inhibitory molecules. The phenomenon of synergistically augmented axon regeneration was observed. In summary, our results indicated a novel local mTOR pathway for fidgetin to impact axon growth and provided a combined strategy in SCI.


Assuntos
Axônios , Traumatismos da Medula Espinal , Ratos , Animais , Axônios/fisiologia , Regeneração Nervosa/fisiologia , Leucina/metabolismo , Leucina/farmacologia , Neurônios/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Mamíferos
5.
Metabolism ; 139: 155372, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-36470472

RESUMO

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.


Assuntos
Diabetes Mellitus Experimental , Hepatopatia Gordurosa não Alcoólica , Camundongos , Animais , Hepatopatia Gordurosa não Alcoólica/metabolismo , Glucose , Homeostase , Trifosfato de Adenosina/metabolismo , Citocinas/metabolismo
6.
Exp Neurol ; 347: 113905, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34699790

RESUMO

Circular RNAs (circRNAs) are important for the development and regeneration of the nervous system. We investigated the differential expression profiles of circRNA induced by spinal cord injury and reported that circRNA_01477 facilitates spinal astrocyte proliferation and migration after injury in rats. In this study, we further clarified the function and possible mechanism of action of circRNA_01477 in neurons. Fluorescence in situ hybridization assay revealed that circRNA_01477 is mainly localized in the neuronal cytoplasm. Knockdown of circRNA_01477 significantly increased axonal length. The circRNA_01477/microRNAs (miRNA)/messenger RNA (mRNA) interaction network was investigated using RNA sequencing. miRNA-3075 showed a remarkable increase after circRNA_01477 depletion, and either overexpression of miRNA-3075 or downregulation of its target gene FosB significantly promoted axonal growth. Luciferase reporter assay showed that miRNA-3075 could directly bind to the 3'UTR of FosB and negatively regulated FosB transcription. Dual silencing of circRNA_01477 and miR-3075 revealed that miR-3075 inhibition rescued the increased axon length caused by siCircRNA_01477. Finally, we verified that the Stat3 pathway was activated after FosB protein depletion in rat spinal neurons, while the NF-κB pathway was not altered. In summary, our study is the first to report that circRNA_01477 contributes to axon growth by functioning as miRNA sponge by regulating the miRNA-3075/FosB/Stat3 axis.


Assuntos
Axônios/metabolismo , MicroRNAs/metabolismo , Proteínas Proto-Oncogênicas c-fos/metabolismo , RNA Circular/metabolismo , Fator de Transcrição STAT3/metabolismo , Animais , Células Cultivadas , Feminino , Gravidez , Ligação Proteica/fisiologia , Ratos , Ratos Sprague-Dawley , Nervos Espinhais/citologia , Nervos Espinhais/metabolismo
7.
Anal Bioanal Chem ; 413(8): 2235-2246, 2021 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-33608751

RESUMO

Lateral flow immunoassays (LFIs) can be used to detect intact bacteria or spores; when gold nanoparticles (AuNPs) are used as the signal reporters, the detection limits are very low. Spore-based surface display has been widely studied for enzyme immobilization and live-nontoxic oral vaccines. In this study, recombinant spores were used to improve the sensitivity of a LFI. We developed a test kit that combines streptavidin-displayed spores with a LFI assay for rapid protein detection. The recombinant spores served as a signal amplifier and AuNPs were used as the signal reporters. For detection of ß-galactosidase, which was used as the model protein, the detection limit was about 10-15 mol, while that of the conventional LFI is about 10-12 mol. In both methods, nanogold was used as the colorimetric signal and could be observed with the naked eye. This method improved LFI sensitivity without sacrificing its advantages. Furthermore, enhanced green fluorescent protein (eGFP) was also displayed on the surface of the streptavidin-displayed spores. Without AuNPs, the fluorescent recombinant spores acted as the signal, which could be detected by a fluorescence detector, such as a fluorescence microscope. The detection limit was 10-16 mol under fluorescence microscopy whose magnification was 25-fold. Therefore, in conclusion, in this proof of concept study, the detection limits of both proposed methods were far superior to those of traditional LFI assay.


Assuntos
Bacillus subtilis/química , Técnicas Biossensoriais/métodos , Ouro/química , Nanopartículas Metálicas/química , Proteínas/análise , Esporos Bacterianos/química , Proteínas de Fluorescência Verde/química , Imunoensaio/métodos , Limite de Detecção , Estreptavidina/química
8.
Diabetes ; 69(6): 1126-1139, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32312868

RESUMO

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.


Assuntos
Citocinas/metabolismo , Doxepina/uso terapêutico , Reposicionamento de Medicamentos , Fígado Gorduroso/tratamento farmacológico , Regulação da Expressão Gênica/efeitos dos fármacos , Hiperglicemia/tratamento farmacológico , Animais , Antidepressivos Tricíclicos/uso terapêutico , Citocinas/genética , Bases de Dados de Produtos Farmacêuticos , Proteína Forkhead Box O1/metabolismo , Células Hep G2 , Fator 4 Nuclear de Hepatócito/genética , Fator 4 Nuclear de Hepatócito/metabolismo , Hepatócitos/metabolismo , Humanos , Masculino , Camundongos , Camundongos Knockout
9.
Cells ; 8(9)2019 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-31480503

RESUMO

The incidence and mortality rate of cancer has been quickly increasing in the past decades. At present, cancer has become the leading cause of death worldwide. Most of the cancers cannot be effectively diagnosed at the early stage. Although there are multiple therapeutic treatments, including surgery, radiotherapy, chemotherapy, and targeted drugs, their effectiveness is still limited. The overall survival rate of malignant cancers is still low. It is necessary to further study the mechanisms for malignant cancers, and explore new biomarkers and targets that are more sensitive and effective for early diagnosis, treatment, and prognosis of cancers than traditional biomarkers and methods. Long non-coding RNAs (lncRNAs) are a class of RNA transcripts with a length greater than 200 nucleotides. Generally, lncRNAs are not capable of encoding proteins or peptides. LncRNAs exert diverse biological functions by regulating gene expressions and functions at transcriptional, translational, and post-translational levels. In the past decade, it has been demonstrated that the dysregulated lncRNA profile is widely involved in the pathogenesis of many diseases, including cancer, metabolic disorders, and cardiovascular diseases. In particular, lncRNAs have been revealed to play an important role in tumor growth and metastasis. Many lncRNAs have been shown to be potential biomarkers and targets for the diagnosis and treatment of cancers. This review aims to briefly discuss the latest findings regarding the roles and mechanisms of some important lncRNAs in the pathogenesis of certain malignant cancers, including lung, breast, liver, and colorectal cancers, as well as hematological malignancies and neuroblastoma.


Assuntos
Biomarcadores Tumorais/genética , Neoplasias/genética , RNA Longo não Codificante/genética , Animais , Biomarcadores Tumorais/metabolismo , Humanos , Neoplasias/etiologia , Neoplasias/patologia , RNA Longo não Codificante/metabolismo
10.
J Cell Mol Med ; 23(5): 3464-3475, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30887707

RESUMO

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.


Assuntos
Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Movimento Celular , Citocinas/metabolismo , Fatores de Transcrição de Choque Térmico/metabolismo , Proteínas de Neoplasias/metabolismo , Transdução de Sinais , Fator de Crescimento Transformador beta/farmacologia , Fator de Transcrição YY1/metabolismo , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Feminino , Inativação Gênica , Humanos , Transdução de Sinais/efeitos dos fármacos , Regulação para Cima/efeitos dos fármacos
11.
J Cell Mol Med ; 23(3): 1746-1758, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30488666

RESUMO

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.


Assuntos
Citocinas/metabolismo , Proteína Forkhead Box O1/metabolismo , Gluconeogênese , Glucose/metabolismo , Hepatócitos/metabolismo , Proteínas de Neoplasias/metabolismo , Animais , Células Cultivadas , Citocinas/genética , Proteína Forkhead Box O1/genética , Regulação da Expressão Gênica , Hepatócitos/citologia , Humanos , Resistência à Insulina , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Obesos , Proteínas de Neoplasias/genética
12.
J Genet Genomics ; 45(7): 389-397, 2018 07 20.
Artigo em Inglês | MEDLINE | ID: mdl-30054214

RESUMO

Enrichment analysis methods, e.g., gene set enrichment analysis, represent one class of important bioinformatical resources for mining patterns in biomedical datasets. However, tools for inferring patterns and rules of a list of drugs are limited. In this study, we developed a web-based tool, DrugPattern, for drug set enrichment analysis. We first collected and curated 7019 drug sets, including indications, adverse reactions, targets, pathways, etc. from public databases. For a list of interested drugs, DrugPattern then evaluates the significance of the enrichment of these drugs in each of the 7019 drug sets. To validate DrugPattern, we employed it for the prediction of the effects of oxidized low-density lipoprotein (oxLDL), a factor expected to be deleterious. We predicted that oxLDL has beneficial effects on some diseases, most of which were supported by evidence in the literature. Because DrugPattern predicted the potential beneficial effects of oxLDL in type 2 diabetes (T2D), animal experiments were then performed to further verify this prediction. As a result, the experimental evidences validated the DrugPattern prediction that oxLDL indeed has beneficial effects on T2D in the case of energy restriction. These data confirmed the prediction accuracy of our approach and revealed unexpected protective roles for oxLDL in various diseases. This study provides a tool to infer patterns and rules in biomedical datasets based on drug set enrichment analysis. DrugPattern is available at http://www.cuilab.cn/drugpattern.


Assuntos
Biologia Computacional/métodos , Diabetes Mellitus Tipo 2/tratamento farmacológico , Animais , Diabetes Mellitus Tipo 2/metabolismo , Humanos , Internet , Lipoproteínas LDL/metabolismo , Camundongos
13.
Diabetes ; 67(4): 581-593, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29382663

RESUMO

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.


Assuntos
Diabetes Mellitus Tipo 2/genética , Galactosiltransferases/metabolismo , Gluconeogênese/genética , Ribonucleoproteína Nuclear Heterogênea A1/genética , Lipogênese/genética , Fígado/metabolismo , Hepatopatia Gordurosa não Alcoólica/metabolismo , RNA Longo não Codificante/genética , Animais , Glicemia/metabolismo , Cálcio/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Modelos Animais de Doenças , Técnicas de Silenciamento de Genes , Glucose/metabolismo , Hepatócitos/metabolismo , Humanos , Insulina/metabolismo , Metabolismo dos Lipídeos , Camundongos , Camundongos Obesos , Proteínas Monoméricas de Montagem de Clatrina/genética , Proteínas Monoméricas de Montagem de Clatrina/metabolismo , Fosfatidilinositol 3-Quinase/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , RNA Mensageiro/metabolismo , Transdução de Sinais , Proteína de Ligação a Elemento Regulador de Esterol 1/metabolismo , Serina-Treonina Quinases TOR/metabolismo
14.
Metabolism ; 81: 71-82, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29221790

RESUMO

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.


Assuntos
Citocinas/fisiologia , Diabetes Mellitus Tipo 2/metabolismo , Hepatopatia Gordurosa não Alcoólica/metabolismo , Animais , Citocinas/sangue , Diabetes Mellitus Tipo 2/etiologia , Glucose/metabolismo , Humanos , Resistência à Insulina , Metabolismo dos Lipídeos , Fígado/metabolismo , Proteínas de Neoplasias/sangue , Hepatopatia Gordurosa não Alcoólica/etiologia , PPAR gama/fisiologia , Transdução de Sinais
15.
Oncotarget ; 8(62): 106038-106049, 2017 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-29285313

RESUMO

FAM3C, a member of FAM3 gene family, has been shown to improve insulin resistance and hyperglycemia in obese mice. This study further determined whether FAM3C functions as a hepatokine to suppress hepatic gluconeogenesis of type 1 diabetic mice. In STZ-induced type 1 diabetic mouse liver, the FAM3C-HSF1-CaM signaling axis was repressed. Hepatic FAM3C overexpression activated HSF1-CaM-Akt pathway to repress gluconeogenic gene expression and ameliorate hyperglycemia of type 1 diabetic mice. Moreover, hepatic HSF1 overexpression also activated CaM-Akt pathway to repress gluconeogenic gene expression and improve hyperglycemia of type 1 diabetic mice. Hepatic FAM3C and HSF1 overexpression had little effect on serum insulin levels in type 1 diabetic mice. In cultured hepatocytes, conditioned medium of Ad-FAM3C-infected cells induced Akt phosphorylation. Moreover, Akt activation and gluconeogenesis repression induced by FAM3C overexpression were reversed by the treatment with anti-FAM3C antibodies. Treatment with recombinant FAM3C protein induced Akt activation in a HSF1- and CaM-dependent manner in cultured hepatocytes. Furthermore, recombinant FAM3C protein repressed gluconeogenic gene expression and gluconeogenesis by inactivating FOXO1 in a HSF1-dependent manner in cultured hepatocytes. In conclusion, FAM3C is a new hepatokine that suppresses hepatic gluconeogenic gene expression and gluconeogenesis independent of insulin by activating HSF1-CaM-Akt pathway.

16.
Oncotarget ; 8(30): 49882-49896, 2017 Jul 25.
Artigo em Inglês | MEDLINE | ID: mdl-28562339

RESUMO

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.


Assuntos
Citocinas/metabolismo , Hepatopatias/metabolismo , Estresse Oxidativo , PPAR gama/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Traumatismo por Reperfusão/metabolismo , Transdução de Sinais , Animais , Apoptose , Sobrevivência Celular , Citocinas/genética , Modelos Animais de Doenças , Progressão da Doença , Expressão Gênica , Hepatócitos/metabolismo , Hepatopatias/patologia , Testes de Função Hepática , Masculino , Camundongos , Camundongos Knockout , Estresse Oxidativo/efeitos dos fármacos , Transporte Proteico/efeitos dos fármacos , Traumatismo por Reperfusão/patologia , Rosiglitazona , Transdução de Sinais/efeitos dos fármacos , Tiazolidinedionas/farmacologia
17.
Diabetes ; 66(7): 1819-1832, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-28411267

RESUMO

Hepatic FAM3A expression is repressed under obese conditions, but the underlying mechanism remains unknown. This study determined the role and mechanism of miR-423-5p in hepatic glucose and lipid metabolism by repressing FAM3A expression. miR-423-5p expression was increased in the livers of obese diabetic mice and in patients with nonalcoholic fatty liver disease (NAFLD) with decreased FAM3A expression. miR-423-5p directly targeted FAM3A mRNA to repress its expression and the FAM3A-ATP-Akt pathway in cultured hepatocytes. Hepatic miR-423-5p inhibition suppressed gluconeogenesis and improved insulin resistance, hyperglycemia, and fatty liver in obese diabetic mice. In contrast, hepatic miR-423-5p overexpression promoted gluconeogenesis and hyperglycemia and increased lipid deposition in normal mice. miR-423-5p inhibition activated the FAM3A-ATP-Akt pathway and repressed gluconeogenic and lipogenic gene expression in diabetic mouse livers. The miR-423 precursor gene was further shown to be a target gene of NFE2, which induced miR-423-5p expression to repress the FAM3A-ATP-Akt pathway in cultured hepatocytes. Hepatic NFE2 overexpression upregulated miR-423-5p to repress the FAM3A-ATP-Akt pathway, promoting gluconeogenesis and lipid deposition and causing hyperglycemia in normal mice. In conclusion, under the obese condition, activation of the hepatic NFE2/miR-423-5p axis plays important roles in the progression of type 2 diabetes and NAFLD by repressing the FAM3A-ATP-Akt signaling pathway.


Assuntos
Gluconeogênese/genética , Hepatócitos/metabolismo , Hiperglicemia/genética , Fígado/metabolismo , MicroRNAs/genética , Subunidade p45 do Fator de Transcrição NF-E2/genética , Hepatopatia Gordurosa não Alcoólica/genética , Trifosfato de Adenosina/metabolismo , Adulto , Animais , Estudos de Casos e Controles , Citocinas/genética , Citocinas/metabolismo , Feminino , Técnicas de Introdução de Genes , Técnicas de Silenciamento de Genes , Teste de Tolerância a Glucose , Células HEK293 , Células Hep G2 , Humanos , Hiperglicemia/metabolismo , Lipólise , Masculino , Camundongos , Pessoa de Meia-Idade , Subunidade p45 do Fator de Transcrição NF-E2/metabolismo , Hepatopatia Gordurosa não Alcoólica/metabolismo , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais
18.
Diabetes ; 66(5): 1185-1197, 2017 05.
Artigo em Inglês | MEDLINE | ID: mdl-28246289

RESUMO

FAM3C is a member of the family with sequence similarity 3 (FAM3) gene family, and this study determined its role and mechanism in regulation of hepatic glucose/lipid metabolism. In obese diabetic mice, FAM3C expression was reduced in the liver, and hepatic FAM3C restoration improved insulin resistance, hyperglycemia, and fatty liver. FAM3C overexpression increased the expression of heat shock factor 1 (HSF1), calmodulin (CaM), and phosphorylated protein kinase B (Akt) and reduced that of gluconeogenic and lipogenic genes in diabetic mouse livers with the suppression of gluconeogenesis and lipid deposition. In cultured hepatocytes, FAM3C overexpression upregulated HSF1 expression, which elevated CaM protein level by inducing CALM1 transcription to activate Akt in a Ca2+- and insulin-independent manner. Furthermore, FAM3C overexpression promoted nuclear exclusion of FOXO1 and repressed gluconeogenic gene expression and gluconeogenesis in a CaM-dependent manner in hepatocytes. Hepatic HSF1 overexpression activated the CaM-Akt pathway to repress gluconeogenic and lipogenic gene expression and improve hyperglycemia and fatty liver in obese diabetic mice. In conclusion, the FAM3C-HSF1-CaM-Akt pathway plays important roles in regulating glucose and lipid metabolism in hepatocytes independent of insulin and calcium. Restoring hepatic FAM3C expression is beneficial for the management of type 2 diabetes and fatty liver.


Assuntos
Glicemia/metabolismo , Calmodulina/genética , Citocinas/genética , Proteínas de Ligação a DNA/metabolismo , Diabetes Mellitus Tipo 2/genética , Fígado/metabolismo , Proteínas de Neoplasias/genética , Obesidade/genética , Fatores de Transcrição/metabolismo , Animais , Calmodulina/metabolismo , Citocinas/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Dieta Hiperlipídica , Modelos Animais de Doenças , Fígado Gorduroso/genética , Fígado Gorduroso/metabolismo , Proteína Forkhead Box O1/metabolismo , Regulação da Expressão Gênica/genética , Gluconeogênese/genética , Fatores de Transcrição de Choque Térmico , Hepatócitos/metabolismo , Resistência à Insulina/genética , Lipogênese/genética , Camundongos , Proteínas de Neoplasias/metabolismo , Obesidade/metabolismo , Fosfoproteínas , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais
19.
Sheng Li Ke Xue Jin Zhan ; 47(3): 194-202, 2016 Jun.
Artigo em Chinês | MEDLINE | ID: mdl-29888883

RESUMO

In the past decades, diabetes, in particular type 2 diabetes (T2D)mainly characterized by global insulin resistance and pancreatic beta cell failure, had become epidemic and a severe public health threat worldwide with the development of economy and change of lifestyle.The interactions between environment factors and genetic background play vital roles in the development and progression of T2D.More recently, it had been revealed that non-coding RNA including microRNA (miRNA)and long noncoding RNA (LncRNA)are widely involved inthe regulation of glucose and lipid metabolism. So far, it had been established that deregulated miRNA and LncRNA profile in main metabolic tissues is tightly associated with T2D,and intensive studies on non-coding RNAs had shed light on understanding the pathogen-esis of T2D.The current review aimed to briefly summarize and discuss the latest findings regarding the role and mechanism of miRNAs and LncRNAs in the regulation hepatic glucose and lipid metabolism.


Assuntos
Metabolismo dos Lipídeos , Diabetes Mellitus Tipo 2 , Glucose , Humanos , Resistência à Insulina , Células Secretoras de Insulina , Fígado , RNA não Traduzido
20.
PLoS One ; 10(7): e0133462, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26221732

RESUMO

Long noncoding RNAs (LncRNAs) have been believed to be the major transcripts in various tissues and organs, and may play important roles in regulation of many biological processes. The current study determined the LncRNA profile in mouse plasma after liver ischemia/reperfusion injury (IRI) using microarray technology. Microarray assays revealed that 64 LncRNAs were upregulated, and 244 LncRNAs were downregulated in the plasma of liver IRI mouse. Among these dysregulated plasma LncRNAs, 59-61% were intergenic, 22-25% were antisense overlap, 8-12% were sense overlap and 6-7% were bidirectional. Ten dysregulated plasma LncRNAs were validated by quantitative PCR assays, confirming the accuracy of microarray analysis result. Comparison analysis between dysregulated plasma and liver LncRNA profile after liver IRI revealed that among the 308 dysregulated plasma LncRNAs, 245 LncRNAs were present in the liver, but remained unchanged. In contrast, among the 98 dysregulated liver LncRNAs after IRI, only 19 were present in the plasma, but remained unchanged. LncRNA AK139328 had been previously reported to be upregulated in the liver after IRI, and silencing of hepatic AK139328 ameliorated liver IRI. Both microarray and RT-PCR analyses failed to detect the presence of AK139328 in mouse plasma. In summary, the current study compared the difference between dysregulated LncRNA profile in mouse plasma and liver after liver IRI, and suggested that a group of dysregulated plasma LncRNAs have the potential of becoming novel biomarkers for evaluation of ischemic liver injury.


Assuntos
Regulação da Expressão Gênica , Fígado/metabolismo , RNA Longo não Codificante/sangue , Traumatismo por Reperfusão/sangue , Animais , Perfilação da Expressão Gênica , Fígado/patologia , Masculino , Camundongos , Análise de Sequência com Séries de Oligonucleotídeos , Traumatismo por Reperfusão/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...