Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
1.
JCI Insight ; 7(4)2022 02 22.
Artigo em Inglês | MEDLINE | ID: mdl-35050901

RESUMO

Lung alveolar type 2 (AT2) cells are progenitors for alveolar type 1 (AT1) cells. Although many factors regulate AT2 cell plasticity, the role of mitochondrial calcium (mCa2+) uptake in controlling AT2 cells remains unclear. We previously identified that the miR-302 family supports lung epithelial progenitor cell proliferation and less differentiated phenotypes during development. Here, we report that a sustained elevation of miR-302 in adult AT2 cells decreases AT2-to-AT1 cell differentiation during the Streptococcus pneumoniae-induced lung injury repair. We identified that miR-302 targets and represses the expression of mitochondrial Ca2+ uptake 1 (MICU1), which regulates mCa2+ uptake through the mCa2+ uniporter channel by acting as a gatekeeper at low cytosolic Ca2+ levels. Our results reveal a marked increase in MICU1 protein expression and decreased mCa2+ uptake during AT2-to-AT1 cell differentiation in the adult lung. Deletion of Micu1 in AT2 cells reduces AT2-to-AT1 cell differentiation during steady-state tissue maintenance and alveolar epithelial regeneration after bacterial pneumonia. These studies indicate that mCa2+ uptake is extensively modulated during AT2-to-AT1 cell differentiation and that MICU1-dependent mCa2+ uniporter channel gating is a prominent mechanism modulating AT2-to-AT1 cell differentiation.


Assuntos
Células Epiteliais Alveolares/metabolismo , Proteínas de Ligação ao Cálcio/genética , Cálcio/metabolismo , Regulação da Expressão Gênica , Proteínas de Transporte da Membrana Mitocondrial/genética , Pneumonia Bacteriana/genética , RNA/genética , Regeneração/genética , Células Epiteliais Alveolares/patologia , Animais , Transporte Biológico , Proteínas de Ligação ao Cálcio/biossíntese , Diferenciação Celular , Plasticidade Celular , Células Cultivadas , Modelos Animais de Doenças , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Proteínas de Transporte da Membrana Mitocondrial/biossíntese , Pneumonia Bacteriana/metabolismo , Pneumonia Bacteriana/patologia
2.
Biomolecules ; 11(8)2021 07 23.
Artigo em Inglês | MEDLINE | ID: mdl-34439753

RESUMO

There is a pressing need for molecular targets and biomarkers in gastric cancer (GC). We aimed at identifying aberrations in L-arginine metabolism with therapeutic and diagnostic potential. Systemic metabolites were quantified using mass spectrometry in 293 individuals and enzymes' gene expression was quantified in 29 paired tumor-normal samples using qPCR and referred to cancer pathology and molecular landscape. Patients with cancer or benign disorders had reduced systemic arginine, citrulline, and ornithine and elevated symmetric dimethylarginine and dimethylamine. Citrulline and ornithine depletion was accentuated in metastasizing cancers. Metabolite diagnostic panel had 91% accuracy in detecting cancer and 70% accuracy in differentiating cancer from benign disorders. Gastric tumors had upregulated NOS2 and downregulated ASL, PRMT2, ORNT1, and DDAH1 expression. NOS2 upregulation was less and ASL downregulation was more pronounced in metastatic cancers. Tumor ASL and PRMT2 expression was inversely related to local advancement. Enzyme up- or downregulation was greater or significant solely in cardia subtype. Metabolic reprogramming in GC includes aberrant L-arginine metabolism, reflecting GC subtype and pathology, and is manifested by altered interplay of its intermediates and enzymes. Exploiting L-arginine metabolic pathways for diagnostic and therapeutic purposes is warranted. Functional studies on ASL, PRMT2, and ORNT1 in GC are needed.


Assuntos
Arginina/metabolismo , Regulação Neoplásica da Expressão Gênica , Neoplasias Gástricas/metabolismo , Idoso , Argininossuccinato Liase/biossíntese , Diferenciação Celular , Citrulina/metabolismo , DNA Complementar/metabolismo , Feminino , Perfilação da Expressão Gênica , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/biossíntese , Masculino , Espectrometria de Massas , Metabolômica , Pessoa de Meia-Idade , Proteínas de Transporte da Membrana Mitocondrial/biossíntese , Metástase Neoplásica , Óxido Nítrico Sintase Tipo II , Ornitina/metabolismo , Reação em Cadeia da Polimerase , Proteína-Arginina N-Metiltransferases/biossíntese , Reprodutibilidade dos Testes , Neoplasias Gástricas/tratamento farmacológico , Transcriptoma
3.
Nat Metab ; 2(11): 1223-1231, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-33106688

RESUMO

Cardiomyocytes rely on metabolic substrates, not only to fuel cardiac output, but also for growth and remodelling during stress. Here we show that mitochondrial pyruvate carrier (MPC) abundance mediates pathological cardiac hypertrophy. MPC abundance was reduced in failing hypertrophic human hearts, as well as in the myocardium of mice induced to fail by angiotensin II or through transverse aortic constriction. Constitutive knockout of cardiomyocyte MPC1/2 in mice resulted in cardiac hypertrophy and reduced survival, while tamoxifen-induced cardiomyocyte-specific reduction of MPC1/2 to the attenuated levels observed during pressure overload was sufficient to induce hypertrophy with impaired cardiac function. Failing hearts from cardiomyocyte-restricted knockout mice displayed increased abundance of anabolic metabolites, including amino acids and pentose phosphate pathway intermediates and reducing cofactors. These hearts showed a concomitant decrease in carbon flux into mitochondrial tricarboxylic acid cycle intermediates, as corroborated by complementary 1,2-[13C2]glucose tracer studies. In contrast, inducible cardiomyocyte overexpression of MPC1/2 resulted in increased tricarboxylic acid cycle intermediates, and sustained carrier expression during transverse aortic constriction protected against cardiac hypertrophy and failure. Collectively, our findings demonstrate that loss of the MPC1/2 causally mediates adverse cardiac remodelling.


Assuntos
Proteínas de Transporte de Ânions/metabolismo , Cardiomegalia/metabolismo , Proteínas de Transporte da Membrana Mitocondrial/metabolismo , Transportadores de Ácidos Monocarboxílicos/metabolismo , Angiotensina II , Animais , Proteínas de Transporte de Ânions/biossíntese , Proteínas de Transporte de Ânions/genética , Cardiomegalia/patologia , Proliferação de Células , Ciclo do Ácido Cítrico , Constrição Patológica , Feminino , Insuficiência Cardíaca/induzido quimicamente , Insuficiência Cardíaca/metabolismo , Insuficiência Cardíaca/patologia , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Mitocôndrias Cardíacas/metabolismo , Proteínas de Transporte da Membrana Mitocondrial/biossíntese , Proteínas de Transporte da Membrana Mitocondrial/genética , Transportadores de Ácidos Monocarboxílicos/biossíntese , Transportadores de Ácidos Monocarboxílicos/genética , Miocárdio/metabolismo , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia , Ácido Pirúvico/metabolismo
4.
Tumour Biol ; 42(8): 1010428320951057, 2020 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-32829673

RESUMO

The exchange of metabolites between mitochondria and cytosol occurs through pores formed by voltage-dependent anion channel proteins. Voltage-dependent anion channels appear to be master regulators of mitochondrial bioenergetics and the intracellular flow of energy. Deregulation of voltage-dependent anion channels expression is thought to be related to mitochondrial dysfunction in cancer. The aim of this study was to investigate the mRNA and protein expression levels of VDAC1, VDAC2, and VDAC3 in relation to clinicopathological characteristics of endometrial cancer as well as the prognostic significance of voltage-dependent anion channels expression for overall survival. VDAC1 and VDAC3 expressions were significantly higher in cancer compared to normal tissues. Kaplan-Meier analysis indicated that high expression of all VDAC genes or high VDAC2 protein level predicted poor overall survival. Multivariate analysis identified the VDAC1 and VDAC2 mRNA levels as well as VDAC2 protein level as independent prognostic factors. Our results suggest that increased expression of voltage-dependent anion channels correlates with tumor progression and may serve as a potential prognostic biomarker in endometrial cancer.


Assuntos
Neoplasias do Endométrio/patologia , Mitocôndrias/patologia , Proteínas de Transporte da Membrana Mitocondrial/genética , Canal de Ânion 1 Dependente de Voltagem/genética , Canal de Ânion 2 Dependente de Voltagem/genética , Canais de Ânion Dependentes de Voltagem/genética , Sequência de Aminoácidos , Biomarcadores Tumorais/genética , Citoplasma/metabolismo , Neoplasias do Endométrio/mortalidade , Feminino , Humanos , Pessoa de Meia-Idade , Mitocôndrias/metabolismo , Proteínas de Transporte da Membrana Mitocondrial/biossíntese , Prognóstico , RNA Mensageiro/genética , Canal de Ânion 1 Dependente de Voltagem/biossíntese , Canal de Ânion 2 Dependente de Voltagem/biossíntese , Canais de Ânion Dependentes de Voltagem/biossíntese
5.
Pathol Oncol Res ; 26(1): 411-418, 2020 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-30382527

RESUMO

The substantial difference between normal cells and cancer cells in terms of their energy metabolism in mitochondria provides an interesting basis for the development of novel therapeutic agents targeting energy machinery of tumour cells. TOMM34 is one of the Tom (translocase of the outer membrane of mitochondria) family that was found to be overexpressed in colorectal, hepatocellular, lung and early invasive breast carcinomas. The expression profile of mitochondrial translocases in bladder cancer compared to normal urinary bladder tissues has not been investigated yet. Therefore, the aim of the current study is to investigate the expression pattern of TOMM34 in bladder cancer tissues and explore its correlation with the clinico-pathological parameters of those cases. Sixty patients who underwent either transurethral resection or radical cystectomy for bladder cancer were included in this study with revision of all their clinicopathological data and tumor slides. Ten histologically normal urothelial biopsies were also included. Immunohistochemical staining for TOMM34 was done and semi-quantitatively scored using the modified H-score. All relations were analysed using established statistical methodologies. TOMM34 overexpression was significantly associated with high tumour stage, muscle invasion and high grade. Significant positive association was observed between TOMM34 expression and poor outcome in terms of shorter disease-specific survival. This study suggests TOMM34 as a biomarker of progression and poor prognosis in urothelial cell carcinoma patients. Furthermore, we suggest a role played by mitochondrial machinery in urothelial cell carcinoma progression, which is a potential target for the newly-discovered vaccine therapy for urothelial cell carcinoma.


Assuntos
Biomarcadores Tumorais/análise , Carcinoma de Células de Transição/patologia , Proteínas de Transporte da Membrana Mitocondrial/biossíntese , Neoplasias da Bexiga Urinária/patologia , Idoso , Idoso de 80 Anos ou mais , Carcinoma de Células de Transição/mortalidade , Progressão da Doença , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Proteínas do Complexo de Importação de Proteína Precursora Mitocondrial , Prognóstico , Neoplasias da Bexiga Urinária/mortalidade
6.
Mol Cell Biochem ; 463(1-2): 189-201, 2020 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-31612353

RESUMO

Human triple negative breast cancer cells, MDA-MB-231, show typical epithelial to mesenchymal transition associated with cancer progression. Mitochondria play a major role in cancer progression, including metastasis. Changes in mitochondrial architecture affect cellular migration, autophagy and apoptosis. Silibinin is reported to have anti-breast cancer effect. We here report that silibinin at lower concentrations (30-90 µM) inhibits epithelial to mesenchymal transition (EMT) of MDA-MB-231, by increasing the expression of epithelial marker, E-cadherin, and decreasing the expression of mesenchymal markers, N-cadherin and vimentin. Besides, silibinin inhibition of cell migration is associated with reduction in the protein expression of matrix metalloproteinases 2 and 9 (MMP2 and MMP9) and paxillin. In addition, silibinin treatment increases mitochondrial fusion through down-regulating the expression of mitochondrial fission-associated protein dynamin-related protein 1 (DRP1) and up-regulating the expression of mitochondrial fusion-associated proteins, optic atrophy 1, mitofusin 1 and mitofusin 2. Silibinin perturbed mitochondrial biogenesis via down-regulating the levels of mitochondrial biogenesis regulators including mitochondrial transcription factor A (TFAM), peroxisome proliferator-activated receptor gamma coactivator (PGC1) and nuclear respiratory factor (NRF2). Moreover, DRP1 knockdown or silibinin inhibited cell migration, and MFN1&2 knockdown restored it. Mitochondrial fusion contributes to silibinin's negative effect on cell migration. Silibinin decreased reactive oxygen species (ROS) generation, leading to inhibition of the NLRP3 inflammasome activation. In addition, knockdown of mitofusin 1&2 (MFN 1&2) relieved silibinin-induced inhibition of NLRP3 inflammasome activation. Repression of ROS contributes to the inhibition of the expression of NLRP3, caspase-1 and IL-ß proteins as well as of cell migration. Taken together, our study provides evidence that silibinin impairs mitochondrial dynamics and biogenesis, resulting in reduced migration and invasion of the MDA-MB-231 breast cancer cells.


Assuntos
Transição Epitelial-Mesenquimal/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Dinâmica Mitocondrial/efeitos dos fármacos , Proteínas de Transporte da Membrana Mitocondrial/biossíntese , Proteínas de Neoplasias/biossíntese , Silibina/farmacologia , Neoplasias de Mama Triplo Negativas/metabolismo , Linhagem Celular Tumoral , Movimento Celular , Transição Epitelial-Mesenquimal/genética , Feminino , Humanos , Dinâmica Mitocondrial/genética , Proteínas de Transporte da Membrana Mitocondrial/genética , Proteínas de Neoplasias/genética , Neoplasias de Mama Triplo Negativas/tratamento farmacológico , Neoplasias de Mama Triplo Negativas/genética , Neoplasias de Mama Triplo Negativas/patologia
7.
Biol Chem ; 401(1): 117-129, 2019 12 18.
Artigo em Inglês | MEDLINE | ID: mdl-31513529

RESUMO

Biogenesis and function of mitochondria depend on the import of about 1000 precursor proteins that are produced on cytosolic ribosomes. The translocase of the outer membrane (TOM) forms the entry gate for most proteins. After passage through the TOM channel, dedicated preprotein translocases sort the precursor proteins into the mitochondrial subcompartments. Many proteins have to be assembled into oligomeric membrane-integrated complexes in order to perform their functions. In this review, we discuss a dual role of mitochondrial preprotein translocases in protein translocation and oligomeric assembly, focusing on the biogenesis of the TOM complex and the respiratory chain. The sorting and assembly machinery (SAM) of the outer mitochondrial membrane forms a dynamic platform for coupling transport and assembly of TOM subunits. The biogenesis of the cytochrome c oxidase of the inner membrane involves a molecular circuit to adjust translation of mitochondrial-encoded core subunits to the availability of nuclear-encoded partner proteins. Thus, mitochondrial protein translocases not only import precursor proteins but can also support their assembly into functional complexes.


Assuntos
Proteínas de Transporte/genética , Mitocôndrias/genética , Proteínas de Transporte da Membrana Mitocondrial/genética , Proteínas Mitocondriais/genética , Proteínas de Transporte/ultraestrutura , Mitocôndrias/metabolismo , Proteínas de Transporte da Membrana Mitocondrial/biossíntese , Membranas Mitocondriais/metabolismo , Membranas Mitocondriais/ultraestrutura , Proteínas do Complexo de Importação de Proteína Precursora Mitocondrial , Proteínas Mitocondriais/biossíntese , Proteínas Mitocondriais/ultraestrutura , Biossíntese de Proteínas/genética , Transporte Proteico/genética , Ribossomos/genética , Ribossomos/metabolismo , Saccharomyces cerevisiae/genética
8.
Hum Pathol ; 84: 81-91, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30261190

RESUMO

Tumor cells frequently evade apoptosis triggered by cellular stress via aberrant regulation of the BCL-2 family members, which are key players in regulating cell death under physiological and pathological situations. Previously, we have identified a novel BH3-only protein of the BCL-2 family, BLM-s (BCL-2-like molecule, short form), that modulates apoptosis of postmitotic immature neurons during corticohistogenesis. Whether BLM-s expression correlates with any subtype of human tumors has not been investigated. Here, via BLM-s immunohistochemistry performed in various kinds of human tumors, we demonstrate that BLM-s is specifically expressed in tumors derived from salivary gland (specificity, 0.76 [95% confidence interval, or CI], 0.65-0.85]; sensitivity, 1 [95% CI, 0.99-1]). Stratification of BLM-s immunointensity and its subcellular localization in correlation with salivary gland tumor subtype shows a statistically significant increase in proportion and in intensity of nuclear staining for adenoid cystic carcinoma (ACC; specificity, 0.92 [95% CI, 0.88-0.95]; sensitivity, 0.82 [95% CI, 0.66-0.92]), a locally aggressive head and neck malignancy. Comparison among salivary ACC in correlation with MYB/MYBL fluorescence in situ hybridization, c-KIT immunohistochemistry, and BLM-s immunohistochemistry shows that BLM-s' nuclear immunoreactivity has lower false-negative detection rate (18.5% compared with 26.3% [MYB/MYBL fluorescence in situ hybridization] and 34.2% [c-KIT], respectively). Intriguingly, ACC derived from other cell origins such as breast shows negative BLM-s immunoreactivity. We thus propose that nuclear localization of BLM-s detected by immunohistochemistry could be potentially used as an ancillary diagnostic marker for ACC originating from the salivary gland, especially when the biopsy specimen is small with an unknown tumor origin.


Assuntos
Carcinoma Adenoide Cístico/diagnóstico , Proteínas de Transporte da Membrana Mitocondrial/biossíntese , Neoplasias das Glândulas Salivares/diagnóstico , Adulto , Idoso , Biomarcadores Tumorais/análise , Núcleo Celular/metabolismo , Feminino , Humanos , Imuno-Histoquímica , Masculino , Pessoa de Meia-Idade , Proteínas de Transporte da Membrana Mitocondrial/análise
9.
Cell Death Dis ; 9(5): 466, 2018 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-29700317

RESUMO

PGC1α acts as a central regulator of mitochondrial metabolism, whose role in cancer progression has been highlighted but remains largely undefined. Especially, it is completely unknown about the effect of PGC1α on cholangiocarcinoma (CCA). Here we showed that PGC1α overexpression had no impact on CCA growth despite the decreased expression of PGC1α in CCA compared with adjacent normal tissue. Instead, PGC1α overexpression-promoted CCA metastasis both in vitro and in vivo. Mechanistically, for the first time, we illuminated that PGC1α reversed the Warburg effect by upregulating the expression of pyruvate dehydrogenase E1 alpha 1 subunit and mitochondrial pyruvate carrier 1 to increase pyruvate flux into the mitochondria for oxidation, whereas simultaneously promoting mitochondrial biogenesis and fusion to mediate the metabolic switch to oxidative phosphorylation. On the one hand, enhanced mitochondrial oxidation metabolism correlated with elevated reactive oxygen species (ROS) production; on the other hand, increased PGC1α expression upregulated the expression levels of mRNA for several ROS-detoxifying enzymes. To this end, the ROS levels, which were elevated but below a critical threshold, did not inhibit CCA cells proliferation. And the moderately increased ROS facilitated metastatic dissemination of CCA cells, which can be abrogated by antioxidants. Our study suggests the potential utility of developing the PGC1α-targeted therapies or blocking PGC1α signaling axis for inhibiting CCA metastasis.


Assuntos
Neoplasias dos Ductos Biliares/metabolismo , Colangiocarcinoma/metabolismo , Regulação Neoplásica da Expressão Gênica , Proteínas de Transporte da Membrana Mitocondrial/biossíntese , Proteínas de Neoplasias/metabolismo , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo/metabolismo , Piruvato Desidrogenase (Lipoamida)/biossíntese , Regulação para Cima , Animais , Neoplasias dos Ductos Biliares/genética , Neoplasias dos Ductos Biliares/patologia , Colangiocarcinoma/genética , Colangiocarcinoma/patologia , Feminino , Glicólise/genética , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Proteínas de Transporte da Membrana Mitocondrial/genética , Transportadores de Ácidos Monocarboxílicos , Metástase Neoplásica , Proteínas de Neoplasias/genética , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo/genética , Piruvato Desidrogenase (Lipoamida)/genética
10.
Int J Mol Med ; 40(1): 21-30, 2017 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-28498397

RESUMO

Dihydroartemisinin (DHA) has been shown to inhibit the viability of various cancer cells. Previous studies have revealed that the mechanisms involved in the inhibitory effects of DHA are based on theactivation of p53 and the mitochondrial-related cell death pathway. However, the exact association between upstream signaling and the activation of cell death pathway remains unclear. In this study, we found that DHA treatment induced the upregulation of caveolin 1 (Cav1) and mitochondrial carrier homolog 2 (MTCH2) in HeLa cells, and this was associated with the DHA-induced inhibition of cell viability and DHA-induced apoptosis. Additionally, the overexpression of Cav1 and MTCH2 in HeLa cells enhanced the inhibitory effects of DHA on cell viability. Moreover, we also found that the upregulation of Cav1 contributed to the DHA-mediated p53 activation and the downregulation of the redox enzyme, NAD(P)H:quinone oxidoreductase 1 (NQO1), which have been reported to contribute to the activation of the cell death pathway. Of note, we also found that DHA induced the nuclear translocation and accumulation of both Cav1 and p53, indicating a novel potential mechanism, namely the regulation of p53 activation by Cav1. On the whole, our study identified Cav1 and MTCH2 as the molecular targets of DHA and revealed a new link between the upstream Cav1/MTCH2 upregulation and the downstream activation of the cell death pathway involved in the DHA-mediated inhibition of cell viability.


Assuntos
Artemisininas/farmacologia , Caveolina 1/biossíntese , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Proteínas de Transporte da Membrana Mitocondrial/biossíntese , NAD(P)H Desidrogenase (Quinona)/biossíntese , Proteína Supressora de Tumor p53/metabolismo , Regulação para Cima/efeitos dos fármacos , Neoplasias do Colo do Útero/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Feminino , Células HeLa , Humanos , Neoplasias do Colo do Útero/patologia
11.
J Am Heart Assoc ; 6(3)2017 Mar 13.
Artigo em Inglês | MEDLINE | ID: mdl-28288978

RESUMO

BACKGROUND: The cardiac microvascular system ischemia/reperfusion injury following percutaneous coronary intervention is a clinical thorny problem. This study explores the mechanisms by which ischemia/reperfusion injury induces cardiac microcirculation collapse. METHODS AND RESULTS: In wild-type mice, mitochondrial fission factor (Mff) expression increased in response to acute microvascular ischemia/reperfusion injury. Compared with wild-type mice, homozygous Mff-deficient (Mffgt) mice exhibited a smaller infarcted area, restored cardiac function, improved blood flow, and reduced microcirculation perfusion defects. Histopathology analysis demonstrated that cardiac microcirculation endothelial cells (CMECs) in Mffgt mice had an intact endothelial barrier, recovered phospho-endothelial nitric oxide synthase production, opened lumen, undivided mitochondrial structures, and less CMEC death. In vitro, Mff-deficient CMECs (derived from Mffgt mice or Mff small interfering RNA-treated) demonstrated less mitochondrial fission and mitochondrial-dependent apoptosis compared with cells derived from wild-type mice. The loss of Mff inhibited mitochondrial permeability transition pore opening via blocking the oligomerization of voltage-dependent anion channel 1 and subsequent hexokinase 2 separation from mitochondria. Moreover, Mff deficiency reduced the cyt-c leakage into the cytoplasm by alleviating cardiolipin oxidation resulting from damage to the electron transport chain complexes and mitochondrial reactive oxygen species overproduction. CONCLUSIONS: This evidence clearly illustrates that microcirculatory ischemia/reperfusion injury can be attributed to Mff-dependent mitochondrial fission via voltage-dependent anion channel 1/hexokinase 2-mediated mitochondrial permeability transition pore opening and mitochondrial reactive oxygen species/cardiolipin involved cyt-c release.


Assuntos
Cardiolipinas/genética , Regulação da Expressão Gênica , Hexoquinase/genética , Proteínas de Membrana/genética , Microcirculação/efeitos dos fármacos , Proteínas Mitocondriais/genética , Traumatismo por Reperfusão Miocárdica/genética , Canal de Ânion 1 Dependente de Voltagem/genética , Animais , Apoptose/efeitos dos fármacos , Western Blotting , Cardiolipinas/biossíntese , Células Cultivadas , Cromatografia Líquida de Alta Pressão , DNA Mitocondrial/genética , Modelos Animais de Doenças , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Hexoquinase/biossíntese , Imuno-Histoquímica , Masculino , Proteínas de Membrana/biossíntese , Camundongos , Microvasos/fisiopatologia , Mitocôndrias Cardíacas/genética , Mitocôndrias Cardíacas/metabolismo , Mitocôndrias Cardíacas/ultraestrutura , Dinâmica Mitocondrial/efeitos dos fármacos , Dinâmica Mitocondrial/genética , Proteínas de Transporte da Membrana Mitocondrial/biossíntese , Proteínas de Transporte da Membrana Mitocondrial/genética , Poro de Transição de Permeabilidade Mitocondrial , Proteínas Mitocondriais/biossíntese , Traumatismo por Reperfusão Miocárdica/metabolismo , Traumatismo por Reperfusão Miocárdica/fisiopatologia , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/ultraestrutura , Oxirredução , Espécies Reativas de Oxigênio/metabolismo , Canal de Ânion 1 Dependente de Voltagem/biossíntese
12.
Indian J Ophthalmol ; 64(7): 500-3, 2016 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-27609161

RESUMO

PURPOSE: The aim of this study is to preliminarily investigate the expression of mitochondrial fusion protein 1 (MFN1) in a lens-induced animal myopia (LIM) model and to explore the relationship between MFN1 and the visual development. MATERIALS AND METHODS: MFN1 gene expression in guinea pigs was examined during the development of minus LIM, 15 tri-colored guinea pigs were obtained, and one eye of each pig was randomly selected and treated with -7.00D lenses. Ocular refraction and axial length were collected before intervention and 1, 2, and 3 weeks after intervention. After the refraction and axial length measurements at 1, 2, and 3 weeks of lens intervention, five guinea pigs were randomly selected. MFN1 expression in the retina of both eyes was tested by immunohistochemistry technique. RESULTS: MFN1-positive cells could be observed in the retina of both eyes. The positive cells in the LIM eyes were staining deeper, and much more positive cells could be observed. Furthermore, MFN1-positive expression could be seen mainly in ganglion cells after 1 week of minus lens intervention, and with time extension, more and more positive cells appeared in the rod-cone cell and bipolar cell layer, and this phenomenon could not be found in the normal control eyes. CONCLUSION: This study suggested that MFN1 might be correlated to the development of myopia.


Assuntos
GTP Fosfo-Hidrolases/genética , Regulação da Expressão Gênica , Dinâmica Mitocondrial/genética , Proteínas de Transporte da Membrana Mitocondrial/genética , Miopia/genética , RNA/genética , Refração Ocular , Animais , Modelos Animais de Doenças , GTP Fosfo-Hidrolases/biossíntese , Cobaias , Imuno-Histoquímica , Proteínas de Transporte da Membrana Mitocondrial/biossíntese , Miopia/metabolismo , Miopia/fisiopatologia , Retina/metabolismo
13.
J Biol Chem ; 291(32): 16448-61, 2016 08 05.
Artigo em Inglês | MEDLINE | ID: mdl-27317664

RESUMO

Selective transport of pyruvate across the inner mitochondrial membrane by the mitochondrial pyruvate carrier (MPC) is a fundamental step that couples cytosolic and mitochondrial metabolism. The recent molecular identification of the MPC complex has revealed two interacting subunits, MPC1 and MPC2. Although in yeast, an additional subunit, MPC3, can functionally replace MPC2, no alternative MPC subunits have been described in higher eukaryotes. Here, we report for the first time the existence of a novel MPC subunit termed MPC1-like (MPC1L), which is present uniquely in placental mammals. MPC1L shares high sequence, structural, and topological homology with MPC1. In addition, we provide several lines of evidence to show that MPC1L is functionally equivalent to MPC1: 1) when co-expressed with MPC2, it rescues pyruvate import in a MPC-deleted yeast strain; 2) in mammalian cells, it can associate with MPC2 to form a functional carrier as assessed by bioluminescence resonance energy transfer; 3) in MPC1 depleted mouse embryonic fibroblasts, MPC1L rescues the loss of pyruvate-driven respiration and stabilizes MPC2 expression; and 4) MPC1- and MPC1L-mediated pyruvate imports show similar efficiency. However, we show that MPC1L has a highly specific expression pattern and is localized almost exclusively in testis and more specifically in postmeiotic spermatids and sperm cells. This is in marked contrast to MPC1/MPC2, which are ubiquitously expressed throughout the organism. To date, the biological importance of this alternative MPC complex during spermatogenesis in placental mammals remains unknown. Nevertheless, these findings open up new avenues for investigating the structure-function relationship within the MPC complex.


Assuntos
Proteínas de Transporte de Ânions/biossíntese , Regulação da Expressão Gênica/fisiologia , Proteínas de Transporte da Membrana Mitocondrial/biossíntese , Espermátides/metabolismo , Espermatogênese/fisiologia , Testículo/metabolismo , Animais , Proteínas de Transporte de Ânions/genética , Feminino , Células HEK293 , Humanos , Masculino , Camundongos , Proteínas de Transporte da Membrana Mitocondrial/genética , Proteínas Mitocondriais/biossíntese , Proteínas Mitocondriais/genética , Transportadores de Ácidos Monocarboxílicos , Espermátides/citologia , Testículo/citologia
14.
PLoS One ; 11(2): e0148311, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26845575

RESUMO

Exercise dynamically changes skeletal muscle protein synthesis to respond and adapt to the external and internal stimuli. Many studies have focused on overall protein synthesis to understand how exercise regulates the muscular adaptation. However, despite the probability that each gene transcript may have its own unique translational characteristics and would be differentially regulated at translational level, little attention has been paid to how exercise affects translational regulation of individual genes at a genome-wide scale. Here, we conducted a genome-wide translational analysis using ribosome profiling to investigate the effect of a single bout of treadmill running (20 m/min for 60 min) on mouse gastrocnemius. Global translational profiles largely differed from those in transcription even at a basal resting condition as well as immediately after exercise. As for individual gene, Slc25a25 (Solute carrier family 25, member 25), localized in mitochondrial inner membrane and maintaining ATP homeostasis and endurance performance, showed significant up-regulation at translational level. However, multiple regression analysis suggests that Slc25a25 protein degradation may also have a role in mediating Slc25a25 protein abundance in the basal and early stages after acute endurance exercise.


Assuntos
Regulação da Expressão Gênica/genética , Proteínas de Transporte da Membrana Mitocondrial/biossíntese , Músculo Esquelético/fisiologia , Condicionamento Físico Animal/fisiologia , Resistência Física/fisiologia , Animais , Sequência de Bases , Proteínas de Ligação ao Cálcio , Teste de Esforço , Estudo de Associação Genômica Ampla , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Proteínas de Transporte da Membrana Mitocondrial/genética , RNA Mensageiro/genética , Ribossomos/genética , Alinhamento de Sequência , Análise de Sequência de RNA , Transcrição Gênica/genética
15.
Syst Biol Reprod Med ; 62(1): 69-76, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26636621

RESUMO

Preimplantation genetic diagnosis (PGD) to select histocompatible siblings to facilitate curative haematopoeitic stem-cell transplantation (HSCT) is now an acceptable option in the absence of an available human leukocyte antigen (HLA) compatible donor. We describe a case where the couple who requested HLA-PGD, were both carriers of two serious haematological diseases, beta-thalassaemia and sideroblastic anaemia. Their daughter, affected with sideroblastic anaemia, was programmed to have HSCT. A multiplex-fluorescent-touchdown-PCR protocol was optimized for the simultaneous amplification of: the two HBB-gene mutated regions (c.118C> T, c.25-26delAA), four short tandem repeats (STRs) in chr11p15.5 linked to the HBB gene, the SLC25A38 gene mutation (c.726C > T), two STRs in chr3p22.1 linked to the SLC25A38 gene, plus eleven informative STRs for HLA-haplotyping (chr6p22.1-21.3). This was followed by real-time nested PCR and high-resolution melting analysis (HRMA) for the detection of HBB and SLC25A38 gene mutations, as well as the analysis of all STRs on an automatic genetic analyzer (sequencer). The couple completed four clinical in vitro fertilization (IVF)/PGD cycles. At least one matched unaffected embryo was identified and transferred in each cycle. A twin pregnancy was established in the fourth PGD cycle and genotyping results at all loci were confirmed by prenatal diagnosis. Two healthy baby girls were delivered at week 38 of pregnancy. The need to exclude two familial disorders for HLA-PGD is rarely encountered. The methodological approach described here is fast, accurate, clinically-validated, and of relatively low cost.


Assuntos
Anemia Sideroblástica/diagnóstico , Anemia Sideroblástica/genética , Teste de Histocompatibilidade/métodos , Diagnóstico Pré-Implantação/métodos , Talassemia beta/diagnóstico , Talassemia beta/genética , Adulto , Anemia Sideroblástica/terapia , Feminino , Fertilização in vitro , Testes Genéticos , Genótipo , Transplante de Células-Tronco Hematopoéticas , Humanos , Recém-Nascido , Masculino , Proteínas de Transporte da Membrana Mitocondrial/biossíntese , Proteínas de Transporte da Membrana Mitocondrial/genética , Mutação/genética , Gravidez , Gravidez de Gêmeos , Talassemia beta/terapia
16.
Mol Cell Biol ; 35(18): 3200-11, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26149385

RESUMO

A special group of mitochondrial outer membrane (MOM) proteins spans the membrane several times via multiple helical segments. Such multispan proteins are synthesized on cytosolic ribosomes before their targeting to mitochondria and insertion into the MOM. Previous work recognized the import receptor Tom70 and the mitochondrial import (MIM) complex, both residents of the MOM, as required for optimal biogenesis of these proteins. However, their involvement is not sufficient to explain either the entire import pathway or its regulation. To identify additional factors that are involved in the biogenesis of MOM multispan proteins, we performed complementary high-throughput visual and growth screens in Saccharomyces cerevisiae. Cardiolipin (CL) synthase (Crd1) appeared as a candidate in both screens. Our results indeed demonstrate lower steady-state levels of the multispan proteins Ugo1, Scm4, and Om14 in mitochondria from crd1Δ cells. Importantly, MOM single-span proteins were not affected by this mutation. Furthermore, organelles lacking Crd1 had a lower in vitro capacity to import newly synthesized Ugo1 and Scm4 molecules. Crd1, which is located in the mitochondrial inner membrane, condenses phosphatidylglycerol together with CDP-diacylglycerol to obtain de novo synthesized CL molecules. Hence, our findings suggest that CL is an important component in the biogenesis of MOM multispan proteins.


Assuntos
Cardiolipinas/biossíntese , Proteínas de Membrana/metabolismo , Membranas Mitocondriais/fisiologia , Saccharomyces cerevisiae/metabolismo , Transferases (Outros Grupos de Fosfato Substituídos)/metabolismo , Diglicerídeos de Citidina Difosfato/metabolismo , Regulação Fúngica da Expressão Gênica , Proteínas de Fluorescência Verde , Proteínas de Membrana/biossíntese , Proteínas de Transporte da Membrana Mitocondrial/biossíntese , Proteínas de Transporte da Membrana Mitocondrial/metabolismo , Proteínas Mitocondriais/biossíntese , Fosfatidilgliceróis/metabolismo , Transporte Proteico/fisiologia , Proteínas de Saccharomyces cerevisiae/biossíntese , Proteínas de Saccharomyces cerevisiae/metabolismo , Fatores de Transcrição/biossíntese
17.
J Cereb Blood Flow Metab ; 34(7): 1205-11, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24756078

RESUMO

Fructose has recently been observed to affect brain metabolism and cognitive function in adults. Yet, possible late-onset effects by gestational fructose exposure have not been examined. We evaluated mitochondrial function in the brain of aging (15 months) male offspring of Fischer F344 rat dams fed a high-fructose diet (50% energy from fructose) during gestation and lactation. Maternal fructose exposure caused a significantly lower body weight of the offspring throughout life after weaning, while birth weight, litter size, and body fat percentage were unaffected. Isolated brain mitochondria displayed a significantly increased state 3 respiration of 8%, with the substrate combinations malate/pyruvate, malate/pyruvate/succinate, and malate/pyruvate/succinate/rotenone, as well as a significant decrease in the P/O2 ratio, compared with the control. Uncoupling protein 5 (UCP5) protein levels increased in the fructose group compared with the control (P=0.03) and both UCP5 mRNA and protein levels were inversely correlated with the P/O2 ratio (P=0.008 and 0.03, respectively), suggesting that UCP5 may have a role in the observed decreased phosphorylation efficiency. In conclusion, maternal high-fructose diet during gestation and lactation has long-term effects (fetal programming) on brain mitochondrial function in aging rats, which appears to be linked to an increase in UCP5 protein levels.


Assuntos
Encéfalo/efeitos dos fármacos , Frutose/toxicidade , Mitocôndrias/efeitos dos fármacos , Proteínas de Transporte da Membrana Mitocondrial/biossíntese , Proteínas do Tecido Nervoso/biossíntese , Efeitos Tardios da Exposição Pré-Natal/metabolismo , Animais , Western Blotting , Encéfalo/metabolismo , Respiração Celular/efeitos dos fármacos , Dieta , Feminino , Frutose/administração & dosagem , Masculino , Mitocôndrias/metabolismo , Proteínas de Desacoplamento Mitocondrial , Análise de Sequência com Séries de Oligonucleotídeos , Fosforilação , Gravidez , Ratos , Ratos Endogâmicos F344 , Reação em Cadeia da Polimerase em Tempo Real
18.
Naunyn Schmiedebergs Arch Pharmacol ; 387(1): 47-58, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24062016

RESUMO

During the past 10 years, the use of methamphetamine (METH) has significantly increased in Iran and around the world. The widespread use of 3,4-methylenedioxymethamphetamine as a recreational drug has been responsible for the incidence of several cases of liver failure in young people. This issue made researchers focus on METH toxicity due to the lack of effective treatment and human health risk assessment. There are several reports showing that its long-term use increases the risk for dopamine depletion, but the toxicity mechanisms of METH in liver are not well understood. Therefore, we aimed to investigate the mitochondrial toxicity mechanisms of METH on isolated mitochondria. Rat liver mitochondria were obtained by differential ultracentrifugation, and the isolated mitochondria were then incubated with different concentrations of METH (2.5-20 µM). Our results showed that this agent could induce oxidative stress via rising in mitochondrial reactive oxygen species (ROS) formation, lipid peroxidation, mitochondrial membrane potential collapse, and mitochondrial swelling. In addition, collapse of mitochondrial membrane potential, mitochondrial swelling, and release of cytochrome c following METH treatment were well inhibited by pretreatment of mitochondria with cyclosporin A and butylated hydroxytoluene. Finally, it is suggested that METH could interact with respiratory complexes (II and III) and METH-induced liver toxicity may be the result of its disruptive effect on mitochondrial respiratory chain that is the obvious cause of ROS formation, mitochondrial membrane potential decline, and cytochrome c expulsion which start cell death signaling.


Assuntos
Estimulantes do Sistema Nervoso Central/toxicidade , Metanfetamina/toxicidade , Mitocôndrias Hepáticas/efeitos dos fármacos , Mitocôndrias Hepáticas/metabolismo , Proteínas de Transporte da Membrana Mitocondrial/biossíntese , Espécies Reativas de Oxigênio/metabolismo , Animais , Células Cultivadas , Relação Dose-Resposta a Droga , Masculino , Poro de Transição de Permeabilidade Mitocondrial , Ratos , Ratos Sprague-Dawley
19.
Int J Mol Sci ; 14(8): 16685-705, 2013 Aug 13.
Artigo em Inglês | MEDLINE | ID: mdl-23945562

RESUMO

Correct and timely folding is critical to the function of all proteins. The importance of this is illustrated in the biogenesis of the mitochondrial intermembrane space (IMS) "small Tim" proteins. Biogenesis of the small Tim proteins is regulated by dedicated systems or pathways, beginning with synthesis in the cytosol and ending with assembly of individually folded proteins into functional complexes in the mitochondrial IMS. The process is mostly centered on regulating the redox states of the conserved cysteine residues: oxidative folding is crucial for protein function in the IMS, but oxidized (disulfide bonded) proteins cannot be imported into mitochondria. How the redox-sensitive small Tim precursor proteins are maintained in a reduced, import-competent form in the cytosol is not well understood. Recent studies suggest that zinc and the cytosolic thioredoxin system play a role in the biogenesis of these proteins. In the IMS, the mitochondrial import and assembly (MIA) pathway catalyzes both import into the IMS and oxidative folding of the small Tim proteins. Finally, assembly of the small Tim complexes is a multistep process driven by electrostatic and hydrophobic interactions; however, the chaperone function of the complex might require destabilization of these interactions to accommodate the substrate. Here, we review how folding of the small Tim proteins is regulated during their biogenesis, from maintenance of the unfolded precursors in the cytosol, to their import, oxidative folding, complex assembly and function in the IMS.


Assuntos
Proteínas de Membrana/metabolismo , Mitocôndrias/metabolismo , Proteínas de Transporte da Membrana Mitocondrial/metabolismo , Dobramento de Proteína , Proteínas de Saccharomyces cerevisiae/metabolismo , Sequência de Aminoácidos , Interações Hidrofóbicas e Hidrofílicas , Proteínas de Membrana/biossíntese , Proteínas de Transporte da Membrana Mitocondrial/biossíntese , Proteínas do Complexo de Importação de Proteína Precursora Mitocondrial , Dados de Sequência Molecular , Oxirredução , Oxirredutases/metabolismo , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/biossíntese , Eletricidade Estática
20.
FEBS J ; 280(20): 4960-9, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23937629

RESUMO

The discovery of the mitochondrial intermembrane space assembly (MIA) pathway was followed by studies that focused mainly on the typical small substrates of this disulfide relay system and the interactions between its two central partners: the oxidoreductase Mia40 and the FAD-protein Erv1. Recent studies have revealed that more complex proteins utilize this pathway, including Mia40 itself. In the present study, we dissect the Mia40 biogenesis in distinct stages, supporting a kinetically coordinated sequence of events, starting with (a) import and insertion through the Tim23 translocon, followed by (b) folding of the core of imported Mia40 assisted by the endogenous Mia40 and (c) final interaction with Erv1. The interaction with endogenous Mia40 and the subsequent interaction with Erv1 represent kinetically distinguishable steps that rely on completely different determinants. Interaction with Mia40 proceeds very early (within 30 s) and is characterized by no Cys-specificity, an increased tolerance to mutations of the hydrophobic substrate-binding cleft and no apparent dependence on glutathione as a proofreading mechanism. All of these features illustrate a very atypical behaviour for the Mia40 precursor compared to other substrates of the MIA pathway. By contrast, interaction with Erv1 occurs after 5 min of import and relies on a more stringent specificity.


Assuntos
Proteínas de Transporte da Membrana Mitocondrial/biossíntese , Domínio Catalítico , Redutases do Citocromo/metabolismo , Glutationa/metabolismo , Mitocôndrias/metabolismo , Proteínas de Transporte da Membrana Mitocondrial/química , Proteínas de Transporte da Membrana Mitocondrial/metabolismo , Proteínas do Complexo de Importação de Proteína Precursora Mitocondrial , Oxirredução , Oxirredutases atuantes sobre Doadores de Grupo Enxofre , Ligação Proteica , Dobramento de Proteína , Transporte Proteico
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA