Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
RNA ; 30(7): 839-853, 2024 Jun 17.
Artículo en Inglés | MEDLINE | ID: mdl-38609156

RESUMEN

Several enzymes of intermediary metabolism have been identified to bind RNA in cells, with potential consequences for the bound RNAs and/or the enzyme. In this study, we investigate the RNA-binding activity of the mitochondrial enzyme malate dehydrogenase 2 (MDH2), which functions in the tricarboxylic acid (TCA) cycle and the malate-aspartate shuttle. We confirmed in cellulo RNA binding of MDH2 using orthogonal biochemical assays and performed enhanced cross-linking and immunoprecipitation (eCLIP) to identify the cellular RNAs associated with endogenous MDH2. Surprisingly, MDH2 preferentially binds cytosolic over mitochondrial RNAs, although the latter are abundant in the milieu of the mature protein. Subcellular fractionation followed by RNA-binding assays revealed that MDH2-RNA interactions occur predominantly outside of mitochondria. We also found that a cytosolically retained N-terminal deletion mutant of MDH2 is competent to bind RNA, indicating that mitochondrial targeting is dispensable for MDH2-RNA interactions. MDH2 RNA binding increased when cellular NAD+ levels (MDH2's cofactor) were pharmacologically diminished, suggesting that the metabolic state of cells affects RNA binding. Taken together, our data implicate an as yet unidentified function of MDH2-binding RNA in the cytosol.


Asunto(s)
Ciclo del Ácido Cítrico , Citosol , Malato Deshidrogenasa , Mitocondrias , Unión Proteica , Malato Deshidrogenasa/metabolismo , Malato Deshidrogenasa/genética , Citosol/metabolismo , Citosol/enzimología , Humanos , Mitocondrias/metabolismo , Mitocondrias/genética , Mitocondrias/enzimología , ARN/metabolismo , ARN/genética , ARN Mitocondrial/metabolismo , ARN Mitocondrial/genética , NAD/metabolismo , Células HEK293 , Proteínas de Unión al ARN/metabolismo , Proteínas de Unión al ARN/genética
2.
J Inherit Metab Dis ; 47(2): 270-279, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38084664

RESUMEN

In this study, we investigated the metabolic signatures of different mitochondrial defects (two different complex I and complex V, and the one MDH2 defect) in human skin fibroblasts (HSF). We hypothesized that using a selective culture medium would cause defect specific adaptation of the metabolome and further our understanding of the biochemical implications for the studied defects. All cells were cultivated under galactose stress condition and compared to glucose-based cell culture condition. We investigated the bioenergetic profile using Seahorse XFe96 cell analyzer and assessed the extracellular metabolic footprints and the intracellular metabolic fingerprints using NMR. The galactose-based culture condition forced a bioenergetic switch from a glycolytic to an oxidative state in all cell lines which improved overall separation of controls from the different defect groups. The extracellular metabolome was discriminative for separating controls from defects but not the specific defects, whereas the intracellular metabolome suggests CI and CV changes and revealed clear MDH2 defect-specific changes in metabolites associated with the TCA cycle, malate aspartate shuttle, and the choline metabolism, which are pronounced under galactose condition.


Asunto(s)
Metabolismo Energético , Galactosa , Humanos , Galactosa/metabolismo , Glucólisis , Espectroscopía de Resonancia Magnética , Complejo I de Transporte de Electrón/metabolismo , Fibroblastos/metabolismo , Malato Deshidrogenasa
3.
Int J Mol Sci ; 25(3)2024 Feb 04.
Artículo en Inglés | MEDLINE | ID: mdl-38339168

RESUMEN

Differentiation-inducing factor 1 (DIF-1), found in Dictyostelium discoideum, has antiproliferative and glucose-uptake-promoting activities in mammalian cells. DIF-1 is a potential lead for the development of antitumor and/or antiobesity/antidiabetes drugs, but the mechanisms underlying its actions have not been fully elucidated. In this study, we searched for target molecules of DIF-1 that mediate the actions of DIF-1 in mammalian cells by identifying DIF-1-binding proteins in human cervical cancer HeLa cells and mouse 3T3-L1 fibroblast cells using affinity chromatography and liquid chromatography-tandem mass spectrometry and found mitochondrial malate dehydrogenase (MDH2) to be a DIF-1-binding protein in both cell lines. Since DIF-1 has been shown to directly inhibit MDH2 activity, we compared the effects of DIF-1 and the MDH2 inhibitor LW6 on the growth of HeLa and 3T3-L1 cells and on glucose uptake in confluent 3T3-L1 cells in vitro. In both HeLa and 3T3-L1 cells, DIF-1 at 10-40 µM dose-dependently suppressed growth, whereas LW6 at 20 µM, but not at 2-10 µM, significantly suppressed growth in these cells. In confluent 3T3-L1 cells, DIF-1 at 10-40 µM significantly promoted glucose uptake, with the strongest effect at 20 µM DIF-1, whereas LW6 at 2-20 µM significantly promoted glucose uptake, with the strongest effect at 10 µM LW6. Western blot analyses showed that LW6 (10 µM) and DIF-1 (20 µM) phosphorylated and, thus, activated AMP kinase in 3T3-L1 cells. Our results suggest that MDH2 inhibition can suppress cell growth and promote glucose uptake in the cells, but appears to promote glucose uptake more strongly than it suppresses cell growth. Thus, DIF-1 may promote glucose uptake, at least in part, via direct inhibition of MDH2 and a subsequent activation of AMP kinase in 3T3-L1 cells.


Asunto(s)
Glucosa , Malato Deshidrogenasa , Animales , Humanos , Ratones , Células 3T3-L1/efectos de los fármacos , Células 3T3-L1/metabolismo , Adenilato Quinasa/metabolismo , Dictyostelium/metabolismo , Glucosa/metabolismo , Células HeLa/efectos de los fármacos , Células HeLa/metabolismo , Malato Deshidrogenasa/antagonistas & inhibidores , Malato Deshidrogenasa/metabolismo , Mamíferos/metabolismo
4.
Mol Ther ; 29(9): 2873-2885, 2021 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-33895326

RESUMEN

Dysregulated long non-coding RNAs (lncRNAs) have been shown to contribute to the pathogenesis of ischemic stroke. However, the potential role of lncRNAs in post-stroke microglial activation remains largely unknown. Here, we uncovered that lncRNA-U90926 was significantly increased in microglia exposed to ischemia/reperfusion both in vivo and in vitro. In addition, adenovirus-associated virus (AAV)-mediated microglial U90926 silencing alleviated neurological deficits and reduced infarct volume in experimental stroke mice. Microglial U90926 knockdown could reduce the infiltration of neutrophils into ischemic lesion site, which might be attributed to the downregulation of C-X-C motif ligand 2 (CXCL2). Mechanistically, U90926 directly bound to malate dehydrogenase 2 (MDH2) and competitively inhibited the binding of MDH2 to the CXCL2 3' untranslated region (UTR), thus protecting against MDH2-mediated decay of CXCL2 mRNA. Taken together, our study demonstrated that microglial U90926 aggravated ischemic brain injury via facilitating neutrophil infiltration, suggesting that U90926 might be a potential biomarker and therapeutic target for ischemic stroke.


Asunto(s)
Quimiocina CXCL2/genética , Accidente Cerebrovascular Isquémico/inmunología , Malato Deshidrogenasa/genética , Microglía/química , ARN Largo no Codificante/genética , Regulación hacia Arriba , Regiones no Traducidas 5' , Animales , Células Cultivadas , Modelos Animales de Enfermedad , Células HEK293 , Humanos , Accidente Cerebrovascular Isquémico/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Infiltración Neutrófila , Cultivo Primario de Células
5.
Cancer Sci ; 112(11): 4501-4514, 2021 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-34424600

RESUMEN

Long non-coding RNA AC020978 (lncRNA AC020978) is an oncogenic regulator of non-small cell lung cancer (NSCLC). However, the function of AC020978 in regulating NSCLC metastasis and the potential molecular mechanism remains largely unknown. In this study, we evaluated the expression levels of AC020978 in a series of NSCLC tissues using FISH assays and found that higher AC020978 expression levels were closely associated with metastasis and unfavorable prognosis. Functional studies showed that AC020978 promoted NSCLC migration and invasion both in vitro and in vivo. Further investigation demonstrated that AC020978 interacted with malate dehydrogenase 2 (MDH2) and maintained MDH2 stability. Knockdown of MDH2 weakened the facilitating effect on cell metastasis and 2-hydroxyglutarate (2-HG) metabolism in AC020978-overexpressed NSCLC cells. RNA sequencing, bioinformatic analysis, and western blotting revealed that AC020978 was associated with the AKT signaling pathway. Taken together, our findings revealed that AC020978 might serve as a prognostic biomarker and activate the AKT pathway by stabilizing MDH2, leading to metastasis and progression of NSCLC.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Progresión de la Enfermedad , Neoplasias Pulmonares/metabolismo , Malato Deshidrogenasa/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , ARN Largo no Codificante/metabolismo , Células A549 , Animales , Carcinoma de Pulmón de Células no Pequeñas/mortalidad , Carcinoma de Pulmón de Células no Pequeñas/patología , Carcinoma de Pulmón de Células no Pequeñas/secundario , Línea Celular Tumoral , Movimiento Celular , Femenino , Glutaratos/metabolismo , Humanos , Hibridación Fluorescente in Situ , Neoplasias Pulmonares/mortalidad , Neoplasias Pulmonares/patología , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Persona de Mediana Edad , Invasividad Neoplásica , Pronóstico
6.
J Mol Evol ; 89(4-5): 214-224, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33604781

RESUMEN

Studying the diversity of energy production pathways is important for understanding the evolutionary relationships between metabolic pathways and their biochemical precursors. The lactate/malate dehydrogenase (LDH/MDH) superfamily has been a model system for structural and functional evolution for a long time. Recently, the type-2 family of LDH/MDH (or LDH2/MDH2 oxidoreductase) has been identified. The LDH2/MDH2 oxidoreductase family is now known to have functionally more diverse enzymes than the LDH/MDH superfamily. In channel catfish, the gene encoding the LDH2/MDH2 oxidoreductase has been found (and was provisionally termed AqE). Homologs of this enzyme are predominantly present in organisms living in an aquatic environment. In this work, we studied the AqE gene distribution among non-tetrapod vertebrates. It was found that the AqE gene is present in the genomes of bony and cartilaginous fish and in the genomes of hagfishes and lampreys. In addition, it has been confirmed that in representatives of Cypriniformes, the AqE gene has been lost. AqE in representatives of Salmoniformes underwent significant deletions, which most likely led to its pseudogenization. In most orders of non-Tetrapoda vertebrates, the AqE gene remains highly conserved, suggesting that the AqE gene in aquatic vertebrates is an essential gene and undergoes rigorous selection. The AqE gene has the highest sequence similarity with the archaeal ComC gene that encodes sulfolactate dehydrogenase (SLDH). Based on the similarity of substrates, the enzyme encoded by the AqE gene is likely involved in the malate-aspartate shuttle mechanism or the biosynthesis of the energy coenzyme M equivalent.


Asunto(s)
Anguila Babosa , Vertebrados , Secuencia de Aminoácidos , Animales , L-Lactato Deshidrogenasa/metabolismo , Lampreas/metabolismo , Vertebrados/genética
7.
J Inherit Metab Dis ; 44(4): 792-808, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33990986

RESUMEN

Over the last few years, various inborn disorders have been reported in the malate aspartate shuttle (MAS). The MAS consists of four metabolic enzymes and two transporters, one of them having two isoforms that are expressed in different tissues. Together they form a biochemical pathway that shuttles electrons from the cytosol into mitochondria, as the inner mitochondrial membrane is impermeable to the electron carrier NADH. By shuttling NADH across the mitochondrial membrane in the form of a reduced metabolite (malate), the MAS plays an important role in mitochondrial respiration. In addition, the MAS maintains the cytosolic NAD+ /NADH redox balance, by using redox reactions for the transfer of electrons. This explains why the MAS is also important in sustaining cytosolic redox-dependent metabolic pathways, such as glycolysis and serine biosynthesis. The current review provides insights into the clinical and biochemical characteristics of MAS deficiencies. To date, five out of seven potential MAS deficiencies have been reported. Most of them present with a clinical phenotype of infantile epileptic encephalopathy. Although not specific, biochemical characteristics include high lactate, high glycerol 3-phosphate, a disturbed redox balance, TCA abnormalities, high ammonia, and low serine, which may be helpful in reaching a diagnosis in patients with an infantile epileptic encephalopathy. Current implications for treatment include a ketogenic diet, as well as serine and vitamin B6 supplementation.


Asunto(s)
Aspartato Aminotransferasas/deficiencia , Ácido Aspártico/metabolismo , Malato Deshidrogenasa/deficiencia , Malatos/metabolismo , Errores Innatos del Metabolismo/patología , Mitocondrias/patología , Animales , Aspartato Aminotransferasas/genética , Respiración de la Célula , Humanos , Lactante , Malato Deshidrogenasa/genética , Errores Innatos del Metabolismo/etiología , Errores Innatos del Metabolismo/metabolismo , Mitocondrias/metabolismo , Espasmos Infantiles/etiología
8.
Appl Microbiol Biotechnol ; 104(11): 4971-4983, 2020 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-32248437

RESUMEN

During alcohol fermentation, Saccharomyces cerevisiae produces organic acids, including succinate, acetate, and malate. Since malate contributes to the pleasant flavor of sake (a Japanese alcoholic beverage), various methods for breeding high-malate-producing yeast have been developed. We previously isolated a high-malate-producing strain and found that a missense mutation in GID4 was responsible for the high-malate-producing phenotype. Gid4 is a component of the GID (glucose-induced degradation-deficient) complex and stimulates the catabolic degradation of gluconeogenic enzymes. In this study, the mechanism by which this mutation led to high malate production in yeast cells was investigated. The evaluation of disruptants and mutants of gluconeogenic enzymes revealed that cytosolic malate dehydrogenase (Mdh2) participated in the malate production. Furthermore, target proteome analysis indicated that an increase in malate production resulted from the accumulation of Mdh2 in gid4 disruptant due to the loss of GID complex-mediated degradation. Next, we investigated the effects of GID protein-coding genes (GID1-GID9) on organic acid production and enzyme expression profiles in yeast. The disruptants of GID1, 2, 3, 4, 5, 8, and 9 exhibited high malate production. Comparison of protein abundance among the GID disruptants revealed variations in protein expression profiles, including in glycolysis and tricarboxylic acid cycle-related enzymes. The high-malate-producing disruptants showed the activation of several glycolytic enzymes and a reduction in enzymes involved in the conversion of pyruvate to ethanol. Our results suggest that high-malate-producing disruptants adapt their metabolism to produce malate in excess via the regulation of protein expression in glucose assimilation and ethanol fermentation. KEY POINTS: An increase in malate level of GID4 mutant resulted from the accumulation of Mdh2. The disruptants of GID1, 2, 3, 4, 5, 8, and 9 showed high malate production. The protein expression profiles in the GID disruptants differed from one another.


Asunto(s)
Malatos/metabolismo , Mutación Missense , Proteínas de Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/enzimología , Saccharomyces cerevisiae/genética , Ciclo del Ácido Cítrico/genética , Alimentos Fermentados/microbiología , Regulación Fúngica de la Expresión Génica , Glucólisis/genética , Malato Deshidrogenasa/metabolismo , Proteómica , Proteínas de Saccharomyces cerevisiae/metabolismo
9.
J Biol Chem ; 292(52): 21457-21465, 2017 12 29.
Artículo en Inglés | MEDLINE | ID: mdl-29122887

RESUMEN

Although it is widely appreciated that the use of global translation inhibitors, such as cycloheximide, in protein degradation assays may result in artefacts, these inhibitors continue to be employed, owing to the absence of robust alternatives. We describe here the promoter reference technique (PRT), an assay for protein degradation with two advantageous features: a reference protein and a gene-specific inhibition of translation. In PRT assays, one measures, during a chase, the ratio of a test protein to a long-lived reference protein, a dihydrofolate reductase (DHFR). The test protein and DHFR are coexpressed, in the yeast Saccharomyces cerevisiae, on a low-copy plasmid from two identical P TDH3 promoters containing additional, previously developed DNA elements. Once transcribed, these elements form 5'-RNA aptamers that bind to the added tetracycline, which represses translation of aptamer-containing mRNAs. The selectivity of repression avoids a global inhibition of translation. This selectivity is particularly important if a component of a relevant proteolytic pathway (e.g. a specific ubiquitin ligase) is itself short-lived. We applied PRT to the Pro/N-end rule pathway, whose substrates include the short-lived Mdh2 malate dehydrogenase. Mdh2 is targeted for degradation by the Gid4 subunit of the GID ubiquitin ligase. Gid4 is also a metabolically unstable protein. Through analyses of short-lived Mdh2 as a target of short-lived Gid4, we illustrate the advantages of PRT over degradation assays that lack a reference and/or involve cycloheximide. In sum, PRT avoids the use of global translation inhibitors during a chase and also provides a "built-in" reference protein.


Asunto(s)
Bioensayo/métodos , Productos Finales de Degradación de Proteínas/análisis , Secuencia de Aminoácidos , Malato Deshidrogenasa , Plásmidos , Regiones Promotoras Genéticas/genética , Inhibidores de la Síntesis de la Proteína , Proteolisis/efectos de los fármacos , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Tetrahidrofolato Deshidrogenasa , Proteínas de Transporte Vesicular
10.
Genet Med ; 20(12): 1652-1662, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30008476

RESUMEN

PURPOSE: MDH2 (malate dehydrogenase 2) has recently been proposed as a novel potential pheochromocytoma/paraganglioma (PPGL) susceptibility gene, but its role in the disease has not been addressed. This study aimed to determine the prevalence of MDH2 pathogenic variants among PPGL patients and determine the associated phenotype. METHODS: Eight hundred thirty patients with PPGLs, negative for the main PPGL driver genes, were included in the study. Interpretation of variants of unknown significance (VUS) was performed using an algorithm based on 20 computational predictions, by implementing cell-based enzymatic and immunofluorescence assays, and/or by using a molecular dynamics simulation approach. RESULTS: Five variants with potential involvement in pathogenicity were identified: three missense (p.Arg104Gly, p.Val160Met and p.Ala256Thr), one in-frame deletion (p.Lys314del), and a splice-site variant (c.429+1G>T). All were germline and those with available biochemical data, corresponded to noradrenergic PPGL. CONCLUSION: This study suggests that MDH2 pathogenic variants may play a role in PPGL susceptibility and that they might be responsible for less than 1% of PPGLs in patients without pathogenic variants in other major PPGL driver genes, a prevalence similar to the one recently described for other PPGL genes. However, more epidemiological data are needed to recommend MDH2 testing in patients negative for other major PPGL genes.


Asunto(s)
Neoplasias de las Glándulas Suprarrenales/genética , Malato Deshidrogenasa/genética , Paraganglioma/genética , Feocromocitoma/genética , Neoplasias de las Glándulas Suprarrenales/patología , Adulto , Femenino , Predisposición Genética a la Enfermedad , Mutación de Línea Germinal , Humanos , Masculino , Persona de Mediana Edad , Mutación Missense , Paraganglioma/patología , Feocromocitoma/patología , Isoformas de Proteínas
11.
Biochim Biophys Acta Mol Basis Dis ; 1863(6): 1492-1499, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28433711

RESUMEN

Voltage-gated sodium channel α-subunit type I (NaV1.1, encoded by SCN1A gene) plays a critical role in the excitability of brain. Downregulation of SCN1A expression is associated with epilepsy, a common neurological disorder characterized by recurrent seizures. Here we reveal a novel role of malate dehydrogenase 2 (MDH2) in the posttranscriptional regulation of SCN1A expression under seizure condition. We identified that MDH2 was an RNA binding protein that could bind two of the four conserved regions in the 3' UTRs of SCN1A. We further showed that knockdown of MDH2 or inactivation of MDH2 activity in HEK-293 cells increased the reporter gene expression through the 3' UTR of SCN1A, and MDH2 overexpression decreased gene expression by affecting mRNA stability. In the hippocampus of seizure mice, the upregulation of MDH2 expression contributed to the decrease of the NaV1.1 levels at posttranscriptional level. In addition, we showed that the H2O2 levels increased in the hippocampus of the seizure mice, and H2O2 could promote the binding of MDH2 to the binding sites of Scn1a gene, whereas ß-mercaptoethanol decreased the binding capability, indicating an important effect of the seizure-induced oxidation on the MDH2-mediated downregulation of Scn1a expression. Taken together, these data suggest that MDH2, functioning as an RNA-binding protein, is involved in the posttranscriptional downregulation of SCN1A expression under seizure condition.


Asunto(s)
Regiones no Traducidas 3' , Regulación hacia Abajo , Malato Deshidrogenasa/metabolismo , Canal de Sodio Activado por Voltaje NAV1.1/biosíntesis , Proteínas de Unión al ARN/metabolismo , Convulsiones/metabolismo , Animales , Células HEK293 , Hipocampo/metabolismo , Hipocampo/patología , Humanos , Malato Deshidrogenasa/genética , Ratones , Canal de Sodio Activado por Voltaje NAV1.1/genética , Proteínas de Unión al ARN/genética , Convulsiones/genética , Convulsiones/patología
12.
Mol Biotechnol ; 2024 Feb 21.
Artículo en Inglés | MEDLINE | ID: mdl-38381377

RESUMEN

The pathogenesis of endometrial cancer (EC) involves the regulation of lactate dehydrogenases. However, the role and mechanism of lactate dehydrogenase-B (LDHB) in EC progression have not been studied. The mRNA levels of LDHB and malate dehydrogenase 2 (MDH2) were detected by quantitative real-time polymerase chain reaction. Protein expression was checked by western blotting and immunohistochemistry assays. Cell proliferation, apoptosis, and invasion were analyzed by 5-Ethynyl-2'-deoxyuridine, transwell, and flow cytometry assay, respectively. Glycolysis was investigated using Glucose Assay Kit, CheKine™ Micro Lactate Assay Kit, and ADP/ATP ratio assay kit. An in vivo tumor formation assay was conducted to disclose the effect of LDHB on tumor growth in vivo. The associations among signal transducer and activator of transcription 3 (STAT3), LDHB, and MDH2 were predicted through JASPAR or GeneMANIA online database and identified by chromatin immunoprecipitation assay, dual-luciferase reporter assay, and co-immunoprecipitation assay. LDHB expression was increased in EC tissues and cells in comparison with normal endometrial tissues and human endometrial stromal cells. LDHB had the potential as a biomarker to predict the prognosis of EC patients. In addition, LDHB knockdown inhibited the proliferation, invasion, and glycolysis and promoted apoptosis of RL95-2 and Ishikawa cells. LDHB knockdown inhibited tumor property of Ishikawa cells in vivo. STAT3 bound to the promoter region of LDHB, and STAT3 silencing-induced effects were relieved after LDHB upregulation. LDHB interacted with and regulated MDH2 expression. Moreover, MDH2 overexpression rescued LDHB knockdown-induced effects on EC cell phenotypes. STAT3-activated LDHB promoted endometrial cancer cell malignancy by inducing MDH2 production.

13.
Biochem Biophys Res Commun ; 441(1): 77-82, 2013 Nov 08.
Artículo en Inglés | MEDLINE | ID: mdl-24134846

RESUMEN

Previously, we identified proteins showing a differential acetylation pattern during adipogenic differentiation. Here, we examined the role of malate dehydrogenase 2 (MDH2) acetylation in the adipogenesis of 3T3-L1 preadipocytes. The acetylation level of MDH2 showed a dramatic increase during adipogenesis. The overexpression of wild-type MDH2 induced the significant acceleration of adipogenic differentiation. On the other hand, the acetylation-block mutant MDH2 showed significantly reduced adipogenic differentiation compared to the wild type. MDH2 acetylation enhances its enzymatic activity and consequently intracellular NADPH level. These results suggest that the acetylation of MDH2 was affected by the cellular energy state and subsequently regulated adipogenic differentiation.


Asunto(s)
Adipogénesis , Diferenciación Celular , Malato Deshidrogenasa/metabolismo , Células 3T3-L1 , Acetilación/efectos de los fármacos , Adipocitos/citología , Adipocitos/efectos de los fármacos , Adipocitos/enzimología , Adipogénesis/efectos de los fármacos , Animales , Diferenciación Celular/efectos de los fármacos , Espacio Extracelular/efectos de los fármacos , Espacio Extracelular/metabolismo , Glucosa/farmacología , Ratones , Mutación/genética , NADP/metabolismo
14.
Cancers (Basel) ; 15(24)2023 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-38136396

RESUMEN

A significant fraction of breast cancer recurs, with lethal outcome, but specific genetic variants responsible have yet to be identified. Five cousin pairs with recurrent breast cancer from pedigrees with a statistical excess of recurrent breast cancer were sequenced to identify rare, shared candidate predisposition variants. The candidates were tested for association with breast cancer risk with UKBiobank data. Additional breast cancer cases were assayed for a subset of candidate variants to test for co-segregation. Three-dimensional protein structure prediction methods were used to investigate how the mutation under consideration is predicted to change structural and electrostatic properties in the mutated protein. One hundred and eighty-one rare candidate predisposition variants were shared in at least one cousin pair from a high-risk pedigree. A rare variant in MDH2 was found to segregate with breast-cancer-affected relatives in one extended pedigree. MDH2 is an estrogen-stimulated gene encoding the protein malate dehydrogenase, which catalyzes the reversible oxidation of malate to oxaloacetate. The molecular simulation results strongly suggest that the mutation changes the NAD+ binding pocket electrostatics of MDH2. This small sequencing study, using a powerful approach based on recurrent breast cancer cases from high-risk pedigrees, identified a set of strong candidate variants for inherited predisposition for breast cancer recurrence, including MDH2, which should be pursued in other resources.

15.
Sci China Life Sci ; 65(10): 2017-2030, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-35366151

RESUMEN

Epithelial ovarian cancer (EOC) exhibits strong dependency on the tricarboxylic acid (TCA) cycle and oxidative phosphorylation to fuel anabolic process. Here, we show that malate dehydrogenase 2 (MDH2), a key enzyme of the TCA cycle, is palmitoylated at cysteine 138 (C138) residue, resulting in increased activity of MDH2. We next identify that ZDHHC18 acts as a palmitoyltransferase of MDH2. Glutamine deprivation enhances MDH2 palmitoylation by increasing the binding between ZDHHC18 and MDH2. MDH2 silencing represses mitochondrial respiration as well as ovarian cancer cell proliferation both in vitro and in vivo. Intriguingly, re-expression of wild-type MDH2, but not its palmitoylation-deficient C138S mutant, sustains mitochondrial respiration and restores the growth as well as clonogenic capability of ovarian cancer cells. Notably, MDH2 palmitoylation level is elevated in clinical cancer samples from patients with high-grade serous ovarian cancer. These observations suggest that MDH2 palmitoylation catalyzed by ZDHHC18 sustains mitochondrial respiration and promotes the malignancy of ovarian cancer, yielding possibilities of targeting ZDHHC18-mediated MDH2 palmitoylation in the treatment of EOC.


Asunto(s)
Malato Deshidrogenasa , Neoplasias Ováricas , Carcinoma Epitelial de Ovario , Línea Celular Tumoral , Cisteína , Femenino , Glutamina , Humanos , Lipoilación , Malato Deshidrogenasa/química , Malato Deshidrogenasa/metabolismo , Respiración , Ácidos Tricarboxílicos
16.
Front Endocrinol (Lausanne) ; 13: 1070074, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36760809

RESUMEN

Introduction: The percentage of patients diagnosed with pheochromocytoma and paraganglioma (altogether PPGL) carrying known germline mutations in one of the over fifteen susceptibility genes identified to date has dramatically increased during the last two decades, accounting for up to 35-40% of PPGL patients. Moreover, the application of NGS to the diagnosis of PPGL detects unexpected co-occurrences of pathogenic allelic variants in different susceptibility genes. Methods: Herein we uncover several cases with dual mutations in NF1 and other PPGL genes by targeted sequencing. We studied the molecular characteristics of the tumours with co-occurrent mutations, using omic tools to gain insight into the role of these events in tumour development. Results: Amongst 23 patients carrying germline NF1 mutations, targeted sequencing revealed additional pathogenic germline variants in DLST (n=1) and MDH2 (n=2), and two somatic mutations in H3-3A and PRKAR1A. Three additional patients, with somatic mutations in NF1 were found carrying germline pathogenic mutations in SDHB or DLST, and a somatic truncating mutation in ATRX. Two of the cases with dual germline mutations showed multiple pheochromocytomas or extra-adrenal paragangliomas - an extremely rare clinical finding in NF1 patients. Transcriptional and methylation profiling and metabolite assessment showed an "intermediate signature" to suggest that both variants had a pathological role in tumour development. Discussion: In conclusion, mutations affecting genes involved in different pathways (pseudohypoxic and receptor tyrosine kinase signalling) co-occurring in the same patient could provide a selective advantage for the development of PPGL, and explain the variable expressivity and incomplete penetrance observed in some patients.


Asunto(s)
Neoplasias de las Glándulas Suprarrenales , Paraganglioma , Feocromocitoma , Humanos , Feocromocitoma/patología , Predisposición Genética a la Enfermedad , Paraganglioma/patología , Mutación , Neoplasias de las Glándulas Suprarrenales/diagnóstico
17.
J Exp Clin Cancer Res ; 41(1): 349, 2022 Dec 17.
Artículo en Inglés | MEDLINE | ID: mdl-36527092

RESUMEN

BACKGROUND: To identify potential targets related to nicotinamide adenine dinucleotide (NAD+) metabolism in gliomas, we used RNA immunoprecipitation to identify a novel long noncoding RNA renamed malate dehydrogenase degradation helper (MDHDH) (NONCODE annotation ID: NONHSAT138800.2, NCBI Reference Sequence: NR_028345), which bound to MDH2 (malate dehydrogenase 2), that is downregulated in glioblastoma multiforme (GBM) and associated with metabolic regulation. However, its underlying mechanisms in the progression of GBM have not been well studied. METHODS: To investigate the clinical significance of MDHDH, we analyzed its expression levels in publicly available datasets and collected clinical samples from Shandong Provincial Hospital, affiliated with Shandong University. Functional assays, including FISH/CISH, CCK8, EdU, wound healing, and transwell assays, were used to determine the cellular/subcellular localization, tissue expression profile and anti-oncogenic role of MDHDH. Furthermore, RNA pulldown, mass spectrometry RNA immunoprecipitation, coimmunoprecipitation, JC-1 probe, and cell energy-production assays were used to determine the mechanisms of MDHDH in the development of GBM. Animal experiments were conducted to determine the antitumorigenic role of MDHDH in GBM in vivo. RESULTS: In public datasets, MDHDH expression was significantly downregulated in GBM and LGG compared with GTEx normal brain tissues. The results of the tissue microarray showed that the MDHDH expression level negatively correlated with the tumor grade. Altered MDHDH expression led to significant changes in the proliferation, migration and invasion of GBM cells both in vitro and in vivo. Mechanistically, we found that MDHDH directly bound to MDH2 and PSMA1 (20S proteasomal core subunit alpha-type 1) as a molecular scaffold and accelerated the degradation of MDH2 by promoting the binding of ubiquitinated MDH2 to the proteasome. The degradation of MDH2 subsequently led to changes in the mitochondrial membrane potential and NAD+/NADH ratio, which impeded glycolysis in glioma cells. CONCLUSIONS: In conclusion, this study broadened our understanding of the functions of lncRNAs in GBM. We demonstrated that the tumor suppressor MDHDH might act as a clinical biomarker and that the overexpression of MDHDH might be a novel synergistic strategy for enhancing metabolism-based, epigenetic-based, and autophagy regulation-based therapies with clinical benefits for glioblastoma multiforme patients.


Asunto(s)
Neoplasias Encefálicas , Glioblastoma , Glioma , ARN Largo no Codificante , Animales , Glioblastoma/patología , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , NAD/genética , NAD/metabolismo , NAD/uso terapéutico , Neoplasias Encefálicas/patología , Malato Deshidrogenasa/genética , Malato Deshidrogenasa/metabolismo , Proliferación Celular/genética , Regulación Neoplásica de la Expresión Génica , Línea Celular Tumoral , Glioma/genética , Autofagia/genética
18.
Front Immunol ; 13: 962986, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36159820

RESUMEN

Focal segmental glomerulosclerosis (FSGS) has an over 30% risk of recurrence after kidney transplantation (Ktx) and is associated with an extremely high risk of graft loss. However, mechanisms remain largely unclear. Thus, this study identifies novel genes related to the recurrence of FSGS (rFSGS). Whole genome-wide sequencing and next-generation RNA sequencing were used to identify the candidate mutant genes associated with rFSGS in peripheral blood mononuclear cells (PBMCs) from patients with biopsy-confirmed rFSGS after KTx. To confirm the functional role of the identified gene with the MDH2 c.26C >T mutation, a homozygous MDH2 c.26C >T mutation in HMy2.CIR cell line was induced by CRISPR/Cas9 and co-cultured with podocytes, mesangial cells, or HK2 cells, respectively, to detect the potential pathogenicity of the c.26C >T variant in MDH2. A total of 32 nonsynonymous single nucleotide polymorphisms (SNPs) and 610 differentially expressed genes (DEGs) related to rFSGS were identified. DEGs are mainly enriched in the immune and metabolomic-related pathways. A variant in MDH2, c.26C >T, was found in all patients with rFSGS, which was also accompanied by lower levels of mRNA expression in PBMCs from relapsed patients compared with patients with remission after KTx. Functionally, co-cultures of HMy2.CIR cells overexpressing the mutant MDH2 significantly inhibited the expression of synaptopodin, podocin, and F-actin by podocytes compared with those co-cultured with WT HMy2.CIR cells or podocytes alone. We identified that MDH2 is a novel rFSGS susceptibility gene in patients with recurrence of FSGS after KTx. Mutation of the MDH2 c.26C >T variant may contribute to progressive podocyte injury in rFSGS patients.


Asunto(s)
Glomeruloesclerosis Focal y Segmentaria , Actinas/genética , Genómica , Glomeruloesclerosis Focal y Segmentaria/genética , Humanos , Leucocitos Mononucleares , Malato Deshidrogenasa/genética , Mutación , ARN Mensajero , Recurrencia , Transcriptoma
19.
Biochim Biophys Acta Bioenerg ; 1863(3): 148532, 2022 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-35063410

RESUMEN

The mitochondrial respiratory chain (RC) enables many metabolic processes by regenerating both mitochondrial and cytosolic NAD+ and ATP. The oxidation by the RC of the NADH metabolically produced in the cytosol involves redox shuttles as the malate-aspartate shuttle (MAS) and is of paramount importance for cell fate. However, the specific metabolic regulations allowing mitochondrial respiration to prioritize NADH oxidation in response to high NADH/NAD+ redox stress have not been elucidated. The recent discovery that complex I (NADH dehydrogenase), and not complex II (Succinate dehydrogenase), can assemble with other respiratory chain complexes to form functional entities called respirasomes, led to the assumption that this supramolecular organization would favour NADH oxidation. Unexpectedly, characterization of heart and liver mitochondria demonstrates that the RC systematically favours electrons provided by the 'respirasome free' complex II. Our results demonstrate that the preferential succinate driven respiration is tightly controlled by OAA levels, and that OAA feedback inhibition of complex II rewires RC fuelling increasing NADH oxidation capacity. This new regulatory mechanism synergistically increases RC's NADH oxidative capacity and rewires MDH2 driven anaplerosis of the TCA, preventing malate production from succinate to favour oxidation of cytosolic malate. This regulatory mechanism synergistically adjusts RC and TCA fuelling in response to extramitochondrial malate produced by the MAS.


Asunto(s)
NAD , Ácido Succínico , Respiración de la Célula , Ciclo del Ácido Cítrico , Transporte de Electrón , NAD/metabolismo
20.
Mol Genet Metab Rep ; 33: 100931, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-36420423

RESUMEN

Malate dehydrogenases (MDH) serve a critical role in maintaining equilibrium of the NAD+/NADH ratio between the mitochondria and cytosol through the catalysis of the oxidation of L-malate to oxaloacetate in a reversible, NADH-dependent manner. MDH2 encodes the mitochondrial isoform, which is integral to the tricarboxylic acid cycle and thus energy homeostasis. Recently, five patients harboring compound heterozygous MDH2 variants have been described, three with early-onset epileptic encephalopathy, one with a stroke-like episode, and one with dilated cardiomyopathy. Here, we describe an additional seven patients with biallelic variants in MDH2, the largest and most neurodevelopmentally and ethnically diverse cohort to-date, including homozygous variants, a sibling pair, non-European patients, and an adult. From these patients, we learn that MDH2 deficiency results in a biochemical signature including elevations of plasma lactate and the lactate:pyruvate ratio with urinary excretion of malate. It also results in a recognizable constellation of neuroimaging findings of anterior-predominant cerebral atrophy, subependymal cysts with ventricular septations. We also recognize MDH2 deficiency as a cause of Leigh syndrome. Taken with existing patient reports, we conclude that MDH2 deficiency is an emerging and likely under-recognized cause of infantile epileptic encephalopathy and provide a framework for medical evaluation of patients identified with biallelic MDH2 variants.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA