Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 186
Filtrar
Más filtros

País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
PLoS Pathog ; 17(1): e1009122, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33411818

RESUMEN

Lactic acidosis and hyperlactatemia are common metabolic disturbances in patients with severe malaria. Lactic acidosis causes physiological adverse effects, which can aggravate the outcome of malaria. Despite its clear association with mortality in malaria patients, the etiology of lactic acidosis is not completely understood. In this review, the possible contributors to lactic acidosis and hyperlactatemia in patients with malaria are discussed. Both increased lactate production and impaired lactate clearance may play a role in the pathogenesis of lactic acidosis. The increased lactate production is caused by several factors, including the metabolism of intraerythrocytic Plasmodium parasites, aerobic glycolysis by activated immune cells, and an increase in anaerobic glycolysis in hypoxic cells and tissues as a consequence of parasite sequestration and anemia. Impaired hepatic and renal lactate clearance, caused by underlying liver and kidney disease, might further aggravate hyperlactatemia. Multiple factors thus participate in the etiology of lactic acidosis in malaria, and further investigations are required to fully understand their relative contributions and the consequences of this major metabolic disturbance.


Asunto(s)
Acidosis Láctica/etiología , Malaria/complicaciones , Plasmodium/fisiología , Acidosis Láctica/patología , Humanos
2.
Ultrastruct Pathol ; 47(3): 227-235, 2023 May 04.
Artículo en Inglés | MEDLINE | ID: mdl-36871223

RESUMEN

A biopsy of gastrocnemius muscle from a patient with mitochondrial encephalomyopathy with lactic acidosis and stroke-like episodes (MELAS) syndrome was studied histologically in semithin sections stained by hematoxylin-and-eosin (H&E) and toluidine blue, and ultrathin sections by transmission electron microscopy (TEM). H&E stain demonstrated typical ragged-red fibers (RRFs) and affected fibers in fascicles. Toluidine-blue stain showed an irregular meshwork in the center of RRFs. TEM demonstrated damaged myofibrils and variations in mitochondrial structure in RRFs and affected fibers. Dense mitochondria were compacted with cristae and pleomorphic electron-dense inclusions. Lucent mitochondria included paracrystalline inclusions with a parking lot appearance. At high magnification, the paracrystalline inclusions were composed of plates that paralleled and connected with mitochondrial cristae. These observations indicated that electron-dense granular and paracrystalline inclusions resulted from cristal degeneration and overlapping in mitochondria in MELAS syndrome.


Asunto(s)
Acidosis Láctica , Síndrome MELAS , Accidente Cerebrovascular , Humanos , Acidosis Láctica/patología , Síndrome MELAS/patología , Accidente Cerebrovascular/patología , Músculo Esquelético/patología , Mitocondrias/patología
3.
Int J Mol Sci ; 24(24)2023 Dec 06.
Artículo en Inglés | MEDLINE | ID: mdl-38139018

RESUMEN

Mitochondrial encephalomyopathy, lactic acidosis, and stroke-like episode (MELAS) syndrome, caused by a single base substitution in mitochondrial DNA (m.3243A>G), is one of the most common maternally inherited mitochondrial diseases accompanied by neuronal damage due to defects in the oxidative phosphorylation system. There is no established treatment. Our previous study reported a superior restoration of mitochondrial function and bioenergetics in mitochondria-deficient cells using highly purified mesenchymal stem cells (RECs). However, whether such exogenous mitochondrial donation occurs in mitochondrial disease models and whether it plays a role in the recovery of pathological neuronal functions is unknown. Here, utilizing induced pluripotent stem cells (iPSC), we differentiated neurons with impaired mitochondrial function from patients with MELAS. MELAS neurons and RECs/mesenchymal stem cells (MSCs) were cultured under contact or non-contact conditions. Both RECs and MSCs can donate mitochondria to MELAS neurons, but RECs are more excellent than MSCs for mitochondrial transfer in both systems. In addition, REC-mediated mitochondrial transfer significantly restored mitochondrial function, including mitochondrial membrane potential, ATP/ROS production, intracellular calcium storage, and oxygen consumption rate. Moreover, mitochondrial function was maintained for at least three weeks. Thus, REC-donated exogenous mitochondria might offer a potential therapeutic strategy for treating neurological dysfunction in MELAS.


Asunto(s)
Acidosis Láctica , Síndrome MELAS , Células Madre Mesenquimatosas , Enfermedades Mitocondriales , Humanos , Síndrome MELAS/genética , Síndrome MELAS/terapia , Mitocondrias/genética , Acidosis Láctica/metabolismo , Acidosis Láctica/patología , ADN Mitocondrial/metabolismo , Enfermedades Mitocondriales/metabolismo , Neuronas/patología , Células Madre Mesenquimatosas/metabolismo
4.
Acta Neurol Taiwan ; 32(2): 74-78, 2023 Jun 30.
Artículo en Inglés | MEDLINE | ID: mdl-37198511

RESUMEN

BACKGROUND: During corona virus pandemic, various neurological complications of COVID-19 have been reported. Recent studies demonstrated different pathophysiology for neurological manifestations of COVID-19 such as mitochondrial dysfunction and damage to cerebral vasculature. In addition, mitochondrial encephalomyopathy, lactic acidosis, and stroke-like episodes (MELAS) syndrome is a mitochondrial disorder with a variety of neurological symptoms. In this study, we aim to assess a potential predisposition in mitochondrial dysfunction of COVID-19, leading to MELAS presentation. METHODS: We studied three previously healthy patients with the first presentation of acute stroke-like symptoms, following COVID-19 infection. We analyzed the patients' clinical data and brain magnetic resonance imaging (MRI) lesions that presented to the neurological center of a university-affiliated hospital in Tehran, Iran, from September 2020 to August 2021. RESULTS: All cases are characterized by a temporoparietal abnormality in imaging studies and electroencephalogram (EEG). Based on electrodiagnostic tests, three patients were diagnosed with myopathy. In two brothers with relatively the same symptoms, one performed muscle biopsy finding myopathic process, and genetic testing confirmed a 3243A>G point mutation in a heteroplasmic state in one of our patients. CONCLUSION: Although MELAS is not a prevalent condition, the recent increase in the number of these patients in our center might indicate the potential role of COVID-19 in triggering the silent pre- existing mitochondrial dysfunction in these patients.


Asunto(s)
Acidosis Láctica , COVID-19 , Síndrome MELAS , Enfermedades del Sistema Nervioso , Accidente Cerebrovascular , Masculino , Humanos , Síndrome MELAS/complicaciones , Síndrome MELAS/genética , Síndrome MELAS/diagnóstico , COVID-19/complicaciones , COVID-19/patología , Irán , Acidosis Láctica/complicaciones , Acidosis Láctica/patología , Accidente Cerebrovascular/etiología , Enfermedades del Sistema Nervioso/complicaciones , Enfermedades del Sistema Nervioso/patología , Mitocondrias/patología
5.
Clin Genet ; 102(1): 56-60, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35246835

RESUMEN

Genetic defect in the nuclear encoded subunits of cytochrome c oxidase are very rare. To date, most deleterious variants affect the mitochondrially encoded subunits of complex IV and the nuclear genes encoded for assembly factors. A biallelic pathogenic variant in the mitochondrial complex IV subunit COX5A was previously reported in a couple of sibs with failure to thrive, lactic acidosis and pulmonary hypertension and a lethal phenotype. Here, we describe a second family with a 11-year-old girl presenting with failure to thrive, lactic acidosis, hypoglycemia and short stature. Clinical exome revealed the homozygous missense variant c.266 T > G in COX5A, which produces a drop of the corresponding protein and a reduction of the COX activity. Compared to the previous observation, this girl showed an attenuated metabolic derangement without involvement of the cardiovascular system and neurodevelopment. Our observation confirms that COX5A recessive variants may cause mitochondrial disease and expands the associated phenotype to less severe presentations.


Asunto(s)
Acidosis Láctica , Enanismo , Hipoglucemia , Acidosis Láctica/genética , Acidosis Láctica/patología , Complejo IV de Transporte de Electrones/genética , Complejo IV de Transporte de Electrones/metabolismo , Insuficiencia de Crecimiento/genética , Homocigoto , Humanos , Hipoglucemia/genética , Fenotipo
6.
J Med Genet ; 58(3): 155-167, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-32439808

RESUMEN

BACKGROUND: Mitochondria provide ATP through the process of oxidative phosphorylation, physically located in the inner mitochondrial membrane (IMM). The mitochondrial contact site and organising system (MICOS) complex is known as the 'mitoskeleton' due to its role in maintaining IMM architecture. APOO encodes MIC26, a component of MICOS, whose exact function in its maintenance or assembly has still not been completely elucidated. METHODS: We have studied a family in which the most affected subject presented progressive developmental delay, lactic acidosis, muscle weakness, hypotonia, weight loss, gastrointestinal and body temperature dysautonomia, repetitive infections, cognitive impairment and autistic behaviour. Other family members showed variable phenotype presentation. Whole exome sequencing was used to screen for pathological variants. Patient-derived skin fibroblasts were used to confirm the pathogenicity of the variant found in APOO. Knockout models in Drosophila melanogaster and Saccharomyces cerevisiae were employed to validate MIC26 involvement in MICOS assembly and mitochondrial function. RESULTS: A likely pathogenic c.350T>C transition was found in APOO predicting an I117T substitution in MIC26. The mutation caused impaired processing of the protein during import and faulty insertion into the IMM. This was associated with altered MICOS assembly and cristae junction disruption. The corresponding mutation in MIC26 or complete loss was associated with mitochondrial structural and functional deficiencies in yeast and D. melanogaster models. CONCLUSION: This is the first case of pathogenic mutation in APOO, causing altered MICOS assembly and neuromuscular impairment. MIC26 is involved in the assembly or stability of MICOS in humans, yeast and flies.


Asunto(s)
Apolipoproteínas/genética , Trastorno Autístico/genética , Disfunción Cognitiva/genética , Proteínas de la Membrana/genética , Miopatías Mitocondriales/genética , Proteínas Mitocondriales/genética , Proteínas de Saccharomyces cerevisiae/genética , Acidosis Láctica/genética , Acidosis Láctica/patología , Animales , Trastorno Autístico/patología , Disfunción Cognitiva/patología , Drosophila melanogaster/genética , Fibroblastos/metabolismo , Enfermedades Genéticas Ligadas al Cromosoma X/genética , Enfermedades Genéticas Ligadas al Cromosoma X/patología , Humanos , Membranas Mitocondriales/metabolismo , Membranas Mitocondriales/patología , Miopatías Mitocondriales/epidemiología , Miopatías Mitocondriales/patología , Unión Proteica , Saccharomyces cerevisiae/genética
7.
Hum Mol Genet ; 28(4): 639-649, 2019 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-30358850

RESUMEN

Dysfunction of mitochondrial translation is an increasingly important molecular cause of human disease, but structural defects of mitochondrial ribosomal subunits are rare. We used next-generation sequencing to identify a homozygous variant in the mitochondrial small ribosomal protein 14 (MRPS14, uS14m) in a patient manifesting with perinatal hypertrophic cardiomyopathy, growth retardation, muscle hypotonia, elevated lactate, dysmorphy and mental retardation. In skeletal muscle and fibroblasts from the patient, there was biochemical deficiency in complex IV of the respiratory chain. In fibroblasts, mitochondrial translation was impaired, and ectopic expression of a wild-type MRPS14 cDNA functionally complemented this defect. Surprisingly, the mutant uS14m was stable and did not affect assembly of the small ribosomal subunit. Instead, structural modeling of the uS14m mutation predicted a disruption to the ribosomal mRNA channel.Collectively, our data demonstrate pathogenic mutations in MRPS14 can manifest as a perinatal-onset mitochondrial hypertrophic cardiomyopathy with a novel molecular pathogenic mechanism that impairs the function of mitochondrial ribosomes during translation elongation or mitochondrial mRNA recruitment rather than assembly.


Asunto(s)
Cardiomiopatía Hipertrófica/genética , Mitocondrias/genética , Enfermedades Mitocondriales/genética , Proteínas Ribosómicas/genética , Acidosis Láctica/genética , Acidosis Láctica/metabolismo , Acidosis Láctica/patología , Secuencia de Aminoácidos/genética , Cardiomiopatía Hipertrófica/metabolismo , Cardiomiopatía Hipertrófica/patología , Niño , Preescolar , Complejo IV de Transporte de Electrones/genética , Femenino , Secuenciación de Nucleótidos de Alto Rendimiento , Homocigoto , Humanos , Lactante , Recién Nacido , Mitocondrias/metabolismo , Enfermedades Mitocondriales/patología , Ribosomas Mitocondriales/metabolismo , Ribosomas Mitocondriales/patología , Mutación , Linaje
8.
Neuropathology ; 41(1): 84-90, 2021 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-33300189

RESUMEN

We report clinicopathological findings of a patient with mitochondrial encephalopathy, lactic acidosis, and stroke-like episodes/Leigh syndrome (MELAS/LS) associated with a novel m.3482A>G mutation in MT-ND1. A 41-year-old woman had experienced multiple stroke-like episodes since age 16. She developed akinetic mutism two months before admission to our hospital. Neurological examination revealed akinetic mutism, bilateral deafness, and muscular atrophy. Cerebrospinal fluid tests revealed elevated pyruvate and lactate levels. Fluid-attenuated inversion recovery images on magnetic resonance imaging showed hyperintense areas in the right frontal and both sides of temporal and occipital lobes, both sides of the striatum, and the midbrain. Muscle biopsy revealed strongly succinate dehydrogenase-reactive blood vessels. L-arginine therapy improved her consciousness and prevented further stroke-like episodes. However, she died from aspiration pneumonia. Postmortem autopsy revealed scattered infarct-like lesions with cavitation in the cerebral cortex and necrotic lesions in the striatum and midbrain. The patient was pathologically confirmed as having MELAS/LS based on two characteristic clinicopathological findings: presenting MELAS/LS overlap phenotype and effectiveness of L-arginine treatment.


Asunto(s)
Acidosis Láctica/patología , Enfermedad de Leigh/patología , Encefalomiopatías Mitocondriales/patología , Mutación , NADH Deshidrogenasa , Accidente Cerebrovascular/patología , Acidosis Láctica/complicaciones , Acidosis Láctica/genética , Adulto , Resultado Fatal , Femenino , Humanos , Enfermedad de Leigh/complicaciones , Enfermedad de Leigh/genética , Encefalomiopatías Mitocondriales/complicaciones , Encefalomiopatías Mitocondriales/genética , Mutación/genética , NADH Deshidrogenasa/genética , Accidente Cerebrovascular/complicaciones , Accidente Cerebrovascular/genética
9.
J Biol Chem ; 294(19): 7810-7820, 2019 05 10.
Artículo en Inglés | MEDLINE | ID: mdl-30923124

RESUMEN

Aerobic glycolysis and mitochondrial dysfunction are key metabolic features of cancer cells, but their interplay during cancer development remains unclear. We previously reported that human hepatoma cells with mitochondrial defects exhibit down-regulated lactate dehydrogenase subunit B (LDHB) expression. Here, using several molecular and biochemical assays and informatics analyses, we investigated how LDHB suppression regulates mitochondrial respiratory activity and contributes to liver cancer progression. We found that transcriptional LDHB down-regulation is an upstream event during suppressed oxidative phosphorylation. We also observed that LDHB knockdown increases inhibitory phosphorylation of pyruvate dehydrogenase (PDH) via lactate-mediated PDH kinase (PDK) activation and thereby attenuates oxidative phosphorylation activity. Interestingly, monocarboxylate transporter 1 was the major lactate transporter in hepatoma cells, and its expression was essential for PDH phosphorylation by modulating intracellular lactate levels. Finally, bioinformatics analysis of the hepatocellular carcinoma cohort from The Cancer Genome Atlas revealed that a low LDHB/LDHA ratio is statistically significantly associated with poor prognostic outcomes. A low ratio was also associated with a significant enrichment in glycolysis genes and negatively correlated with PDK1 and 2 expression, supporting a close link between LDHB suppression and the PDK-PDH axis. These results suggest that LDHB suppression is a key mechanism that enhances glycolysis and is critically involved in the maintenance and propagation of mitochondrial dysfunction via lactate release in liver cancer progression.


Asunto(s)
Acidosis Láctica/enzimología , Carcinoma Hepatocelular/enzimología , Regulación hacia Abajo , Regulación Enzimológica de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Lactato Deshidrogenasas/biosíntesis , Neoplasias Hepáticas/enzimología , Mitocondrias Hepáticas/enzimología , Proteínas de Neoplasias/sangre , Fosforilación Oxidativa , Acidosis Láctica/genética , Acidosis Láctica/patología , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/patología , Línea Celular Tumoral , Humanos , Lactato Deshidrogenasas/genética , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patología , Mitocondrias Hepáticas/genética , Mitocondrias Hepáticas/patología , Proteínas de Neoplasias/genética
10.
Br J Cancer ; 122(9): 1298-1308, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32152504

RESUMEN

BACKGROUND: Akt signalling regulates glycolysis and drives the Warburg effect in cancer, thus decreased glucose utilisation is a pharmacodynamic marker of Akt inhibition. However, cancer cells can utilise alternative nutrients to glucose for energy such as lactate, which is often elevated in tumours together with increased acidity. We therefore hypothesised that lactic acidosis may confer resistance to Akt inhibition. METHODS: The effect of the pan-Akt inhibitor uprosertib (GSK2141795), on HCT116 and LS174T colon cancer cells was evaluated in the presence and absence of lactic acid in vitro. Expression of downstream Akt signalling proteins was determined using a phosphokinase array and immunoblotting. Metabolism was assessed using 1H nuclear magnetic resonance spectroscopy, stable isotope labelling and gas chromatography-mass spectrometry. RESULTS: Lactic acid-induced resistance to uprosertib was characterised by increased cell survival and reduced apoptosis. Uprosertib treatment reduced Akt signalling and glucose uptake irrespective of lactic acid supplementation. However, incorporation of lactate carbon and enhanced respiration was maintained in the presence of uprosertib and lactic acid. Inhibiting lactate transport or oxidative phosphorylation was sufficient to potentiate apoptosis in the presence of uprosertib. CONCLUSIONS: Lactic acidosis confers resistance to uprosertib, which can be reversed by inhibiting lactate transport or oxidative metabolism.


Asunto(s)
Acidosis Láctica/tratamiento farmacológico , Neoplasias del Colon/tratamiento farmacológico , Resistencia a Antineoplásicos/genética , Proteína Oncogénica v-akt/genética , Acidosis Láctica/genética , Acidosis Láctica/metabolismo , Acidosis Láctica/patología , Inhibidores de la Angiogénesis/farmacología , Apoptosis/efectos de los fármacos , Neoplasias del Colon/genética , Neoplasias del Colon/metabolismo , Neoplasias del Colon/patología , Diaminas/farmacología , Glucosa/metabolismo , Glucólisis/efectos de los fármacos , Células HCT116 , Humanos , Ácido Láctico/farmacología , Proteína Oncogénica v-akt/antagonistas & inhibidores , Fosforilación Oxidativa/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología , Pirazoles/farmacología , Transducción de Señal/efectos de los fármacos
11.
J Hum Genet ; 65(3): 231-240, 2020 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-31907385

RESUMEN

TOM70 is a member of the TOM complex that transports cytosolic proteins into mitochondria. Here, we identified two compound heterozygous variants in TOMM70 [c.794C>T (p.T265M) and c.1745C>T (p.A582V)] from a patient with severe anemia, lactic acidosis, and developmental delay. Patient-derived immortalized lymphocytes showed decreased TOM70 expression, oligomerized TOM70 complex, and TOM 20/22/40 complex compared with expression in control lymphocytes. Functional analysis revealed that patient-derived cells exhibited multi-oxidative phosphorylation system (OXPHOS) complex defects, with complex IV being primarily affected. As a result, patient-derived cells grew slower in galactose medium and generated less ATP and more extracellular lactic acid than did control cells. In vitro cell model compensatory experiments confirmed the pathogenicity of TOMM70 variants since only wild-type TOM70, but not mutant TOM70, could restore the complex IV defect and TOM70 expression in TOM70 knockdown U2OS cells. Altogether, we report the first case of mitochondrial disease-causing mutations in TOMM70 and demonstrate that TOM70 is essential for multi-OXPHOS assembly. Mutational screening of TOMM70 should be employed to identify mitochondrial disease-causing gene mutations in the future.


Asunto(s)
Acidosis Láctica/genética , Anemia/genética , Discapacidades del Desarrollo/genética , Proteínas de Transporte de Membrana Mitocondrial/genética , Acidosis Láctica/patología , Anemia/patología , Niño , Discapacidades del Desarrollo/patología , Humanos , Masculino , Mitocondrias/genética , Enfermedades Mitocondriales/genética , Enfermedades Mitocondriales/patología , Proteínas del Complejo de Importación de Proteínas Precursoras Mitocondriales , Mutación/genética , Fosforilación Oxidativa , Secuenciación del Exoma
12.
J Inherit Metab Dis ; 43(5): 1024-1036, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32160317

RESUMEN

Hydrogen sulfide, a signaling molecule formed mainly from cysteine, is catabolized by sulfide:quinone oxidoreductase (gene SQOR). Toxic hydrogen sulfide exposure inhibits complex IV. We describe children of two families with pathogenic variants in SQOR. Exome sequencing identified variants; SQOR enzyme activity was measured spectrophotometrically, protein levels evaluated by western blotting, and mitochondrial function was assayed. In family A, following a brief illness, a 4-year-old girl presented comatose with lactic acidosis and multiorgan failure. After stabilization, she remained comatose, hypotonic, had neurostorming episodes, elevated lactate, and Leigh-like lesions on brain imaging. She died shortly after. Her 8-year-old sister presented with a rapidly fatal episode of coma with lactic acidosis, and lesions in the basal ganglia and left cortex. Muscle and liver tissue had isolated decreased complex IV activity, but normal complex IV protein levels and complex formation. Both patients were homozygous for c.637G > A, which we identified as a founder mutation in the Lehrerleut Hutterite with a carrier frequency of 1 in 13. The resulting p.Glu213Lys change disrupts hydrogen bonding with neighboring residues, resulting in severely reduced SQOR protein and enzyme activity, whereas sulfide generating enzyme levels were unchanged. In family B, a boy had episodes of encephalopathy and basal ganglia lesions. He was homozygous for c.446delT and had severely reduced fibroblast SQOR enzyme activity and protein levels. SQOR dysfunction can result in hydrogen sulfide accumulation, which, consistent with its known toxicity, inhibits complex IV resulting in energy failure. In conclusion, SQOR deficiency represents a new, potentially treatable, cause of Leigh disease.


Asunto(s)
Sulfuro de Hidrógeno/metabolismo , Enfermedad de Leigh/enzimología , Mitocondrias/metabolismo , Oxidorreductasas actuantes sobre Donantes de Grupos Sulfuro/genética , Quinona Reductasas/fisiología , Acidosis Láctica/patología , Encefalopatías/patología , Preescolar , Complejo IV de Transporte de Electrones/metabolismo , Familia , Femenino , Homocigoto , Humanos , Sulfuro de Hidrógeno/química , Cinética , Enfermedad de Leigh/metabolismo , Imagen por Resonancia Magnética , Masculino , Oxidación-Reducción , Quinona Reductasas/química
13.
Kidney Blood Press Res ; 45(4): 523-531, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32663831

RESUMEN

BACKGROUND: The etiology of acute metabolic acidosis (aMA) is heterogeneous, and the consequences are potentially life-threatening. The aim of this article was to summarize the causes and management of aMA from a clinician's perspective. SUMMARY: We performed a systematic search on PubMed, applying the following search terms: "acute metabolic acidosis," "lactic acidosis," "metformin" AND "acidosis," "unbalanced solutions" AND "acidosis," "bicarbonate" AND "acidosis" AND "outcome," "acute metabolic acidosis" AND "management," and "acute metabolic acidosis" AND "renal replacement therapy (RRT)/dialysis." The literature search did not consider diabetic ketoacidosis at all. Lactic acidosis evolves from various conditions, either with or without systemic hypoxia. The incidence of metformin-associated aMA is actually quite low. Unbalanced electrolyte preparations can induce hyperchloremic aMA. The latter potentially worsens kidney-related outcome parameters. Nevertheless, prospective and controlled data are missing at the moment. Recently, bicarbonate has been shown to improve clinically relevant endpoints in the critically ill, even if higher pH values (>7.3) are targeted. New therapeutics for aMA control are under development, since bicarbonate treatment can induce serious side effects. Key Messages: aMA is a frequent and potentially life-threatening complication of various conditions. Lactic acidosis might occur even in the absence of systemic hypoxia. The incidence of metformin-associated aMA is comparably low. Unbalanced electrolyte solutions induce hyperchloremic aMA, which most likely worsens the renal prognosis of critically ill patients. Bicarbonate, although potentially deleterious due to increased carbon dioxide production with subsequent intracellular acidosis, improves kidney-related endpoints in the critically ill.


Asunto(s)
Acidosis/etiología , Acidosis/terapia , Acidosis/patología , Acidosis Láctica/etiología , Acidosis Láctica/patología , Acidosis Láctica/terapia , Enfermedad Aguda , Animales , Bicarbonatos/uso terapéutico , Manejo de la Enfermedad , Electrólitos/efectos adversos , Humanos , Hipoglucemiantes/efectos adversos , Metformina/efectos adversos
14.
Cell Commun Signal ; 17(1): 169, 2019 12 17.
Artículo en Inglés | MEDLINE | ID: mdl-31847904

RESUMEN

BACKGROUND: Tumor endothelial cells (TECs) perform tumor angiogenesis, which is essential for tumor growth and metastasis. Tumor cells produce large amounts of lactic acid from glycolysis; however, the mechanism underlying the survival of TECs to enable tumor angiogenesis under high lactic acid conditions in tumors remains poorly understood. METHODOLOGY: The metabolomes of TECs and normal endothelial cells (NECs) were analyzed by capillary electrophoresis time-of-flight mass spectrometry. The expressions of pH regulators in TECs and NECs were determined by quantitative reverse transcription-PCR. Cell proliferation was measured by the MTS assay. Western blotting and ELISA were used to validate monocarboxylate transporter 1 and carbonic anhydrase 2 (CAII) protein expression within the cells, respectively. Human tumor xenograft models were used to access the effect of CA inhibition on tumor angiogenesis. Immunohistochemical staining was used to observe CAII expression, quantify tumor microvasculature, microvessel pericyte coverage, and hypoxia. RESULTS: The present study shows that, unlike NECs, TECs proliferate in lactic acidic. TECs showed an upregulated CAII expression both in vitro and in vivo. CAII knockdown decreased TEC survival under lactic acidosis and nutrient-replete conditions. Vascular endothelial growth factor A and vascular endothelial growth factor receptor signaling induced CAII expression in NECs. CAII inhibition with acetazolamide minimally reduced tumor angiogenesis in vivo. However, matured blood vessel number increased after acetazolamide treatment, similar to bevacizumab treatment. Additionally, acetazolamide-treated mice showed decreased lung metastasis. CONCLUSION: These findings suggest that due to their effect on blood vessel maturity, pH regulators like CAII are promising targets of antiangiogenic therapy. Video Abstract.


Asunto(s)
Acidosis Láctica/metabolismo , Anhidrasa Carbónica II/metabolismo , Células Neoplásicas Circulantes/metabolismo , Microambiente Tumoral , Acidosis Láctica/patología , Animales , Anhidrasa Carbónica II/genética , Proliferación Celular , Supervivencia Celular , Células Endoteliales/metabolismo , Femenino , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Células Neoplásicas Circulantes/patología , Transducción de Señal , Células Tumorales Cultivadas
15.
Klin Mikrobiol Infekc Lek ; 25(3): 92-96, 2019 Sep.
Artículo en Checo | MEDLINE | ID: mdl-31904104

RESUMEN

Lactic acidosis is a rare but serious adverse event linked to treatment with linezolid, an oxazolidinone antibiotic. Presented is a case of a 67-year-old man treated for 26 days with linezolid for staphylococcal osteomyelitis of the right foot with subsequent sepsis. During the course of treatment, severe lactic acidosis developed, requiring hospitalization in an intensive care unit. The likely mechanism of this potentially life-threatening complication is discussed.


Asunto(s)
Acidosis Láctica , Osteomielitis , Oxazolidinonas , Sepsis , Infecciones Estafilocócicas , Acidosis Láctica/diagnóstico , Acidosis Láctica/etiología , Acidosis Láctica/patología , Anciano , Antibacterianos/efectos adversos , Antibacterianos/uso terapéutico , Humanos , Linezolid/efectos adversos , Masculino , Osteomielitis/complicaciones , Osteomielitis/tratamiento farmacológico , Osteomielitis/etiología , Sepsis/etiología , Infecciones Estafilocócicas/complicaciones , Infecciones Estafilocócicas/tratamiento farmacológico , Infecciones Estafilocócicas/etiología , Resultado del Tratamiento
16.
Hum Mol Genet ; 25(23): 5178-5187, 2016 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-28007899

RESUMEN

ISCU myopathy is an inherited disease that primarily affects individuals of northern Swedish descent who share a single point mutation in the fourth intron of the ISCU gene. The current study shows correction of specific phenotypes associated with disease following treatment with an antisense oligonucleotide (ASO) targeted to the site of the mutation. We have shown that ASO treatment diminished aberrant splicing and increased ISCU protein levels in both patient fibroblasts and patient myotubes in a concentration dependent fashion. Upon ASO treatment, levels of SDHB in patient myotubular cell lines increased to levels observed in control myotubular cell lines. Additionally, we have shown that both patient fibroblast and myotubular cell lines displayed an increase in complex II activity with a concomitant decrease in succinate levels in patient myotubular cell lines after ASO treatment. Mitochondrial and cytosolic aconitase activities increased significantly following ASO treatment in patient myotubes. The current study suggests that ASO treatment may serve as a viable approach to correcting ISCU myopathy in patients.


Asunto(s)
Acidosis Láctica/congénito , Proteínas Hierro-Azufre/genética , Enfermedades Musculares/congénito , Oligonucleótidos Antisentido/genética , Succinato Deshidrogenasa/genética , Acidosis Láctica/genética , Acidosis Láctica/patología , Acidosis Láctica/terapia , Femenino , Humanos , Intrones/genética , Masculino , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Mitocondrias/patología , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/patología , Enfermedades Musculares/genética , Enfermedades Musculares/patología , Enfermedades Musculares/terapia , Oligonucleótidos Antisentido/uso terapéutico , Fenotipo , Mutación Puntual , Empalme del ARN/efectos de los fármacos , Empalme del ARN/genética , Succinato Deshidrogenasa/biosíntesis
17.
Clin Genet ; 93(5): 1097-1102, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29327420

RESUMEN

SLC25A42 gene encodes an inner mitochondrial membrane protein that imports Coenzyme A into the mitochondrial matrix. A mutation in this gene was recently reported in a subject born to consanguineous parents who presented with mitochondrial myopathy with muscle weakness and lactic acidosis. In this report, we present 12 additional individuals with the same founder mutation who presented with variable manifestations ranging from asymptomatic lactic acidosis to a severe phenotype characterized by developmental regression and epilepsy. Our report confirms the link between SLC25A42 and mitochondrial disease in humans, and suggests that pathogenic variants in SLC25A42 should be interpreted with the understanding that the associated phenotype may be highly variable.


Asunto(s)
Acidosis Láctica/genética , Encefalomiopatías Mitocondriales/genética , Miopatías Mitocondriales/genética , Proteínas de Transporte de Nucleótidos/genética , Acidosis Láctica/patología , Adolescente , Adulto , Niño , Preescolar , ADN Mitocondrial , Femenino , Humanos , Lactante , Masculino , Mitocondrias/genética , Mitocondrias/patología , Encefalomiopatías Mitocondriales/complicaciones , Encefalomiopatías Mitocondriales/patología , Miopatías Mitocondriales/patología , Linaje , Fenotipo , Mutación Puntual , Adulto Joven
18.
Hum Mutat ; 38(6): 692-703, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28247525

RESUMEN

COX5A is a nuclear-encoded subunit of mitochondrial respiratory chain complex IV (cytochrome c oxidase). We present patients with a homozygous pathogenic variant in the COX5A gene. Clinical details of two affected siblings suffering from early-onset pulmonary arterial hypertension, lactic acidemia, failure to thrive, and isolated complex IV deficiency are presented. We show that the variant lies within the evolutionarily conserved COX5A/COX4 interface domain, suggesting that it alters the interaction between these two subunits during complex IV biogenesis. In patient skin fibroblasts, the enzymatic activity and protein levels of complex IV and several of its subunits are reduced. Lentiviral complementation rescues complex IV deficiency. The monomeric COX1 assembly intermediate accumulates demonstrating a function of COX5A in complex IV biogenesis. A potential therapeutic lead is demonstrated by showing that copper supplementation leads to partial rescue of complex IV deficiency in patient fibroblasts.


Asunto(s)
Acidosis Láctica/genética , Ciclooxigenasa 1/genética , Grupo Citocromo c/genética , Insuficiencia de Crecimiento/genética , Hipertensión Pulmonar/genética , Acidosis Láctica/patología , Núcleo Celular/genética , Ciclooxigenasa 1/química , Grupo Citocromo c/química , Deficiencia de Citocromo-c Oxidasa , Complejo IV de Transporte de Electrones , Insuficiencia de Crecimiento/patología , Fibroblastos , Predisposición Genética a la Enfermedad , Homocigoto , Humanos , Hipertensión Pulmonar/patología , Mitocondrias/genética , Mutación , Subunidades de Proteína/genética
19.
J Transl Med ; 15(1): 129, 2017 06 07.
Artículo en Inglés | MEDLINE | ID: mdl-28592308

RESUMEN

BACKGROUND: The pursuit for clarity in diagnostic and treatment pathways for the complex, chronic condition of myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS) continues. This systematic review raises a novel question to explore possible overlapping aetiology in two distinct conditions. Similar neurocognitive symptoms and evidence of D-lactate producing bacteria in ME/CFS raise questions about shared mechanisms with the acute condition of D-lactic acidosis (D-la). METHODS: D-la case reports published between 1965 and March 2016 were reviewed for episodes describing both neurological symptoms and high D-lactate levels. Fifty-nine D-la episodes were included in the qualitative synthesis comparing D-la symptoms with ME/CFS diagnostic criteria. A narrative review of D-la mechanisms and relevance for ME/CFS was provided. RESULTS: The majority of neurological disturbances reported in D-la episodes overlapped with ME/CFS symptoms. Of these, the most frequently reported D-la symptoms were motor disturbances that appear more prominent during severe presentations of ME/CFS. Both patient groups shared a history of gastrointestinal abnormalities and evidence of bacterial dysbiosis, although only preliminary evidence supported the role of lactate-producing bacteria in ME/CFS. LIMITATIONS: Interpretation of results are constrained by both the breadth of symptoms included in ME/CFS diagnostic criteria and the conservative methodology used for D-la symptom classification. Several pathophysiological mechanisms in ME/CFS were not examined. CONCLUSIONS: Shared symptomatology and underlying microbiota-gut-brain interactions raise the possibility of a continuum of acute (D-la) versus chronic (ME/CFS) presentations related to D-lactate absorption. Measurement of D-lactate in ME/CFS is needed to effectively evaluate whether subclinical D-lactate levels affect neurological symptoms in this clinical population.


Asunto(s)
Acidosis Láctica/patología , Síndrome de Fatiga Crónica/patología , Adulto , Niño , Femenino , Humanos , Masculino
20.
Hum Mol Genet ; 23(1): 24-39, 2014 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-23943793

RESUMEN

Iron-sulfur (Fe-S) clusters are ancient enzyme cofactors found in virtually all life forms. We evaluated the physiological effects of chronic Fe-S cluster deficiency in human skeletal muscle, a tissue that relies heavily on Fe-S cluster-mediated aerobic energy metabolism. Despite greatly decreased oxidative capacity, muscle tissue from patients deficient in the Fe-S cluster scaffold protein ISCU showed a predominance of type I oxidative muscle fibers and higher capillary density, enhanced expression of transcriptional co-activator PGC-1α and increased mitochondrial fatty acid oxidation genes. These Fe-S cluster-deficient muscles showed a dramatic up-regulation of the ketogenic enzyme HMGCS2 and the secreted protein FGF21 (fibroblast growth factor 21). Enhanced muscle FGF21 expression was reflected by elevated circulating FGF21 levels in the patients, and robust FGF21 secretion could be recapitulated by respiratory chain inhibition in cultured myotubes. Our findings reveal that mitochondrial energy starvation elicits a coordinated response in Fe-S-deficient skeletal muscle that is reflected systemically by increased plasma FGF21 levels.


Asunto(s)
Acidosis Láctica/congénito , Factores de Crecimiento de Fibroblastos/metabolismo , Hidroximetilglutaril-CoA Sintasa/metabolismo , Proteínas Hierro-Azufre/metabolismo , Músculo Esquelético/metabolismo , Enfermedades Musculares/congénito , Factores de Transcripción/genética , Acidosis Láctica/genética , Acidosis Láctica/metabolismo , Acidosis Láctica/patología , Adulto , Anciano , Estudios de Casos y Controles , Células Cultivadas , Metabolismo Energético , Femenino , Factores de Crecimiento de Fibroblastos/genética , Regulación de la Expresión Génica , Humanos , Hidroximetilglutaril-CoA Sintasa/genética , Proteínas Hierro-Azufre/genética , Masculino , Persona de Mediana Edad , Mitocondrias Musculares/metabolismo , Mitocondrias Musculares/patología , Enfermedades Musculares/genética , Enfermedades Musculares/metabolismo , Enfermedades Musculares/patología , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma , Factores de Transcripción/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA