Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Int J Mol Sci ; 22(12)2021 Jun 15.
Article in English | MEDLINE | ID: mdl-34203775

ABSTRACT

Mitochondrial disorders (MD) comprise a group of heterogeneous clinical disorders for which non-invasive diagnosis remains a challenge. Two protein biomarkers have so far emerged for MD detection, FGF-21 and GDF-15, but the identification of additional biomarkers capable of improving their diagnostic accuracy is highly relevant. Previous studies identified Gelsolin as a regulator of cell survival adaptations triggered by mitochondrial defects. Gelsolin presents a circulating plasma isoform (pGSN), whose altered levels could be a hallmark of mitochondrial dysfunction. Therefore, we investigated the diagnostic performance of pGSN for MD relative to FGF-21 and GDF-15. Using ELISA assays, we quantified plasma levels of pGSN, FGF-21, and GDF-15 in three age- and gender-matched adult cohorts: 60 genetically diagnosed MD patients, 56 healthy donors, and 41 patients with unrelated neuromuscular pathologies (non-MD). Clinical variables and biomarkers' plasma levels were compared between groups. Discrimination ability was calculated using the area under the ROC curve (AUC). Optimal cut-offs and the following diagnostic parameters were determined: sensitivity, specificity, positive and negative predictive values, positive and negative likelihood ratios, and efficiency. Comprehensive statistical analyses revealed significant discrimination ability for the three biomarkers to classify between MD and healthy individuals, with the best diagnostic performance for the GDF-15/pGSN combination. pGSN and GDF-15 preferentially discriminated between MD and non-MD patients under 50 years, whereas FGF-21 best classified older subjects. Conclusion: pGSN improves the diagnosis accuracy for MD provided by FGF-21 and GDF-15.


Subject(s)
Fibroblast Growth Factors/blood , Gelsolin/blood , Growth Differentiation Factor 15/blood , Mitochondrial Diseases/blood , Mitochondrial Diseases/diagnosis , Adult , Biomarkers/blood , Case-Control Studies , Female , Humans , Male , Middle Aged , Phenotype
2.
Cells ; 9(9)2020 08 19.
Article in English | MEDLINE | ID: mdl-32824961

ABSTRACT

Mitochondrial oxidative phosphorylation (OXPHOS) defects are the primary cause of inborn errors of energy metabolism. Despite considerable progress on their genetic basis, their global pathophysiological consequences remain undefined. Previous studies reported that OXPHOS dysfunction associated with complex III deficiency exacerbated the expression and mitochondrial location of cytoskeletal gelsolin (GSN) to promote cell survival responses. In humans, besides the cytosolic isoform, GSN presents a plasma isoform secreted to extracellular environments. We analyzed the interplay between both GSN isoforms in human cellular and clinical models of OXPHOS dysfunction. Regardless of its pathogenic origin, OXPHOS dysfunction induced the physiological upregulation of cytosolic GSN in the mitochondria (mGSN), in parallel with a significant downregulation of plasma GSN (pGSN) levels. Consequently, significantly high mGSN-to-pGSN ratios were associated with OXPHOS deficiency both in human cells and blood. In contrast, control cells subjected to hydrogen peroxide or staurosporine treatments showed no correlation between oxidative stress or cell death induction and the altered levels and subcellular location of GSN isoforms, suggesting their specificity for OXPHOS dysfunction. In conclusion, a high mitochondrial-to-plasma GSN ratio represents a useful cellular indicator of OXPHOS defects, with potential use for future research of a wide range of clinical conditions with mitochondrial involvement.


Subject(s)
Gelsolin/metabolism , Mitochondria/metabolism , Protein Isoforms/metabolism , Adult , Female , Humans , Male , Middle Aged , Oxidative Phosphorylation , Young Adult
3.
Hum Mol Genet ; 26(13): 2493-2506, 2017 07 01.
Article in English | MEDLINE | ID: mdl-28431142

ABSTRACT

Despite considerable knowledge on the genetic basis of mitochondrial disorders, their pathophysiological consequences remain poorly understood. We previously used two-dimensional difference gel electrophoresis analyses to define a protein profile characteristic for respiratory chain complex III-deficiency that included a significant overexpression of cytosolic gelsolin (GSN), a cytoskeletal protein that regulates the severing and capping of the actin filaments. Biochemical and immunofluorescence assays confirmed a specific increase of GSN levels in the mitochondria from patients' fibroblasts and from transmitochondrial cybrids with complex III assembly defects. A similar effect was obtained in control cells upon treatment with antimycin A in a dose-dependent manner, showing that the enzymatic inhibition of complex III is sufficient to promote the mitochondrial localization of GSN. Mitochondrial subfractionation showed the localization of GSN to the mitochondrial outer membrane, where it interacts with the voltage-dependent anion channel protein 1 (VDAC1). In control cells, VDAC1 was present in five stable oligomeric complexes, which showed increased levels and a modified distribution pattern in the complex III-deficient cybrids. Downregulation of GSN expression induced cell death in both cell types, in parallel with the specific accumulation of VDAC1 dimers and the release of mitochondrial cytochrome c into the cytosol, indicating a role for GSN in the oligomerization of VDAC complexes and in the prevention of apoptosis. Our results demonstrate that respiratory chain complex III dysfunction induces the physiological upregulation and mitochondrial location of GSN, probably to promote cell survival responses through the modulation of the oligomeric state of the VDAC complexes.


Subject(s)
Electron Transport/physiology , Gelsolin/metabolism , Voltage-Dependent Anion Channel 1/metabolism , Actin Cytoskeleton/metabolism , Actins/metabolism , Antimycin A/metabolism , Apoptosis/genetics , Cell Line, Tumor , Cell Survival , Cytochromes c/metabolism , Fibroblasts/metabolism , Gelsolin/genetics , HeLa Cells , Humans , Mitochondria/metabolism , Mitochondrial Diseases/metabolism , Mitochondrial Membranes/metabolism , Two-Dimensional Difference Gel Electrophoresis/methods , Voltage-Dependent Anion Channel 1/physiology
4.
Cell Rep ; 16(9): 2387-98, 2016 08 30.
Article in English | MEDLINE | ID: mdl-27545886

ABSTRACT

Mitochondrial respiratory chain (MRC) complexes I, III, and IV associate into a variety of supramolecular structures known as supercomplexes and respirasomes. While COX7A2L was originally described as a supercomplex-specific factor responsible for the dynamic association of complex IV into these structures to adapt MRC function to metabolic variations, this role has been disputed. Here, we further examine the functional significance of COX7A2L in the structural organization of the mammalian respiratory chain. As in the mouse, human COX7A2L binds primarily to free mitochondrial complex III and, to a minor extent, to complex IV to specifically promote the stabilization of the III2+IV supercomplex without affecting respirasome formation. Furthermore, COX7A2L does not affect the biogenesis, stabilization, and function of the individual oxidative phosphorylation complexes. These data show that independent regulatory mechanisms for the biogenesis and turnover of different MRC supercomplex structures co-exist.


Subject(s)
Electron Transport Complex III/metabolism , Electron Transport Complex IV/metabolism , Electron Transport Complex I/metabolism , Mitochondria, Heart/metabolism , Mitochondrial Membranes/metabolism , Oxidative Phosphorylation , Animals , Electron Transport , Electron Transport Complex I/genetics , Electron Transport Complex III/genetics , Electron Transport Complex IV/genetics , Gene Expression , HEK293 Cells , HeLa Cells , Humans , Mice , Mitochondria, Heart/chemistry , Myocardium/cytology , Myocardium/metabolism , Protein Binding , Protein Stability
5.
J Proteomics ; 113: 38-56, 2015 Jan 15.
Article in English | MEDLINE | ID: mdl-25239759

ABSTRACT

We have analyzed the cellular pathways and metabolic adaptations that take place in primary skin fibroblasts from patients with mutations in BCS1L, a major genetic cause of mitochondrial complex III enzyme deficiency. Mutant fibroblasts exhibited low oxygen consumption rates and intracellular ATP levels, indicating that the main altered molecular event probably is a limited respiration-coupled ATP production through the OXPHOS system. Two-dimensional DIGE and MALDI-TOF/TOF mass spectrometry analyses unambiguously identified 39 proteins whose expression was significantly altered in complex III-deficient fibroblasts. Extensive statistical and cluster analyses revealed a protein profile characteristic for the BCS1L mutant fibroblasts that included alterations in energy metabolism, cell signaling and gene expression regulation, cytoskeleton formation and maintenance, and intracellular stress responses. The physiological validation of the predicted functional adaptations of human cultured fibroblasts to complex III deficiency confirmed the up-regulation of glycolytic enzyme activities and the accumulation of branched-chain among other amino acids, suggesting the activation of anaerobic glycolysis and cellular catabolic states, in particular protein catabolism, together with autophagy as adaptive responses to mitochondrial respiratory chain dysfunction and ATP deficiency. Our data point to an overall metabolic and genetic reprogramming that could contribute to explain the clinical manifestations of complex III deficiency in patients. BIOLOGICAL SIGNIFICANCE: Despite considerable knowledge about their genetic origins, the pathophysiological mechanisms that contribute to the clinical manifestations of mitochondrial disorders remain poorly understood. We have investigated the molecular pathways and metabolic adaptations that take place in primary skin fibroblasts from patients with mutations in the BCS1L gene, a primary cause of mitochondrial complex III enzyme deficiency. Two-dimensional DIGE together with MALDI-TOF/TOF mass spectrometry and physiological validation analyses revealed a significant metabolic and genetic reprogramming as an adaptive response to mitochondrial respiratory chain dysfunction. Our data provide information about specific protein targets that regulate the transmitochondrial functional responses to complex III deficiency, thereby opening new doors for future research.


Subject(s)
Electron Transport Complex III/deficiency , Fibroblasts/metabolism , Genetic Diseases, Inborn/metabolism , Mitochondria/metabolism , Mitochondrial Proteins/deficiency , Proteomics , ATPases Associated with Diverse Cellular Activities , Cells, Cultured , Cytoskeleton/genetics , Cytoskeleton/metabolism , Cytoskeleton/pathology , Energy Metabolism/genetics , Female , Fibroblasts/pathology , Gene Expression Regulation/genetics , Genetic Diseases, Inborn/genetics , Humans , Male , Mitochondria/genetics , Mitochondria/pathology , Signal Transduction/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...