Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 10.629
Filtrer
1.
Sci Adv ; 10(31): eadp0443, 2024 Aug 02.
Article de Anglais | MEDLINE | ID: mdl-39093974

RÉSUMÉ

Mitochondrial fusion and fission accompany adaptive responses to stress and altered metabolic demands. Inner membrane fusion and cristae morphogenesis depends on optic atrophy 1 (Opa1), which is expressed in different isoforms and is cleaved from a membrane-bound, long to a soluble, short form. Here, we have analyzed the physiological role of Opa1 isoforms and Opa1 processing by generating mouse lines expressing only one cleavable Opa1 isoform or a non-cleavable variant thereof. Our results show that expression of a single cleavable or non-cleavable Opa1 isoform preserves embryonic development and the health of adult mice. Opa1 processing is dispensable under metabolic and thermal stress but prolongs life span and protects against mitochondrial cardiomyopathy in OXPHOS-deficient Cox10-/- mice. Mechanistically, loss of Opa1 processing disturbs the balance between mitochondrial biogenesis and mitophagy, suppressing cardiac hypertrophic growth in Cox10-/- hearts. Our results highlight the critical regulatory role of Opa1 processing, mitochondrial dynamics, and metabolism for cardiac hypertrophy.


Sujet(s)
Cardiomyopathies , dGTPases , Animaux , dGTPases/métabolisme , dGTPases/génétique , Souris , Cardiomyopathies/métabolisme , Cardiomyopathies/génétique , Cardiomyopathies/anatomopathologie , Dynamique mitochondriale , Mitophagie/génétique , Souris knockout , Isoformes de protéines/métabolisme , Isoformes de protéines/génétique , Mitochondries/métabolisme , Modèles animaux de maladie humaine , Développement embryonnaire/génétique
2.
Cell Mol Biol (Noisy-le-grand) ; 70(7): 8-14, 2024 Jul 28.
Article de Anglais | MEDLINE | ID: mdl-39097901

RÉSUMÉ

Alzheimer's disease (AD) is the most common neurodegenerative disorder, and amyloid beta oligomers (AßO), which are pathological markers of AD, are known to be highly toxic. AßO increase mitochondrial dysfunction, which is accompanied by a decrease in mitochondrial fusion. Although mitofusin (Mfn) 1 and Mfn2 are mitochondrial fusion proteins, Mfn2 is known to regulate endoplasmic reticulum (ER) function, as it is located in the ER. Several studies have shown that AßO exacerbates ER stress, however, the exact mechanism requires further elucidation. In this study, we used mouse neuroblastoma cells stably overexpressing the amyloid precursor protein (APP) with the Swedish mutation (N2a APPswe cells) to investigate the role of Mfn in ER stress. Our results revealed that  amyloid beta (Aß) caused cellular toxicity in N2a APPswe cells, upregulated ER stress-related proteins, and promoted ER expansion. The AßO-mediated ER stress was reduced when Mfn1 and Mfn2 were overexpressed. Moreover, Mfn1 and Mfn2 overexpressed resulted in reduced apoptosis of N2a APPswe cells. In conclusion, our results indicate that both Mfn1 and Mfn2 reduce ER stress and apoptosis. Our data provide a foundation for future studies on the roles of Mfn1 and Mfn2 in the molecular mechanisms underlying AßO-mediated ER stress and the pathogenesis of AD.


Sujet(s)
Peptides bêta-amyloïdes , Apoptose , Stress du réticulum endoplasmique , dGTPases , Animaux , Humains , Souris , Maladie d'Alzheimer/métabolisme , Maladie d'Alzheimer/génétique , Maladie d'Alzheimer/anatomopathologie , Peptides bêta-amyloïdes/métabolisme , Précurseur de la protéine bêta-amyloïde/génétique , Précurseur de la protéine bêta-amyloïde/métabolisme , Apoptose/génétique , Lignée cellulaire tumorale , Réticulum endoplasmique/métabolisme , Stress du réticulum endoplasmique/génétique , dGTPases/métabolisme , dGTPases/génétique , Mitochondries/métabolisme
3.
Int J Mol Sci ; 25(14)2024 Jul 16.
Article de Anglais | MEDLINE | ID: mdl-39063023

RÉSUMÉ

Mitochondrial fission and fusion are vital dynamic processes for mitochondrial quality control and for the maintenance of cellular respiration; they also play an important role in the formation and maintenance of cells with high energy demand including cardiomyocytes and neurons. The DNM1L (dynamin-1 like) gene encodes for the DRP1 protein, an evolutionary conserved member of the dynamin family that is responsible for the fission of mitochondria; it is ubiquitous but highly expressed in the developing neonatal heart. De novo heterozygous pathogenic variants in the DNM1L gene have been previously reported to be associated with neonatal or infantile-onset encephalopathy characterized by hypotonia, developmental delay and refractory epilepsy. However, cardiac involvement has been previously reported only in one case. Next-Generation Sequencing (NGS) was used to genetically assess a baby girl characterized by developmental delay with spastic-dystonic, tetraparesis and hypertrophic cardiomyopathy of the left ventricle. Histochemical analysis and spectrophotometric determination of electron transport chain were performed to characterize the muscle biopsy; moreover, the morphology of mitochondria and peroxisomes was evaluated in cultured fibroblasts as well. Herein, we expand the phenotype of DNM1L-related disorder, describing the case of a girl with a heterozygous mutation in DNM1L and affected by progressive infantile encephalopathy, with cardiomyopathy and fatal paroxysmal vomiting correlated with bulbar transitory abnormal T2 hyperintensities and diffusion-weighted imaging (DWI) restriction areas, but without epilepsy. In patients with DNM1L mutations, careful evaluation for cardiac involvement is recommended.


Sujet(s)
Cardiomyopathies , Dynamines , Mutation , Humains , Femelle , Dynamines/génétique , Cardiomyopathies/génétique , Mutation/génétique , Nourrisson , Issue fatale , Encéphalopathies/génétique , Encéphalopathies/anatomopathologie , dGTPases/génétique
4.
Mol Cell ; 84(14): 2732-2746.e5, 2024 Jul 25.
Article de Anglais | MEDLINE | ID: mdl-38981483

RÉSUMÉ

Metabolic enzymes can adapt during energy stress, but the consequences of these adaptations remain understudied. Here, we discovered that hexokinase 1 (HK1), a key glycolytic enzyme, forms rings around mitochondria during energy stress. These HK1-rings constrict mitochondria at contact sites with the endoplasmic reticulum (ER) and mitochondrial dynamics protein (MiD51). HK1-rings prevent mitochondrial fission by displacing the dynamin-related protein 1 (Drp1) from mitochondrial fission factor (Mff) and mitochondrial fission 1 protein (Fis1). The disassembly of HK1-rings during energy restoration correlated with mitochondrial fission. Mechanistically, we identified that the lack of ATP and glucose-6-phosphate (G6P) promotes the formation of HK1-rings. Mutations that affect the formation of HK1-rings showed that HK1-rings rewire cellular metabolism toward increased TCA cycle activity. Our findings highlight that HK1 is an energy stress sensor that regulates the shape, connectivity, and metabolic activity of mitochondria. Thus, the formation of HK1-rings may affect mitochondrial function in energy-stress-related pathologies.


Sujet(s)
Dynamines , Métabolisme énergétique , Hexokinase , Mitochondries , Dynamique mitochondriale , Protéines mitochondriales , Hexokinase/métabolisme , Hexokinase/génétique , Humains , Mitochondries/métabolisme , Mitochondries/génétique , Mitochondries/enzymologie , Dynamines/métabolisme , Dynamines/génétique , Protéines mitochondriales/métabolisme , Protéines mitochondriales/génétique , Animaux , Adénosine triphosphate/métabolisme , Stress physiologique , Réticulum endoplasmique/métabolisme , Protéines membranaires/métabolisme , Protéines membranaires/génétique , Cycle citrique , Glucose-6-phosphate/métabolisme , Souris , Cellules HeLa , Cellules HEK293 , dGTPases/métabolisme , dGTPases/génétique , Mutation
5.
Cell Signal ; 121: 111283, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-38960059

RÉSUMÉ

It has been demonstrated that circular RNAs (circRNAs) are associated with the development of diabetic retinopathy (DR). Nevertheless, the function of circSLC16A10 in the development of DR remains unclear. In order to investigate the role of circSLC16A10, we employed cell and animal models of DR. An analysis of a public database revealed that hsa_circSLC16A10 was expressed at lower levels in DR patients than in diabetic patients without DR or healthy controls. Additionally, the level of hsa_circSLC16A10 was lower in high glucose (HG)-exposed ARPE-19 cells and diabetic mice. hsa_circSLC16A10 was observed to be mainly distributed in the cytoplasm. Moreover, overexpression of hsa_circSLC16A10 alleviated HG-induced endoplasmic reticulum stress and cell apoptosis in vitro. Furthermore, overexpression of hsa_circSLC16A10 ameliorated HG-induced mitochondrial dysfunction, as evidenced by improvements in mitochondrial structure and function. hsa_circSLC16A10 acted as a hsa-miR-761-5p sponge to increase MFN2 expression. MFN2 knockdown or hsa-miR-761-5p overexpression partially reversed the protective effect of hsa_circSLC16A10 in vitro. The protective effect of mmu_circSLC16A10 against DR was confirmed in an animal model of DR. These findings indicate that circSLC16A10 may regulate DR progression by improving mitochondrial function via the miR-761-5p/MFN2 axis.


Sujet(s)
Rétinopathie diabétique , dGTPases , microARN , Mitochondries , ARN circulaire , Rétinopathie diabétique/génétique , Rétinopathie diabétique/métabolisme , Rétinopathie diabétique/anatomopathologie , ARN circulaire/génétique , ARN circulaire/métabolisme , Animaux , microARN/métabolisme , microARN/génétique , Humains , Souris , Mitochondries/métabolisme , dGTPases/métabolisme , dGTPases/génétique , Diabète expérimental/métabolisme , Diabète expérimental/génétique , Mâle , Apoptose , Souris de lignée C57BL , Protéines mitochondriales/métabolisme , Protéines mitochondriales/génétique , Stress du réticulum endoplasmique , Lignée cellulaire
6.
Dis Model Mech ; 17(8)2024 Aug 01.
Article de Anglais | MEDLINE | ID: mdl-38966981

RÉSUMÉ

Inherited retinal diseases encompass a genetically diverse group of conditions caused by variants in genes critical to retinal function, including handful of ribosome-associated genes. This study focuses on the HBS1L gene, which encodes for the HBS1-like translational GTPase that is crucial for ribosomal rescue. We have reported a female child carrying biallelic HBS1L variants, manifesting with poor growth and neurodevelopmental delay. Here, we describe the ophthalmologic findings in the patient and in Hbs1ltm1a/tm1a hypomorph mice and describe the associated microscopic and molecular perturbations. The patient has impaired visual function, showing dampened amplitudes of a- and b-waves in both rod- and cone-mediated responses. Hbs1ltm1a/tm1a mice exhibited profound thinning of the entire retina, specifically of the outer photoreceptor layer, due to extensive photoreceptor cell apoptosis. Loss of Hbs1l resulted in comprehensive proteomic alterations by mass spectrometry analysis, with an increase in the levels of 169 proteins and a decrease in the levels of 480 proteins, including rhodopsin (Rho) and peripherin 2 (Prph2). Gene Ontology biological process and gene set enrichment analyses reveal that the downregulated proteins are primarily involved in phototransduction, cilium assembly and photoreceptor cell development. These findings underscore the importance of ribosomal rescue proteins in maintaining retinal health, particularly in photoreceptor cells.


Sujet(s)
Modèles animaux de maladie humaine , Dystrophies rétiniennes , Animaux , Dystrophies rétiniennes/anatomopathologie , Dystrophies rétiniennes/génétique , Femelle , Humains , Souris , Cellules photoréceptrices/métabolisme , Cellules photoréceptrices/anatomopathologie , Apoptose , Protéines G/métabolisme , Protéines G/déficit , Protéines G/génétique , dGTPases/déficit , dGTPases/métabolisme , dGTPases/génétique , Enfant
7.
Nat Commun ; 15(1): 5797, 2024 Jul 10.
Article de Anglais | MEDLINE | ID: mdl-38987236

RÉSUMÉ

The basal structure of the bacterial flagellum includes a membrane embedded MS-ring (formed by multiple copies of FliF) and a cytoplasmic C-ring (composed of proteins FliG, FliM and FliN). The SRP-type GTPase FlhF is required for directing the initial flagellar protein FliF to the cell pole, but the mechanisms are unclear. Here, we show that FlhF anchors developing flagellar structures to the polar landmark protein HubP/FimV, thereby restricting their formation to the cell pole. Specifically, the GTPase domain of FlhF interacts with HubP, while a structured domain at the N-terminus of FlhF binds to FliG. FlhF-bound FliG subsequently engages with the MS-ring protein FliF. Thus, the interaction of FlhF with HubP and FliG recruits a FliF-FliG complex to the cell pole. In addition, the modulation of FlhF activity by the MinD-type ATPase FlhG controls the interaction of FliG with FliM-FliN, thereby regulating the progression of flagellar assembly at the pole.


Sujet(s)
Protéines bactériennes , Flagelles , Flagelles/métabolisme , Protéines bactériennes/métabolisme , Protéines bactériennes/composition chimique , Protéines bactériennes/génétique , Liaison aux protéines , dGTPases/métabolisme , dGTPases/composition chimique , dGTPases/génétique , Particule de reconnaissance du signal/métabolisme , Particule de reconnaissance du signal/composition chimique , Protéines G monomériques/métabolisme , Protéines G monomériques/composition chimique , Protéines G monomériques/génétique , Protéines membranaires
8.
J Pineal Res ; 76(5): e12991, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-39039850

RÉSUMÉ

Although rapid progression and a poor prognosis in influenza A virus (IAV) infection-induced acute exacerbation of chronic obstructive pulmonary disease (AECOPD) are frequently associated with metabolic energy disorders, the underlying mechanisms and rescue strategies remain unknown. We herein demonstrated that the level of resting energy expenditure increased significantly in IAV-induced AECOPD patients and that cellular energy exhaustion emerged earlier and more significantly in IAV-infected primary COPD bronchial epithelial (pDHBE) cells. The differentially expressed genes were enriched in the oxidative phosphorylation (OXPHOS) pathway; additionally, we consistently uncovered much earlier ATP exhaustion, more severe mitochondrial structural destruction and dysfunction, and OXPHOS impairment in IAV-inoculated pDHBE cells, and these changes were rescued by melatonin. The level of OMA1-dependent cleavage of OPA1 in the mitochondrial inner membrane and the shift in energy metabolism from OXPHOS to glycolysis were significantly increased in IAV-infected pDHBE cells; however, these changes were rescued by OMA1-siRNA or melatonin further treatment. Collectively, our data revealed that melatonin rescued IAV-induced cellular energy exhaustion via OMA1-OPA1-S to improve the clinical prognosis in COPD. This treatment may serve as a potential therapeutic agent for patients in which AECOPD is induced by IAV.


Sujet(s)
Métabolisme énergétique , dGTPases , Virus de la grippe A , Mélatonine , Broncho-pneumopathie chronique obstructive , Humains , Métabolisme énergétique/effets des médicaments et des substances chimiques , dGTPases/métabolisme , dGTPases/génétique , Virus de la grippe A/effets des médicaments et des substances chimiques , Grippe humaine/métabolisme , Grippe humaine/traitement médicamenteux , Mélatonine/pharmacologie , Metalloendopeptidases , Phosphorylation oxydative/effets des médicaments et des substances chimiques , Broncho-pneumopathie chronique obstructive/métabolisme , Broncho-pneumopathie chronique obstructive/traitement médicamenteux
9.
FASEB J ; 38(14): e23816, 2024 Jul 31.
Article de Anglais | MEDLINE | ID: mdl-39072779

RÉSUMÉ

Acetaminophen (APAP) is one of the most clinically relevant medications associated with acute liver damage. A prolific deal of research validated the hepatoprotective effect of empagliflozin (EMPA); however, its effect on APAP-induced hepatotoxicity has still not been investigated. In this study, the prospective hepatoprotective impact of EMPA against APAP-induced hepatotoxicity was investigated. Twenty-eight Balb-C mice were assigned to four groups: control, APAP, EMPA10/APAP, and EMPA25/APAP. At the end of the experiment, serum hepatotoxicity biomarkers, MDA level, and GSH content were estimated. Hepatic mitofusin-2 (MFN2), optic atrophy 1 (OPA1), dynamin-related protein 1 (Drp1), and mitochondrial fission 1 protein (FIS1) were immunoassayed. PGC-1α, cGAS, and STING mRNA expression were assessed by real-time PCR. Histopathological changes and immunohistochemistry of INF-ß, p-NF-κB, and iNOS were evaluated. APAP treatment caused significant hepatic functional impairment and increased hepatic MDA levels, as well as a concomitant decrease in GSH content. Marked elevation in Drp1 and FIS1 levels, INF-ß, p-NF-κB, and iNOS immunoreactivity, and reduction in MFN2 and OPA1 levels in the APAP-injected group, PGC-1α downregulation, and high expression of cGAS and STING were also documented. EMPA effectively ameliorated APAP-generated structural and functional changes in the liver, restored redox homeostasis and mitochondrial dynamics balance, and enhanced mitochondrial biogenesis, remarkably diminished hepatic expression of cGAS and STING, and elicited a reduction in hepatic inflammation. Moreover, the computational modeling data support the interaction of APAP with antioxidant system-related proteins as well as the interactions of EMPA against Drp1, cGAS, IKKA, and iNOS proteins. Our findings demonstrated for the first time that EMPA has an ameliorative impact against APAP-induced hepatotoxicity in mice via modulation of mitochondrial dynamics, biogenesis, and cGAS/STING-dependent inflammation. Thus, this study concluded that EMPA could be a promising therapeutic modality for acute liver toxicity.


Sujet(s)
Acétaminophène , Composés benzhydryliques , Lésions hépatiques dues aux substances , Dynamines , dGTPases , Glucosides , Protéines membranaires , Dynamique mitochondriale , Nucleotidyltransferases , Animaux , Mâle , Souris , Acétaminophène/toxicité , Acétaminophène/effets indésirables , Composés benzhydryliques/pharmacologie , Composés benzhydryliques/toxicité , Lésions hépatiques dues aux substances/métabolisme , Lésions hépatiques dues aux substances/traitement médicamenteux , Dynamines/métabolisme , Dynamines/génétique , Glucosides/pharmacologie , dGTPases/métabolisme , Foie/métabolisme , Foie/effets des médicaments et des substances chimiques , Foie/anatomopathologie , Protéines membranaires/métabolisme , Protéines membranaires/génétique , Souris de lignée BALB C , Dynamique mitochondriale/effets des médicaments et des substances chimiques , Protéines mitochondriales/métabolisme , Facteur de transcription NF-kappa B/métabolisme , Nucleotidyltransferases/métabolisme , Biogenèse des organelles , Coactivateur 1-alpha du récepteur gamma activé par les proliférateurs de peroxysomes/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques
10.
Proc Natl Acad Sci U S A ; 121(30): e2313609121, 2024 Jul 23.
Article de Anglais | MEDLINE | ID: mdl-39012824

RÉSUMÉ

Mitofusins (Mfn1 and Mfn2) are the mitochondrial outer-membrane fusion proteins in mammals and belong to the dynamin superfamily of multidomain GTPases. Recent structural studies of truncated variants lacking alpha helical transmembrane domains suggested that Mfns dimerize to promote the approximation and the fusion of the mitochondrial outer membranes upon the hydrolysis of guanine 5'-triphosphate disodium salt (GTP). However, next to the presence of GTP, the fusion activity seems to require multiple regulatory factors that control the dynamics and kinetics of mitochondrial fusion through the formation of Mfn1-Mfn2 heterodimers. Here, we purified and reconstituted the full-length murine Mfn2 protein into giant unilamellar vesicles (GUVs) with different lipid compositions. The incubation with GTP resulted in the fusion of Mfn2-GUVs. High-speed video-microscopy showed that the Mfn2-dependent membrane fusion pathway progressed through a zipper mechanism where the formation and growth of an adhesion patch eventually led to the formation of a membrane opening at the rim of the septum. The presence of physiological concentration (up to 30 mol%) of dioleoyl-phosphatidylethanolamine (DOPE) was shown to be a requisite to observe GTP-induced Mfn2-dependent fusion. Our observations show that Mfn2 alone can promote the fusion of micron-sized DOPE-enriched vesicles without the requirement of regulatory cofactors, such as membrane curvature, or the assistance of other proteins.


Sujet(s)
dGTPases , Fusion membranaire , Animaux , dGTPases/métabolisme , dGTPases/génétique , Souris , Fusion membranaire/physiologie , Liposomes unilamellaires/métabolisme , Liposomes unilamellaires/composition chimique , Guanosine triphosphate/métabolisme , Phosphatidyléthanolamine/métabolisme , Membranes mitochondriales/métabolisme , Protéines mitochondriales/métabolisme , Protéines mitochondriales/génétique , Mitochondries/métabolisme
11.
Biomed Pharmacother ; 177: 116953, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38955087

RÉSUMÉ

The second most common mutation in melanoma occurs in NRAS oncogene, being a more aggressive disease that has no effective approved treatment. Besides, cellular plasticity limits better outcomes of the advanced and therapy-resistant patients. Peroxiredoxins (PRDXs) control cellular processes through direct hydrogen peroxide oxidation or by redox-relaying processes. Here, we demonstrated that PRDX2 could act as a modulator of multiple EMT markers in NRAS-mutated melanomas. PRDX2 knockdown lead to phenotypic changes towards invasion in human reconstructed skin and the treatment with a PRDX mimetic (gliotoxin), decreased migration in PRDX2-deficient cells. We also confirmed the favorable clinical outcome of patients expressing PRDX2 in a large primary melanoma cohort. This study contributes to our knowledge about genes involved in phenotype switching and opens a new perspective for PRDX2 as a biomarker and target in NRAS-mutated melanomas.


Sujet(s)
Transition épithélio-mésenchymateuse , dGTPases , Mélanome , Protéines membranaires , Mutation , Invasion tumorale , Peroxirédoxines , Humains , Mélanome/génétique , Mélanome/anatomopathologie , Mélanome/traitement médicamenteux , Mélanome/métabolisme , Peroxirédoxines/génétique , Peroxirédoxines/métabolisme , Lignée cellulaire tumorale , dGTPases/génétique , dGTPases/métabolisme , Protéines membranaires/génétique , Protéines membranaires/métabolisme , Transition épithélio-mésenchymateuse/génétique , Transition épithélio-mésenchymateuse/effets des médicaments et des substances chimiques , Mouvement cellulaire/effets des médicaments et des substances chimiques , Mouvement cellulaire/génétique , Tumeurs cutanées/génétique , Tumeurs cutanées/anatomopathologie , Tumeurs cutanées/métabolisme , Tumeurs cutanées/traitement médicamenteux , Femelle , Marqueurs biologiques tumoraux/génétique , Marqueurs biologiques tumoraux/métabolisme , Régulation de l'expression des gènes tumoraux
12.
Nagoya J Med Sci ; 86(2): 326-332, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38962422

RÉSUMÉ

We previously reported the Marimo cell line, which was established from the bone marrow cells of a patient with essential thrombocythemia (ET) at the last stage after transformation to acute myeloid leukemia (AML). This cell line is widely used for the biological analysis of ET because it harbors CALR mutation. However, genetic processes during disease progression in the original patient were not analyzed. We sequentially analyzed the genetic status in the original patient samples during disease progression. The ET clone had already acquired CALR and MPL mutations, and TP53 and NRAS mutations affected the disease progression from ET to AML in this patient. Particularly, the variant allele frequency of the NRAS mutation increased along with the disease progression after transformation, and the NRAS-mutated clone selectively proliferated in vitro, resulting in the establishment of the Marimo cell line. Although CALR and MPL mutations co-existed, MPL was not expressed in Marimo cells or any clinical samples. Furthermore, mitogen-activated protein kinase (MAPK) but not the JAK2-STAT pathway was activated. These results collectively indicate that MAPK activation is mainly associated with the proliferation ability of Marimo cells.


Sujet(s)
Calréticuline , Évolution clonale , Leucémie aigüe myéloïde , Mutation , Récepteurs à la thrombopoïétine , Thrombocytémie essentielle , Humains , Thrombocytémie essentielle/génétique , Thrombocytémie essentielle/anatomopathologie , Leucémie aigüe myéloïde/génétique , Leucémie aigüe myéloïde/anatomopathologie , Calréticuline/génétique , Calréticuline/métabolisme , Récepteurs à la thrombopoïétine/génétique , Évolution clonale/génétique , Kinase Janus-2/génétique , Kinase Janus-2/métabolisme , Protéines membranaires/génétique , Protéines membranaires/métabolisme , dGTPases/génétique , Protéine p53 suppresseur de tumeur/génétique , Protéine p53 suppresseur de tumeur/métabolisme , Mâle , Évolution de la maladie , Femelle , Lignée cellulaire tumorale , Sujet âgé , Adulte d'âge moyen
13.
Sci Rep ; 14(1): 16607, 2024 07 18.
Article de Anglais | MEDLINE | ID: mdl-39025927

RÉSUMÉ

Immune checkpoint inhibitors (ICI) have emerged as a promising therapeutic option for melanoma, which demonstrating improved clinical outcomes in melanoma patients regardless of specific genetic mutations. However, the identification of reliable biomarkers for predicting immunotherapy response and prognosis remains a challenge. In this study, we performed genetic profiling of the melanoma patients with different subtypes and evaluated the efficacy of ICI treatment. A total of 221 melanoma patients were included in our cohort, consisting primarily of acral lentiginous melanoma (ALM), cutaneous malignant melanoma (CMM), and mucosal malignant melanoma (MMM). Genetic analysis revealed BRAF mutations was predominant in CMM and NRAS mutations was prevalent in ALM. Copy number variants (CNVs) and structural variants (SV) were also detected, with CCND1 and CDK4 being the most affected genes in CNV and BRAF, ALK and RAF1 being the druggable targets in SV. Furthermore, NRAS mutations were associated with a poor prognosis in ALM, while TERT mutations were linked to unfavorable outcomes in CMM after receiving PD-1 therapy. Additionally, ALK expression exhibited improved outcomes in both ALM and CMM subtypes. Our study provides a comprehensive genomic and pathological profiling of Chinese melanoma patients, shedding light on the molecular landscape of the disease. Furthermore, numbers of gene mutations and ALK expression were identified as prognostic indicators. These findings contribute to the understanding of melanoma genetics in the Chinese population and have implications for personalized treatment approaches.


Sujet(s)
Immunothérapie , Mélanome , Adulte , Sujet âgé , Femelle , Humains , Mâle , Adulte d'âge moyen , Marqueurs biologiques tumoraux/génétique , Chine , Variations de nombre de copies de segment d'ADN , Peuples d'Asie de l'Est/génétique , dGTPases/génétique , Inhibiteurs de points de contrôle immunitaires/usage thérapeutique , Immunothérapie/méthodes , Mélanome/diagnostic , Mélanome/traitement médicamenteux , Mélanome/génétique , Protéines membranaires/génétique , Mutation , Pronostic , Protéines proto-oncogènes B-raf/génétique , Tumeurs cutanées/diagnostic , Tumeurs cutanées/traitement médicamenteux , Tumeurs cutanées/génétique
14.
Int J Mol Sci ; 25(13)2024 Jun 30.
Article de Anglais | MEDLINE | ID: mdl-39000346

RÉSUMÉ

Autosomal dominant optic atrophy (ADOA) is a rare progressive disease mainly caused by mutations in OPA1, a nuclear gene encoding for a mitochondrial protein that plays an essential role in mitochondrial dynamics, cell survival, oxidative phosphorylation, and mtDNA maintenance. ADOA is characterized by the degeneration of retinal ganglion cells (RGCs). This causes visual loss, which can lead to legal blindness in many cases. Nowadays, there is no effective treatment for ADOA. In this article, we have established an isogenic human RGC model for ADOA using iPSC technology and the genome editing tool CRISPR/Cas9 from a previously generated iPSC line of an ADOA plus patient harboring the pathogenic variant NM_015560.3: c.1861C>T (p.Gln621Ter) in heterozygosis in OPA1. To this end, a protocol based on supplementing the iPSC culture media with several small molecules and defined factors trying to mimic embryonic development has been employed. Subsequently, the created model was validated, confirming the presence of a defect of intergenomic communication, impaired mitochondrial respiration, and an increase in apoptosis and ROS generation. Finally, we propose the analysis of OPA1 expression by qPCR as an easy read-out method to carry out future drug screening studies using the created RGC model. In summary, this model provides a useful platform for further investigation of the underlying pathophysiological mechanisms of ADOA plus and for testing compounds with potential pharmacological action.


Sujet(s)
dGTPases , Cellules souches pluripotentes induites , Atrophie optique autosomique dominante , Cellules ganglionnaires rétiniennes , Humains , Atrophie optique autosomique dominante/génétique , Atrophie optique autosomique dominante/anatomopathologie , Atrophie optique autosomique dominante/métabolisme , Cellules souches pluripotentes induites/métabolisme , dGTPases/génétique , dGTPases/métabolisme , Cellules ganglionnaires rétiniennes/métabolisme , Cellules ganglionnaires rétiniennes/anatomopathologie , Systèmes CRISPR-Cas , Édition de gène/méthodes , Mutation , Apoptose/génétique , Espèces réactives de l'oxygène/métabolisme , Mitochondries/métabolisme , Mitochondries/génétique
15.
Clin Nutr ; 43(7): 1816-1831, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38870662

RÉSUMÉ

BACKGROUND: Optical atrophy 1 (OPA1), a protein accountable for mitochondrial fusion, facilitates the restoration of mitochondrial structure and function following cerebral ischemia/reperfusion (I/R) injury. The OPA1-conferred mitochondrial protection involves its expression and activity, which can be improved by SIRT3 in non-cerebral ischemia. Nevertheless, it remains obscure whether SIRT3 enhances the expression and activity of OPA1 after cerebral I/R injury. METHODS: Mature male Sprague Dawley rats were intracranially injected with adeno-associated viral-Sirtuin-3(AAV-SIRT3) and AAV-sh_OPA1, followed by a 90-min temporary blockage of the middle cerebral artery and subsequent restoration of blood flow. Cultured cortical neurons of rats were transfected with LV-SIRT3 or LV-sh_OPA1 before a 2-h oxygen-glucose deprivation and reoxygenation. The rats and neurons were subsequently treated with a selective OPA1 activity inhibitor (MYLS22). The interaction between SIRT3 and OPA1 was assessed by molecular dynamics simulation technology and co-immunoprecipitation. The expression, function, and specific protective mechanism of SIRT3 were examined by various analyses. RESULTS: SIRT3 interacted with OPA1 in the rat cerebral cortex before and after cerebral I/R. After cerebral I/R damage, SIRT3 upregulation increased the OPA1 expression, which enhanced deacetylation and OPA1 activity, thus alleviating cerebral infarct volume, neuronal apoptosis, oxidative pressure, and impairment in mitochondrial energy production; SIRT3 upregulation also improved neuromotor performance, repaired mitochondrial ultrastructure and membrane composition, and promoted the mitochondrial biogenesis. These neuroprotective effects were partly reversed by OPA1 expression interference and OPA1 activity inhibitor MYLS22. CONCLUSION: In rats, SIRT3 enhances the expression and activity of OPA1, facilitating the repair of mitochondrial structure and functional recovery following cerebral I/R injury. These findings highlight that regulating SIRT3 may be a promising therapeutic strategy for ischemic stroke.


Sujet(s)
dGTPases , Accident vasculaire cérébral ischémique , Mitochondries , Rat Sprague-Dawley , Sirtuine-3 , Animaux , Mâle , dGTPases/métabolisme , dGTPases/génétique , Sirtuine-3/métabolisme , Sirtuine-3/génétique , Rats , Mitochondries/métabolisme , Accident vasculaire cérébral ischémique/métabolisme , Neurones/métabolisme , Lésion d'ischémie-reperfusion/métabolisme , Modèles animaux de maladie humaine , Récupération fonctionnelle , Sirtuines
16.
Cell Death Dis ; 15(6): 399, 2024 Jun 07.
Article de Anglais | MEDLINE | ID: mdl-38849335

RÉSUMÉ

The loss of dopaminergic neurons in the substantia nigra is a hallmark of pathology in Parkinson's disease (PD). Dimethylarginine dimethylaminohydrolase-1 (DDAH-1) is the critical enzyme responsible for the degradation of asymmetric dimethylarginine (ADMA) which inhibits nitric oxide (NO) synthase and has been implicated in neurodegeneration. Mitochondrial dysfunction, particularly in the mitochondria-associated endoplasmic reticulum membrane (MAM), plays a critical role in this process, although the specific molecular target has not yet been determined. This study aims to examine the involvement of DDAH-1 in the nigrostriatal dopaminergic pathway and PD pathogenesis. The distribution of DDAH-1 in the brain and its colocalization with dopaminergic neurons were observed. The loss of dopaminergic neurons and aggravated locomotor disability after rotenone (ROT) injection were showed in the DDAH-1 knockout rat. L-arginine (ARG) and NO donors were employed to elucidate the role of NO respectively. In vitro, we investigated the effects of DDAH-1 knockdown or overexpression on cell viability and mitochondrial functions, as well as modulation of ADMA/NO levels using ADMA or ARG. MAM formation was assessed by the Mitofusin2 oligomerization and the mitochondrial ubiquitin ligase (MITOL) phosphorylation. We found that DDAH-1 downregulation resulted in enhanced cell death and mitochondrial dysfunctions, accompanied by elevated ADMA and reduced NO levels. However, the recovered NO level after the ARG supplement failed to exhibit a protective effect on mitochondrial functions and partially restored cell viability. DDAH-1 overexpression prevented ROT toxicity, while ADMA treatment attenuated these protective effects. The declines of MAM formation in ROT-treated cells were exacerbated by DDAH-1 downregulation via reduced MITOL phosphorylation, which was reversed by DDAH-1 overexpression. Together, the abundant expression of DDAH-1 in nigral dopaminergic neurons may exert neuroprotective effects by maintaining MAM formation and mitochondrial function probably via ADMA, indicating the therapeutic potential of targeting DDAH-1 for PD.


Sujet(s)
Amidohydrolases , Arginine , Neurones dopaminergiques , Réticulum endoplasmique , Mitochondries , Monoxyde d'azote , Maladie de Parkinson , Neurones dopaminergiques/métabolisme , Neurones dopaminergiques/effets des médicaments et des substances chimiques , Neurones dopaminergiques/anatomopathologie , Animaux , Amidohydrolases/métabolisme , Amidohydrolases/génétique , Mitochondries/métabolisme , Mitochondries/effets des médicaments et des substances chimiques , Maladie de Parkinson/métabolisme , Maladie de Parkinson/anatomopathologie , Maladie de Parkinson/génétique , Arginine/métabolisme , Arginine/analogues et dérivés , Réticulum endoplasmique/métabolisme , Réticulum endoplasmique/effets des médicaments et des substances chimiques , Rats , Monoxyde d'azote/métabolisme , Mâle , Rat Sprague-Dawley , Humains , dGTPases/métabolisme , dGTPases/génétique , Roténone/pharmacologie , Protéines mitochondriales/métabolisme ,
17.
Cell Commun Signal ; 22(1): 332, 2024 Jun 17.
Article de Anglais | MEDLINE | ID: mdl-38886790

RÉSUMÉ

BACKGROUND: HRASKO/NRASKO double knockout mice exhibit exceedingly high rates of perinatal lethality due to respiratory failure caused by a significant lung maturation delay. The few animals that reach adulthood have a normal lifespan, but present areas of atelectasis mixed with patches of emphysema and normal tissue in the lung. METHODS: Eight double knockout and eight control mice were analyzed using micro-X-ray computerized tomography and a Small Animal Physiological Monitoring system. Tissues and samples from these mice were analyzed using standard histological and Molecular Biology methods and the significance of the results analyzed using a Student´s T-test. RESULTS: The very few double knockout mice surviving up to adulthood display clear craniofacial abnormalities reminiscent of those seen in RASopathy mouse models, as well as thrombocytopenia, bleeding anomalies, and reduced platelet activation induced by thrombin. These surviving mice also present heart and spleen hyperplasia, and elevated numbers of myeloid-derived suppressor cells in the spleen. Mechanistically, we observed that these phenotypic alterations are accompanied by increased KRAS-GTP levels in heart, platelets and primary mouse embryonic fibroblasts from these animals. CONCLUSIONS: Our data uncovers a new, previously unidentified mechanism capable of triggering a RASopathy phenotype in mice as a result of the combined removal of HRAS and NRAS.


Sujet(s)
dGTPases , Souris knockout , Phénotype , Protéines proto-oncogènes p21(ras) , Animaux , Protéines proto-oncogènes p21(ras)/génétique , Protéines proto-oncogènes p21(ras)/métabolisme , Souris , dGTPases/génétique , dGTPases/métabolisme , Protéines membranaires/génétique , Protéines membranaires/métabolisme , Activation plaquettaire/génétique , Rate/anatomopathologie , Rate/métabolisme , Protéines G monomériques
18.
Sci Adv ; 10(26): eadk2913, 2024 Jun 28.
Article de Anglais | MEDLINE | ID: mdl-38941455

RÉSUMÉ

The blood-brain barrier (BBB) acts as the crucial physical filtration structure in the central nervous system. Here, we investigate the role of a specific subset of astrocytes in the regulation of BBB integrity. We showed that Dmp1-expressing astrocytes transfer mitochondria to endothelial cells via their endfeet for maintaining BBB integrity. Deletion of the Mitofusin 2 (Mfn2) gene in Dmp1-expressing astrocytes inhibited the mitochondrial transfer and caused BBB leakage. In addition, the decrease of MFN2 in astrocytes contributes to the age-associated reduction of mitochondrial transfer efficiency and thus compromises the integrity of BBB. Together, we describe a mechanism in which astrocytes regulate BBB integrity through mitochondrial transfer. Our findings provide innnovative insights into the cellular framework that underpins the progressive breakdown of BBB associated with aging and disease.


Sujet(s)
Astrocytes , Barrière hémato-encéphalique , Cellules endothéliales , Mitochondries , Astrocytes/métabolisme , Barrière hémato-encéphalique/métabolisme , Animaux , Mitochondries/métabolisme , Souris , Cellules endothéliales/métabolisme , dGTPases/métabolisme , dGTPases/génétique
19.
Cancer Control ; 31: 10732748241262179, 2024.
Article de Anglais | MEDLINE | ID: mdl-38875469

RÉSUMÉ

OBJECTIVES: The present study aimed to evaluate the frequencies of KRAS, NRAS, and BRAF mutations and their possible associations with clinicopathological features in 249 Moroccan patients with colorectal cancer (CRC). METHODS: A retrospective investigation of a cohort of formalin-fixed paraffin-embedded tissues of 249 patients with CRC was screened for KRAS/NRAS/BRAF mutations using Idylla™ technology and pyrosequencing. RESULTS: KRAS, NRAS, and BRAF mutations were revealed in 46.6% (116/249), 5.6% (14/249), and 2.4% (6/249) of patients. KRAS exon 2 mutations were identified in 87.9% of patients (102/116). KRAS G12D and G12 C were the most frequent, at 32.8% and 12.93%, respectively. Among the patients with KRAS exon 2 wild-type (wt), 27.6% (32/116) harbored additional KRAS mutations. Concurrent KRAS mutations were identified in 9.5% (11/116); including six in codon 146 (A146P/T/V), three in codon 61 (Q61H/L/R), one in codon 12 (G12 A and Q61H), and one in codon 13 (G13D and Q61 L). Among the NRAS exon 2 wt patients, 64.3% (9/14) harbored additional NRAS mutations. Concurrent NRAS mutations were identified in 28.6% (4/14) of NRAS-mutant patients. Since 3.2% wt KRAS were identified with NRAS mutations, concomitant KRAS and NRAS mutations were identified in 2.4% (6/249) of patients. KRAS mutations were higher in the >50-year-old age-group (P = .031), and the tumor location was revealed to be significantly associated with KRAS mutations (P = .028) predominantly in left colon (27.5%) and colon (42.2%) locations. NRAS mutations were most prevalent in the left colon (42.8%) and in well-differentiated tumors (64.2%). CONCLUSION: Detection of KRAS mutations, particularly the G12 C subtype, may be significant for patients with CRC and has possible therapeutic implications. However, rare KRAS concomitant mutations in CRC patients suggest that each individual may present distinct therapeutic responses. KRAS testing alongside the identification of other affected genes in the same patient will make the treatments even more personalized by contributing more accurately to the clinical decision process. Overall, early diagnosis using novel molecular techniques may improve the management of CRC by providing the most efficient therapies for Moroccan patients.


Sujet(s)
Tumeurs colorectales , dGTPases , Protéines membranaires , Mutation , Protéines proto-oncogènes B-raf , Protéines proto-oncogènes p21(ras) , Humains , Protéines proto-oncogènes B-raf/génétique , Tumeurs colorectales/génétique , Tumeurs colorectales/anatomopathologie , Mâle , Femelle , Protéines proto-oncogènes p21(ras)/génétique , Protéines membranaires/génétique , Adulte d'âge moyen , dGTPases/génétique , Maroc/épidémiologie , Études rétrospectives , Sujet âgé , Adulte , Sujet âgé de 80 ans ou plus , Analyse de mutations d'ADN
20.
Chem Biol Interact ; 398: 111090, 2024 Aug 01.
Article de Anglais | MEDLINE | ID: mdl-38825057

RÉSUMÉ

Mitochondrial dysfunction and endoplasmic reticulum (ER) stress play pivotal roles in the pathology of cerebral ischemia. In this study, we investigated whether phelligridimer A (PA), an active compound isolated from the medicinal and edible fungus Phellinus igniarius, ameliorates ischemic cerebral injury by restoring mitochondrial function and restricting ER stress. An in vitro cellular model of ischemic stroke-induced neuronal damage was established by exposing HT-22 neuronal cells to oxygen-glucose deprivation/reoxygenation (OGD/R). An in vivo animal model was established in rats subjected to middle cerebral artery occlusion/reperfusion (MCAO/R). The results showed that PA (1-10 µM) dose-dependently increased HT-22 cell viability, reduced OGD/R-induced lactate dehydrogenase release, and reversed OGD/R-induced apoptosis. PA reduced OGD/R-induced accumulation of reactive oxygen species, restored mitochondrial membrane potential, and increased ATP levels. Additionally, PA reduced the expression of the 78-kDa glucose-regulated protein (GRP78) and the phosphorylation of inositol-requiring enzyme-1α (p-IRE1α) and eukaryotic translation-initiation factor 2α (p-eIF2α). PA also inhibited the activation of the mitogen-activated protein kinase (MAPK) pathway in the OGD/R model. Moreover, treatment with PA restored the expression of mitofusin 2 (Mfn-2), a protein linking mitochondria and ER. The silencing of Mfn-2 abolished the protective effects of PA. The results from the animal study showed that PA (3-10 mg/kg) significantly reduced the volume of cerebral infarction and neurological deficits, which were accompanied by an increased level of Mfn-2, and decreased activation of the ER stress in the penumbra of the ipsilateral side after MCAO/R in rats. Taken together, these results indicate that PA counteracts cerebral ischemia-induced injury by restoring mitochondrial function and reducing ER stress. Therefore, PA might be a novel protective agent to prevent ischemia stroke-induced neuronal injury.


Sujet(s)
Encéphalopathie ischémique , Stress du réticulum endoplasmique , dGTPases , Rat Sprague-Dawley , Espèces réactives de l'oxygène , Lésion d'ischémie-reperfusion , Animaux , Mâle , Souris , Rats , Apoptose/effets des médicaments et des substances chimiques , Encéphalopathie ischémique/métabolisme , Encéphalopathie ischémique/traitement médicamenteux , Lignée cellulaire , Survie cellulaire/effets des médicaments et des substances chimiques , Chaperonne BiP du réticulum endoplasmique/métabolisme , Stress du réticulum endoplasmique/effets des médicaments et des substances chimiques , Facteur-2 d'initiation eucaryote/métabolisme , Glucose/métabolisme , dGTPases/métabolisme , Protéines du choc thermique/métabolisme , Infarctus du territoire de l'artère cérébrale moyenne/métabolisme , Infarctus du territoire de l'artère cérébrale moyenne/traitement médicamenteux , Potentiel de membrane mitochondriale/effets des médicaments et des substances chimiques , Mitochondries/effets des médicaments et des substances chimiques , Mitochondries/métabolisme , Protéines mitochondriales/métabolisme , Protéines mitochondriales/génétique , Neurones/effets des médicaments et des substances chimiques , Neurones/métabolisme , Neurones/anatomopathologie , Neuroprotecteurs/pharmacologie , Protein-Serine-Threonine Kinases/métabolisme , Protein-Serine-Threonine Kinases/génétique , Espèces réactives de l'oxygène/métabolisme , Lésion d'ischémie-reperfusion/métabolisme , Lésion d'ischémie-reperfusion/prévention et contrôle , Lésion d'ischémie-reperfusion/traitement médicamenteux
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE