Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 45
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Cell ; 167(7): 1705-1718.e13, 2016 Dec 15.
Article in English | MEDLINE | ID: mdl-27984722

ABSTRACT

Metformin has utility in cancer prevention and treatment, though the mechanisms for these effects remain elusive. Through genetic screening in C. elegans, we uncover two metformin response elements: the nuclear pore complex (NPC) and acyl-CoA dehydrogenase family member-10 (ACAD10). We demonstrate that biguanides inhibit growth by inhibiting mitochondrial respiratory capacity, which restrains transit of the RagA-RagC GTPase heterodimer through the NPC. Nuclear exclusion renders RagC incapable of gaining the GDP-bound state necessary to stimulate mTORC1. Biguanide-induced inactivation of mTORC1 subsequently inhibits growth through transcriptional induction of ACAD10. This ancient metformin response pathway is conserved from worms to humans. Both restricted nuclear pore transit and upregulation of ACAD10 are required for biguanides to reduce viability in melanoma and pancreatic cancer cells, and to extend C. elegans lifespan. This pathway provides a unified mechanism by which metformin kills cancer cells and extends lifespan, and illuminates potential cancer targets. PAPERCLIP.


Subject(s)
Metformin/pharmacology , Acyl-CoA Dehydrogenase/genetics , Aging , Animals , Body Size , Caenorhabditis elegans/growth & development , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , Cell Line, Tumor , DNA-Binding Proteins/metabolism , Humans , Longevity , Mechanistic Target of Rapamycin Complex 1 , Mitochondria/metabolism , Monomeric GTP-Binding Proteins/metabolism , Multiprotein Complexes/metabolism , Neoplasms/drug therapy , Nuclear Pore/metabolism , Oxidative Phosphorylation , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/metabolism , Transcription Factors/metabolism
2.
Cell Mol Life Sci ; 81(1): 367, 2024 Aug 22.
Article in English | MEDLINE | ID: mdl-39174697

ABSTRACT

Hydroxylated fatty acids are important intermediates in lipid metabolism and signaling. Surprisingly, the metabolism of 4-hydroxy fatty acids remains largely unexplored. We found that both ACAD10 and ACAD11 unite two enzymatic activities to introduce these metabolites into mitochondrial and peroxisomal ß-oxidation, respectively. First, they phosphorylate 4-hydroxyacyl-CoAs via a kinase domain, followed by an elimination of the phosphate to form enoyl-CoAs catalyzed by an acyl-CoA dehydrogenase (ACAD) domain. Studies in knockout cell lines revealed that ACAD10 preferentially metabolizes shorter chain 4-hydroxy fatty acids than ACAD11 (i.e. 6 carbons versus 10 carbons). Yet, recombinant proteins showed comparable activity on the corresponding 4-hydroxyacyl-CoAs. This suggests that the localization of ACAD10 and ACAD11 to mitochondria and peroxisomes, respectively, might influence their physiological substrate spectrum. Interestingly, we observed that ACAD10 is cleaved internally during its maturation generating a C-terminal part consisting of the ACAD domain, and an N-terminal part comprising the kinase domain and a haloacid dehalogenase (HAD) domain. HAD domains often exhibit phosphatase activity, but negligible activity was observed in the case of ACAD10. Yet, inactivation of a presumptive key residue in this domain significantly increased the kinase activity, suggesting that this domain might have acquired a regulatory function to prevent accumulation of the phospho-hydroxyacyl-CoA intermediate. Taken together, our work reveals that 4-hydroxy fatty acids enter mitochondrial and peroxisomal fatty acid ß-oxidation via two enzymes with an overlapping substrate repertoire.


Subject(s)
Fatty Acids , Oxidation-Reduction , Peroxisomes , Fatty Acids/metabolism , Humans , Peroxisomes/metabolism , Mitochondria/metabolism , Acyl-CoA Dehydrogenases/metabolism , Acyl-CoA Dehydrogenases/genetics , Animals , HEK293 Cells
3.
Diabetes Obes Metab ; 26(5): 1731-1745, 2024 May.
Article in English | MEDLINE | ID: mdl-38351663

ABSTRACT

AIM: Acyl-coenzyme A dehydrogenase family member 10 (ACAD10) is a mitochondrial protein purported to be involved in the fatty acid oxidation pathway. Metformin is the most prescribed therapy for type 2 diabetes; however, its precise mechanisms of action(s) are still being uncovered. Upregulation of ACAD10 is a requirement for metformin's ability to inhibit growth in cancer cells and extend lifespan in Caenorhabditis elegans. However, it is unknown whether ACAD10 plays a role in metformin's metabolic actions. MATERIALS AND METHODS: We assessed the role for ACAD10 on whole-body metabolism and metformin action by generating ACAD10KO mice on a C57BL/6J background via CRISPR-Cas9 technology. In-depth metabolic phenotyping was conducted in both sexes on a normal chow and high fat-high sucrose diet. RESULTS: Compared with wildtype mice, we detected no difference in body composition, energy expenditure or glucose tolerance in male or female ACAD10KO mice, on a chow diet or high-fat, high-sucrose diet (p ≥ .05). Hepatic mitochondrial function and insulin signalling was not different between genotypes under basal or insulin-stimulated conditions (p ≥ .05). Glucose excursions following acute administration of metformin before a glucose tolerance test were not different between genotypes nor was body composition or energy expenditure altered after 4 weeks of daily metformin treatment (p ≥ .05). Despite the lack of a metabolic phenotype, liver lipidomic analysis suggests ACAD10 depletion influences the abundance of specific ceramide species containing very long chain fatty acids, while metformin treatment altered clusters of cholesterol ester, plasmalogen, phosphatidylcholine and ceramide species. CONCLUSIONS: Loss of ACAD10 does not alter whole-body metabolism or impact the acute or chronic metabolic actions of metformin in this model.


Subject(s)
Diabetes Mellitus, Type 2 , Metformin , Male , Female , Mice , Animals , Diabetes Mellitus, Type 2/metabolism , Mice, Inbred C57BL , Metformin/pharmacology , Glucose/metabolism , Insulin , Ceramides , Sucrose , Diet, High-Fat/adverse effects
4.
Bioessays ; 44(8): e2200056, 2022 08.
Article in English | MEDLINE | ID: mdl-35708204

ABSTRACT

A decade ago I postulated that ROS formation in mitochondria was influenced by different FADH2 /NADH (F/N) ratios of catabolic substrates. Thus, fatty acid oxidation (FAO) would give higher ROS formation than glucose oxidation. Both the emergence of peroxisomes and neurons not using FAO, could be explained thus. ROS formation in NADH:ubiquinone oxidoreductase (Complex I) comes about by reverse electron transport (RET) due to high QH2 levels, and scarcity of its electron-acceptor (Q) during FAO. The then new, unexpected, finding of an FAO enzyme, ACAD9, being involved in complex I biogenesis, hinted at connections in line with the hypothesis. Recent findings about ACAD9's role in regulation of respiration fit with predictions the model makes: cementing connections between ROS production and F/N ratios. I describe how ACAD9 might be central to reversing the oxidative damage in complex I resulting from FAO. This seems to involve two distinct, but intimately connected, ACAD9 characteristics: (i) its upregulation of complex I biogenesis, and (ii) releasing FADH2 , with possible conversion into FMN, the crucial prosthetic group of complex I. Also see the video abstract here: https://youtu.be/N7AT_HBNumg.


Subject(s)
Mitochondria , NAD , Electron Transport , Electron Transport Complex I/metabolism , Mitochondria/metabolism , NAD/metabolism , Oxidation-Reduction , Reactive Oxygen Species/metabolism
5.
J Biol Phys ; 50(1): 89-118, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38103157

ABSTRACT

Acyl-CoA dehydrogenase deficiency (ACAD) is an inherited and potentially fatal disorder with variable clinical symptoms. The relationship between pathogenicity and deleterious point mutations is investigated here in ACAD structures of short (SCAD) and medium-chain (MCAD) types. Structures and dynamic features of native and mutant forms of enzymes models were compared. A total of 2.88 µs molecular dynamics simulations were performed at four different temperatures. Total energy, RMSD, protein ligand interactions and affinity, RMSF measures, secondary structure changes, and important interactions were studied. Mutations in the three main domains of ACADs are pathogenic, while those located at linker turns are not. Mutations affect mostly tetramer formations, secondary structures, and many contacts and interactions. In R206H (MCAD mutant) which is experimentally known to cause a huge turnover decrease, the lack of a single H-bond between substrate and FAD was observed. Secondary structures showed temperature-dependent changes, and SCAD activity was found to be highly correlated to the enzyme helix 3-10 content. Finally, RMSF patterns pointed to one important loop that maintains the substrate close to the active site and is a cause of substrate wobbling upon mutation. Despite similar structure, function, and cellular location, SCAD and MCAD may have different optimum temperatures that are related to the structure taken at that specific temperature. In conclusion, new insight has been provided on the effect of various SCAD and MCAD pathogenic mutations on the structure and dynamical features of the enzymes.


Subject(s)
Lipid Metabolism, Inborn Errors , Point Mutation , Humans , Virulence , Acyl-CoA Dehydrogenase/chemistry , Acyl-CoA Dehydrogenase/genetics , Lipid Metabolism, Inborn Errors/genetics , Protein Structure, Secondary
6.
J Pak Med Assoc ; 74(3): 566-569, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38591299

ABSTRACT

This study aims to assess the correl ation between t he resilience level of dental students (preclinical and clinical years) and its effects on their academic performance. It is a correlational research study that was carried out on second, third, and final-yea r denta l students at Lahore Medical & Dental College, Lahore. Academic resilience was judged by using the academic resilience scale (ARS-30). The correlation between resilience and academic performance was e s tablished by appl ying the bivar iate Pea rso n correlation. The mean age of the stude nt s was 2 1.49±1.39 years. Among 196 dental students from different years, 132 (67.35%) were females and 64(32.65%) were males. A strong p ositive co rrelati on was obser ved bet ween the academic performance and resilience of denta l students, i.e. r=0.774. From the results, it can be concluded that there is a positive correlation between academic resilience and academic performance among dental students.


Subject(s)
Academic Performance , Resilience, Psychological , Male , Female , Humans , Young Adult , Adult , Students, Dental
7.
Int J Geriatr Psychiatry ; 38(5): e5940, 2023 05.
Article in English | MEDLINE | ID: mdl-37198727

ABSTRACT

OBJECTIVES: This study aimed to examine the mediating effect of perceived social support in the relationship between loneliness and social isolation among Chinese older adults relocated for poverty relief. METHODS: We surveyed 128 older migrants from four resettlement areas in Guizhou Province, southwest China. A general information questionnaire, the Lubben Social Network Scale-6, the Perceived Social Support Rating Scale, and the Single Item Loneliness Scale were used in our study. We also used the SPSS macro PROCESS to test a mediation model and the Bootstrap method to assess its significance. RESULTS: The prevalence of social isolation among older relocators was 85.9%; the mediation model showed that loneliness had a direct negative effect on social isolation (B = -1.25, p < 0.01), and that perceived social support fully mediated this effect (-1.18), with a total effect of -1.25 (p < 0.01) and a mediating proportion of 94.4%. CONCLUSIONS: Older relocators in poverty alleviation areas experienced high levels of social isolation. Perceived social support might buffer the negative impact of loneliness on social isolation. We suggest that interventions should be designed to enhance perceived social support and reduce social isolation among this vulnerable population.


Subject(s)
East Asian People , Loneliness , Humans , Aged , Cross-Sectional Studies , Social Isolation , Social Support , Poverty
8.
Angew Chem Int Ed Engl ; 60(9): 4689-4697, 2021 02 23.
Article in English | MEDLINE | ID: mdl-33320993

ABSTRACT

Fatty acid ß-oxidation (FAO) and oxidative phosphorylation (OXPHOS) are mitochondrial redox processes that generate ATP. The biogenesis of the respiratory Complex I, a 1 MDa multiprotein complex that is responsible for initiating OXPHOS, is mediated by assembly factors including the mitochondrial complex I assembly (MCIA) complex. However, the organisation and the role of the MCIA complex are still unclear. Here we show that ECSIT functions as the bridging node of the MCIA core complex. Furthermore, cryo-electron microscopy together with biochemical and biophysical experiments reveal that the C-terminal domain of ECSIT directly binds to the vestigial dehydrogenase domain of the FAO enzyme ACAD9 and induces its deflavination, switching ACAD9 from its role in FAO to an MCIA factor. These findings provide the structural basis for the MCIA complex architecture and suggest a unique molecular mechanism for coordinating the regulation of the FAO and OXPHOS pathways to ensure an efficient energy production.


Subject(s)
Electron Transport Complex I/chemistry , Flavin-Adenine Dinucleotide/metabolism , Mitochondria/metabolism , Acyl-CoA Dehydrogenases/genetics , Acyl-CoA Dehydrogenases/metabolism , Adaptor Proteins, Signal Transducing/chemistry , Adaptor Proteins, Signal Transducing/metabolism , Cryoelectron Microscopy , Electron Transport Complex I/metabolism , Energy Metabolism , Flavin-Adenine Dinucleotide/chemistry , Humans , Oxidative Phosphorylation , Protein Interaction Domains and Motifs , Protein Structure, Tertiary , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification
9.
J Cell Biochem ; 120(2): 1773-1782, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30206977

ABSTRACT

Metformin exhibits antiproliferative and proapoptotic effects in a variety of diseases, characterized by malignant and nonmalignant hyperplastic cells; however, the underlying molecular mechanism of metformin in psoriasis has not been elucidated. In the current study, we found that after metformin treatment the proliferation of human immortalized keratinocytes (HaCaT) was significantly inhibited, while cell apoptosis was increased in a dose-dependent manner, accompanied with enhanced protein expression of acyl-coenzyme A dehydrogenase 10 (ACAD10). Furthermore, mechanism analysis revealed that ACAD10 expression is induced by downregulated activities of mechanistic target of rapamycin 1 (mTORC1) signaling rather than AMP-activated protein kinase signaling. The inactivation of mTORC1 by rapamycin pretreatment or rotenone-induced mitochondrial complex inhibition showed a similar effect because of the metformin treatment on the proliferation and apoptosis of HaCaT keratinocytes. Overexpression of mTORC1 almost reversed the antiproliferation and proapoptosis effects induced by metformin. This study showed that the metformin treatment inhibited HaCaT cells proliferation and promoted apoptosis by affecting the mitochondrial-mTORC1 signaling and elevated the ACAD10 expression. Hence, metformin can be used as a potential therapeutic agent for psoriasis.

10.
Cell Commun Signal ; 17(1): 129, 2019 10 17.
Article in English | MEDLINE | ID: mdl-31623618

ABSTRACT

BACKGROUND: While regulated WNT activity is required for normal development and stem cell maintenance, mutations that lead to constitutive activation of the WNT pathway cause cellular transformation and drive colorectal cancer. Activation of the WNT pathway ultimately leads to the nuclear translocation of ß-catenin which, in complex with TCF/LEF factors, promotes the transcription of genes necessary for growth. The proto-oncogene MYC is one of the most critical genes activated downstream the WNT pathway in colon cancer. Here, we investigate the converse regulation of the WNT pathway by MYC. METHODS: We performed RNA-seq analyses to identify genes regulated in cells expressing MYC. We validated the regulation of genes in the WNT pathway including LEF1 by MYC using RT-qPCR, Western blotting, and ChIP-seq. We investigated the importance of LEF1 for the viability of MYC-expressing cells in in fibroblasts, epithelial cells, and colon cells. Bioinformatic analyses were utilized to define the expression of MYC-regulated genes in human colon cancer and metabolomics analyses were used to identify pathways regulated by LEF1 in MYC expressing cells. RESULTS: MYC regulates the levels of numerous WNT-related genes, including the ß-catenin co-transcription factor LEF1. MYC activates the transcription of LEF1 and is required for LEF1 expression in colon cancer cells and in primary colonic cells transformed by APC loss of function, a common mutation in colon cancer patients. LEF1 caused the retention of ß-catenin in the nucleus, leading to the activation of the WNT pathway in MYC-expressing cells. Consequently, MYC-expressing cells were sensitive to LEF1 inhibition. Moreover, we describe two examples of genes induced in MYC-expressing cells that require LEF1 activity: the peroxisome proliferator activated receptor delta (PPARδ) and the Acyl CoA dehydrogenase 9 (ACAD9). CONCLUSIONS: We demonstrated that MYC is a transcriptional regulator of LEF1 in colonic cells. Our work proposes a novel pathway by which MYC regulates proliferation through activating LEF1 expression which in turn activates the WNT pathway.


Subject(s)
Lymphoid Enhancer-Binding Factor 1/genetics , Proto-Oncogene Proteins c-myc/metabolism , Transcriptional Activation , Wnt Signaling Pathway , Acyl-CoA Dehydrogenases/genetics , Cell Line , Cell Proliferation , Colonic Neoplasms/pathology , Gene Knockdown Techniques , Humans , Lymphoid Enhancer-Binding Factor 1/deficiency , PPAR delta/genetics , Proto-Oncogene Mas , beta Catenin/metabolism
11.
Mol Genet Metab ; 121(3): 224-226, 2017 07.
Article in English | MEDLINE | ID: mdl-28529009

ABSTRACT

Patients carrying Acyl-CoA dehydrogenase 9 (ACAD9) mutations reported to date mainly present with severe hypertrophic cardiomyopathy and isolated complex I (CI) dysfunction. Here we report a novel ACAD9 mutation in a young girl presenting with severe hypertrophic cardiomyopathy, isolated CI deficiency and interestingly multiple respiratory chain complexes assembly defects. We show that ACAD9 analysis has to be performed in first intention in patients presenting with cardiac hypertrophy even in the presence of multiple assembly defects.


Subject(s)
Acyl-CoA Dehydrogenases/genetics , Cardiomyopathy, Hypertrophic/genetics , Cardiomyopathy, Hypertrophic/metabolism , Electron Transport Complex I/deficiency , Mutation , Acyl-CoA Dehydrogenase/genetics , Acyl-CoA Dehydrogenases/blood , Child , Electron Transport , Electron Transport Complex I/blood , Electron Transport Complex I/genetics , Electron Transport Complex I/metabolism , Female , Humans , Infant
12.
Muscle Nerve ; 55(6): 919-922, 2017 06.
Article in English | MEDLINE | ID: mdl-27438479

ABSTRACT

INTRODUCTION: Acyl-coenzyme A dehydrogenase 9 (ACAD9) has a role in mitochondrial complex I (CI) assembly. Only a few patients who carry ACAD9 mutations have been reported. They mainly present with severe hypertrophic cardiomyopathy, although a minority have only mild isolated myopathy. Although the secondary factors influencing disease severity have not been elucidated, conservation of CI assembly and residual enzymatic activity have been suggested as explanations for the mild phenotypes associated with ACAD9 mutations. METHODS: We report a novel homozygous ACAD9 mutation (c.1240C>T; p.Arg414Cys) in a 34-year-old woman who presented with non-progressive myopathy. RESULTS: We show that this ACAD9 mutation led to a severe defect in CI assembly in the patient's muscle. Furthermore, the impact of CI deficiency is confirmed by accumulation of mitochondrial DNA deletions. CONCLUSION: Our data suggest that a major defect of CI assembly is not responsible for a severe phenotype. Muscle Nerve 55: 919-922, 2017.


Subject(s)
Acyl-CoA Dehydrogenases/metabolism , Acyl-CoA Dehydrogenases/genetics , Adult , Consanguinity , DNA Mutational Analysis , DNA, Mitochondrial/genetics , Electron Transport Complex I/genetics , Electron Transport Complex I/metabolism , Female , Humans , Muscular Diseases/genetics , Muscular Diseases/pathology , Mutation/genetics
13.
Br J Nutr ; 118(9): 641-650, 2017 Nov.
Article in English | MEDLINE | ID: mdl-29185933

ABSTRACT

Suboptimal vitamin B2 status is encountered globally. Riboflavin deficiency depresses growth and results in a fatty liver. The underlying mechanisms remain to be established and an overview of molecular alterations is lacking. We investigated hepatic proteome changes induced by riboflavin deficiency to explain its effects on growth and hepatic lipid metabolism. In all, 360 1-d-old Pekin ducks were divided into three groups of 120 birds each, with twelve replicates and ten birds per replicate. For 21 d, the ducks were fed ad libitum a control diet (CAL), a riboflavin-deficient diet (RD) or were pair-fed with the control diet to the mean daily intake of the RD group (CPF). When comparing RD with CAL and CPF, growth depression, liver enlargement, liver lipid accumulation and enhanced liver SFA (C6 : 0, C12 : 0, C16 : 0, C18 : 0) were observed. In RD, thirty-two proteins were enhanced and thirty-one diminished (>1·5-fold) compared with CAL and CPF. Selected proteins were confirmed by Western blotting. The diminished proteins are mainly involved in fatty acid ß-oxidation and the mitochondrial electron transport chain (ETC), whereas the enhanced proteins are mainly involved in TAG and cholesterol biosynthesis. RD causes liver lipid accumulation and growth depression probably by impairing fatty acid ß-oxidation and ETC. These findings contribute to our understanding of the mechanisms of liver lipid metabolic disorders due to RD.


Subject(s)
Ducks/blood , Liver/metabolism , Proteome/genetics , Riboflavin Deficiency/blood , Alanine Transaminase/blood , Animals , Aspartate Aminotransferases/blood , Blood Glucose/metabolism , Diet , Electron Transport Chain Complex Proteins/genetics , Electron Transport Chain Complex Proteins/metabolism , Lipid Metabolism , Male , Mitochondria, Liver/metabolism , Proteome/metabolism , Riboflavin/blood , Serum Albumin/metabolism
14.
Metab Brain Dis ; 32(4): 967-970, 2017 08.
Article in English | MEDLINE | ID: mdl-28374236

ABSTRACT

Reports on magnetic resonance imaging findings in patients with short chain acyl -Coenzyme A dehydrogenase (SCAD) deficiency, an autosomal recessive disorder caused by mutations in the acyl-Coenzyme A dehydrogenase (ACADS), are limited. Many asymptomatic carriers of ACAD variants have also been described necessitating careful evaluation of clinical and biochemical findings for an accurate diagnosis. Here we report a an infant with short chain acyl -Coenzyme A dehydrogenase (SCAD) deficiency diagnosed based on the characteristic biochemical findings and confirmed by genetic testing. He presented with refractory seizures and neuro regression at 4 months of age. His metabolic work up revealed elevated butyryl carnitine in plasma and ethyl malonic acid in urine. Magnetic resonance imaging of the brain showed cortical and basal ganglia signal changes with cortical swelling. Serial scans showed progression of the lesions resulting in cystic leukomalacia with brain atrophy. Exome sequencing revealed a novel homozygous nonsense variation, c.1146C > G (p.Y382Ter) in exon ten of ACADS which was further validated by Sanger sequencing. Both parents were heterozygous carriers. Follow up at 15 months showed gross psychomotor retardation and refractory seizures despite being on optimal doses of anti-epileptic medications, carnitine and multivitamin supplementation. This report expands the phenotypic and genotypic spectrum of SCAD deficiency.


Subject(s)
Acyl-CoA Dehydrogenase/deficiency , Brain/diagnostic imaging , Leukomalacia, Periventricular/diagnostic imaging , Lipid Metabolism, Inborn Errors/diagnostic imaging , Atrophy/diagnostic imaging , Disease Progression , Humans , Infant , Magnetic Resonance Imaging , Male , Seizures/diagnostic imaging
15.
Mol Genet Metab ; 118(3): 185-189, 2016 07.
Article in English | MEDLINE | ID: mdl-27233227

ABSTRACT

Acyl-CoA dehydrogenase 9 (ACAD9) is a mitochondrial protein involved in oxidative phosphorylation complex I biogenesis. This protein also exhibits acyl-CoA dehydrogenase (ACAD) activity. ACAD9-mutated patients have been reported to suffer from primarily heart, muscle, liver, and nervous system disorders. ACAD9 mutation is suspected in cases of elevated lactic acid levels combined with complex I deficiency, and confirmed by ACAD9 gene analysis. At least 18 ACAD9-mutated patients have previously been reported, usually displaying severe cardiac involvement. We retrospectively studied nine additional patients from three unrelated families with a wide spectrum of cardiac involvement between the families as well as the patients from the same families. All patients exhibited elevated lactate levels. Deleterious ACAD9 mutations were identified in all patients except one for whom it was not possible to recover DNA. To our knowledge, this is one of the first reports on isolated mild ventricular hypertrophy due to ACAD9 mutation in a family with moderate symptoms during adolescence. This report also confirms that dilated cardiomyopathy may occur in conjunction with ACAD9 mutation and that some patients may respond clinically to riboflavin treatment. Of note, several patients suffered from patent ductus arteriosus (PDA), with one exhibiting a complex congenital heart defect. It is yet unknown whether these cardiac manifestations were related to ACAD9 mutation. In conclusion, this disorder should be suspected in the presence of lactic acidosis, complex I deficiency, and any cardiac involvement, even mild.


Subject(s)
Acyl-CoA Dehydrogenases/genetics , Heart Diseases/genetics , Lactic Acid/blood , Mutation , Acyl-CoA Dehydrogenases/metabolism , Adult , Child , Female , Genetic Predisposition to Disease , Heart Diseases/drug therapy , Humans , Infant , Infant, Newborn , Male , Pedigree , Retrospective Studies , Riboflavin/therapeutic use , Treatment Outcome , Young Adult
16.
Biochim Biophys Acta ; 1834(12): 2750-60, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24140568

ABSTRACT

Hypoxia inducible factors (HIFs) are important mediators of the cellular adaptive response during acute hypoxia. The role of HIF-1 and HIF-2 during prolonged periods of hypoxia, i.e. chronic hypoxia is less defined. Therefore, we used human THP-1 macrophages with a knockdown of either HIF-1α, HIF-2α, or both HIFα-subunits, incubated them for several days under hypoxia (1% O2), and analyzed responses to hypoxia using 2D-DIGE coupled to MS/MS-analysis. Chronic hypoxia was defined as a time point when the early but transient accumulation of HIFα-subunits and mRNA expression of classical HIF target genes returned towards basal levels, with a new steady state that was constant from 72h onwards. From roughly 800 spots, that were regulated comparing normoxia to chronic hypoxia, about 100 proteins were unambiguously assigned during MS/MS-analysis. Interestingly, a number of glycolytic proteins were up-regulated, while a number of inner mitochondrial membrane proteins were down-regulated independently of HIF-1α or HIF-2α. Chronic hypoxic conditions depleted the mitochondrial mass by autophagy, which occurred independently of HIF proteins. Macrophages tolerate periods of chronic hypoxia very well and adaptive responses occur, at least in part, independently of HIF-1α and/or HIF-2α and comprise mitophagy as a pathway of particular importance.


Subject(s)
Autophagy , Macrophages/metabolism , Mitochondria/metabolism , Mitochondrial Proteins/biosynthesis , Mitophagy , Up-Regulation , Basic Helix-Loop-Helix Transcription Factors/biosynthesis , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Hypoxia/genetics , Cell Line, Tumor , Glycolysis/genetics , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/biosynthesis , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Macrophages/pathology , Mitochondria/genetics , Mitochondria/pathology , Mitochondrial Proteins/genetics
17.
Mitochondrion ; 78: 101905, 2024 May 24.
Article in English | MEDLINE | ID: mdl-38797357

ABSTRACT

Pathogenic ACAD9 variants cause complex I deficiency. Patients presenting in infancy unresponsive to riboflavin have high mortality. A six-month-old infant presented with riboflavin unresponsive lactic acidosis and life-threatening cardiomyopathy. Treatment with high dose bezafibrate and nicotinamide riboside resulted in marked clinical improvement including reduced lactate and NT-pro-brain type natriuretic peptide levels, with stabilized echocardiographic measures. After a long stable period, the child succumbed from cardiac failure with infection at 10.5 months. Therapy was well tolerated. Peak bezafibrate levels exceeded its EC50. The clinical improvement with this treatment illustrates its potential, but weak PPAR agonist activity of bezafibrate limited its efficacy.

18.
Mol Genet Metab ; 110 Suppl: S31-9, 2013.
Article in English | MEDLINE | ID: mdl-24206932

ABSTRACT

During the last two decades the realization has emerged that the phenotype of the majority of inherited genetic diseases, including inborn errors of metabolism, cannot be predicted by the genotype identified in patients. This is true for PKU and in the majority of fatty acid oxidation (FAO) defects, where the genotypes identified in patients may be allocated into two groups. One comprising big deletions and small out-of-frame deletions/insertions as well as severe splice and stop codon changes, generally giving rise to no or very little protein product, and the other group, comprising small in-frame deletions/insertions and missense variations, resulting in misfolding proteins with varying stability. In all cases of FAO defects the pathophysiology may be due to energy insufficiency as well as toxic effects from accumulated enzyme substrates. In patients carrying missense variations, it may in addition be caused by the presence of misfolding proteins. A common effect of accumulated substrates and misfolding proteins is chronic oxidative stress, the severeness of which may depend on a complex interplay of modifying factors, including genetic, cellular, environmental and dietary. In this review we will discuss the hypothesis that especially the amounts of reactive oxygen species (ROS) and reactive nitrogen species (RNS), created in connection with the electron transport chain (ETC), are the driving forces in the balance between cell survival and death. In young and healthy cells small amounts of ROS function as signaling molecules, activating cell protection systems, such as protein quality networks, antioxidant enzymes and metabolic shift from ATP production by the ETC to glycolysis. In the sick and old cell, containing misfolding and damaged proteins, the dynamic range of these protecting systems are narrowed, and cells develop a state of chronic stress, which easier than young and healthy cells may initiate cell death programs like apoptosis and necrosis. We will discuss a wealth of literature that support this hypothesis, which - if supported by studies - is important for new treatment strategies. We conclude that crude antioxidant treatment may not be beneficial, since it may inhibit the survival stress responses. We discuss the ongoing studies to enhance the residual activity of mild misfolding enzyme proteins by cofactor or chemical chaperones or by inducing the transcription of FAO enzyme proteins by bezafibrate with respect to misfolding/distorted conformational proteins ability to create ROS, and the need to know the exact pathophysiological mechanisms in order to suggest new treatment regimes.


Subject(s)
Antioxidants/therapeutic use , Fatty Acids/metabolism , Lipid Metabolism, Inborn Errors/genetics , Lipid Metabolism, Inborn Errors/metabolism , Oxidative Stress , Antioxidants/metabolism , Apoptosis/drug effects , Apoptosis/genetics , Bezafibrate/pharmacology , Electron Transport Chain Complex Proteins/chemistry , Electron Transport Chain Complex Proteins/metabolism , Genetic Variation , Genotype , Humans , Necrosis/genetics , Necrosis/pathology , Oxidative Stress/drug effects , Oxidative Stress/genetics , Phenotype , Protein Folding , Reactive Nitrogen Species/chemistry , Reactive Nitrogen Species/metabolism , Reactive Oxygen Species/chemistry , Reactive Oxygen Species/metabolism
19.
Methods Mol Biol ; 2643: 1-12, 2023.
Article in English | MEDLINE | ID: mdl-36952174

ABSTRACT

Sophisticated organelle fractionation strategies were the workhorse of early peroxisome research and led to the characterization of the principal functions of the organelle. However, even in the era of molecular biology and "omics" technologies, they are still of importance to unravel peroxisome-specific proteomes, confirm the localization of still uncharacterized proteins, analyze peroxisome metabolism or lipid composition, or study their protein import mechanism. To isolate and analyze peroxisomes for these purposes, density gradient centrifugation still represents a highly reliable and reproducible technique. This article describes two protocols to purify peroxisomes from either liver tissue or the HepG2 hepatoma cell line. The protocol for liver enables purification of peroxisome fractions with high purity (95%) and is therefore suitable to study low-abundant peroxisomal proteins or analyze their lipid composition, for example. The protocol presented for HepG2 cells is not suitable to gain highly pure peroxisomal fractions but is intended to be used for gradient profiling experiments and allows easier manipulation of the peroxisomal compartment, e.g., by gene knockdown or protein overexpression for functional studies. Both purification methods therefore represent complementary tools to be used to analyze different aspects of peroxisome physiology. Please note that this is an updated version of a protocol, which has been published in a former volume of Methods in Molecular Biology.


Subject(s)
Liver , Peroxisomes , Animals , Peroxisomes/metabolism , Cell Fractionation/methods , Liver/metabolism , Mammals , Centrifugation, Density Gradient/methods , Lipids
20.
Mol Syndromol ; 14(2): 101-108, 2023 Apr.
Article in English | MEDLINE | ID: mdl-37064341

ABSTRACT

Introduction: Antenatal presentation of hypertrophic cardiomyopathy (HCM) is rare. We describe familial recurrence of antenatal HCM associated with intrauterine growth restriction and the diagnostic process undertaken. Methods: Two pregnancies with antenatal HCM were followed up. Biological assessment including metabolic analyses, genetic analyses, and respiratory chain study was performed. We describe the clinical course of these two pregnancies, antenatal manifestations as well as specific histopathological findings, and review the literature. Results: The assessment revealed a deficiency in complex I of the respiratory chain and two likely pathogenic variations in the ACAD9 gene. Discussion and Conclusion: Antenatal HCM is rare and a diagnosis is not always made. In pregnancies presenting with cardiomyopathy and intrauterine growth restriction, ACAD9 deficiency should be considered as one of the potential underlying diagnoses, and ACAD9 molecular testing should be included among other prenatal investigations.

SELECTION OF CITATIONS
SEARCH DETAIL