Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 404
Filter
1.
Clinics (Sao Paulo) ; 79: 100410, 2024.
Article in English | MEDLINE | ID: mdl-38901133

ABSTRACT

BACKGROUND: Cuproptosis is known to regulate diverse physiological functions in many diseases, but its role in regulating Myocardial Ischemia-Reperfusion Injury (MI/RI) remains unclear. METHODS: For this purpose, the MI/RI microarray datasets GSE61592 were downloaded from the Gene Expression Omnibus (GEO) database, and the Differently Expressed Genes (DEGs) in MI/RI were identified using R software. Moreover, the MI/RI mice model was established to confirm further the diagnostic value of Pyruvate Dehydrogenase B (Pdhb), Dihydrolipoamide S-acetyltransferase (Dlat), and Pyruvate dehydrogenase E1 subunit alpha 1 (Pdhα1). RESULTS: The analysis of microarray datasets GSE61592 revealed that 798 genes were upregulated and 768 were downregulated in the myocardial tissue of the ischemia-reperfusion injury mice. Furthermore, Dlat, Pdhb, Pdhα1, and cuproptosis-related genes belonged to the downregulated genes. The receiver operating characteristics curve analysis results indicated that the Dlat, Pdhb, and Pdhα1 levels were downregulated in MI/RI and were found to be potential biomarkers for MI/RI diagnosis and prognosis. Similarly, analysis of Dlat, Pdhb, and Pdhα1 levels in the MI/RI mice revealed Pdhb being the key diagnostic marker. CONCLUSIONS: This study demonstrated the prognostic value of cuproptosis-related genes (Dlat, Pdhb, and Pdhα1), especially Pdhb, MI/RI, providing new insight into the MI/RI treatment.


Subject(s)
Computational Biology , Myocardial Reperfusion Injury , Animals , Myocardial Reperfusion Injury/genetics , Mice , Down-Regulation/genetics , Male , Disease Models, Animal , Up-Regulation , Mice, Inbred C57BL , Gene Expression Profiling/methods , Pyruvate Dehydrogenase (Lipoamide)/genetics , Biomarkers/analysis , Acetyltransferases/genetics
2.
Clin Neurol Neurosurg ; 241: 108307, 2024 06.
Article in English | MEDLINE | ID: mdl-38701546

ABSTRACT

Pyruvate dehydrogenase complex (PDC) deficiency is a genetic mitochondrial disease mostly associated with severe lactic acidosis, rapid progression of neurological symptoms and death during childhood. We present a 33-year-old male with PDC deficiency caused by a Val262Leu mutation in PDHA1gene. He demonstrated generalized dystonia affecting trunk and upper extremities and paraparesis as the most significant features, with onset of symptoms at age 8. Brain MRI showed bilaterally increased signal within the globus pallidus, typical of Leigh syndrome. A periodic lactate increase in serum and cerebrospinal fluid was detected. We describe a case of pyruvate dehydrogenase deficiency being diagnosed only 25 years after the onset of symptoms and highlight PDHC deficiency as a possible cause of treatable dystonia in childhood, which may respond well to thiamine and levodopa treatment.


Subject(s)
Pyruvate Dehydrogenase (Lipoamide) , Pyruvate Dehydrogenase Complex Deficiency Disease , Humans , Male , Adult , Pyruvate Dehydrogenase Complex Deficiency Disease/genetics , Pyruvate Dehydrogenase Complex Deficiency Disease/complications , Pyruvate Dehydrogenase (Lipoamide)/genetics , Dystonia/genetics , Dystonia/etiology , Levodopa/therapeutic use , Dystonic Disorders/genetics , Dystonic Disorders/diagnosis , Magnetic Resonance Imaging , Mutation
3.
Eur J Neurol ; 31(7): e16283, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38497591

ABSTRACT

BACKGROUND AND PURPOSE: Pyruvate dehydrogenase complex deficiency is in up to 90% caused by pathogenic variants in the X-linked PDHA1 gene. We aimed to investigate female relatives of index patients with PDHA1-related disease to (i) describe the prevalence of female PDHA1 carriers, (ii) determine whether they had symptoms and signs, and (iii) delineate the associated phenotype. METHODS: In a national population-based study, we identified 37 patients with pathogenic variants in PDHA1. Sanger sequencing for the presence of the pathogenic variant was performed in their mothers and female relatives. The identified female carriers were clinically assessed, and their medical records were reviewed. RESULTS: The proportion carrying a de novo variant was 86%. We identified seven female PDHA1 carriers from five families. Five of them exhibited clinical features of the disease and were previously undiagnosed; all had signs of peripheral axonal neuropathy, four presented with strokelike episodes including two with Leigh-like lesions, and three had facial stigmata. CONCLUSIONS: PDHA1-related disease is underrecognized in heterozygous female carriers. Peripheral axonal neuropathy, strokelike and Leigh-like changes, and facial dysmorphism should raise suspicion of the disorder. Genetic analysis and clinical examination of potential female carriers are important for genetic counseling and have implications for treatment.


Subject(s)
Heterozygote , Pyruvate Dehydrogenase (Lipoamide) , Pyruvate Dehydrogenase Complex Deficiency Disease , Humans , Female , Pyruvate Dehydrogenase (Lipoamide)/genetics , Pyruvate Dehydrogenase Complex Deficiency Disease/genetics , Adult , Middle Aged , Adolescent , Young Adult , Phenotype , Child , Pedigree
4.
Diabetes Metab J ; 48(3): 405-417, 2024 May.
Article in English | MEDLINE | ID: mdl-38311057

ABSTRACT

BACKGRUOUND: Reactive oxygen species (ROS) and inflammation are reported to have a fundamental role in the pathogenesis of ischemia-reperfusion (IR) injury, a leading cause of acute kidney injury. The present study investigated the role of pyruvate dehydrogenase kinase 4 (PDK4) in ROS production and inflammation following IR injury. METHODS: We used a streptozotocin-induced diabetic C57BL6/J mouse model, which was subjected to IR by clamping both renal pedicles. Cellular apoptosis and inflammatory markers were evaluated in NRK-52E cells and mouse primary tubular cells after hypoxia and reoxygenation using a hypoxia work station. RESULTS: Following IR injury in diabetic mice, the expression of PDK4, rather than the other PDK isoforms, was induced with a marked increase in pyruvate dehydrogenase E1α (PDHE1α) phosphorylation. This was accompanied by a pronounced ROS activation, as well as tumor necrosis factor-α (TNF-α), interleukin-6 (IL-6), interleukin-1ß (IL-1ß), and monocyte chemoattractant protein-1 (MCP-1) production. Notably, sodium dichloroacetate (DCA) attenuated renal IR injury-induced apoptosis which can be attributed to reducing PDK4 expression and PDHE1α phosphorylation levels. DCA or shPdk4 treatment reduced oxidative stress and decreased TNF-α, IL-6, IL-1ß, and MCP-1 production after IR or hypoxia-reoxygenation injury. CONCLUSION: PDK4 inhibition alleviated renal injury with decreased ROS production and inflammation, supporting a critical role for PDK4 in IR mediated damage. This result indicates another potential target for reno-protection during IR injury; accordingly, the role of PDK4 inhibition needs to be comprehensively elucidated in terms of mitochondrial function during renal IR injury.


Subject(s)
Apoptosis , Diabetes Mellitus, Experimental , Inflammation , Mice, Inbred C57BL , Oxidative Stress , Pyruvate Dehydrogenase Acetyl-Transferring Kinase , Reactive Oxygen Species , Reperfusion Injury , Animals , Reperfusion Injury/metabolism , Reperfusion Injury/prevention & control , Oxidative Stress/drug effects , Mice , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/metabolism , Male , Reactive Oxygen Species/metabolism , Inflammation/metabolism , Pyruvate Dehydrogenase Acetyl-Transferring Kinase/metabolism , Pyruvate Dehydrogenase Acetyl-Transferring Kinase/antagonists & inhibitors , Apoptosis/drug effects , Acute Kidney Injury/metabolism , Acute Kidney Injury/prevention & control , Acute Kidney Injury/etiology , Kidney/pathology , Kidney/metabolism , Pyruvate Dehydrogenase (Lipoamide)/metabolism , Dichloroacetic Acid/pharmacology , Phosphorylation/drug effects , Diabetic Nephropathies/metabolism
5.
Int J Mol Sci ; 25(3)2024 Feb 05.
Article in English | MEDLINE | ID: mdl-38339218

ABSTRACT

The previous publication identified that pyruvate dehydrogenase E1 (PDHE1) positively regulated the process of male reproduction in M. nipponense through affecting the expressions of insulin-like androgenic gland hormone. The present study aimed to identify the potential male-reproduction-related genes that were regulated by PDHE1 through performing the transcriptome profiling analysis in the testis and androgenic gland after the knockdown of the expressions of PDHE1 by the injection of dsPDHE1. Both RNA-Seq and qPCR analysis identified the significant decreases in PDHE1 expressions in the testis and androgenic gland in dsPDHE1-injected prawns compared to those in dsGFP-injected prawns, indicating the efficiency of dsPDHE1 in the present study. Transcriptome profiling analysis identified 56 and 127 differentially expressed genes (DEGs) in the testis and androgenic gland, respectively. KEGG analysis revealed that the energy-metabolism-related pathways represented the main enriched metabolic pathways of DEGs in both the testis and androgenic gland, including pyruvate metabolism, the Citrate cycle (TCA cycle), Glycolysis/Gluconeogenesis, and the Glucagon signaling pathway. Thus, it is predicted that these metabolic pathways and the DEGs from these metabolic pathways regulated by PDHE1 may be involved in the regulation of male reproduction in M. nipponense. Furthermore, four genes were found to be differentially expressed in both the testis and androgenic gland, of which ribosomal protein S3 was down-regulated and uncharacterized protein LOC113829596 was up-regulated in both the testis and androgenic gland in dsPDHE1-injected prawns. The present study provided valuable evidence for the establishment of an artificial technique to regulate the process of male reproduction in M. nipponense.


Subject(s)
Palaemonidae , Animals , Male , Palaemonidae/genetics , Testis/metabolism , Pyruvate Dehydrogenase (Lipoamide)/genetics , Androgens/metabolism , Gene Expression Profiling/methods , Reproduction , Transcriptome
8.
Article in English | MEDLINE | ID: mdl-37730160

ABSTRACT

BACKGROUND: The altered cellular metabolism is one of the hallmarks of the cancer cells, favoring the process of aerobic glycolysis, known as the Warburg effect. The pyruvate dehydrogenase (PDH) complex is one of the elements involved in this metabolic process. The present study aims to evaluate the relationship between the transcriptional expression of PDHB and the risk of local recurrence in patients with oral cavity carcinomas. METHODS: We determined the transcriptional expression of PDHB in biopsies from 41 patients with oral cavity carcinomas treated with surgery. The PDHB expression was categorized according to the local control of the disease with a recursive partitioning analysis. RESULTS: During the follow-up period 13 patients (31.7%) had a local recurrence of the tumor. Considering local disease control as the dependent variable, the recursive partitioning analysis classified the patients in two categories according to high (n=16, 39.0%) or low (n=25, 61.0%) PDHB expression. Five-year local recurrence-free survival for patients with high PDHB expression was 84.8% (95% CI: 65.2-100%), and for patients with low expression it was 54.3% (95% CI: 34.3-74.2 %) (P=0.034). The results of multivariate analysis showed that patients with a low PDHB expression had a 4.90 times higher risk of local recurrence of the tumor (95% CI: 1.02-22.68, P=0.042). CONCLUSION: There is a relationship between the metabolic characteristics of the tumor and its aggressiveness. According to our results, patients with oral cavity carcinomas with low transcriptional expression levels of PDHB have a significantly higher risk of local tumor recurrence.


Subject(s)
Carcinoma , Pyruvate Dehydrogenase (Lipoamide) , Humans , Pyruvate Dehydrogenase (Lipoamide)/genetics , Pyruvate Dehydrogenase (Lipoamide)/metabolism , Mouth/metabolism , Pyruvates
9.
Exp Neurol ; 363: 114368, 2023 05.
Article in English | MEDLINE | ID: mdl-36863478

ABSTRACT

Key metabolic enzymes not only regulate Glucose, lipid, amino acid metabolism to serve the cellular energy needs, but also modulate noncanonical or nonmetabolic signaling pathway such as gene expression, cell-cycle progression, DNA repair, apoptosis and cell proliferation in regulating the pathologic progression of disease. However, the role of glycometabolism in peripheral nerve axon regeneration is little known. In this study, we investigated the expression of Pyruvate dehydrogenase E1(PDH), a key enzyme linking glycolysis and the tricarboxylic acid (TCA) cycle, with qRT-PCR and found that pyruvate dehydrogenase beta subunit (Pdhb) is up-regulated at the early stage during peripheral nerve injury. The knockdown of Pdhb inhibits neurite outgrowth of primary DRG neurons in vitro and restrains axon regeneration of sciatic nerve after crush injury. Pdhb overexpression promoting axonal regeneration is reversed by knockdown of Monocarboxylate transporter 2(Mct2), a transporter involved in the transport and metabolism of lactate, indicating Pdhb promoting axon regeneration depends on lactate for energy supply. Given the nucleus-localization of Pdhb, further analysis revealed that Pdhb enhances the acetylation of H3K9 and affecting the expression of genes involved in arachidonic acid metabolism and Ras signaling pathway, such as Rsa-14-44 and Pla2g4a, thereby promoting axon regeneration. Collectively, our data indicates that Pdhb is a positive dual modulator of energy generation and gene expression in regulating peripheral axon regeneration.


Subject(s)
Axons , Nerve Regeneration , Axons/pathology , Nerve Regeneration/physiology , Pyruvate Dehydrogenase (Lipoamide)/genetics , Pyruvate Dehydrogenase (Lipoamide)/metabolism , Gene Expression , Lactates/metabolism , Pyruvates/metabolism
10.
Org Biomol Chem ; 21(8): 1755-1763, 2023 02 22.
Article in English | MEDLINE | ID: mdl-36723268

ABSTRACT

Suppression of pyruvate dehydrogenase complex (PDHc) is a mechanism for cancer cells to manifest the Warburg effect. However, recent evidence suggests that whether PDHc activity is suppressed or activated depends on the type of cancer. The PDHc E1 subunit (PDH E1) is a thiamine pyrophosphate (TPP)-dependent enzyme, catalysing the first and rate-limiting step of PDHc; thus, there is a need for selective PDH E1 inhibitors. There is, however, inadequate understanding of the structure-activity relationship (SAR) and a lack of inhibitors specific for mammalian PDH E1. Our group have reported TPP analogues as TPP-competitive inhibitors to study the family of TPP-dependent enzymes. Most of these TPP analogues cannot be used to study PDHc in cells because (a) they inhibit all members of the family and (b) they are membrane-impermeable. Here we report derivatives of thiamine/TPP analogues that identify elements distinctive to PDH E1 for selectivity. Based on our SAR findings, we developed a series of furan-based thiamine analogues as potent, selective and membrane-permeable inhibitors of mammalian PDH E1. We envision that our SAR findings and inhibitors will aid work on using chemical inhibition to understand the oncogenic role of PDHc.


Subject(s)
Thiamine Pyrophosphate , Thiamine , Animals , Thiamine Pyrophosphate/metabolism , Structure-Activity Relationship , Pyruvate Dehydrogenase (Lipoamide)/metabolism , Diphosphates , Pyruvates , Pyruvate Dehydrogenase Complex/metabolism , Mammals/metabolism
11.
J Cell Mol Med ; 26(19): 5078-5094, 2022 10.
Article in English | MEDLINE | ID: mdl-36071546

ABSTRACT

Abnormal nuclear structure caused by dysregulation of skeletal proteins is a common phenomenon in tumour cells. However, how skeletal proteins promote tumorigenesis remains uncovered. Here, we revealed the mechanism by which skeletal protein Emerin (EMD) promoted glucose metabolism to induce lung adenocarcinoma (LUAD). Firstly, we identified that EMD was highly expressed and promoted the malignant phenotypes in LUAD. The high expression of EMD might be due to its low level of ubiquitination. Additionally, the ISGylation at lysine 37 of EMD inhibited lysine 36 ubiquitination and upregulated EMD stability. We further explored that EMD could inhibit aerobic oxidation and stimulate glycolysis. Mechanistically, via its ß-catenin interaction domain, EMD bound with PDHA, stimulated serine 293 and 300 phosphorylation and inhibited PDHA expression, facilitated glycolysis of glucose that should enter the aerobic oxidation pathway, and EMD ISGylation was essential for EMD-PDHA interaction. In clinical LUAD specimens, EMD was negatively associated with PDHA, while positively associated with EMD ISGylation, tumour stage and diameter. In LUAD with higher glucose level, EMD expression and ISGylation were higher. Collectively, EMD was a stimulator for LUAD by inhibiting aerobic oxidation via interacting with PDHA. Restricting cancer-promoting role of EMD might be helpful for LUAD treatment.


Subject(s)
Adenocarcinoma of Lung , Lung Neoplasms , Adenocarcinoma of Lung/genetics , Glucose , Humans , Lung Neoplasms/pathology , Lysine , Membrane Proteins , Nuclear Proteins , Pyruvate Dehydrogenase (Lipoamide) , Serine , beta Catenin
12.
Zh Nevrol Psikhiatr Im S S Korsakova ; 122(9. Vyp. 2): 27-31, 2022.
Article in Russian | MEDLINE | ID: mdl-36170095

ABSTRACT

Deficiency of the pyruvate dehydrogenase complex E1-alpha subunit is a rare genetic disease with X-linked dominant inheritance. The clinical spectrum of the disease is extremely wide: from lethal forms in children of the first year of life with lactic acidosis to chronic neurological manifestations with structural changes in the central nervous system without increasing the level of lactate in the blood. The authors report a case of this disease in a preschool child and present the results of laboratory and instrumental studies. The importance of early diagnosis of the disease is emphasized.


Subject(s)
Epilepsy , Movement Disorders , Pyruvate Dehydrogenase Complex Deficiency Disease , Child, Preschool , Epilepsy/complications , Epilepsy/diagnosis , Epilepsy/genetics , Humans , Lactates , Movement Disorders/diagnosis , Movement Disorders/genetics , Mutation , Pyruvate Dehydrogenase (Lipoamide)/genetics , Pyruvate Dehydrogenase Complex Deficiency Disease/complications , Pyruvate Dehydrogenase Complex Deficiency Disease/diagnosis , Pyruvate Dehydrogenase Complex Deficiency Disease/genetics
13.
Br J Biomed Sci ; 79: 10382, 2022.
Article in English | MEDLINE | ID: mdl-35996497

ABSTRACT

Pyruvate dehydrogenase (PDH) deficiency is caused by a number of pathogenic variants and the most common are found in the PDHA1 gene. The PDHA1 gene encodes one of the subunits of the PDH enzyme found in a carbohydrate metabolism pathway involved in energy production. Pathogenic variants of PDHA1 gene usually impact the α-subunit of PDH causing energy reduction. It potentially leads to increased mortality in sufferers. Potential treatments for this disease include dichloroacetate and phenylbutyrate, previously used for other diseases such as cancer and maple syrup urine disease. However, not much is known about their efficacy in treating PDH deficiency. Effective treatment for PDH deficiency is crucial as carbohydrate is needed in a healthy diet and rice is the staple food for a large portion of the Asian population. This review analysed the efficacy of dichloroacetate and phenylbutyrate as potential treatments for PDH deficiency caused by PDHA1 pathogenic variants. Based on the findings of this review, dichloroacetate will have an effect on most PDHA1 pathogenic variant and can act as a temporary treatment to reduce the lactic acidosis, a common symptom of PDH deficiency. Phenylbutyrate can only be used on patients with certain pathogenic variants (p.P221L, p.R234G, p.G249R, p.R349C, p.R349H) on the PDH protein. It is hoped that the review would provide an insight into these treatments and improve the quality of lives for patients with PDH deficiency.


Subject(s)
Pyruvate Dehydrogenase Complex Deficiency Disease , Humans , Phenylbutyrates/therapeutic use , Pyruvate Dehydrogenase (Lipoamide)/genetics , Pyruvate Dehydrogenase (Lipoamide)/metabolism , Pyruvate Dehydrogenase Complex Deficiency Disease/diagnosis , Pyruvate Dehydrogenase Complex Deficiency Disease/drug therapy , Pyruvate Dehydrogenase Complex Deficiency Disease/genetics
14.
J Chem Inf Model ; 62(14): 3463-3475, 2022 07 25.
Article in English | MEDLINE | ID: mdl-35797142

ABSTRACT

Pyruvate dehydrogenase complex (PDC) deficiency is a major cause of primary lactic acidemia resulting in high morbidity and mortality, with limited therapeutic options. The E1 component of the mitochondrial multienzyme PDC (PDC-E1) is a symmetric dimer of heterodimers (αß/α'ß') encoded by the PDHA1 and PDHB genes, with two symmetric active sites each consisting of highly conserved phosphorylation loops A and B. PDHA1 mutations are responsible for 82-88% of cases. Greater than 85% of E1α residues with disease-causing missense mutations (DMMs) are solvent-inaccessible, with ∼30% among those involved in subunit-subunit interface contact (SSIC). We performed molecular dynamics simulations of wild-type (WT) PDC-E1 and E1 variants with E1α DMMs at R349 and W185 (residues involved in SSIC), to investigate their impact on human PDC-E1 structure. We evaluated the change in E1 structure and dynamics and examined their implications on E1 function with the specific DMMs. We found that the dynamics of phosphorylation Loop A, which is crucial for E1 biological activity, changes with DMMs that are at least about 15 Å away. Because communication is essential for PDC-E1 activity (with alternating active sites), we also investigated the possible communication network within WT PDC-E1 via centrality analysis. We observed that DMMs altered/disrupted the communication network of PDC-E1. Collectively, these results indicate allosteric effect in PDC-E1, with implications for the development of novel small-molecule therapeutics for specific recurrent E1α DMMs such as replacements of R349 responsible for ∼10% of PDC deficiency due to E1α DMMs.


Subject(s)
Pyruvate Dehydrogenase (Lipoamide) , Pyruvate Dehydrogenase Complex Deficiency Disease , Humans , Mitochondria , Mutation , Pyruvate Dehydrogenase (Lipoamide)/chemistry , Pyruvate Dehydrogenase (Lipoamide)/genetics , Pyruvate Dehydrogenase Complex/chemistry , Pyruvate Dehydrogenase Complex/genetics , Pyruvate Dehydrogenase Complex Deficiency Disease/genetics
15.
J Clin Endocrinol Metab ; 107(9): 2556-2570, 2022 08 18.
Article in English | MEDLINE | ID: mdl-35731579

ABSTRACT

CONTEXT: Nephropathy is a severe complication of type 1 diabetes (T1DM). However, the interaction between the PDHA1-regulated mechanism and CD4+ T cells in the early stage of kidney tubular injury remains unknown. OBJECTIVE: To evaluate the role of PDHA1 in the regulation of tubular cells and CD4+ T cells and further to study its interaction in tubular cell injury in T1DM. METHODS: Plasma and total RNA were collected from T cells of T1DM patients (n = 35) and healthy donors (n = 33) and evaluated for neutrophil gelatinase-associated lipocalin (NGAL), kidney injury molecule-1, PDHA1, and biomarkers of CD4+ T cells including T helper 1 cells (Th1) and regulatory T cells (Treg) markers. HK-2 cells cocultured with CD4+ T cells from T1DM patients or healthy donors (HDs) to evaluate the interaction with CD4+ T cells. RESULTS: Increased PDHA1 gene expression levels in CD4+ T cells were positively associated with the plasma level of NGAL in T1DM patients and HDs. Our data demonstrated that the Th1/Treg subsets skewed Th1 in T1DM. Knockdown of PDHA1 in kidney tubular cells decreased ATP/ROS production, NAD/NADH ratio, mitochondrial respiration, and cell apoptosis. Furthermore, PDHA1 depletion induced impaired autophagic flux. Coculture of tubular cells and T1DM T cells showed impaired CPT1A, upregulated FASN, and induced kidney injury. CONCLUSION: Our findings indicate that Th1 cells induced tubular cell injury through dysregulated metabolic reprogramming and autophagy, thereby indicating a new therapeutic approach for kidney tubular injury in T1DM.


Subject(s)
Diabetes Mellitus, Type 1 , Autophagy , Biomarkers/metabolism , Diabetes Mellitus, Type 1/metabolism , Humans , Kidney/metabolism , Kidney Tubules/metabolism , Lipocalin-2 , Pyruvate Dehydrogenase (Lipoamide) , T-Lymphocytes
16.
Sci Rep ; 12(1): 2283, 2022 02 10.
Article in English | MEDLINE | ID: mdl-35145193

ABSTRACT

Chronic thromboembolic pulmonary hypertension (CTEPH) and pulmonary arterial hypertension (PAH) are two forms of pulmonary hypertension (PH) characterized by obstructive vasculopathy. Endothelial dysfunction along with metabolic changes towards increased glycolysis are important in PAH pathophysiology. Less is known about such abnormalities in endothelial cells (ECs) from CTEPH patients. This study provides a systematic metabolic comparison of ECs derived from CTEPH and PAH patients. Metabolic gene expression was studied using qPCR in cultured CTEPH-EC and PAH-EC. Western blot analyses were done for HK2, LDHA, PDHA1, PDK and G6PD. Basal viability of CTEPH-EC and PAH-EC with the incubation with metabolic inhibitors was measured using colorimetric viability assays. Human pulmonary artery endothelial cells (HPAEC) were used as healthy controls. Whereas PAH-EC showed significant higher mRNA levels of GLUT1, HK2, LDHA, PDHA1 and GLUD1 metabolic enzymes compared to HPAEC, CTEPH-EC did not. Oxidative phosphorylation associated proteins had an increased expression in PAH-EC compared to CTEPH-EC and HPAEC. PAH-EC, CTEPH-EC and HPAEC presented similar HOXD macrovascular gene expression. Metabolic inhibitors showed a dose-dependent reduction in viability in all three groups, predominantly in PAH-EC. A different metabolic profile is present in CTEPH-EC compared to PAH-EC and suggests differences in molecular mechanisms important in the disease pathology and treatment.


Subject(s)
Endothelial Cells/metabolism , Pulmonary Arterial Hypertension/genetics , Pulmonary Arterial Hypertension/metabolism , Pulmonary Embolism/genetics , Pulmonary Embolism/metabolism , Adult , Aged , Cells, Cultured , Chronic Disease , Female , Gene Expression , Glutamate Dehydrogenase/genetics , Glutamate Dehydrogenase/metabolism , Glycolysis/genetics , Hexokinase/genetics , Hexokinase/metabolism , Humans , L-Lactate Dehydrogenase/genetics , L-Lactate Dehydrogenase/metabolism , Male , Middle Aged , Oxidative Phosphorylation , Pulmonary Artery/cytology , Pyruvate Dehydrogenase (Lipoamide)/genetics , Pyruvate Dehydrogenase (Lipoamide)/metabolism
18.
J Inherit Metab Dis ; 45(3): 557-570, 2022 05.
Article in English | MEDLINE | ID: mdl-35038180

ABSTRACT

Pyruvate dehydrogenase complex deficiency is a major cause of primary lactic acidemia resulting in high morbidity and mortality, with limited therapeutic options. PDHA1 mutations are responsible for >82% of cases. The E1 component of PDC is a symmetric dimer of heterodimers (αß/α'ß') encoded by PDHA1 and PDHB. We measured solvent accessibility surface area (SASA), utilized nearest-neighbor analysis, incorporated sequence changes using mutagenesis tool in PyMOL, and performed molecular modeling with SWISS-MODEL, to investigate the impact of residues with disease-causing missense variants (DMVs) on E1 structure and function. We reviewed 166 and 13 genetically resolved cases due to PDHA1 and PDHB, respectively, from variant databases. We expanded on 102 E1α and 13 E1ß nonduplicate DMVs. DMVs of E1α Arg112-Arg224 stretch (exons 5-7) and of E1α Arg residues constituted 40% and 39% of cases, respectively, with invariant Arg349 accounting for 22% of arginine replacements. SASA analysis showed that 86% and 84% of residues with nonduplicate DMVs of E1α and E1ß, respectively, are solvent inaccessible ("buried"). Furthermore, 30% of E1α buried residues with DMVs are deleterious through perturbation of subunit-subunit interface contact (SSIC), with 73% located in the Arg112-Arg224 stretch. E1α Arg349 represented 74% of buried E1α Arg residues involved in SSIC. Structural perturbations resulting from residue replacements in some matched neighboring pairs of amino acids on different subunits involved in SSIC at 2.9-4.0 Å interatomic distance apart, exhibit similar clinical phenotype. Collectively, this work provides insight for future target-based advanced molecular modeling studies, with implications for development of novel therapeutics for specific recurrent DMVs of E1α.


Subject(s)
Pyruvate Dehydrogenase Complex Deficiency Disease , Humans , Mutation , Mutation, Missense , Pyruvate Dehydrogenase (Lipoamide)/chemistry , Pyruvate Dehydrogenase (Lipoamide)/genetics , Pyruvate Dehydrogenase (Lipoamide)/metabolism , Pyruvate Dehydrogenase Complex/chemistry , Pyruvate Dehydrogenase Complex/genetics , Pyruvate Dehydrogenase Complex/metabolism , Pyruvate Dehydrogenase Complex Deficiency Disease/genetics , Solvents
19.
J Inherit Metab Dis ; 45(2): 248-263, 2022 03.
Article in English | MEDLINE | ID: mdl-34873726

ABSTRACT

The vast clinical and radiological spectrum of pyruvate dehydrogenase complex (PDHc) deficiency continues to pose challenges both in diagnostics and disease monitoring. Prompt diagnosis is important to enable early initiation of ketogenic diet. The patients were recruited from an ongoing population-based study in Sweden. All patients with a genetically confirmed diagnosis who had been investigated with an MRI of the brain were included. Repeated investigations were assessed to study the evolution of the MRI changes. Sixty-two MRI investigations had been performed in 34 patients (23 females). The genetic cause was mutations in PDHA1 in 29, PDHX and DLAT in 2 each, and PDHB in 1. The lesions were prenatal developmental in 16, prenatal clastic in 18, and postnatal clastic in 15 individuals. Leigh-like lesions with predominant involvement of globus pallidus were present in 12, while leukoencephalopathy was present in 6 and stroke-like lesions in 3 individuals. A combination of prenatal developmental and clastic lesions was present in 15 individuals. In addition, one male with PDHA1 also had postnatal clastic lesions. The most common lesions found in our study were agenesis or hypoplasia of corpus callosum, ventriculomegaly, or Leigh-like lesions. Furthermore, we describe a broad spectrum of other MRI changes that include leukoencephalopathy and stroke-like lesions. We argue that a novel important clue, suggesting the possibility of PDHc deficiency on MRI scans, is the simultaneous presence of multiple lesions on MRI that have occurred during different phases of brain development.


Subject(s)
Leukoencephalopathies , Pyruvate Dehydrogenase Complex Deficiency Disease , Stroke , Brain/pathology , Female , Humans , Leukoencephalopathies/diagnostic imaging , Leukoencephalopathies/pathology , Magnetic Resonance Imaging , Male , Pregnancy , Pyruvate Dehydrogenase (Lipoamide)/genetics , Pyruvate Dehydrogenase Complex/genetics , Pyruvate Dehydrogenase Complex/metabolism , Stroke/pathology
20.
Front Endocrinol (Lausanne) ; 12: 752501, 2021.
Article in English | MEDLINE | ID: mdl-34790171

ABSTRACT

Pyruvate dehydrogenase E1 (PDHE1) is thought to play essential roles in energy metabolism, and a previous study suggested that it also has potential regulatory roles in male sexual development in the oriental river prawn, Macrobrachium nipponense. In this study, we used rapid amplification of cDNA ends, quantitative polymerase chain reaction (qPCR), in situ hybridization, western blotting, RNA interference (RNAi), and histological analyses to assess the potential functions of Mn-PDHE1 in the sexual development of male M. nipponense. The full cDNA sequence of Mn-PDHE1 was 1,614 base pairs long, including a 1,077 base pair open reading frame that encodes 358 amino acids. qPCR analysis revealed the regulatory functions of PDHE1 in male sexual development in M. nipponense and in the metamorphosis process. In situ hybridization and western blot results indicated that PDHE1 was involved in testis development, and RNAi analysis showed that PDHE1 positively regulated the expression of insulin-like androgenic gland factor in M. nipponense. Compared with the cell types in the testes of control prawns, histological analysis showed that the number of sperm was dramatically lower after test subjects were injected with Mn-PDHE1 dsRNA, whereas the numbers of spermatogonia and spermatocytes were higher. Sperm constituted only 1% of cells at 14 days after injection in the RNAi group. This indicated that knockdown of the expression of PDHE1 delayed testis development. Thus, PDHE1 has positive effects on male sexual development in M. nipponense. This study highlights the functions of PDHE1 in M. nipponense and its essential roles in the regulation of testis development.


Subject(s)
Palaemonidae/metabolism , Pyruvate Dehydrogenase (Lipoamide)/genetics , Animals , DNA, Complementary/biosynthesis , DNA, Complementary/genetics , Female , Gene Expression Regulation, Developmental , Male , Metamorphosis, Biological/genetics , Open Reading Frames/genetics , Pyruvate Dehydrogenase (Lipoamide)/metabolism , RNA Interference , Sex Characteristics , Sexual Maturation , Spermatocytes/metabolism , Spermatogonia/metabolism , Testis/enzymology , Testis/growth & development
SELECTION OF CITATIONS
SEARCH DETAIL