Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 125
1.
J Proteomics ; 298: 105144, 2024 04 30.
Article En | MEDLINE | ID: mdl-38431085

Effective therapies of chronic kidney disease (CKD) are lacking due to the unclear molecular pathogenesis. Previous single omics-studies have described potential molecular regulation mechanism of CKD only at the level of transcription or translation. Therefore, this study generated an integrated transcriptomic and proteomic profile to provide deep insights into the continuous transcription-translation process during CKD. The comprehensive datasets identified 14,948 transcripts and 6423 proteins, 233 up-regulated and 364 down-regulated common differentially expressed genes of transcriptome and proteome were selected to further combined bioinformatics analysis. The obtained results revealed reactive oxygen species (ROS) metabolism and antioxidant system due to imbalance of mitochondria and peroxisomes were significantly repressed in CKD. Overall, this study presents a valuable multi-omics analysis that sheds light on the molecular mechanisms underlying CKD. SIGNIFICANCE: Chronic kidney disease (CKD) is a progressive and irreversible condition that results in abnormal kidney function and structure, and is ranked 18th among the leading causes of death globally, leading to a significant societal burden. Hence, there is an urgent need for research to detect new, sensitive, and specific biomarkers. Omics-based studies offer great potential to identify underlying disease mechanisms, aid in clinical diagnosis, and develop novel treatment strategies for CKD. Previous studies have mainly focused on the regulation of gene expression or protein synthesis in CKD, thereby compelling us to conduct a meticulous analysis of transcriptomic and proteomic data from the UUO mouse model. Here, we have performed a unified analysis of CKD model by integrating transcriptomes and protein suites for the first time. Our study contributes to a deeper understanding of the pathogenesis of CKD and provides a basis for subsequent disease management and drug development.


Renal Insufficiency, Chronic , Ureteral Obstruction , Mice , Animals , Transcriptome , Oxidative Phosphorylation , Proteomics , Peroxisomes/metabolism , Peroxisomes/pathology , Gene Expression Profiling/methods , Renal Insufficiency, Chronic/metabolism , Fibrosis , Ureteral Obstruction/genetics , Ureteral Obstruction/metabolism , Ureteral Obstruction/pathology , Kidney/metabolism
2.
Sci Transl Med ; 16(736): eadf9874, 2024 Feb 28.
Article En | MEDLINE | ID: mdl-38416843

Targeting aromatase deprives ER+ breast cancers of estrogens and is an effective therapeutic approach for these tumors. However, drug resistance is an unmet clinical need. Lipidomic analysis of long-term estrogen-deprived (LTED) ER+ breast cancer cells, a model of aromatase inhibitor resistance, revealed enhanced intracellular lipid storage. Functional metabolic analysis showed that lipid droplets together with peroxisomes, which we showed to be enriched and active in the LTED cells, controlled redox homeostasis and conferred metabolic adaptability to the resistant tumors. This reprogramming was controlled by acetyl-CoA-carboxylase-1 (ACC1), whose targeting selectively impaired LTED survival. However, the addition of branched- and very long-chain fatty acids reverted ACC1 inhibition, a process that was mediated by peroxisome function and redox homeostasis. The therapeutic relevance of these findings was validated in aromatase inhibitor-treated patient-derived samples. Last, targeting ACC1 reduced tumor growth of resistant patient-derived xenografts, thus identifying a targetable hub to combat the acquisition of estrogen independence in ER+ breast cancers.


Breast Neoplasms , Humans , Female , Breast Neoplasms/pathology , Aromatase Inhibitors/pharmacology , Aromatase Inhibitors/therapeutic use , Peroxisomes/metabolism , Peroxisomes/pathology , Acetyl-CoA Carboxylase , Lipid Droplets/metabolism , Lipid Droplets/pathology , Cell Line, Tumor , Estrogens/metabolism , Drug Resistance, Neoplasm
3.
Acta Neuropathol Commun ; 11(1): 80, 2023 05 11.
Article En | MEDLINE | ID: mdl-37170361

Peroxisomes are eukaryotic organelles that rapidly change in number depending on the metabolic requirement of distinct cell types and tissues. In the brain, these organelles are essential for neuronal migration and myelination during development and their dysfunction is associated with age-related neurodegenerative diseases. Except for one study analysing ABCD3-positive peroxisomes in neurons of the frontal neocortex of Alzheimer disease (AD) patients, no data on other brain regions or peroxisomal proteins are available. In the present morphometric study, we quantified peroxisomes labelled with PEX14, a metabolism-independent peroxisome marker, in 13 different brain areas of 8 patients each either with low, intermediate or high AD neuropathological changes compared to 10 control patients. Classification of patient samples was based on the official ABC score. During AD-stage progression, the peroxisome density decreased in the area entorhinalis, parietal/occipital neocortex and cerebellum, it increased and in later AD-stage patients decreased in the subiculum and hippocampal CA3 region, frontal neocortex and pontine gray and it remained unchanged in the gyrus dentatus, temporal neocortex, striatum and inferior olive. Moreover, we investigated the density of catalase-positive peroxisomes in a subset of patients (> 80 years), focussing on regions with significant alterations of PEX14-positive peroxisomes. In hippocampal neurons, only one third of all peroxisomes contained detectable levels of catalase exhibiting constant density at all AD stages. Whereas the density of all peroxisomes in neocortical neurons was only half of the one of the hippocampus, two thirds of them were catalase-positive exhibiting increased levels at higher ABC scores. In conclusion, we observed spatiotemporal differences in the response of peroxisomes to different stages of AD-associated pathologies.


Alzheimer Disease , Neocortex , Humans , Alzheimer Disease/pathology , Peroxisomes/metabolism , Peroxisomes/pathology , Catalase/metabolism , Pilot Projects , Neocortex/pathology
4.
J Immunol Res ; 2022: 6084589, 2022.
Article En | MEDLINE | ID: mdl-35935579

Lung adenocarcinoma (LUAD) has been the major cause of tumor-associated mortality in recent years and exhibits a poor outcome. New data revealed that peroxisomes have a function in the regulation of the development and progression of several tumors. However, the prognostic values of peroxisome-related genes (PRGs) were rarely reported. Genomic sequence, mutation, and clinical data of 535 LUAD tissues were obtained from TCGA data sets. Within the TCGA cohort, a multigene signature was constructed with the assistance of the LASSO Cox regression model. Three GEO data sets, including GSE3141, GSE31210, and GSE72094, were obtained as validation cohorts. ROC assays, Kaplan-Meier methods, and multivariate assays were applied to examine the prognostic capacities of the novel signature. Gene Set Enrichment Analysis (GSEA) was performed to further understand the underlying molecular mechanisms. In this study, we identified 47 differentially expressed peroxisome-related genes (PRGs), including 25 increased and 22 decreased PRGs. A prognostic model of six PRGs was established. The univariate and multivariate Cox analyses both showed that the p value of risk score was less than 0.05. In LUAD patients, the strong connection between the risk score and overall survival was further verified in three other GEO data sets. TMB and cancer stem cell infiltration were shown to be significantly higher in the high-risk group in comparison to the low-risk group. The TIDE score of the group with the low risk was considerably greater than that of the group with the high risk. Several drugs, targeting PRG-related genes, were available for the treatments of LUAD. Overall, we developed a novel peroxisome-related prognostic signature for LUAD patients. This signature could successfully indicate LUAD patients' chances of survival as well as their immune system's responsiveness to treatments. In addition, it has the potential to serve as immunotherapeutic targets for LUAD patients.


Adenocarcinoma of Lung , Lung Neoplasms , Adenocarcinoma of Lung/genetics , Adenocarcinoma of Lung/therapy , Humans , Kaplan-Meier Estimate , Lung Neoplasms/genetics , Lung Neoplasms/therapy , Peroxisomes/pathology , Prognosis , Tumor Microenvironment/genetics
5.
Orphanet J Rare Dis ; 17(1): 286, 2022 07 19.
Article En | MEDLINE | ID: mdl-35854306

BACKGROUND: Pathogenic variants in PEX-genes can affect peroxisome assembly and function and cause Zellweger spectrum disorders (ZSDs), characterized by variable phenotypes in terms of disease severity, age of onset and clinical presentations. So far, defects in at least 15 PEX-genes have been implicated in Mendelian diseases, but in some of the ultra-rare ZSD subtypes genotype-phenotype correlations and disease mechanisms remain elusive. METHODS: We report five families carrying biallelic variants in PEX13. The identified variants were initially evaluated by using a combination of computational approaches. Immunofluorescence and complementation studies on patient-derived fibroblasts were performed in two patients to investigate the cellular impact of the identified mutations. RESULTS: Three out of five families carried a recurrent p.Arg294Trp non-synonymous variant. Individuals affected with PEX13-related ZSD presented heterogeneous clinical features, including hypotonia, developmental regression, hearing/vision impairment, progressive spasticity and brain leukodystrophy. Computational predictions highlighted the involvement of the Arg294 residue in PEX13 homodimerization, and the analysis of blind docking predicted that the p.Arg294Trp variant alters the formation of dimers, impairing the stability of the PEX13/PEX14 translocation module. Studies on muscle tissues and patient-derived fibroblasts revealed biochemical alterations of mitochondrial function and identified mislocalized mitochondria and a reduced number of peroxisomes with abnormal PEX13 concentration. CONCLUSIONS: This study expands the phenotypic and mutational spectrum of PEX13-related ZSDs and also highlight a variety of disease mechanisms contributing to PEX13-related clinical phenotypes, including the emerging contribution of secondary mitochondrial dysfunction to the pathophysiology of ZSDs.


Zellweger Syndrome , Genetic Association Studies , Humans , Membrane Proteins/genetics , Mutation/genetics , Peroxisomes/genetics , Peroxisomes/pathology , Zellweger Syndrome/genetics , Zellweger Syndrome/pathology
6.
Annu Rev Anal Chem (Palo Alto Calif) ; 15(1): 1-16, 2022 06 13.
Article En | MEDLINE | ID: mdl-35303775

Cellular organelles are highly specialized compartments with distinct functions. With the increasing resolution of detection methods, it is becoming clearer that same organelles may have different functions or properties not only within different cell populations of a tissue but also within the same cell. Dysfunction or altered function affects the organelle itself and may also lead to malignancies or undesirable cell death. To understand cellular function or dysfunction, it is therefore necessary to analyze cellular components at the single-organelle level. Here, we review the recent advances in analyzing cellular function at single-organelle resolution using high-parameter flow cytometry or multicolor confocal microscopy. We focus on the analysis of mitochondria, as they are organelles at the crossroads of various cellular signaling pathways and functions. However, most of the applied methods/technologies are transferable to any other organelle, such as the endoplasmic reticulum, lysosomes, or peroxisomes.


Endoplasmic Reticulum , Mitochondria , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum/pathology , Lysosomes/metabolism , Lysosomes/pathology , Microscopy, Confocal , Mitochondria/metabolism , Mitochondria/pathology , Peroxisomes/metabolism , Peroxisomes/pathology
7.
Clin Res Hepatol Gastroenterol ; 46(10): 101835, 2022 12.
Article En | MEDLINE | ID: mdl-34798303

BACKGROUND AND AIM: A prominent hallmark of tumors is aberrant lipid metabolism, and various peroxisome-related genes (PRGs) are associated with aberrant tumoral metabolic signaling. However, the influence of PRGs on the prognosis of hepatocellular carcinoma (HCC) patients remains debatable. Thus, the current study was designed to evaluate the effect of PRGs on HCC and construct a prognostic model for predicting survival. METHODS: We initially acquired HCC-related gene expression profiles from the Cancer Genome Atlas and International Cancer Genome Consortium databases. We then utilized Cox analysis and Lasso regression to identify suitable PRGs for the risk model. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses were conducted to clarify the functional roles of PRGs. Single-sample gene set enrichment analysis (ssGSEA) was conducted to confirm the relationship between PRGs and immunity. RESULTS: Four PRGs were correlated with HCC patient survival: 2 risk genes (MPV17, and ABCD1) and 2 protective genes (ACSL1 and ACSL6). We derived risk scores based on PRGs to construct a predictive model that could accurately predict overall survival (OS) among HCC patients. Furthermore, GO and KEGG analyses revealed that these PRGs were potentially involved in lipid metabolism and ferroptosis in HCC. Moreover, ssGSEA results demonstrated that high PRG scores were associated with immune suppressor activation, which caused the suppression of immune effectors (CD8+ T-cells, B cells, and NK cells) and the attenuation of the immune-mediated antitumor effect. CONCLUSION: PRGs act as key regulators in tumorigenesis and tumor progression by affecting lipid synthesis and utilization, which we used to predict the outcome of HCC patients. Moreover, PRGs have been shown to promote tumoral immune resistance by serving as a vital bridge between metabolism and immunity. Thus, a personalized treatment approach targeting PRGs would clinically benefit patients by blocking the interaction between tumor metabolism and immunity.


Carcinoma, Hepatocellular , Liver Neoplasms , Humans , Carcinoma, Hepatocellular/pathology , Liver Neoplasms/pathology , Peroxisomes/metabolism , Peroxisomes/pathology , CD8-Positive T-Lymphocytes/metabolism , Gene Expression Regulation, Neoplastic , Biomarkers, Tumor/metabolism , Prognosis
8.
Biomed Pharmacother ; 143: 112223, 2021 Nov.
Article En | MEDLINE | ID: mdl-34649350

Ferroptosis is a recently recognized process of cell death characterized by accumulation of iron-dependent lipid peroxides. Herein, we demonstrate that peroxisome proliferator-activated receptor δ (PPARδ) inhibits ferroptosis of mouse embryonic fibroblasts (MEFs) derived from cysteine/glutamate transporter (xCT)-knockout mice. Activation of PPARδ by the specific ligand GW501516 led to a dose-dependent decrease in ferroptotic cell death triggered by xCT deficiency, along with decreased levels of intracellular iron accumulation and lipid peroxidation. These effects of GW501516 were abolished by PPARδ-targeting small interfering RNA (siRNA) and the PPARδ inhibitor GSK0660, indicating that PPARδ inhibits xCT deficiency-induced ferroptosis. In addition, GW501516-activated PPARδ time- and dose-dependently upregulated catalase expression at both the mRNA and protein levels. This PPARδ-mediated upregulation of catalase was markedly attenuated in cells treated with PPARδ-targeting siRNA and GSK0660, indicating that expression of catalase is dependent on PPARδ. Consistently, the effects of GW501516 on ferroptosis of xCT-deficient MEFs were counteracted in the presence of 3-amino-1,2,4-triazole, a specific inhibitor of catalase, suggesting that catalase is essential for the effect of PPARδ on ferroptosis triggered by xCT deficiency. GW501516-activated PPARδ stabilized peroxisomes through catalase upregulation by targeting peroxisomal hydrogen peroxide-mediated lysosomal rupture, which led to ferroptosis of xCT-deficient MEFs. Collectively, these results demonstrate that PPARδ modulates ferroptotic signals in xCT-deficient MEFs by regulating catalase expression.


Amino Acid Transport System y+/deficiency , Ferroptosis , Fibroblasts/metabolism , PPAR gamma/metabolism , Peroxisomes/metabolism , Amino Acid Transport System y+/genetics , Animals , Catalase/biosynthesis , Catalase/genetics , Cells, Cultured , Enzyme Induction , Ferroptosis/drug effects , Fibroblasts/drug effects , Fibroblasts/pathology , Hydrogen Peroxide/metabolism , Lipid Peroxidation , Mice, Knockout , Oxidative Stress , PPAR gamma/agonists , PPAR gamma/genetics , Peroxisomes/drug effects , Peroxisomes/genetics , Peroxisomes/pathology , Signal Transduction , Thiazoles/pharmacology
9.
Oxid Med Cell Longev ; 2021: 7726058, 2021.
Article En | MEDLINE | ID: mdl-34471469

An adverse intrauterine environment impairs the development of pancreatic islets in the fetus and leads to insufficient ß cell mass and ß cell dysfunction. We previously reported that Pex14, a peroxin protein involved in the biogenesis and degradation of peroxisomes, is markedly reduced in the pancreas of an intrauterine growth restriction fetus and last into adulthood. Peroxisomes function in a wide range of metabolic processes including fatty acid oxidization, ROS detoxification, and anti-inflammatory responses. To elucidate the impact of downregulation of the Pex14 gene on ß cell, Pex14 was knocked down by siRNA in INS-1 cells. Pex14 knockdown disturbed peroxisomal biogenesis and dysregulated fatty acid metabolism and lipid storage capability, thereby increased ROS level and blunted insulin secretion. Moreover, Pex14 knockdown upregulated inflammation factors and regulators of endoplasmic reticulum stress. The lipotoxicity of fatty acid (including palmitic acid and linoleic acid) in ß cells was exacerbated by knockdown of Pex14, as indicated by H2O2 accumulation and increased programmed cell death. The present results demonstrate the vital role of Pex14 in maintaining normal peroxisome function and ß cell viability and highlight the importance of a functional peroxisomal metabolism for the detoxification of excess FAs in ß cells.


Fetal Growth Retardation/physiopathology , Lipid Metabolism/immunology , Membrane Proteins/metabolism , Peroxisomes/pathology , Repressor Proteins/metabolism , Animals , Apoptosis , B-Lymphocytes , Humans , Rats , Transfection
10.
Mol Genet Metab ; 133(3): 307-323, 2021 07.
Article En | MEDLINE | ID: mdl-34016526

In Zellweger syndrome (ZS), lack of peroxisome function causes physiological and developmental abnormalities in many organs such as the brain, liver, muscles, and kidneys, but little is known about the exact pathogenic mechanism. By disrupting the zebrafish pex2 gene, we established a disease model for ZS and found that it exhibits pathological features and metabolic changes similar to those observed in human patients. By comprehensive analysis of the fatty acid profile, we found organ-specific accumulation and reduction of distinct fatty acid species, such as an accumulation of ultra-very-long-chain polyunsaturated fatty acids (ultra-VLC-PUFAs) in the brains of pex2 mutant fish. Transcriptome analysis using microarray also revealed mutant-specific gene expression changes that might lead to the symptoms, including reduction of crystallin, troponin, parvalbumin, and fatty acid metabolic genes. Our data indicated that the loss of peroxisomes results in widespread metabolic and gene expression changes beyond the causative peroxisomal function. These results suggest the genetic and metabolic basis of the pathology of this devastating human disease.


Fatty Acids/metabolism , Gene Expression , Peroxisomes/pathology , Zellweger Syndrome/genetics , Zellweger Syndrome/physiopathology , Animals , Disease Models, Animal , Fatty Acids/analysis , Fatty Acids/classification , Female , Gene Expression Profiling , Humans , Liver/pathology , Male , Peroxins/genetics , Zebrafish/genetics
11.
Mol Biol Cell ; 32(14): 1273-1282, 2021 07 01.
Article En | MEDLINE | ID: mdl-34010015

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is a novel coronavirus that has triggered global health and economic crises. Here we report the effects of SARS-CoV-2 infection on peroxisomes of human cell lines Huh-7 and SK-N-SH. Peroxisomes undergo dramatic changes in morphology in SARS-CoV-2-infected cells. Rearrangement of peroxisomal membranes is followed by redistribution of peroxisomal matrix proteins to the cytosol, resulting in a dramatic decrease in the number of mature peroxisomes. The SARS-CoV-2 ORF14 protein was shown to interact physically with human PEX14, a peroxisomal membrane protein required for matrix protein import and peroxisome biogenesis. Given the important roles of peroxisomes in innate immunity, SARS-CoV-2 may directly target peroxisomes, resulting in loss of peroxisome structural integrity, matrix protein content and ability to function in antiviral signaling.


Peroxisomes/virology , Animals , Cell Line , Cell Membrane/pathology , Chlorocebus aethiops , Coronavirus Nucleocapsid Proteins/metabolism , Extracellular Matrix Proteins/metabolism , Humans , Membrane Proteins/metabolism , Peroxisomes/metabolism , Peroxisomes/pathology , Phosphoproteins/metabolism , Repressor Proteins/metabolism , SARS-CoV-2/metabolism , Vero Cells
12.
Am J Med Genet A ; 185(4): 1236-1241, 2021 04.
Article En | MEDLINE | ID: mdl-33427402

Peroxisomes play an essential role in lipid metabolism via interaction with other intracellular organelles. The information about the role of the Acyl-CoA-binding domain containing-protein 5 (ACBD5) in these interactions in human cells is emerging. Moreover, a few patients with retinal dystrophy and leukodystrophy caused by pathogenic variants in ACBD5 have been recently introduced. Here, we present a 36-year-old female with retinal dystrophy, leukodystrophy, and psychomotor regression due to a novel homozygous variant in ACBD5. Our study adds to the growing knowledge of this peroxisomal disorder by providing phenotypic details of the first adult patient.


Adaptor Proteins, Signal Transducing/genetics , Genetic Predisposition to Disease , Lipid Metabolism/genetics , Membrane Proteins/genetics , Retinal Dystrophies/genetics , Adult , Female , Homozygote , Humans , Peroxisomes/genetics , Peroxisomes/pathology , Retinal Dystrophies/metabolism , Retinal Dystrophies/pathology
13.
Neurotox Res ; 39(3): 986-1006, 2021 Jun.
Article En | MEDLINE | ID: mdl-33400183

Peroxisomes are versatile cell organelles that exhibit a repertoire of organism and cell-type dependent functions. The presence of oxidases and antioxidant enzymes is a characteristic feature of these organelles. The role of peroxisomes in various cell types in human health and disease is under investigation. Defects in the biogenesis of the organelle and its function lead to severe debilitating disorders. In this manuscript, we discuss the distribution and functions of peroxisomes in the nervous system and especially in the brain cells. The important peroxisomal functions in these cells and their role in the pathology of associated disorders such as neurodegeneration are highlighted in recent studies. Although the cause of the pathogenesis of these disorders is still not clearly understood, emerging evidence supports a crucial role of peroxisomes. In this review, we discuss research highlighting the role of peroxisomes in brain development and its function. We also provide an overview of the major findings in recent years that highlight the role of peroxisome dysfunction in various associated diseases.


Brain Diseases/metabolism , Brain/metabolism , Peroxisomal Disorders/metabolism , Peroxisomes/metabolism , Animals , Brain/pathology , Brain Diseases/pathology , Humans , Peroxisomal Disorders/pathology , Peroxisomes/pathology , Reactive Oxygen Species/metabolism
14.
Biomolecules ; 10(11)2020 11 14.
Article En | MEDLINE | ID: mdl-33202661

The changing accessibility of nutrient resources induces the reprogramming of cellular metabolism in order to adapt the cell to the altered growth conditions. The nutrient-depending signaling depends on the kinases mTOR (mechanistic target of rapamycin), which is mainly activated by nitrogen-resources, and PKA (protein kinase A), which is mainly activated by glucose, as well as both of their associated factors. These systems promote protein synthesis and cell proliferation, while they inhibit degradation of cellular content by unselective bulk autophagy. Much less is known about their role in selective autophagy pathways, which have a more regulated cellular function. Especially, we were interested to analyse the central Ras2-module of the PKA-pathway in the context of peroxisome degradation. Yeast Ras2 is homologous to the mammalian Ras proteins, whose mutant forms are responsible for 33% of human cancers. In the present study, we were able to demonstrate a context-dependent role of Ras2 activity depending on the type of mTOR-inhibition and glucose-sensing situation. When mTOR was inhibited directly via the macrolide rapamycin, peroxisome degradation was still partially suppressed by Ras2, while inactivation of Ras2 resulted in an enhanced degradation of peroxisomes, suggesting a role of Ras2 in the inhibition of peroxisome degradation in glucose-grown cells. In contrast, the inhibition of mTOR by shifting cells from oleate-medium, which lacks glucose, to pexophagy-medium, which contains glucose and is limited in nitrogen, required Ras2-activity for efficient pexophagy, strongly suggesting that the role of Ras2 in glucose sensing-associated signaling is more important in this context than its co-function in mTOR-related autophagy-inhibition.


Autophagy/physiology , Peroxisomes/metabolism , Saccharomyces cerevisiae Proteins/metabolism , ras Proteins/metabolism , Glucose/metabolism , Peroxisomes/pathology , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics , ras Proteins/genetics
15.
Aging (Albany NY) ; 12(21): 21890-21903, 2020 11 06.
Article En | MEDLINE | ID: mdl-33159023

Proteomics have long been applied into characterization of molecular signatures in aging. Due to different methods and instrumentations employed for proteomic analysis, inter-dataset validation needs to be performed to identify potential biomarkers for aging. In this study, we used comparative proteomics analysis to profile age-associated changes in proteome and glutathionylome in mouse kidneys. We identified 108 proteins that were differentially expressed in young and aged mouse kidneys in three different datasets; from these, 27 proteins were identified as potential renal aging biomarkers, including phosphoenolpyruvate carboxykinase (Pck1), CD5 antigen-like protein (Cd5l), aldehyde dehydrogenase 1 (Aldh1a1), and uromodulin. Our results also showed that peroxisomal proteins were significantly downregulated in aged mice, whereas IgGs were upregulated, suggesting that peroxisome deterioration might be a hallmark for renal aging. Glutathionylome analysis demonstrated that downregulation of catalase and glutaredoxin-1 (Glrx1) significantly increased protein glutathionylation in aged mice. In addition, nicotinamide mononucleotide (NMN) administration significantly increased the number of peroxisomes in aged mouse kidneys, indicating that NMN enhanced peroxisome biogenesis, and suggesting that it might be beneficial to reduce kidney injuries. Together, our data identify novel potential biomarkers for renal aging, and provide a valuable resource for understanding the age-associated changes in kidneys.


Aging/metabolism , Kidney/metabolism , Peroxisomes/metabolism , Proteome , Proteomics , Age Factors , Aging/pathology , Animals , Biomarkers/metabolism , Chromatography, Reverse-Phase , Databases, Protein , Kidney/drug effects , Kidney/pathology , Mice , Mice, Inbred C57BL , Nicotinamide Mononucleotide/pharmacology , Peroxisomes/drug effects , Peroxisomes/pathology , Proteostasis , Tandem Mass Spectrometry
16.
BMC Med Genet ; 21(1): 229, 2020 11 19.
Article En | MEDLINE | ID: mdl-33213396

BACKGROUND: Peroxisome biogenesis disorders (PBDs) are a group of metabolic diseases caused by dysfunction of peroxisomes. Different forms of PBDs are described; the most severe one is the Zellweger syndrome (ZS). We report on an unusual presentation of Zellweger syndrome manifesting in a newborn with severe and fulminant sepsis, causing death during the neonatal period. CASE PRESENTATION: A term male Caucasian neonate presented at birth with hypotonia and poor feeding associated with dysmorphic craniofacial features and skeletal abnormalities. Blood tests showed progressive leukopenia; ultrasounds revealed cerebral and renal abnormalities. He died on the fourth day of life because of an irreversible Gram-negative sepsis. Post-mortem tests on blood and urine samples showed biochemical alterations suggestive of ZS confirmed by genetic test. CONCLUSIONS: ZS is an early and severe forms of PBDs. Peroxisomes are known to be involved in lipid metabolism, but recent studies suggest their fundamental role in modulating immune response and inflammation. In case of clinical suspicion of ZS it is important to focus the attention on the prevention and management of infections that can rapidly progress to death.


ATPases Associated with Diverse Cellular Activities/genetics , Gram-Negative Bacterial Infections/genetics , Mutation , Peroxisomes/immunology , Sepsis/genetics , Zellweger Syndrome/genetics , ATPases Associated with Diverse Cellular Activities/deficiency , ATPases Associated with Diverse Cellular Activities/immunology , Fatal Outcome , Gene Expression , Gram-Negative Bacterial Infections/immunology , Gram-Negative Bacterial Infections/microbiology , Gram-Negative Bacterial Infections/pathology , Humans , Immunity, Innate , Infant, Newborn , Male , Peroxisomes/microbiology , Peroxisomes/pathology , Sepsis/immunology , Sepsis/microbiology , Sepsis/pathology , Zellweger Syndrome/immunology , Zellweger Syndrome/microbiology , Zellweger Syndrome/pathology
17.
Biochim Biophys Acta Mol Basis Dis ; 1866(10): 165882, 2020 10 01.
Article En | MEDLINE | ID: mdl-32565019

Peroxisomes are organelles, abundant in the liver, involved in a variety of cellular functions, including fatty acid metabolism, plasmalogen synthesis and metabolism of reactive oxygen species. Several inherited disorders are associated with peroxisomal dysfunction; increasingly many are associated with hepatic pathologies. The liver plays a principal role in regulation of iron metabolism. In this study we examined the possibility of a relationship between iron homeostasis and peroxisomal integrity. We examined the effect of deleting Pex13 in mouse liver on systemic iron homeostasis. We also used siRNA-mediated knock-down of PEX13 in a human hepatoma cell line (HepG2/C3A) to elucidate the mechanisms of PEX13-mediated regulation of hepcidin. We demonstrate that transgenic mice lacking hepatocyte Pex13 have defects in systemic iron homeostasis. The ablation of Pex13 expression in hepatocytes leads to a significant reduction in hepatic hepcidin levels. Our results also demonstrate that a deficiency of PEX13 gene expression in HepG2/C3A cells leads to decreased hepcidin expression, which is mediated through an increase in the signalling protein SMAD7, and endoplasmic reticulum (ER) stress. This study identifies a novel role for a protein involved in maintaining peroxisomal integrity and function in iron homeostasis. Loss of Pex13, a protein important for peroxisomal function, in hepatocytes leads to a significant increase in ER stress, which if unresolved, can affect liver function. The results from this study have implications for the management of patients with peroxisomal disorders and the liver-related complications they may develop.


Hepatocytes/metabolism , Iron/metabolism , Membrane Proteins/deficiency , Peroxisomes/pathology , Animals , Bone Morphogenetic Proteins/metabolism , Cell Membrane/pathology , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum/pathology , Endoplasmic Reticulum Stress , Female , Gene Knockdown Techniques , Hep G2 Cells , Hepcidins/metabolism , Humans , Iron/blood , Liver/cytology , Liver/metabolism , Liver/pathology , Membrane Proteins/genetics , Mice , Mice, Knockout , Models, Animal , Peroxisomal Disorders/pathology , Peroxisomes/metabolism , RNA, Small Interfering/metabolism , Smad7 Protein/metabolism
18.
J Biol Chem ; 295(30): 10168-10179, 2020 07 24.
Article En | MEDLINE | ID: mdl-32493774

Feeding of rapeseed (canola) oil with a high erucic acid concentration is known to cause hepatic steatosis in animals. Mitochondrial fatty acid oxidation plays a central role in liver lipid homeostasis, so it is possible that hepatic metabolism of erucic acid might decrease mitochondrial fatty acid oxidation. However, the precise mechanistic relationship between erucic acid levels and mitochondrial fatty acid oxidation is unclear. Using male Sprague-Dawley rats, along with biochemical and molecular biology approaches, we report here that peroxisomal ß-oxidation of erucic acid stimulates malonyl-CoA formation in the liver and thereby suppresses mitochondrial fatty acid oxidation. Excessive hepatic uptake and peroxisomal ß-oxidation of erucic acid resulted in appreciable peroxisomal release of free acetate, which was then used in the synthesis of cytosolic acetyl-CoA. Peroxisomal metabolism of erucic acid also remarkably increased the cytosolic NADH/NAD+ ratio, suppressed sirtuin 1 (SIRT1) activity, and thereby activated acetyl-CoA carboxylase, which stimulated malonyl-CoA biosynthesis from acetyl-CoA. Chronic feeding of a diet including high-erucic-acid rapeseed oil diminished mitochondrial fatty acid oxidation and caused hepatic steatosis and insulin resistance in the rats. Of note, administration of a specific peroxisomal ß-oxidation inhibitor attenuated these effects. Our findings establish a cross-talk between peroxisomal and mitochondrial fatty acid oxidation. They suggest that peroxisomal oxidation of long-chain fatty acids suppresses mitochondrial fatty acid oxidation by stimulating malonyl-CoA formation, which might play a role in fatty acid-induced hepatic steatosis and related metabolic disorders.


Erucic Acids/metabolism , Fatty Liver/metabolism , Liver/metabolism , Malonyl Coenzyme A/biosynthesis , Mitochondria, Liver/metabolism , Peroxisomes/metabolism , Animals , Fatty Liver/pathology , Insulin Resistance , Liver/pathology , Male , Mitochondria, Liver/pathology , Oxidation-Reduction , Peroxisomes/pathology , Rats , Rats, Sprague-Dawley
19.
Hum Genet ; 139(10): 1325-1343, 2020 Oct.
Article En | MEDLINE | ID: mdl-32399598

Perrault syndrome is a rare heterogeneous condition characterised by sensorineural hearing loss and premature ovarian insufficiency. Additional neuromuscular pathology is observed in some patients. There are six genes in which variants are known to cause Perrault syndrome; however, these explain only a minority of cases. We investigated the genetic cause of Perrault syndrome in seven affected individuals from five different families, successfully identifying the cause in four patients. This included previously reported and novel causative variants in known Perrault syndrome genes, CLPP and LARS2, involved in mitochondrial proteolysis and mitochondrial translation, respectively. For the first time, we show that pathogenic variants in PEX6 can present clinically as Perrault syndrome. PEX6 encodes a peroxisomal biogenesis factor, and we demonstrate evidence of peroxisomal dysfunction in patient serum. This study consolidates the clinical overlap between Perrault syndrome and peroxisomal disorders, and highlights the need to consider ovarian function in individuals with atypical/mild peroxisomal disorders. The remaining patients had variants in candidate genes such as TFAM, involved in mtDNA transcription, replication, and packaging, and GGPS1 involved in mevalonate/coenzyme Q10 biosynthesis and whose enzymatic product is required for mouse folliculogenesis. This genomic study highlights the diverse molecular landscape of this poorly understood syndrome.


ATPases Associated with Diverse Cellular Activities/genetics , Amino Acyl-tRNA Synthetases/genetics , DNA-Binding Proteins/genetics , Dimethylallyltranstransferase/genetics , Endopeptidase Clp/genetics , Farnesyltranstransferase/genetics , Genetic Predisposition to Disease , Geranyltranstransferase/genetics , Gonadal Dysgenesis, 46,XX/genetics , Hearing Loss, Sensorineural/genetics , Mitochondrial Proteins/genetics , Transcription Factors/genetics , Adolescent , Adult , Base Sequence , Child , DNA, Mitochondrial/genetics , Female , Gene Expression , Gonadal Dysgenesis, 46,XX/diagnosis , Gonadal Dysgenesis, 46,XX/pathology , Hearing Loss, Sensorineural/diagnosis , Hearing Loss, Sensorineural/pathology , High-Throughput Nucleotide Sequencing , Humans , Male , Ovary/metabolism , Ovary/pathology , Pedigree , Peroxisomes/metabolism , Peroxisomes/pathology
20.
Mol Vis ; 26: 216-225, 2020.
Article En | MEDLINE | ID: mdl-32214787

Purpose: The aim of the present work is the molecular diagnosis of three patients with deafness and retinal degeneration. Methods: Three patients from two unrelated families were initially analyzed with custom gene panels for Usher genes, non-syndromic hearing loss, or inherited syndromic retinopathies and further investigated by means of clinical or whole exome sequencing. Results: The study allowed us to detect likely pathogenic variants in PEX6, a gene typically involved in peroxisomal biogenesis disorders (PBDs). Beside deaf-blindness, both families showed additional features: Siblings from Family 1 showed enamel alteration and abnormal peroxisome. In addition, the brother had mild neurodevelopmental delay and nephrolithiasis. The case II:1 from Family 2 showed intellectual disability, enamel alteration, and dysmorphism. Conclusions: We have reported three new cases with pathogenic variants in PEX6 presenting with milder forms of the Zellweger spectrum disorders (ZSD). The three cases showed distinct clinical features. Thus, expanding the phenotypic spectrum of PBDs and ascertaining exome sequencing is an effective strategy for an accurate diagnosis of clinically overlapping and genetically heterogeneous disorders such as deafness-blindness association.


ATPases Associated with Diverse Cellular Activities/genetics , Hearing Loss, Sensorineural/genetics , Retinitis Pigmentosa/genetics , Zellweger Syndrome/genetics , Adult , Child , Craniofacial Abnormalities/genetics , Dental Enamel/abnormalities , Female , Humans , Intellectual Disability/genetics , Male , Mutation , Nephrolithiasis/genetics , Neurodevelopmental Disorders/genetics , Pedigree , Peroxisomes/genetics , Peroxisomes/metabolism , Peroxisomes/pathology , Exome Sequencing
...