1
|
Glatt H, Meinl W. Sulphotransferase-mediated toxification of chemicals in mouse models: effect of knockout or humanisation of SULT genes. Essays Biochem 2024; 68:523-539. [PMID: 39611595 PMCID: PMC11625864 DOI: 10.1042/ebc20240030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 11/11/2024] [Accepted: 11/18/2024] [Indexed: 11/30/2024]
Abstract
Cytosolic sulphotransferase (SULT) enzymes catalyse reactions involved in xenobiotic elimination and hormone regulation. However, SULTs can also generate electrophilic reactive intermediates from certain substrates, including the activation of carcinogens. Here, we review toxicological studies of mouse strains with SULT status altered by genetic modification. Knockout mouse strains have been constructed for the enzymes Sult1a1, 1d1, 1e1, 2b1 and 4a1. In addition, transgenic strains are available for human SULT1A1/2. Among SULT knockout mouse strains, reduced fertility (Sult1e1) and early postnatal death (Sult4a1) were observed. In contrast, Sult1a1 or Sult1d1 knockouts and SULT1A1/2 transgenics were healthy and showed no obvious deficiencies. These strains were used in toxicological studies with 13 chemicals. Manipulation of the SULT system altered dramatically the adverse effects of many compounds; thus, very large differences in levels of DNA adducts formed in the liver or other tissues were seen with some chemicals - up to 99.2% decreases in knockouts and 83-fold increases in SULT1A1/2 transgenics. In many cases, these changes were restricted to the tissues in which the corresponding enzymes are expressed, arguing for local activation. However, with some compounds, the kidney was an important target tissue, due to the active transfer to that organ, via the circulation, of reactive sulphuric acid esters.
Collapse
Affiliation(s)
- Hansruedi Glatt
- Federal Institute for Risk Assessment (BfR), Department Food Safety, Max-Dohrn-Strasse 8-10, 10589 Berlin, Germany
- German Institute of Human Nutrition (DIfE) Potsdam-Rehbrücke, Department of Nutritional Toxicology (HG & WM) and Department of Molecular Toxicology (WM), Arthur-Scheunert-Allee 114-116, 14558 Nuthetal, Germany
| | - Walter Meinl
- German Institute of Human Nutrition (DIfE) Potsdam-Rehbrücke, Department of Nutritional Toxicology (HG & WM) and Department of Molecular Toxicology (WM), Arthur-Scheunert-Allee 114-116, 14558 Nuthetal, Germany
| |
Collapse
|
2
|
Kolb A, Corridon P, Ullah M, Pfaffenberger ZJ, Xu WM, Winfree S, Sandoval RH, Hato T, Witzmann FA, Mohallem R, Franco J, Aryal UK, Atkinson SJ, Basile DP, Bacallao RL. Sulfotransferase 1C2 Increases Mitochondrial Respiration by Converting Mitochondrial Membrane Cholesterol to Cholesterol Sulfate. Biochemistry 2024; 63:2310-2322. [PMID: 39194960 PMCID: PMC11411706 DOI: 10.1021/acs.biochem.3c00344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Revised: 08/16/2024] [Accepted: 08/21/2024] [Indexed: 08/29/2024]
Abstract
HYPOTHESIS In this communication, we test the hypothesis that sulfotransferase 1C2 (SULT1C2, UniProt accession no. Q9WUW8) can modulate mitochondrial respiration by increasing state-III respiration. METHODS AND RESULTS Using freshly isolated mitochondria, the addition of SULT1C2 and 3-phosphoadenosine 5 phosphosulfate (PAPS) results in an increased maximal respiratory capacity in response to the addition of succinate, ADP, and rotenone. Lipidomics and thin-layer chromatography of mitochondria treated with SULT1C2 and PAPS showed an increase in the level of cholesterol sulfate. Notably, adding cholesterol sulfate at nanomolar concentration to freshly isolated mitochondria also increases maximal respiratory capacity. In vivo studies utilizing gene delivery of SULT1C2 expression plasmids to kidneys result in increased mitochondrial membrane potential and confer resistance to ischemia/reperfusion injury. Mitochondria isolated from gene-transduced kidneys have elevated state-III respiration as compared with controls, thereby recapitulating results obtained with mitochondrial fractions treated with SULT1C2 and PAPS. CONCLUSION SULT1C2 increases mitochondrial respiratory capacity by modifying cholesterol, resulting in increased membrane potential and maximal respiratory capacity. This finding uncovers a unique role of SULT1C2 in cellular physiology and extends the role of sulfotransferases in modulating cellular metabolism.
Collapse
Affiliation(s)
- Alexander
J. Kolb
- Department
of Biology, School of Science, Indiana University-Indianapolis, Indianapolis, Indiana 46202, United States
| | - Peter Corridon
- Khalifa
University of Science and Technology, P.O. Box 127788, Abu Dhabi, United Arab Emirates
| | - Mahbub Ullah
- Department
of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, Indiana 46202, United States
| | | | - Wei Min Xu
- Division
of Nephrology, Indiana Center for Biological Microscopy, Indiana University School of Medicine, Indianapolis, Indiana 46202, United States
| | - Seth Winfree
- Department
of Pathology and Microbiology, University
of Nebraska Medical Center, Omaha, Nebraska 68198, United States
| | - Ruben H. Sandoval
- Division
of Nephrology, Indiana Center for Biological Microscopy, Indiana University School of Medicine, Indianapolis, Indiana 46202, United States
| | - Takeshi Hato
- Division
of Nephrology, Indiana Center for Biological Microscopy, Indiana University School of Medicine, Indianapolis, Indiana 46202, United States
| | - Frank A. Witzmann
- Department
of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, Indiana 46202, United States
| | - Rodrigo Mohallem
- Department
of Comparative Pathobiology, College of Veterinary Medicine, Bindley
Bioscience Center, Purdue University, West Lafayette, Indiana 47907, United States
| | | | - Uma K. Aryal
- Department
of Comparative Pathobiology, College of Veterinary Medicine, Bindley
Bioscience Center, Purdue University, West Lafayette, Indiana 47907, United States
- Purdue
Proteomics Facility, Bindley Biosciences Center, Purdue University, West Lafayette, Indiana 47907, United States
| | - Simon J. Atkinson
- Department
of Neuroscience, Physiology and Behavior, University of California, Davis, California 95616, United States
| | - David P. Basile
- Department
of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, Indiana 46202, United States
| | - Robert L. Bacallao
- Division
of Nephrology, Indiana Center for Biological Microscopy, Indiana University School of Medicine, Indianapolis, Indiana 46202, United States
- Division
of Nephrology, Richard Roudebush
VA Medical Center, Indianapolis, Indiana 46202, United States
| |
Collapse
|
3
|
Xu H, Guo Y, Liu XJ, Liu Y, Yin S, Bao QY, Peng R, Tian WB, Xia YY, Gao L, Liu JM. Idebenone Antagonizes P53-Mediated Neuronal Oxidative Stress Injury by Regulating CD38-SIRT3 Protein Level. Neurochem Res 2024; 49:2491-2504. [PMID: 38862726 PMCID: PMC11310240 DOI: 10.1007/s11064-024-04189-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 05/04/2024] [Accepted: 06/03/2024] [Indexed: 06/13/2024]
Abstract
Idebenone, an antioxidant used in treating oxidative damage-related diseases, has unclear neuroprotective mechanisms. Oxidative stress affects cell and mitochondrial membranes, altering Adp-ribosyl cyclase (CD38) and Silent message regulator 3 (SIRT3) protein expression and possibly impacting SIRT3's ability to deacetylate Tumor protein p53 (P53). This study explores the relationship between CD38, SIRT3, and P53 in H2O2-injured HT22 cells treated with Idebenone. Apoptosis was detected using flow cytometry and terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling (TUNEL) staining after determining appropriate H2O2 and Idebenone concentrations.In this study, Idebenone was found to reduce apoptosis and decrease P53 and Caspase3 expression in H2O2-injured HT22 cells by detecting apoptosis-related protein expression. Through bioinformatics methods, CD38 was identified as the target of Idebenone, and it further demonstrated that Idebenone decreased the expression of CD38 and increased the level of SIRT3. An increased NAD+/NADH ratio was detected, suggesting Idebenone induces SIRT3 expression and protects HT22 cells by decreasing apoptosis-related proteins. Knocking down SIRT3 downregulated acetylated P53 (P53Ac), indicating SIRT3's importance in P53 deacetylation.These results supported that CD38 was used as a target of Idebenone to up-regulate SIRT3 to deacetylate activated P53, thereby protecting HT22 cells from oxidative stress injury. Thus, Idebenone is a drug that may show great potential in protecting against reactive oxygen species (ROS) induced diseases such as Parkinson's disease, and Alzheimer's disease. And it might be able to compensate for some of the defects associated with CD38-related diseases.
Collapse
Affiliation(s)
- Hao Xu
- Department of Histology and Embryology, College of Basic Medical Sciences, Jilin University, Changchun, P.R. China
| | - Ying Guo
- Department of Histology and Embryology, College of Basic Medical Sciences, Jilin University, Changchun, P.R. China
| | - Xiao-Jun Liu
- China-Japan Union Hospital of Jilin University, Changchun, 130033, Jilin, China
| | - Ying Liu
- Department of Histology and Embryology, College of Basic Medical Sciences, Jilin University, Changchun, P.R. China
| | - Shi Yin
- Department of Histology and Embryology, College of Basic Medical Sciences, Jilin University, Changchun, P.R. China
| | - Qi-Ying Bao
- Department of Histology and Embryology, College of Basic Medical Sciences, Jilin University, Changchun, P.R. China
| | - Ru Peng
- Jiangsu Health Vocational College, Nanjing, P.R. China
| | | | - Ying-Yan Xia
- Bethune Second Clinical School of Medicine, Jilin University, Changchun, P.R. China
| | - Ling Gao
- Basic medical department of Changchun Medical College, Changchun, P.R. China.
| | - Jia-Mei Liu
- Department of Histology and Embryology, College of Basic Medical Sciences, Jilin University, Changchun, P.R. China.
| |
Collapse
|
4
|
van Waardenburg RCAM, Falany CN. Sulfotransferase 4A1 Coding Sequence and Protein Structure Are Highly Conserved in Vertebrates. Genes (Basel) 2024; 15:914. [PMID: 39062693 PMCID: PMC11275347 DOI: 10.3390/genes15070914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 07/08/2024] [Accepted: 07/10/2024] [Indexed: 07/28/2024] Open
Abstract
Cytosolic sulfotransferases (SULTs) are Phase 2 drug-metabolizing enzymes that catalyze the conjugation of sulfonate to endogenous and xenobiotic compounds, increasing their hydrophilicity and excretion from cells. To date, 13 human SULTs have been identified and classified into five families. SULT4A1 mRNA encodes two variants: (1) the wild type, encoding a 284 amino acid, ~33 kDa protein, and (2) an alternative spliced variant resulting from a 126 bp insert between exon 6 and 7, which introduces a premature stop codon that enhances nonsense-mediated decay. SULT4A1 is classified as an SULT based on sequence and structural similarities, including PAPS-domains, active-site His, and the dimerization domain; however, the catalytic pocket lid 'Loop 3' size is not conserved. SULT4A1 is uniquely expressed in the brain and localized in the cytosol and mitochondria. SULT4A1 is highly conserved, with rare intronic polymorphisms that have no outward manifestations. However, the SULT4A1 haplotype is correlated with Phelan-McDermid syndrome and schizophrenia. SULT4A1 knockdown revealed potential SULT4A1 functions in photoreceptor signaling and knockout mice display hampered neuronal development and behavior. Mouse and yeast models revealed that SULT4A1 protects the mitochondria from endogenously and exogenously induced oxidative stress and stimulates cell division, promoting dendritic spines' formation and synaptic transmission. To date, no physiological enzymatic activity has been associated with SULT4A1.
Collapse
|
5
|
Li Y, Minic Z, Hüttmann N, Khraibah A, Storey KB, Berezovski MV. Proteomic analysis of Rana sylvatica reveals differentially expressed proteins in liver in response to anoxia, dehydration or freezing stress. Sci Rep 2024; 14:15388. [PMID: 38965296 PMCID: PMC11224343 DOI: 10.1038/s41598-024-65417-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Accepted: 06/20/2024] [Indexed: 07/06/2024] Open
Abstract
Ectothermic animals that live in seasonally cold regions must adapt to seasonal variation and specific environmental conditions. During the winter, some amphibians hibernate on land and encounter limited environmental water, deficient oxygen, and extremely low temperatures that can cause the whole body freezing. These stresses trigger physiological and biochemical adaptations in amphibians that allow them to survive. Rana sylvatica, commonly known as the wood frog, shows excellent freeze tolerance. They can slow their metabolic activity to a near halt and endure freezing of 65-70% of their total body water as extracellular ice during hibernation, returning to normal when the temperatures rise again. To investigate the molecular adaptations of freeze-tolerant wood frogs, a comprehensive proteomic analysis was performed on frog liver tissue after anoxia, dehydration, or freezing exposures using a label-free LC-MS/MS proteomic approach. Quantitative proteomic analysis revealed that 87, 118, and 86 proteins were significantly upregulated in dehydrated, anoxic, and frozen groups, suggesting potential protective functions. The presence of three upregulated enzymes, glutathione S-transferase (GST), aldolase (ALDOA), and sorbitol dehydrogenase (SORD), was also validated. For all enzymes, the specific enzymatic activity was significantly higher in the livers of frozen and anoxic groups than in the controls. This study reveals that GST, ALDOA, and SORD might participate in the freeze tolerance mechanism by contributing to regulating cellular detoxification and energy metabolism.
Collapse
Affiliation(s)
- Yingxi Li
- Department of Chemistry and Biomolecular Sciences, University of Ottawa, Ottawa, ON, K1N 6N5, Canada
- John L. Holmes Mass Spectrometry Facility, Faculty of Science, University of Ottawa, Ottawa, ON, K1N 6N5, Canada
| | - Zoran Minic
- John L. Holmes Mass Spectrometry Facility, Faculty of Science, University of Ottawa, Ottawa, ON, K1N 6N5, Canada
| | - Nico Hüttmann
- John L. Holmes Mass Spectrometry Facility, Faculty of Science, University of Ottawa, Ottawa, ON, K1N 6N5, Canada
| | - Abdullah Khraibah
- Department of Chemistry and Biomolecular Sciences, University of Ottawa, Ottawa, ON, K1N 6N5, Canada
| | - Kenneth B Storey
- Department of Biology, Carleton University, 1125 Colonel By Drive, Ottawa, ON, K1S 5B6, Canada
| | - Maxim V Berezovski
- Department of Chemistry and Biomolecular Sciences, University of Ottawa, Ottawa, ON, K1N 6N5, Canada.
- John L. Holmes Mass Spectrometry Facility, Faculty of Science, University of Ottawa, Ottawa, ON, K1N 6N5, Canada.
| |
Collapse
|
6
|
López-Martínez M, Armengol MP, Pey I, Farré X, Rodríguez-Martínez P, Ferrer M, Porrini E, Luis-Lima S, Díaz-Martín L, Rodríguez-Rodríguez AE, Cruz-Perera C, Alcalde M, Navarro-Díaz M. Integrated miRNA-mRNA Analysis Reveals Critical miRNAs and Targets in Diet-Induced Obesity-Related Glomerulopathy. Int J Mol Sci 2024; 25:6437. [PMID: 38928144 PMCID: PMC11204096 DOI: 10.3390/ijms25126437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 06/02/2024] [Accepted: 06/08/2024] [Indexed: 06/28/2024] Open
Abstract
This study aimed to investigate obesity-related glomerulopathy (ORG) at cellular, structural, and transcriptomic levels. Thirty Wistar rats were randomized into two groups: 15 rats were fed with a standard diet (SD-rats), and 15 rats were fed with a high-fat diet (HFD-rats). After 10 weeks, the weight, kidney function, histological features, and transcriptomic changes were assessed. HFD-rats gained significantly more weight (55.8% vs. 29.2%; p < 0.001) and albuminuria (10,384.04 ng/mL vs. 5845.45 ng/mL; p < 0.001) compared to SD-rats. HFD-rats exhibited early stages of ORG, with predominant mesangial matrix increase and podocyte hypertrophy (PH). These lesions correlated with differentially expressed (DE) genes and miRNAs. Functional analysis showed that miR-205, which was DE in both the kidneys and urine of HFD-rats, negatively regulated the PTEN gene, promoting lipid endocytosis in podocytes. The downregulation of PTEN was proved through a higher PTEN/nephrin ratio in the SD-rats and the presence of lipid vacuoles in HFD-podocytes. This study has found a specific targetome of miRNAs and gene expression in early stages of ORG. Also, it emphasizes the potential value of miR-205 as a urinary biomarker for detecting podocyte injury in ORG, offering a tool for early diagnosis, and opening new avenues for future therapeutic research of obesity-related glomerulopathy.
Collapse
Affiliation(s)
- Marina López-Martínez
- CSUR National Unit of Expertise for Complex Glomerular Diseases of Spain, Nephrology Department, Vall d’Hebron University Hospital, Vall d’Hebron Institute of Research, 08035 Barcelona, Spain
- Department of Medicine, Universitat Autònoma de Barcelona, Bellaterra, 08913 Barcelona, Spain
| | - Maria Pilar Armengol
- Genomic Platform, Germans Trias i Pujol’s Research Institute, Badalona, 08916 Barcelona, Spain
| | - Irina Pey
- Genomic Platform, Germans Trias i Pujol’s Research Institute, Badalona, 08916 Barcelona, Spain
| | - Xavier Farré
- Genomic Platform, Germans Trias i Pujol’s Research Institute, Badalona, 08916 Barcelona, Spain
| | | | - Mireia Ferrer
- Statistics and Bioinformatics Unit, Vall d’Hebron Research Institute, 08035 Barcelona, Spain
| | - Esteban Porrini
- Laboratory of Renal Function (LFR), Faculty of Medicine, University of La Laguna, Complejo Hospitalario Universitario de Canarias, 38320 La Laguna, Spain (L.D.-M.)
- Instituto de Tecnologías Biomédicas (ITB), Faculty of Medicine, University of La Laguna, La Laguna, 38320 Tenerife, Spain
| | - Sergio Luis-Lima
- Laboratory of Renal Function (LFR), Faculty of Medicine, University of La Laguna, Complejo Hospitalario Universitario de Canarias, 38320 La Laguna, Spain (L.D.-M.)
- Department of Laboratory Medicine, Complejo Hospitalario Universitario de Canarias, La Laguna, 38320 Tenerife, Spain
| | - Laura Díaz-Martín
- Laboratory of Renal Function (LFR), Faculty of Medicine, University of La Laguna, Complejo Hospitalario Universitario de Canarias, 38320 La Laguna, Spain (L.D.-M.)
| | - Ana Elena Rodríguez-Rodríguez
- Research Unit, Hospital Universitario de Canarias, La Laguna, 38320 Tenerife, Spain
- Fundación General de la Universidad, University of La Laguna,38320 Tenerife, Spain
| | - Coriolano Cruz-Perera
- Laboratory of Renal Function (LFR), Faculty of Medicine, University of La Laguna, Complejo Hospitalario Universitario de Canarias, 38320 La Laguna, Spain (L.D.-M.)
| | - Marta Alcalde
- Comparative Medicine and Bioimage Centre of Catalonia (CMCiB), Fundació Institut d’Investigació en Ciències de la Salut Germans Trias i Pujol, Badalona, 08916 Barcelona, Spain
- Pharmaco and Device Epidemiology Group, CSM, NDORMS, University of Oxford, Oxford OX1 3PT, UK
| | - Maruja Navarro-Díaz
- Genomic Platform, Germans Trias i Pujol’s Research Institute, Badalona, 08916 Barcelona, Spain
- Nephrology Department, Sant Joan Despí Moisès Broggi Hospital, Sant Joan Despí, 08970 Barcelona, Spain
| |
Collapse
|
7
|
Mitz AR, Boccuto L, Thurm A. Evidence for common mechanisms of pathology between SHANK3 and other genes of Phelan-McDermid syndrome. Clin Genet 2024; 105:459-469. [PMID: 38414139 PMCID: PMC11025605 DOI: 10.1111/cge.14503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 01/18/2024] [Accepted: 02/02/2024] [Indexed: 02/29/2024]
Abstract
Chromosome 22q13.3 deletion (Phelan-McDermid) syndrome (PMS, OMIM 606232) is a rare genetic condition that impacts neurodevelopment. PMS most commonly results from heterozygous contiguous gene deletions that include the SHANK3 gene or likely pathogenic variants of SHANK3 (PMS-SHANK3 related). Rarely, chromosomal rearrangements that spare SHANK3 share the same general phenotype (PMS-SHANK3 unrelated). Very recent human and model system studies of genes that likely contribute to the PMS phenotype point to overlap in gene functions associated with neurodevelopment, synaptic formation, stress/inflammation and regulation of gene expression. In this review of recent findings, we describe the functional overlaps between SHANK3 and six partner genes of 22q13.3 (PLXNB2, BRD1, CELSR1, PHF21B, SULT4A1, and TCF20), which suggest a model that explains the commonality between PMS-SHANK3 related and PMS-SHANK3 unrelated classes of PMS. These genes are likely not the only contributors to neurodevelopmental impairments in the region, but they are the best documented to date. The review provides evidence for the overlapping and likely synergistic contributions of these genes to the PMS phenotype.
Collapse
Affiliation(s)
- Andrew R. Mitz
- Laboratory of Neuropsychology, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| | - Luigi Boccuto
- Healthcare Genetics and Genomics Interdisciplinary Doctoral Program, School of Nursing, College of Behavioral, Social and Health Sciences, Clemson University, Clemson, SC, USA
| | - Audrey Thurm
- Neurodevelopmental and Behavioral Phenotyping Service, Office of the Clinical Director, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
8
|
Salman M, Stayton AS, Parveen K, Parveen A, Puchowicz MA, Parvez S, Bajwa A, Ishrat T. Intranasal Delivery of Mitochondria Attenuates Brain Injury by AMPK and SIRT1/PGC-1α Pathways in a Murine Model of Photothrombotic Stroke. Mol Neurobiol 2024; 61:2822-2838. [PMID: 37946007 DOI: 10.1007/s12035-023-03739-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Accepted: 10/18/2023] [Indexed: 11/12/2023]
Abstract
Ischemic stroke is one of the major causes of morbidity and mortality worldwide. Mitochondria play a vital role in the pathological processes of cerebral ischemic injury, but its transplantation and underlying mechanisms remain unclear. In the present study, we examined the effects of mitochondrial therapy on the modulation of AMPK and SIRT1/PGC-1α signaling pathway, oxidative stress, and NLRP3 inflammasome activation after photothrombotic ischemic stroke (pt-MCAO). The adult male mice were subjected to the pt-MCAO in which the proximal-middle cerebral artery was exposed with a 532-nm laser beam for 4 min by retro-orbital injection of a photosensitive dye (Rose Bengal: 15 mg/kg) before the laser light exposure and isolated mitochondria (100 μg protein) were administered intranasally at 30 min, 24 h, and 48 h following post-stroke. After 72 h, mice were tested for neurobehavioral outcomes and euthanized for infarct volume, brain edema, and molecular analysis. First, we found that mitochondria therapy significantly decreased brain infarct volume and brain edema, improved neurological dysfunction, attenuated ischemic stroke-induced oxidative stress, and neuroinflammation. Second, mitochondria treatment inhibited NLRP3 inflammasome activation. Finally, mitochondria therapy accelerated p-AMPKα(Thr172) and PGC-1α expression and resorted SIRT1 protein expression levels in pt-MCAO mice. In conclusion, our results demonstrate that mitochondria therapy exerts neuroprotective effects by inhibiting oxidative damage and inflammation, mainly dependent on the heightening activation of the AMPK and SIRT1/PGC-1α signaling pathway. Thus, intranasal delivery of mitochondria might be considered a new therapeutic strategy for ischemic stroke treatment.
Collapse
Affiliation(s)
- Mohd Salman
- Department of Anatomy and Neurobiology, College of Medicine, The University of Tennessee Health Science Center, 875 Monroe Avenue, Wittenborg Bldg, Room-231, Memphis, TN, 38163, USA.
- Department of Anatomy and Neurobiology, College of Medicine, The University of Tennessee Health Science Center, 875 Monroe Avenue, Wittenborg Bldg, Room-228, Memphis, TN, 38163, USA.
| | - Amanda S Stayton
- Transplant Research Institute, College of Medicine, The University of Tennessee Health Science Center, 71 S Manassas St, Room 418H, Memphis, TN, 38103, USA
| | - Kehkashan Parveen
- Department of Pediatrics, The University of Tennessee Health Science Center, Memphis, TN, 38163, USA
| | - Arshi Parveen
- Department of Anatomy and Neurobiology, College of Medicine, The University of Tennessee Health Science Center, 875 Monroe Avenue, Wittenborg Bldg, Room-231, Memphis, TN, 38163, USA
| | - Michelle A Puchowicz
- Department of Pediatrics, The University of Tennessee Health Science Center, Memphis, TN, 38163, USA
| | - Suhel Parvez
- Department of Medical Elementology and Toxicology, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi, 110062, India
| | - Amandeep Bajwa
- Transplant Research Institute, College of Medicine, The University of Tennessee Health Science Center, 71 S Manassas St, Room 418H, Memphis, TN, 38103, USA.
- Department of Genetics, Genomics, and Informatics, College of Medicine, The University of Tennessee Health Science Center, Memphis, TN, USA.
- Department of Microbiology, Immunology, and Biochemistry, College of Medicine, The University of Tennessee Health Science Center, Memphis, TN, USA.
| | - Tauheed Ishrat
- Department of Anatomy and Neurobiology, College of Medicine, The University of Tennessee Health Science Center, 875 Monroe Avenue, Wittenborg Bldg, Room-231, Memphis, TN, 38163, USA.
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Tennessee Health Science Center, Memphis, TN, USA.
- Neuroscience Institute, The University of Tennessee Health Science Center, Memphis, TN, USA.
| |
Collapse
|
9
|
Hossain MI, Lee JH, Gagné JP, Khan J, Poirier GG, King PH, Dawson VL, Dawson TM, Andrabi SA. Poly(ADP-ribose) mediates bioenergetic defects and redox imbalance in neurons following oxygen and glucose deprivation. FASEB J 2024; 38:e23556. [PMID: 38498348 DOI: 10.1096/fj.202302559r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 02/16/2024] [Accepted: 02/29/2024] [Indexed: 03/20/2024]
Abstract
PARP-1 over-activation results in cell death via excessive PAR generation in different cell types, including neurons following brain ischemia. Glycolysis, mitochondrial function, and redox balance are key cellular processes altered in brain ischemia. Studies show that PAR generated after PARP-1 over-activation can bind hexokinase-1 (HK-1) and result in glycolytic defects and subsequent mitochondrial dysfunction. HK-1 is the neuronal hexokinase and catalyzes the first reaction of glycolysis, converting glucose to glucose-6-phosphate (G6P), a common substrate for glycolysis, and the pentose phosphate pathway (PPP). PPP is critical in maintaining NADPH and GSH levels via G6P dehydrogenase activity. Therefore, defects in HK-1 will not only decrease cellular bioenergetics but will also cause redox imbalance due to the depletion of GSH. In brain ischemia, whether PAR-mediated inhibition of HK-1 results in bioenergetics defects and redox imbalance is not known. We used oxygen-glucose deprivation (OGD) in mouse cortical neurons to mimic brain ischemia in neuronal cultures and observed that PARP-1 activation via PAR formation alters glycolysis, mitochondrial function, and redox homeostasis in neurons. We used pharmacological inhibition of PARP-1 and adenoviral-mediated overexpression of wild-type HK-1 (wtHK-1) and PAR-binding mutant HK-1 (pbmHK-1). Our data show that PAR inhibition or overexpression of HK-1 significantly improves glycolysis, mitochondrial function, redox homeostasis, and cell survival in mouse cortical neurons exposed to OGD. These results suggest that PAR binding and inhibition of HK-1 during OGD drive bioenergetic defects in neurons due to inhibition of glycolysis and impairment of mitochondrial function.
Collapse
Affiliation(s)
- M Iqbal Hossain
- Department of Pharmacology and Toxicology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Jun Hee Lee
- Department of Pharmacology and Toxicology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Jean-Philippe Gagné
- Department of Molecular Biology, Medical Biochemistry and Pathology, Faculty of Medicine, Université Laval, Québec City, Quebec, Canada
- Oncology Division, Centre de Recherche du Centre Hospitalier Universitaire de Québec-Université Laval, Québec City, Quebec, Canada
| | - Junaid Khan
- Department of Pharmacology and Toxicology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Guy G Poirier
- Department of Molecular Biology, Medical Biochemistry and Pathology, Faculty of Medicine, Université Laval, Québec City, Quebec, Canada
- Oncology Division, Centre de Recherche du Centre Hospitalier Universitaire de Québec-Université Laval, Québec City, Quebec, Canada
| | - Peter H King
- Department of Neurology, University of Alabama at Birmingham, Birmingham, Alabama, USA
- Birmingham Veterans Affairs Medical Center, Birmingham, Alabama, USA
- Center for Neurodegeneration and Experimental Therapeutics, The University of Alabama at Birmingham, Birmingham, Alabama, USA
- Cell, Developmental, and Integrative Biology, The University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Valina L Dawson
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Ted M Dawson
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Shaida A Andrabi
- Department of Pharmacology and Toxicology, University of Alabama at Birmingham, Birmingham, Alabama, USA
- Department of Neurology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| |
Collapse
|
10
|
Kurogi K, Suiko M, Sakakibara Y. Evolution and multiple functions of sulfonation and cytosolic sulfotransferases across species. Biosci Biotechnol Biochem 2024; 88:368-380. [PMID: 38271594 DOI: 10.1093/bbb/zbae008] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Accepted: 01/19/2024] [Indexed: 01/27/2024]
Abstract
Organisms have conversion systems for sulfate ion to take advantage of the chemical features. The use of biologically converted sulfonucleotides varies in an evolutionary manner, with the universal use being that of sulfonate donors. Sulfotransferases have the ability to transfer the sulfonate group of 3'-phosphoadenosine 5'-phosphosulfate to a variety of molecules. Cytosolic sulfotransferases (SULTs) play a role in the metabolism of low-molecular-weight compounds in response to the host organism's living environment. This review will address the diverse functions of the SULT in evolution, including recent findings. In addition to the diversity of vertebrate sulfotransferases, the molecular aspects and recent studies on bacterial and plant sulfotransferases are also addressed.
Collapse
Affiliation(s)
- Katsuhisa Kurogi
- Department of Biochemistry and Applied Biosciences, University of Miyazaki, Miyazaki, Japan
| | - Masahito Suiko
- Department of Biochemistry and Applied Biosciences, University of Miyazaki, Miyazaki, Japan
| | - Yoichi Sakakibara
- Department of Biochemistry and Applied Biosciences, University of Miyazaki, Miyazaki, Japan
| |
Collapse
|
11
|
Chik MW, Hazalin NAMN, Singh GKS. Regulation of phase I and phase II neurosteroid enzymes in the hippocampus of an Alzheimer's disease rat model: A focus on sulphotransferases and UDP-glucuronosyltransferases. Steroids 2022; 184:109035. [PMID: 35405201 DOI: 10.1016/j.steroids.2022.109035] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 03/27/2022] [Accepted: 04/05/2022] [Indexed: 11/17/2022]
Abstract
Neurosteroids have been associated with neurodegenerative diseases because they are involved in the modulation of neurotransmitter, neurotropic and neuroprotective actions. Emerging evidence suggests that the enzymes responsible for the synthesis of neurosteroids change during the progression of Alzheimer's disease (AD). The present study aimed to assess the changes in phase I and II enzymes involved in the metabolism of neurosteroids of the progestogen, androgenic and estrogenic steroidogenic pathways and the possibility that the neurosteroids are actively converted into the most abundant metabolites (i.e. glucuronides and sulphates). The gene expression for the phase I and II neurosteroid biosynthetic enzymes were studied in the hippocampus of streptozotocin AD rat model. Male Sprague-Dawley rats were randomly divided into control, sham (saline injected into the hippocampus) and 3 and 12 weeks post-STZ administration (STZ-G3w and STZ-G12w, respectively) groups. Behavioral assessments showed memory impairment in both STZ-injected groups, whereas the formation of amyloid-beta was more pronounced in the STZ-12w group. Gene expression of the hippocampus revealed that glucuronidation and sulphation enzymes transcript of the phase I metabolites were upregulated at the late stage of the disease progression (Hsd17b10, Hsd3b1, Akr1c3 and Cyp19a1) except for Sts. The phase II Sult and Ugt enzymes were mostly upregulated in the STZ-G12w rats (Sult1a1, Sult1e1, Ugt1a1, Ugt1a7c, Ugt1a6, Ugt2b35 and Ugt2b17) and normally expressed in the STZ-G3w group (Sult2a2, Sult2a6, Sult2b1, Ugt2b7, Sult4a1 and Ugt1a7c). In conclusion, changes occur in the phase I and II enzymes transcript of the progestogen, androgenic and estrogenic steroidogenic pathways during the progression of AD.
Collapse
Affiliation(s)
- Mazzura Wan Chik
- Department of Pharmaceutical Life Sciences, Faculty of Pharmacy, Universiti Teknologi MARA (UiTM), Selangor Branch, Puncak Alam Campus, 42300 Bandar Puncak Alam, Selangor, Malaysia
| | - Nurul Aqmar Mohd Nor Hazalin
- Department of Pharmaceutical Life Sciences, Faculty of Pharmacy, Universiti Teknologi MARA (UiTM), Selangor Branch, Puncak Alam Campus, 42300 Bandar Puncak Alam, Selangor, Malaysia; Integrative Pharmacogenomics Institute (iPROMiSE), Level 7, FF3, Universiti Teknologi MARA, Selangor Branch, Puncak Alam Campus, 42300 Bandar Puncak Alam, Selangor, Malaysia
| | - Gurmeet Kaur Surindar Singh
- Department of Pharmaceutical Life Sciences, Faculty of Pharmacy, Universiti Teknologi MARA (UiTM), Selangor Branch, Puncak Alam Campus, 42300 Bandar Puncak Alam, Selangor, Malaysia; Brain Degeneration and Therapeutics Group, Universiti Teknologi MARA (UiTM), 40450 Shah Alam, Selangor, Malaysia.
| |
Collapse
|
12
|
Boccuto L, Mitz A, Abenavoli L, Sarasua SM, Bennett W, Rogers C, DuPont B, Phelan K. Phenotypic Variability in Phelan–McDermid Syndrome and Its Putative Link to Environmental Factors. Genes (Basel) 2022; 13:genes13030528. [PMID: 35328081 PMCID: PMC8950073 DOI: 10.3390/genes13030528] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2022] [Revised: 03/14/2022] [Accepted: 03/15/2022] [Indexed: 02/01/2023] Open
Abstract
Phelan–McDermid syndrome (PMS) is a multi-systemic disorder characterized by both genetic and phenotypic variability. Genetic abnormalities causing PMS span from pathogenic variants of the SHANK3 gene to chromosomal rearrangements affecting the 22q13 region and leading to the loss of up to over nine megabases. The clinical presentation of individuals with PMS includes intellectual disability, neonatal hypotonia, delayed or absent speech, developmental delay, and minor dysmorphic facial features. Several other features may present with differences in age of onset and/or severity: seizures, autism, regression, sleep disorders, gastrointestinal problems, renal disorders, dysplastic toenails, and disrupted thermoregulation. Among the causes of this phenotypic variability, the size of the 22q13 deletion has effects that may be influenced by environmental factors interacting with haploinsufficiency or hemizygous variants of certain genes. Another mechanism linking environmental factors and phenotypic variability in PMS involves the loss of one copy of genes like BRD1 or CYP2D6, located at 22q13 and involved in the regulation of genomic methylation or pharmacokinetics, which are also influenced by external agents, such as diet and drugs. Overall, several non-mutually exclusive genetic and epigenetic mechanisms interact with environmental factors and may contribute to the clinical variability observed in individuals with PMS. Characterization of such factors will help to better manage this disorder.
Collapse
Affiliation(s)
- Luigi Boccuto
- Healthcare Genetics Program, School of Nursing, College of Behavioral, Social and Health Sciences, Clemson University, Clemson, SC 29634, USA;
- Correspondence: ; Tel.: +1-864-6561437
| | - Andrew Mitz
- Laboratory of Neuropsychology, National Institute of Mental Health, National Institutes of Health, Bethesda, MD 20892, USA;
| | - Ludovico Abenavoli
- Department of Health Sciences, University Magna Graecia, 88100 Catanzaro, Italy;
| | - Sara M. Sarasua
- Healthcare Genetics Program, School of Nursing, College of Behavioral, Social and Health Sciences, Clemson University, Clemson, SC 29634, USA;
| | - William Bennett
- Division of Pediatric Gastroenterology, Hepatology, and Nutrition, Indiana University School of Medicine/Riley Hospital for Children, Indianapolis, IN 46202, USA;
| | - Curtis Rogers
- Greenwood Genetic Center, Greenwood, SC 29646, USA; (C.R.); (B.D.)
| | - Barbara DuPont
- Greenwood Genetic Center, Greenwood, SC 29646, USA; (C.R.); (B.D.)
| | - Katy Phelan
- Genetics Laboratory, Florida Cancer Specialists &Research Institute, Fort Myers, FL 33916, USA;
| |
Collapse
|
13
|
Brettrager EJ, Meehan AW, Falany CN, van Waardenburg RCAM. Sulfotransferase 4A1 activity facilitates sulfate-dependent cellular protection to oxidative stress. Sci Rep 2022; 12:1625. [PMID: 35102205 PMCID: PMC8803991 DOI: 10.1038/s41598-022-05582-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Accepted: 01/13/2022] [Indexed: 11/17/2022] Open
Abstract
Sulfotransferase 4A1 (SULT4A1) is an orphan member of the cytosolic SULT superfamily that contains enzymes that catalyze the sulfonation of hydrophobic drugs and hormones. SULT4A1 has been assessed through all classical SULT approaches yet no SULT activity has been reported. To ascertain SULT4A1 function and activity, we utilized Saccharomyces cerevisiae as a model system, which exhibits no endogenous SULT activity nor possesses SULT-related genes. We observed that ectopic SULT4A1 expression in yeast displays similar subcellular localization as reported in mouse neurons and observed that SULT4A1 is associated with the outer mitochondria membrane. SULT4A1 expression stimulates colony formation and protects these cells from hydrogen peroxide and metabolism-associated oxidative stress. These SULT4A1-mediated phenotypes are dependent on extracellular sulfate that is converted in yeast to PAPS, the universal sulfonate donor for SULT activity. Thus, heterologous SULT4A1 expression in yeast is correctly distributed and functional, and SULT4A1 antioxidant activity is sulfate dependent supporting the concept that SULT4A1 has sulfate-associated activity.
Collapse
Affiliation(s)
- Evan J Brettrager
- Department of Pharmacology and Toxicology, University of Alabama at Birmingham, 155 Volker Hall, 1720 2nd Ave S., Birmingham, AL, 35294-0019, USA
| | - Arthur W Meehan
- Department of Pharmacology and Toxicology, University of Alabama at Birmingham, 155 Volker Hall, 1720 2nd Ave S., Birmingham, AL, 35294-0019, USA
| | - Charles N Falany
- Department of Pharmacology and Toxicology, University of Alabama at Birmingham, 155 Volker Hall, 1720 2nd Ave S., Birmingham, AL, 35294-0019, USA
| | - Robert C A M van Waardenburg
- Department of Pharmacology and Toxicology, University of Alabama at Birmingham, 155 Volker Hall, 1720 2nd Ave S., Birmingham, AL, 35294-0019, USA.
| |
Collapse
|
14
|
Ye Y, Wang X, Yang Z, Xu Q, Zhang B. Hsp22 Inhibits Oxidative Stress-Induced Endplate Chondrocyte Apoptosis by Regulating Mitochondrial Pathway. J BIOMATER TISS ENG 2021. [DOI: 10.1166/jbt.2021.2763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Background: Facet joint degeneration (FJD), which is also called facet joint syndrome (FJS), has become one of the most commonly seen etiological factors for lumbago. Cartilage lesion triggered by lumbar facet joint (LFJ) degeneration might be related to mitochondrial impairment,
but the its underlying mechanism remains unclear. Materials and methods: The endplate chondrocytes were induced by hydrogen peroxide (H2O2) to mimic the pathological conditions of oxidative stress. Enzyme linked immunosorbent assay (ELISA) were used for the evaluation
of reactive oxygen species (ROS). Adenosine-triphosphate (ATP) level was assessed using ATP detection, along with the detection the expression of cytochrome C in mitochondria (mito-cyt c) and in cell cytoplasm (cyto-cyt c) and cleaved caspase 3 by Western blot analysis. TUNEL assay was conducted
for the measurement of cell apoptosis in endplate chondrocytes. Reverse transcription-polymerase chain reaction (RT-qPCR) was used to verify the expression of heat shock protein 22 (HSP22) and the transfection efficiency of HSP22 interference plasmid. Results: It was found that H2O2
promoted the mitochondrial dysfunction, ROS generation and cell apoptosis in endplate chondrocytes. Moreover, HSP22 was down-regulated in H2O2-induced endplate chondrocytes, and interference of HSP22 decreased the ROS production, increased the ATP level and promoted the
cell apoptosis, resulting in the enhanced impairment of endplate chondrocytes. Additionally, mitochondrial ROS inhibitor (Mito-TEMPO) ameliorated the injury effects of HSP22 silencing in the H2O2-induced endplate chondrocytes. Conclusion: In conclusion, HSP22 inhibits
oxidative stress-induced endplate chondrocyte apoptosis by regulating mitochondrial pathway, possibly providing novel guidance direction for the treatment of LFJ degeneration.
Collapse
Affiliation(s)
- Yi Ye
- Department of Orthopedics, Ningbo Fourth Hospital, Xiang Shan County, Zhejiang 315700, P. R. China
| | - Xucan Wang
- Department of Orthopedics, Ningbo Fourth Hospital, Xiang Shan County, Zhejiang 315700, P. R. China
| | - Zhenqing Yang
- Department of Orthopedics, Ningbo Fourth Hospital, Xiang Shan County, Zhejiang 315700, P. R. China
| | - Qian Xu
- Department of Orthopedics, Ningbo Fourth Hospital, Xiang Shan County, Zhejiang 315700, P. R. China
| | - Bo Zhang
- Department of Orthopedics, Hwa Mei Hospital, University of Chinese Academy of Sciences, Ningbo Institute of Life and Health Industry, Ningbo, Zhejiang 315010, P. R.China
| |
Collapse
|
15
|
Ricciardello A, Tomaiuolo P, Persico AM. Genotype-phenotype correlation in Phelan-McDermid syndrome: A comprehensive review of chromosome 22q13 deleted genes. Am J Med Genet A 2021; 185:2211-2233. [PMID: 33949759 PMCID: PMC8251815 DOI: 10.1002/ajmg.a.62222] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 03/28/2021] [Accepted: 04/04/2021] [Indexed: 12/19/2022]
Abstract
Phelan‐McDermid syndrome (PMS, OMIM #606232), also known as chromosome 22q13 deletion syndrome, is a rare genetic disorder characterized by intellectual disability, hypotonia, delayed or absent speech, motor impairment, autism spectrum disorder, behavioral anomalies, and minor aspecific dysmorphic features. Haploinsufficiency of SHANK3, due to intragenic deletions or point mutations, is sufficient to cause many neurobehavioral features of PMS. However, several additional genes located within larger 22q13 deletions can contribute to the great interindividual variability observed in the PMS phenotype. This review summarizes the phenotypic contributions predicted for 213 genes distributed along the largest 22q13.2‐q13.33 terminal deletion detected in our sample of 63 PMS patients by array‐CGH analysis, spanning 9.08 Mb. Genes have been grouped into four categories: (1) genes causing human diseases with an autosomal dominant mechanism, or (2) with an autosomal recessive mechanism; (3) morphogenetically relevant genes, either involved in human diseases with additive co‐dominant, polygenic, and/or multifactorial mechanisms, or implicated in animal models but not yet documented in human pathology; (4) protein coding genes either functionally nonrelevant, with unknown function, or pathogenic through mechanisms other than haploinsufficiency; piRNAs, noncoding RNAs, miRNAs, novel transcripts and pseudogenes. Our aim is to understand genotype–phenotype correlations in PMS patients and to provide clinicians with a conceptual framework to promote evidence‐based genetic work‐ups, clinical assessments, and therapeutic interventions.
Collapse
Affiliation(s)
- Arianna Ricciardello
- Interdepartmental Program "Autism 0-90", "Gaetano Martino" University Hospital, University of Messina, Messina, Italy
| | - Pasquale Tomaiuolo
- Interdepartmental Program "Autism 0-90", "Gaetano Martino" University Hospital, University of Messina, Messina, Italy
| | - Antonio M Persico
- Interdepartmental Program "Autism 0-90", "Gaetano Martino" University Hospital, University of Messina, Messina, Italy
| |
Collapse
|
16
|
Sun Y, Machalz D, Wolber G, Parr MK, Bureik M. Functional Expression of All Human Sulfotransferases in Fission Yeast, Assay Development, and Structural Models for Isoforms SULT4A1 and SULT6B1. Biomolecules 2020; 10:E1517. [PMID: 33171978 PMCID: PMC7694633 DOI: 10.3390/biom10111517] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 11/03/2020] [Accepted: 11/04/2020] [Indexed: 11/29/2022] Open
Abstract
Cytosolic sulfotransferases (SULTs) catalyze phase II (conjugation) reactions of drugs and endogenous compounds. A complete set of recombinant fission yeast strains each expressing one of the 14 human SULTs was generated, including SULT4A1 and SULT6B1. Sulfation of test substrates by whole-cell biotransformation was successfully demonstrated for all enzymes for which substrates were previously known. The results proved that the intracellular production of the cofactor 3'-phosphoadenosine 5'-phosphosulfate (PAPS) necessary for SULT activity in fission yeast is sufficiently high to support metabolite production. A modified variant of sulfotransferase assay was also developed that employs permeabilized fission yeast cells (enzyme bags). Using this approach, SULT4A1-dependent sulfation of 1-naphthol was observed. Additionally, a new and convenient SULT activity assay is presented. It is based on the sulfation of a proluciferin compound, which was catalyzed by SULT1E1, SULT2A1, SULT4A1, and SULT6B1. For the latter two enzymes this study represents the first demonstration of their enzymatic functionality. Furthermore, the first catalytically competent homology models for SULT4A1 and SULT6B1 in complex with PAPS are reported. Through mechanistic molecular modeling driven by substrate docking, we pinned down the increased activity levels of these two isoforms to optimized substrate binding.
Collapse
Affiliation(s)
- Yanan Sun
- School of Pharmaceutical Science and Technology, Health Sciences Platform, Tianjin University, Tianjin 300072, China;
- Pharmaceutical and Medicinal Chemistry (Pharmaceutical Analyses), Institute of Pharmacy, Freie Universitaet Berlin, 14195 Berlin, Germany
| | - David Machalz
- Pharmaceutical and Medicinal Chemistry (Computer-Aided Drug Design), Institute of Pharmacy, Freie Universitaet Berlin, 14195 Berlin, Germany; (D.M.); (G.W.)
| | - Gerhard Wolber
- Pharmaceutical and Medicinal Chemistry (Computer-Aided Drug Design), Institute of Pharmacy, Freie Universitaet Berlin, 14195 Berlin, Germany; (D.M.); (G.W.)
| | - Maria Kristina Parr
- Pharmaceutical and Medicinal Chemistry (Pharmaceutical Analyses), Institute of Pharmacy, Freie Universitaet Berlin, 14195 Berlin, Germany
| | - Matthias Bureik
- School of Pharmaceutical Science and Technology, Health Sciences Platform, Tianjin University, Tianjin 300072, China;
| |
Collapse
|
17
|
Idris M, Mitchell DJ, Gordon R, Sidharthan NP, Butcher NJ, Minchin RF. Interaction of the Brain-Selective Sulfotransferase SULT4A1 with Other Cytosolic Sulfotransferases: Effects on Protein Expression and Function. Drug Metab Dispos 2020; 48:337-344. [PMID: 32152050 DOI: 10.1124/dmd.119.089714] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2019] [Accepted: 01/28/2020] [Indexed: 12/14/2022] Open
Abstract
Sulfotransferase (SULT) 4A1 is a brain-selective sulfotransferase-like protein that has recently been shown to be essential for normal neuronal development in mice. In the present study, SULT4A1 was found to colocalize with SULT1A1/3 in human brain neurons. Using immunoprecipitation, SULT4A1 was shown to interact with both SULT1A1 and SULT1A3 when expressed in human cells. Mutation of the conserved dimerization motif located in the C terminus of the sulfotransferases prevented this interaction. Both ectopically expressed and endogenous SULT4A1 decreased SULT1A1/3 protein levels in neuronal cells, and this was also prevented by mutation of the dimerization motif. During differentiation of neuronal SH-SY5Y cells, there was a loss in SULT1A1/3 protein but an increase in SULT4A1 protein. This resulted in an increase in the toxicity of dopamine, a substrate for SULT1A3. Inhibition of SULT4A1 using small interference RNA abrogated the loss in SULT1A1/3 and reversed dopamine toxicity. These results show a reciprocal relationship between SULT4A1 and the other sulfotransferases, suggesting that it may act as a chaperone to control the expression of SULT1A1/3 in neuronal cells. SIGNIFICANCE STATEMENT: The catalytically inactive sulfotransferase (SULT) 4A1 may regulate the function of other SULTs by interacting with them via a conserved dimerization motif. In neuron-like cells, SULT4A1 is able to modulate dopamine toxicity by interacting with SULT1A3, potentially decreasing the metabolism of dopamine.
Collapse
Affiliation(s)
- Misgana Idris
- School of Biomedical Sciences, University of Queensland, St Lucia, Queensland, Australia
| | - Deanne J Mitchell
- School of Biomedical Sciences, University of Queensland, St Lucia, Queensland, Australia
| | - Richard Gordon
- School of Biomedical Sciences, University of Queensland, St Lucia, Queensland, Australia
| | - Neelima P Sidharthan
- School of Biomedical Sciences, University of Queensland, St Lucia, Queensland, Australia
| | - Neville J Butcher
- School of Biomedical Sciences, University of Queensland, St Lucia, Queensland, Australia
| | - Rodney F Minchin
- School of Biomedical Sciences, University of Queensland, St Lucia, Queensland, Australia
| |
Collapse
|