1
|
den Bakker E, Smith DEC, Finken MJJ, Wamelink MMC, Salomons GS, van de Kamp JM, Bökenkamp A. Sulfate: a neglected (but potentially highly relevant) anion. Essays Biochem 2024; 68:391-399. [PMID: 38639060 DOI: 10.1042/ebc20230097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 03/28/2024] [Accepted: 03/28/2024] [Indexed: 04/20/2024]
Abstract
Sulfate is an important anion as sulfonation is essential in modulation of several compounds, such as exogens, polysaccharide chains of proteoglycans, cholesterol or cholesterol derivatives and tyrosine residues of several proteins. Sulfonation requires the presence of both the sulfate donor 3'-phosphoadenosine-5'-phosphosulfate (PAPS) and a sulfotransferase. Genetic disorders affecting sulfonation, associated with skeletal abnormalities, impaired neurological development and endocrinopathies, demonstrate the importance of sulfate. Yet sulfate is not measured in clinical practice. This review addresses sulfate metabolism and consequences of sulfonation defects, how to measure sulfate and why we should measure sulfate more often.
Collapse
Affiliation(s)
- Emil den Bakker
- Department of Pediatric Nephrology, Emma Childrens Hospital, Amsterdam UMC, Amsterdam, the Netherlands
| | - Desiree E C Smith
- Department of Metabolic Diseases, Amsterdam UMC, Amsterdam, the Netherlands
| | - Martijn J J Finken
- Department of Pediatric Endocrinology, Emma Childrens Hospital, Amsterdam UMC, Amsterdam, the Netherlands
| | | | - Gajja S Salomons
- Department of Metabolic Diseases, Amsterdam UMC, Amsterdam, the Netherlands
- Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam UMC, Amsterdam, the Netherlands
| | - Jiddeke M van de Kamp
- Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam UMC, Amsterdam, the Netherlands
- Amsterdam Reproduction and Development, Amsterdam UMC, Amsterdam, the Netherlands
- Department of Human Genetics, Amsterdam UMC, Amsterdam, the Netherlands
| | - Arend Bökenkamp
- Department of Pediatric Nephrology, Emma Childrens Hospital, Amsterdam UMC, Amsterdam, the Netherlands
| |
Collapse
|
2
|
Chen X, Zhang Y, Yin J, Liu C, Xie M, Wang Y, Chen M, Zhang R, Yuan X, Li D, Chen X, Gao X, Cai G, Zhang S, Zhou B, Yang M. Structural basis for the reaction cycle and transport mechanism of human Na +-sulfate cotransporter NaS1 (SLC13A1). SCIENCE ADVANCES 2024; 10:eado6778. [PMID: 39576865 PMCID: PMC11584011 DOI: 10.1126/sciadv.ado6778] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Accepted: 10/23/2024] [Indexed: 11/24/2024]
Abstract
Sulfate (SO42-) is a pivotal inorganic anion with essential roles in mammalian physiology. NaS1, a member of solute carrier 13 family and divalent anion/sodium symporter family, functions as a Na+-sulfate cotransporter, facilitating sulfate (re)absorption across renal proximal tubule and small intestine epithelia. While previous studies have linked several human disorders to mutations in the NaS1 gene, its transport mechanism remains unclear. Here, we report the cryo-electron microscopy structures of five distinct conformations of the human NaS1 at resolutions of 2.7 to 3.3 angstroms, revealing the substrates recognition mechanism and the conformational change of NaS1 during the Na+-sulfate cotransport cycle. Our studies delineate the molecular basis of the detailed dynamic transport cycle of NaS1. These findings advance the current understanding of the Na+-sulfate cotransport mechanism, human sulfate (re)absorption, and the implications of disease-associated NaS1 mutations.
Collapse
Affiliation(s)
- Xudong Chen
- Ministry of Education Key Laboratory of Protein Science, Tsinghua-Peking Center for Life Sciences, Beijing Advanced Innovation Center for Structural Biology, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Youqi Zhang
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Diseases Research, Beijing 100853, China
| | - Jian Yin
- Ministry of Education Key Laboratory of Protein Science, Tsinghua-Peking Center for Life Sciences, Beijing Advanced Innovation Center for Structural Biology, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Chang Liu
- Beijing Life Science Academy, Beijing 102209, China
| | - Min Xie
- Ministry of Education Key Laboratory of Protein Science, Tsinghua-Peking Center for Life Sciences, Beijing Advanced Innovation Center for Structural Biology, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Yixue Wang
- Ministry of Education Key Laboratory of Protein Science, Tsinghua-Peking Center for Life Sciences, Beijing Advanced Innovation Center for Structural Biology, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Meiying Chen
- Ministry of Education Key Laboratory of Protein Science, Tsinghua-Peking Center for Life Sciences, Beijing Advanced Innovation Center for Structural Biology, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Rui Zhang
- Department of Cardiology, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing 102218, China
| | - Xinyi Yuan
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Hangzhou Normal University, Hangzhou 311121, China
| | - De Li
- Radioisotope Laboratory of Center of Biomedical Analysis, Tsinghua University, Beijing 100084, China
| | - Xiangmei Chen
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Diseases Research, Beijing 100853, China
| | - Xin Gao
- Computer Science Program, Computer, Electrical and Mathematical Sciences and Engineering Division, King Abdullah University of Science and Technology (KAUST), Thuwal 23955-6900, Kingdom of Saudi Arabia
- Computational Bioscience Research Center, King Abdullah University of Science and Technology, Thuwal 23955-6900, Kingdom of Saudi Arabia
| | - Guangyan Cai
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Diseases Research, Beijing 100853, China
| | - Sensen Zhang
- Ministry of Education Key Laboratory of Protein Science, Tsinghua-Peking Center for Life Sciences, Beijing Advanced Innovation Center for Structural Biology, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Boda Zhou
- Department of Cardiology, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing 102218, China
| | - Maojun Yang
- Ministry of Education Key Laboratory of Protein Science, Tsinghua-Peking Center for Life Sciences, Beijing Advanced Innovation Center for Structural Biology, School of Life Sciences, Tsinghua University, Beijing 100084, China
- Cryo-EM Facility Center, Southern University of Science and Technology, Shenzhen, China
| |
Collapse
|
3
|
Zorkoltseva IV, Elgaeva EE, Belonogova NM, Kirichenko AV, Svishcheva GR, Freidin MB, Williams FMK, Suri P, Tsepilov YA, Axenovich TI. Multi-Trait Exome-Wide Association Study of Back Pain-Related Phenotypes. Genes (Basel) 2023; 14:1962. [PMID: 37895311 PMCID: PMC10606006 DOI: 10.3390/genes14101962] [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: 09/21/2023] [Revised: 10/16/2023] [Accepted: 10/18/2023] [Indexed: 10/29/2023] Open
Abstract
Back pain (BP) is a major contributor to disability worldwide, with heritability estimated at 40-60%. However, less than half of the heritability is explained by common genetic variants identified by genome-wide association studies. More powerful methods and rare and ultra-rare variant analysis may offer additional insight. This study utilized exome sequencing data from the UK Biobank to perform a multi-trait gene-based association analysis of three BP-related phenotypes: chronic back pain, dorsalgia, and intervertebral disc disorder. We identified the SLC13A1 gene as a contributor to chronic back pain via loss-of-function (LoF) and missense variants. This gene has been previously detected in two studies. A multi-trait approach uncovered the novel FSCN3 gene and its impact on back pain through LoF variants. This gene deserves attention because it is only the second gene shown to have an effect on back pain due to LoF variants and represents a promising drug target for back pain therapy.
Collapse
Affiliation(s)
- Irina V. Zorkoltseva
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences, 630090 Novosibirsk, Russia; (I.V.Z.); (E.E.E.); (N.M.B.); (A.V.K.); (G.R.S.); (Y.A.T.)
| | - Elizaveta E. Elgaeva
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences, 630090 Novosibirsk, Russia; (I.V.Z.); (E.E.E.); (N.M.B.); (A.V.K.); (G.R.S.); (Y.A.T.)
- Department of Natural Sciences, Novosibirsk State University, 630090 Novosibirsk, Russia
| | - Nadezhda M. Belonogova
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences, 630090 Novosibirsk, Russia; (I.V.Z.); (E.E.E.); (N.M.B.); (A.V.K.); (G.R.S.); (Y.A.T.)
| | - Anatoliy V. Kirichenko
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences, 630090 Novosibirsk, Russia; (I.V.Z.); (E.E.E.); (N.M.B.); (A.V.K.); (G.R.S.); (Y.A.T.)
| | - Gulnara R. Svishcheva
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences, 630090 Novosibirsk, Russia; (I.V.Z.); (E.E.E.); (N.M.B.); (A.V.K.); (G.R.S.); (Y.A.T.)
- Vavilov Institute of General Genetics, Russian Academy of Sciences, 119333 Moscow, Russia
| | - Maxim B. Freidin
- Department of Biology, School of Biological and Behavioural Sciences, Queen Mary University of London, London EC1M 6BQ, UK;
| | - Frances M. K. Williams
- Department of Twin Research and Genetic Epidemiology, King’s College London, London SE1 7EH, UK;
| | - Pradeep Suri
- Seattle Epidemiologic Research and Information Center, VA Puget Sound Health Care System, Seattle, WA 98108, USA
- Division of Rehabilitation Care Services, Seattle, WA 98208, USA
- Clinical Learning, Evidence, and Research Center, University of Washington, Seattle, WA 98195, USA
- Department of Rehabilitation Medicine, University of Washington, Seattle, WA 98195, USA
| | - Yakov A. Tsepilov
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences, 630090 Novosibirsk, Russia; (I.V.Z.); (E.E.E.); (N.M.B.); (A.V.K.); (G.R.S.); (Y.A.T.)
| | - Tatiana I. Axenovich
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences, 630090 Novosibirsk, Russia; (I.V.Z.); (E.E.E.); (N.M.B.); (A.V.K.); (G.R.S.); (Y.A.T.)
| |
Collapse
|
4
|
Yu M, Wu M, Secundo F, Liu Z. Detection, production, modification, and application of arylsulfatases. Biotechnol Adv 2023; 67:108207. [PMID: 37406746 DOI: 10.1016/j.biotechadv.2023.108207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 06/16/2023] [Accepted: 06/30/2023] [Indexed: 07/07/2023]
Abstract
Arylsulfatase is a subset of sulfatase which catalyzes the hydrolysis of aryl sulfate ester. Arylsulfatase is widely distributed among microorganisms, mammals and green algae, but the arylsulfatase-encoding gene has not yet been found in the genomes of higher plants so far. Arylsulfatase plays an important role in the sulfur flows between nature and organisms. In this review, we present the maturation and catalytic mechanism of arylsulfatase, and the recent literature on the expression and production of arylsulfatase in wild-type and engineered microorganisms, as well as the modification of arylsulfatase by genetic engineering are summarized. We focus on arylsulfatases from microbial origin and give an overview of different assays and substrates used to determine the arylsulfatase activity. Furthermore, the researches about arylsulfatase application on the field of agar desulfation, soil sulfur cycle and soil evaluation are also discussed. Finally, the perspectives concerning the future research on arylsulfatase are prospected.
Collapse
Affiliation(s)
- Mengjiao Yu
- Qingdao Key Laboratory of Food Biotechnology, College of Food Science and Engineering, Ocean University of China, Qingdao 266404, PR China; Key Laboratory of Biological Processing of Aquatic Products, China National Light Industry, Qingdao 266404, PR China
| | - Meixian Wu
- Qingdao Key Laboratory of Food Biotechnology, College of Food Science and Engineering, Ocean University of China, Qingdao 266404, PR China; Key Laboratory of Biological Processing of Aquatic Products, China National Light Industry, Qingdao 266404, PR China
| | - Francesco Secundo
- Istituto di Scienze e Tecnologie Chimiche "Giulio Natta", Consiglio Nazionale delle Ricerche, via Mario Bianco 9, Milan 20131, Italy
| | - Zhen Liu
- Qingdao Key Laboratory of Food Biotechnology, College of Food Science and Engineering, Ocean University of China, Qingdao 266404, PR China; Key Laboratory of Biological Processing of Aquatic Products, China National Light Industry, Qingdao 266404, PR China.
| |
Collapse
|
5
|
Hurrion EM, Badawi N, Boyd RN, Morgan C, Gibbons K, Hennig S, Koorts P, Chauhan M, Bowling F, Flenady V, Kumar S, Dawson PA. SuPreme Study: a protocol to study the neuroprotective potential of sulfate among very/extremely preterm infants. BMJ Open 2023; 13:e076130. [PMID: 37451710 PMCID: PMC10351292 DOI: 10.1136/bmjopen-2023-076130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Accepted: 06/30/2023] [Indexed: 07/18/2023] Open
Abstract
INTRODUCTION Antenatal maternal magnesium sulfate (MgSO4) administration is a proven efficacious neuroprotective treatment reducing the risk of cerebral palsy (CP) among infants born preterm. Identification of the neuroprotective component with target plasma concentrations could lead to neonatal treatment with greater efficacy and accessibility. METHODS AND ANALYSIS This is a prospective observational cohort study, in three tertiary Australian centres. Participants are preterm infants, irrespective of antenatal MgSO4 exposure, born in 2013-2020 at 24+0 to 31+6 weeks gestation, and followed up to 2 years corrected age (CA) (to September 2023). 1595 participants are required (allowing for 17% deaths/loss to follow-up) to detect a clinically significant reduction (30% relative risk reduction) in CP when sulfate concentration at 7 days of age is 1 SD above the mean.A blood sample is collected on day 7 of age for plasma sulfate and magnesium measurement. In a subset of participants multiple blood and urine samples are collected for pharmacokinetic studies, between days 1-28, and in a further subset mother/infant blood is screened for genetic variants of sulfate transporter genes.The primary outcome is CP. Surviving infants are assessed for high risk of CP at 12-14 weeks CA according to Prechtl's Method to assess General Movements. Follow-up at 2 years CA includes assessments for CP, cognitive, language and motor development, and social/behavioural difficulties.Multivariate analyses will examine the association between day 7 plasma sulfate/magnesium concentrations with adverse neurodevelopmental outcomes. A population pharmacokinetic model for sulfate in the preterm infant will be created using non-linear mixed-effects modelling. ETHICS AND DISSEMINATION The study has been approved by Mater Misericordiae Ltd Human Research Ethics Committee (HREC/14/MHS/188). Results will be disseminated in peer-reviewed journal publications, and provided to the funding bodies. Using consumer input, a summary will be prepared for participants and consumer groups.
Collapse
Affiliation(s)
- Elizabeth M Hurrion
- Department of Newborn Services, Mater Mothers' Hospital, Brisbane, Queensland, Australia
- Mater Research Institute The University of Queensland, South Brisbane, Queensland, Australia
| | - Nadia Badawi
- Cerebral Palsy Alliance, The University of Sydney, Sydney, New South Wales, Australia
| | - Roslyn N Boyd
- Queensland Cerebral Palsy and Rehabilitation Research Centre, The University of Queensland, Saint Lucia, Queensland, Australia
| | - Catherine Morgan
- Cerebral Palsy Alliance, The University of Sydney, Sydney, New South Wales, Australia
| | - Kristen Gibbons
- Child Health Research Centre, Mater Research Institute The University of Queensland, South Brisbane, Queensland, Australia
| | - Stefanie Hennig
- School of Clinical Sciences, Queensland University of Technology Faculty of Health, Kelvin Grove, Queensland, Australia
- Integrated Drug Development, Certara Strategic Consulting, Certara LP, Princeton, New Jersey, USA
| | - Pieter Koorts
- Grantley Stable Neonatal Unit, Royal Brisbane and Women's Hospital, Herston, Queensland, Australia
| | - Manbir Chauhan
- Department of Newborn Care, Gold Coast University Hospital, Southport, Queensland, Australia
| | - Francis Bowling
- Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria, Australia
| | - Vicki Flenady
- Mater Research Institute The University of Queensland, South Brisbane, Queensland, Australia
| | - Sailesh Kumar
- Mater Research Institute The University of Queensland, South Brisbane, Queensland, Australia
| | - Paul A Dawson
- Mater Research Institute The University of Queensland, South Brisbane, Queensland, Australia
| |
Collapse
|
6
|
Ong WSY, Ji K, Pathiranage V, Maydew C, Baek K, Villones RLE, Meloni G, Walker AR, Dodani SC. Rational Design of the β-Bulge Gate in a Green Fluorescent Protein Accelerates the Kinetics of Sulfate Sensing. Angew Chem Int Ed Engl 2023; 62:e202302304. [PMID: 37059690 PMCID: PMC10330437 DOI: 10.1002/anie.202302304] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 04/11/2023] [Accepted: 04/12/2023] [Indexed: 04/16/2023]
Abstract
Detection of anions in complex aqueous media is a fundamental challenge with practical utility that can be addressed by supramolecular chemistry. Biomolecular hosts such as proteins can be used and adapted as an alternative to synthetic hosts. Here, we report how the mutagenesis of the β-bulge residues (D137 and W138) in mNeonGreen, a bright, monomeric fluorescent protein, unlocks and tunes the anion preference at physiological pH for sulfate, resulting in the turn-off sensor SulfOFF-1. This unprecedented sensing arises from an enhancement in the kinetics of binding, largely driven by position 138. In line with these data, molecular dynamics (MD) simulations capture how the coordinated entry and gating of sulfate into the β-barrel is eliminated upon mutagenesis to facilitate binding and fluorescence quenching.
Collapse
Affiliation(s)
- Whitney S. Y. Ong
- Department of Chemistry and Biochemistry, The University of Texas at Dallas, 800 W. Campbell Road, Richardson, Texas 75080-3021, USA
| | - Ke Ji
- Department of Chemistry and Biochemistry, The University of Texas at Dallas, 800 W. Campbell Road, Richardson, Texas 75080-3021, USA
| | - Vishaka Pathiranage
- Department of Chemistry, Wayne State University, 42 W. Warren Ave. Detroit, MI 48202, USA
| | - Caden Maydew
- Department of Chemistry and Biochemistry, The University of Texas at Dallas, 800 W. Campbell Road, Richardson, Texas 75080-3021, USA
| | - Kiheon Baek
- Department of Chemistry and Biochemistry, The University of Texas at Dallas, 800 W. Campbell Road, Richardson, Texas 75080-3021, USA
| | - Rhiza Lyne E. Villones
- Department of Chemistry and Biochemistry, The University of Texas at Dallas, 800 W. Campbell Road, Richardson, Texas 75080-3021, USA
| | - Gabriele Meloni
- Department of Chemistry and Biochemistry, The University of Texas at Dallas, 800 W. Campbell Road, Richardson, Texas 75080-3021, USA
| | - Alice R. Walker
- Department of Chemistry, Wayne State University, 42 W. Warren Ave. Detroit, MI 48202, USA
| | - Sheel C. Dodani
- Department of Chemistry and Biochemistry, The University of Texas at Dallas, 800 W. Campbell Road, Richardson, Texas 75080-3021, USA
| |
Collapse
|
7
|
Soprano LL, Ferrero MR, Jacobs T, Couto AS, Duschak VG. Hallmarks of the relationship between host and Trypanosoma cruzi sulfated glycoconjugates along the course of Chagas disease. Front Cell Infect Microbiol 2023; 13:1028496. [PMID: 37256110 PMCID: PMC10225527 DOI: 10.3389/fcimb.2023.1028496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Accepted: 04/17/2023] [Indexed: 06/01/2023] Open
Abstract
American Trypanosomiasis or Chagas disease (ChD), a major problem that is still endemic in large areas of Latin America, is caused by Trypanosoma cruzi. This agent holds a major antigen, cruzipain (Cz). Its C-terminal domain (C-T) is retained in the glycoprotein mature form and bears several post-translational modifications. Glycoproteins containing sulfated N-linked oligosaccharides have been mostly implicated in numerous specific procedures of molecular recognition. The presence of sulfated oligosaccharides was demonstrated in Cz, also in a minor abundant antigen with serine-carboxypeptidase (SCP) activity, as well as in parasite sulfatides. Sulfate-bearing glycoproteins in Trypanosomatids are targets of specific immune responses. T. cruzi chronically infected subjects mount specific humoral immune responses to sulfated Cz. Unexpectedly, in the absence of infection, mice immunized with C-T, but not with sulfate-depleted C-T, showed ultrastructural heart anomalous pathological effects. Moreover, the synthetic anionic sugar conjugate GlcNAc6SO3-BSA showed to mimic the N-glycan-linked sulfated epitope (sulfotope) humoral responses that natural Cz elicits. Furthermore, it has been reported that sulfotopes participate via the binding of sialic acid Ig-like-specific lectins (Siglecs) to sulfosialylated glycoproteins in the immunomodulation by host-parasite interaction as well as in the parasite infection process. Strikingly, recent evidence involved Cz-sulfotope-specific antibodies in the immunopathogenesis and infection processes during the experimental ChD. Remarkably, sera from chronically T. cruzi-infected individuals with mild disease displayed higher levels of IgG2 antibodies specific for sulfated glycoproteins and sulfatides than those with more severe forms of the disease, evidencing that T. cruzi sulfotopes are antigenic independently of the sulfated glycoconjugate type. Ongoing assays indicate that antibodies specific for sulfotopes might be considered biomarkers of human cardiac ChD progression, playing a role as predictors of stability from the early mild stages of chronic ChD.
Collapse
Affiliation(s)
- Luciana L. Soprano
- Area of Protein Biochemistry and Parasite Glycobiology, Research Department National Institute of Parasitology (INP)”Dr. Mario Fatala Chaben”, National Administration of Health Institutes (ANLIS)-Malbrán, National Health Department, National Council of Scientific and Technical Research (CONICET), Buenos Aires, Argentina
| | - Maximiliano R. Ferrero
- Max-Planck Heart and Lung Laboratory, Research Institute in Biomedicine in Buenos Aires (IBioBA), Argentine-Department of Internal Medicine II, University Medical Center Giessen and Marburg, Giessen, Germany
| | - Thomas Jacobs
- Immunology Department, Bernhard Notch Institute of Tropical Medicine, Hamburg, Germany
| | - Alicia S. Couto
- Faculty in Exact and Natural Sciences (FCEN), Chemical Organic Department-National Council of Scientific and Technical Research (CONICET), Center of CarboHydrates (CHIHIDECAR), University of Buenos Aires, Buenos Aires, Argentina
| | - Vilma G. Duschak
- Area of Protein Biochemistry and Parasite Glycobiology, Research Department National Institute of Parasitology (INP)”Dr. Mario Fatala Chaben”, National Administration of Health Institutes (ANLIS)-Malbrán, National Health Department, National Council of Scientific and Technical Research (CONICET), Buenos Aires, Argentina
| |
Collapse
|
8
|
Bovonratwet P, Kulm S, Kolin DA, Song J, Morse KW, Cunningham ME, Albert TJ, Sandhu HS, Kim HJ, Iyer S, Elemento O, Qureshi SA. Identification of Novel Genetic Markers for the Risk of Spinal Pathologies: A Genome-Wide Association Study of 2 Biobanks. J Bone Joint Surg Am 2023:00004623-990000000-00758. [PMID: 36927824 DOI: 10.2106/jbjs.22.00872] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/18/2023]
Abstract
BACKGROUND Identifying genetic risk factors for spinal disorders may lead to knowledge regarding underlying molecular mechanisms and the development of new treatments. METHODS Cases of lumbar spondylolisthesis, spinal stenosis, degenerative disc disease, and pseudarthrosis after spinal fusion were identified from the UK Biobank. Controls were patients without the diagnosis. Whole-genome regressions were used to test for genetic variants potentially implicated in the occurrence of each phenotype. External validation was performed in FinnGen. RESULTS A total of 389,413 participants were identified from the UK Biobank. A locus on chromosome 2 spanning GFPT1, NFU1, AAK1, and LOC124906020 was implicated in lumbar spondylolisthesis. Two loci on chromosomes 2 and 12 spanning genes GFPT1, NFU1, and PDE3A were implicated in spinal stenosis. Three loci on chromosomes 6, 10, and 15 spanning genes CHST3, LOC102723493, and SMAD3 were implicated in degenerative disc disease. Finally, 2 novel loci on chromosomes 5 and 9, with the latter corresponding to the LOC105376270 gene, were implicated in pseudarthrosis. Some of these variants associated with spinal stenosis and degenerative disc disease were also replicated in FinnGen. CONCLUSIONS This study revealed nucleotide variations in select genetic loci that were potentially implicated in 4 different spinal pathologies, providing potential insights into the pathological mechanisms. LEVEL OF EVIDENCE Prognostic Level III. See Instructions for Authors for a complete description of levels of evidence.
Collapse
Affiliation(s)
| | - Scott Kulm
- Caryl and Israel Englander Institute of Precision Medicine, Weill Cornell Medicine, New York, NY
| | - David A Kolin
- Department of Spine Surgery, Hospital for Special Surgery, New York, NY
| | - Junho Song
- Department of Spine Surgery, Hospital for Special Surgery, New York, NY
| | - Kyle W Morse
- Department of Spine Surgery, Hospital for Special Surgery, New York, NY
| | | | - Todd J Albert
- Department of Spine Surgery, Hospital for Special Surgery, New York, NY
| | | | - Han Jo Kim
- Department of Spine Surgery, Hospital for Special Surgery, New York, NY
| | - Sravisht Iyer
- Department of Spine Surgery, Hospital for Special Surgery, New York, NY
| | - Olivier Elemento
- Caryl and Israel Englander Institute of Precision Medicine, Weill Cornell Medicine, New York, NY
| | - Sheeraz A Qureshi
- Department of Spine Surgery, Hospital for Special Surgery, New York, NY
| |
Collapse
|
9
|
Genome-wide characterization of the cytosolic sulfotransferase 1B member 1 (SULT1B1) family and its expression responses to sulfide stress in the razor clam Sinonovacula constricta. Gene 2023; 856:147136. [PMID: 36572072 DOI: 10.1016/j.gene.2022.147136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 12/16/2022] [Accepted: 12/20/2022] [Indexed: 12/24/2022]
Abstract
The razor clam (Sinonovacula constricta), a typical burrowing organism in the intertidal zones, is often exposed to sulfide environment and shows strong sulfide tolerance. Located downstream of the sulfur metabolism pathway, cytosolic sulfotransferase family 1B member 1 (SULT1B1) is a key enzyme catalysing the sulfonation reaction, and plays an important role in the biotransformation of endogenous substances such as thyroid hormones (THs). To investigate their roles in sulfide resistance, a systematic analysis of S. constricta SULT1B1s (ScSULT1B1s), including genomic distribution, phylogenetic relationships, gene structure, conserved motifs, and expression profiles under sulfide stress, was performed. A total of 10 ScSULT1B1 genes were found in the S. constricta genome. Sequence analysis showed that ScSULT1B1 gene family encoded 155-425 amino acids, containing four catalytic active sites (K, N, H, and S), one PAPS binding domain at the N-terminus, and one PAPS binding and dimerization domain at the C-terminus. The spatial-temporal expression patterns of ScSULT1B1s were further estimated by quantitative real-time PCR (qRT-PCR). Among them, partial ScSULT1B1s showed significantly high expression in the gill, hepatopancreas, and siphon. Furthermore, the response expression of certain ScSULT1B1s significantly fluctuated under sulfide stress. Together, our results suggest that ScSULT1B1s, by mediating the sulfonation reaction, may regulate THs levels to maintain basic metabolic and immune functions, making S. constricta highly sulfide tolerant.
Collapse
|
10
|
Rhodes AML, Ali S, Minnion M, Lee LH, Joseph BM, Ndzo J, Clarke NMP, Feelisch M, Aarvold A. An Explorative Study into the Aetiology of Developmental Dysplasia of the Hip Using Targeted Urine Metabolomics. Antioxidants (Basel) 2023; 12:antiox12030538. [PMID: 36978785 PMCID: PMC10045260 DOI: 10.3390/antiox12030538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 02/09/2023] [Accepted: 02/17/2023] [Indexed: 02/23/2023] Open
Abstract
Developmental dysplasia of the hip (DDH) is the most prevalent congenital musculoskeletal disorder, yet its cause remains unknown. Adequate nutrient provision and coordinated electron exchange (redox) processes are critical for foetal growth and tissue development. This novel study sought to explore specific biochemical pathways in skeletal development for potential involvement in the aetiology of DDH. Spot urine samples were collected from infants, aged 13–61 days, with and without DDH. Ion chromatography-mass spectrometry was used to quantify thiosulphate, sulphate, nitrate, and phosphate, whilst nitrite was quantified using high-performance liquid chromato-graphy. Thiobarbituric acid reactive substances (TBARS) were measured as markers of lipid peroxidation. Creatinine and osmolality were determined by a 96-well plate assay and micro-osmometer to potentially normalise values for renal function, lean body mass, and hydration status. Urine samples were analysed from 99 babies: 30 with DDH and 69 age-matched non-DDH controls. Thiosulphate, TBARS, and creatinine concentrations differed between the DDH group and the controls (p = 0.025, 0.015, and 0.004 respectively). Urine osmolality was significantly lower in DDH compared to the controls (p = 0.036), indicative of the production of a more diluted urine in DDH infants. Following adjustment for osmolality, significant differences became apparent in urinary sulphate levels in DDH (p = 0.035) whereas all other parameters were similar between the groups. This is the first study to assess the potential role of these inorganic anions in DDH. The higher levels of sulphate found in infants with DDH suggests either enhanced intake from milk, increased endogenous formation, or impaired renal reabsorption. This investigation demonstrates the power of urine metabolomics and highlights the importance of normalisation for hydration status to disentangle developmental disorders. Our results strongly suggest that DDH is a systemic disease associated with altered uptake, formation, or handling of sulphate. There is potential for new opportunities in the prevention or treatment of DDH via nutritional intervention.
Collapse
Affiliation(s)
- Amanda M. L. Rhodes
- Orthopaedic Surgery, University Hospital Southampton, Southampton SO16 6YD, UK
| | - Sehrish Ali
- Clinical and Experimental Sciences, Faculty of Medicine, University Hospital Southampton, Southampton SO16 6YD, UK
| | - Magdalena Minnion
- Clinical and Experimental Sciences, Faculty of Medicine, University Hospital Southampton, Southampton SO16 6YD, UK
| | - Ling H. Lee
- Southampton Children’s Hospital, University Hospital Southampton, Southampton SO16 6YD, UK
| | - Brijil M. Joseph
- Southampton Children’s Hospital, University Hospital Southampton, Southampton SO16 6YD, UK
| | - Judwin Ndzo
- Southampton Children’s Hospital, University Hospital Southampton, Southampton SO16 6YD, UK
| | - Nicholas M. P. Clarke
- Department of Paediatric Orthopaedics, University of Southampton, Southampton SO16 6YD, UK
| | - Martin Feelisch
- Clinical and Experimental Sciences, Faculty of Medicine, University Hospital Southampton, Southampton SO16 6YD, UK
- Correspondence: (M.F.); (A.A.)
| | - Alexander Aarvold
- Southampton Children’s Hospital, University Hospital Southampton, Southampton SO16 6YD, UK
- Correspondence: (M.F.); (A.A.)
| |
Collapse
|
11
|
Pfau A, López-Cayuqueo KI, Scherer N, Wuttke M, Wernstedt A, González Fassrainer D, Smith DE, van de Kamp JM, Ziegeler K, Eckardt KU, Luft FC, Aronson PS, Köttgen A, Jentsch TJ, Knauf F. SLC26A1 is a major determinant of sulfate homeostasis in humans. J Clin Invest 2023; 133:e161849. [PMID: 36719378 PMCID: PMC9888379 DOI: 10.1172/jci161849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Accepted: 11/29/2022] [Indexed: 02/01/2023] Open
Abstract
Sulfate plays a pivotal role in numerous physiological processes in the human body, including bone and cartilage health. A role of the anion transporter SLC26A1 (Sat1) for sulfate reabsorption in the kidney is supported by the observation of hyposulfatemia and hypersulfaturia in Slc26a1-knockout mice. The impact of SLC26A1 on sulfate homeostasis in humans remains to be defined. By combining clinical genetics, functional expression assays, and population exome analysis, we identify SLC26A1 as a sulfate transporter in humans and experimentally validate several loss-of-function alleles. Whole-exome sequencing from a patient presenting with painful perichondritis, hyposulfatemia, and renal sulfate wasting revealed a homozygous mutation in SLC26A1, which has not been previously described to the best of our knowledge. Whole-exome data analysis of more than 5,000 individuals confirmed that rare, putatively damaging SCL26A1 variants were significantly associated with lower plasma sulfate at the population level. Functional expression assays confirmed a substantial reduction in sulfate transport for the SLC26A1 mutation of our patient, which we consider to be novel, as well as for the additional variants detected in the population study. In conclusion, combined evidence from 3 complementary approaches supports SLC26A1 activity as a major determinant of sulfate homeostasis in humans. In view of recent evidence linking sulfate homeostasis with back pain and intervertebral disc disorder, our study identifies SLC26A1 as a potential target for modulation of musculoskeletal health.
Collapse
Affiliation(s)
- Anja Pfau
- Department of Nephrology and Medical Intensive Care, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Karen I. López-Cayuqueo
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP) and Max-Delbrück-Centrum für Molekulare Medizin (MDC), Berlin, Germany
| | - Nora Scherer
- Institute of Genetic Epidemiology, Faculty of Medicine and Medical Center and
- Spemann Graduate School of Biology and Medicine (SGBM), University of Freiburg, Freiburg, Germany
| | - Matthias Wuttke
- Institute of Genetic Epidemiology, Faculty of Medicine and Medical Center and
| | | | | | - Desiree E.C. Smith
- Metabolic Laboratory, Department of Clinical Chemistry, Amsterdam Neuroscience and
| | - Jiddeke M. van de Kamp
- Department of Human Genetics, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Katharina Ziegeler
- Department of Radiology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Kai-Uwe Eckardt
- Department of Nephrology and Medical Intensive Care, Charité-Universitätsmedizin Berlin, Berlin, Germany
- Department of Nephrology and Hypertension, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Friedrich C. Luft
- Department of Nephrology and Medical Intensive Care, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Peter S. Aronson
- Department of Internal Medicine, Section of Nephrology, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Anna Köttgen
- Institute of Genetic Epidemiology, Faculty of Medicine and Medical Center and
- CIBSS – Centre for Integrative Biological Signalling Studies, Albert-Ludwigs-University Freiburg, Freiburg, Germany
| | - Thomas J. Jentsch
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP) and Max-Delbrück-Centrum für Molekulare Medizin (MDC), Berlin, Germany
- NeuroCure Cluster of Excellence, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Felix Knauf
- Department of Nephrology and Medical Intensive Care, Charité-Universitätsmedizin Berlin, Berlin, Germany
- Department of Internal Medicine, Section of Nephrology, Yale University School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
12
|
van de Kamp JM, Bökenkamp A, Smith DEC, Wamelink MMC, Jansen EEW, Struys EA, Waisfisz Q, Verkleij M, Hartmann MF, Wang R, Wudy SA, Paganini C, Rossi A, Finken MJJ. Biallelic variants in the SLC13A1 sulfate transporter gene cause hyposulfatemia with a mild spondylo-epi-metaphyseal dysplasia. Clin Genet 2023; 103:45-52. [PMID: 36175384 PMCID: PMC10092256 DOI: 10.1111/cge.14239] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 09/01/2022] [Accepted: 09/25/2022] [Indexed: 12/14/2022]
Abstract
Sulfate is the fourth most abundant anion in human plasma but is not measured in clinical practice and little is known about the consequences of sulfate deficiency. Nevertheless, sulfation plays an essential role in the modulation of numerous compounds, including proteoglycans and steroids. We report the first patient with a homozygous loss-of-function variant in the SLC13A1 gene, encoding a renal and intestinal sulfate transporter, which is essential for maintaining plasma sulfate levels. The homozygous (Arg12Ter) variant in SLC13A1 was found by exome sequencing performed in a patient with unexplained skeletal dysplasia. The main clinical features were enlargement of joints and spondylo-epi-metaphyseal radiological abnormalities in early childhood, which improved with age. In addition, autistic features were noted. We found profound hyposulfatemia due to complete loss of renal sulfate reabsorption. Cholesterol sulfate was reduced. Intravenous N-acetylcysteine administration temporarily restored plasma sulfate levels. We conclude that loss of the SLC13A1 gene leads to profound hypersulfaturia and hyposulfatemia, which is mainly associated with abnormal skeletal development, possibly predisposing to degenerative bone and joint disease. The diagnosis might be easily missed and more frequent.
Collapse
Affiliation(s)
- Jiddeke M van de Kamp
- Department of Human Genetics, Amsterdam UMC location Vrije Universiteit Amsterdam, Amsterdam, The Netherlands.,Amsterdam Reproduction and Development, Amsterdam, The Netherlands.,Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam, The Netherlands
| | - Arend Bökenkamp
- Emma Children's Hospital, Amsterdam UMC location Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Desiree E C Smith
- Department of Clinical Chemistry, Metabolic Laboratory, Amsterdam UMC location Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Mirjam M C Wamelink
- Department of Clinical Chemistry, Metabolic Laboratory, Amsterdam UMC location Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Erwin E W Jansen
- Department of Clinical Chemistry, Metabolic Laboratory, Amsterdam UMC location Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Eduard A Struys
- Department of Clinical Chemistry, Metabolic Laboratory, Amsterdam UMC location Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Quinten Waisfisz
- Department of Human Genetics, Amsterdam UMC location Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Marieke Verkleij
- Department of Pediatric Psychology, Amsterdam UMC location Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Michaela F Hartmann
- Steroid Research & Mass Spectrometry Unit, Laboratory for Translational Hormone Analytics in Pediatric Endocrinology, Division of Pediatric Endocrinology & Diabetology, Center of Child and Adolescent Medicine, Justus Liebig University, Giessen, Germany
| | - Rong Wang
- Steroid Research & Mass Spectrometry Unit, Laboratory for Translational Hormone Analytics in Pediatric Endocrinology, Division of Pediatric Endocrinology & Diabetology, Center of Child and Adolescent Medicine, Justus Liebig University, Giessen, Germany
| | - Stefan A Wudy
- Steroid Research & Mass Spectrometry Unit, Laboratory for Translational Hormone Analytics in Pediatric Endocrinology, Division of Pediatric Endocrinology & Diabetology, Center of Child and Adolescent Medicine, Justus Liebig University, Giessen, Germany
| | - Chiara Paganini
- Department of Molecular Medicine, Unit of Biochemistry, University of Pavia, Pavia, Italy
| | - Antonio Rossi
- Department of Molecular Medicine, Unit of Biochemistry, University of Pavia, Pavia, Italy
| | - Martijn J J Finken
- Emma Children's Hospital, Amsterdam UMC location Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
13
|
Atcheson RJ, Burne THJ, Dawson PA. Serum sulfate level and Slc13a1 mRNA expression remain unaltered in a mouse model of moderate vitamin D deficiency. Mol Cell Biochem 2022:10.1007/s11010-022-04634-7. [PMID: 36566486 DOI: 10.1007/s11010-022-04634-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 12/05/2022] [Indexed: 12/26/2022]
Abstract
Sulfate is essential for healthy foetal growth and neurodevelopment. The SLC13A1 sulfate transporter is primarily expressed in the kidney where it mediates sulfate reabsorption and maintains circulating sulfate levels. To meet foetal demands, maternal sulfate levels increase by twofold in pregnancy via upregulated SLC13A1 expression. Previous studies found hyposulfataemia and reduced renal Slc13a1 mRNA expression in rodent models with either severe vitamin D deficiency or perturbed vitamin D signalling. Here we investigated a mouse model of moderate vitamin D deficiency. However, serum sulfate level and renal Slc13a1 mRNA expression was not decreased by a moderate reduction in circulating vitamin D level. We confirmed that the mouse Slc13a1 5'-flanking region was upregulated by 1,25(OH)2D3 using luciferase assays in a cultured renal OK cell line. These results support the presence of a functional VDRE in the mouse Slc13a1 but suggests that moderate vitamin D deficiency does not impact on sulfate homeostasis. As sulfate biology is highly conserved between rodents and humans, we proposed that human SLC13A1 would be under similar transcriptional regulation by 1,25(OH)2D3. Using an online prediction tool we identified a putative VDRE in the SLC13A1 5'-flanking region but unlike the mouse Slc13a1 sequence, the human sequence did not confer a significant response to 1,25(OH)2D3 in vitro. Overall, this study suggests that moderate vitamin D deficiency may not alter sulfate homeostasis. This needs to be confirmed in humans, particularly during pregnancy when vitamin D and sulfate levels need to be maintained at high levels for healthy maternal and child outcomes.
Collapse
Affiliation(s)
- Ranita J Atcheson
- Mater Research Institute, The University of Queensland, Translational Research Institute, 37 Kent St, Woolloongabba, QLD, 4102, Australia
| | - Thomas H J Burne
- Queensland Brain Institute, The University of Queensland, St. Lucia, QLD, 4072, Australia.,Queensland Centre for Mental Health Research, The Park Centre for Mental Health, Wacol, QLD, 4076, Australia
| | - Paul A Dawson
- Mater Research Institute, The University of Queensland, Translational Research Institute, 37 Kent St, Woolloongabba, QLD, 4102, Australia.
| |
Collapse
|
14
|
Tzertzinis G, Baker B, Benner J, Brown E, Corrêa IR, Ettwiller L, McClung C, Schildkraut I. Coelenterazine sulfotransferase from Renilla muelleri. PLoS One 2022; 17:e0276315. [PMID: 36251663 PMCID: PMC9576082 DOI: 10.1371/journal.pone.0276315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Accepted: 10/04/2022] [Indexed: 11/19/2022] Open
Abstract
The luciferin sulfokinase (coelenterazine sulfotransferase) of Renilla was previously reported to activate the storage form, luciferyl sulfate (coelenterazine sulfate) to luciferin (coelenterazine), the substrate for the luciferase bioluminescence reaction. The gene coding for the coelenterazine sulfotransferase has not been identified. Here we used a combined proteomic/transcriptomic approach to identify and clone the sulfotransferase cDNA. Multiple isoforms of coelenterazine sulfotransferase were identified from the anthozoan Renilla muelleri by intersecting its transcriptome with the LC-MS/MS derived peptide sequences of coelenterazine sulfotransferase purified from Renilla. Two of the isoforms were expressed in E. coli, purified, and partially characterized. The encoded enzymes display sulfotransferase activity that is comparable to that of the native sulfotransferase isolated from Renilla reniformis that was reported in 1970. The bioluminescent assay for sensitive detection of 3'-phosphoadenosine 5'-phosphate (PAP) using the recombinant sulfotransferase is demonstrated.
Collapse
Affiliation(s)
- George Tzertzinis
- New England Biolabs, Ipswich, Massachusetts, United States of America
| | - Brenda Baker
- New England Biolabs, Ipswich, Massachusetts, United States of America
| | - Jack Benner
- New England Biolabs, Ipswich, Massachusetts, United States of America
| | - Elizabeth Brown
- New England Biolabs, Ipswich, Massachusetts, United States of America
| | - Ivan R. Corrêa
- New England Biolabs, Ipswich, Massachusetts, United States of America
| | | | - Colleen McClung
- New England Biolabs, Ipswich, Massachusetts, United States of America
| | - Ira Schildkraut
- New England Biolabs, Ipswich, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
15
|
Sorrell JM, Caplan AI. Heparan Sulfate: A Regulator of White Adipocyte Differentiation and of Vascular/Adipocyte Interactions. Biomedicines 2022; 10:biomedicines10092115. [PMID: 36140217 PMCID: PMC9495464 DOI: 10.3390/biomedicines10092115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2022] [Revised: 08/16/2022] [Accepted: 08/20/2022] [Indexed: 11/30/2022] Open
Abstract
White adipose tissues are major endocrine organs that release factors, termed adipokines, which affect other major organ systems. The development and functions of adipose tissues depend largely upon the glycosaminoglycan heparan sulfate. Heparan sulfate proteoglycans (HSPGs) surround both adipocytes and vascular structures and facilitate the communication between these two components. This communication mediates the continued export of adipokines from adipose tissues. Heparan sulfates regulate cellular physiology and communication through a sulfation code that ionically interacts with heparan-binding regions on a select set of proteins. Many of these proteins are growth factors and chemokines that regulate tissue function and inflammation. Cells regulate heparan sulfate sulfation through the release of heparanases and sulfatases. It is now possible to tissue engineer vascularized adipose tissues that express heparan sulfate proteoglycans. This makes it possible to use these tissue constructs to study the role of heparan sulfates in the regulation of adipokine production and release. It is possible to regulate the production of heparanases and sulfatases in order to fine-tune experimental studies.
Collapse
|
16
|
Simultaneous Quantification of Steroid Hormones Using hrLC-MS in Endocrine Tissues of Male Rats and Human Samples. Metabolites 2022; 12:metabo12080714. [PMID: 36005586 PMCID: PMC9414922 DOI: 10.3390/metabo12080714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 07/22/2022] [Accepted: 07/28/2022] [Indexed: 12/03/2022] Open
Abstract
Steroid hormones play a vital role in the regulation of cellular processes, and dysregulation of these metabolites can provoke or aggravate pathological issues, such as autoimmune diseases and cancer. Regulation of steroid hormones involves different organs and biological compartments. Therefore, it is important to accurately determine their levels in tissues and biofluids to monitor changes after challenge or during disease. In this work, we have developed and optimized the extraction and quantification of 11 key members of the different steroid classes, including androgens, estrogens, progestogens and corticoids. The assay consists of a liquid/liquid extraction step and subsequent quantification by high-resolution liquid chromatography coupled time-of-flight mass spectrometry. The recoveries range between 74.2 to 126.9% and 54.9 to 110.7%, using a cell culture or urine as matrix, respectively. In general, the signal intensity loss due to matrix effect is no more than 30%. The method has been tested in relevant steroidogenic tissues in rat models and it has also been tested in human urine samples. Overall, this assay measures 11 analytes simultaneously in 6 min runtime and it has been applied in adrenal gland, testis, prostate, brain and serum from rats, and urine and extracellular vesicles from humans.
Collapse
|
17
|
Pedersen LC, Yi M, Pedersen LG, Kaminski AM. From Steroid and Drug Metabolism to Glycobiology, Using Sulfotransferase Structures to Understand and Tailor Function. Drug Metab Dispos 2022; 50:1027-1041. [PMID: 35197313 PMCID: PMC10753775 DOI: 10.1124/dmd.121.000478] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Accepted: 12/06/2021] [Indexed: 11/22/2022] Open
Abstract
Sulfotransferases are ubiquitous enzymes that transfer a sulfo group from the universal cofactor donor 3'-phosphoadenosine 5'-phosphosulfate to a broad range of acceptor substrates. In humans, the cytosolic sulfotransferases are involved in the sulfation of endogenous compounds such as steroids, neurotransmitters, hormones, and bile acids as well as xenobiotics including drugs, toxins, and environmental chemicals. The Golgi associated membrane-bound sulfotransferases are involved in post-translational modification of macromolecules from glycosaminoglycans to proteins. The sulfation of small molecules can have profound biologic effects on the functionality of the acceptor, including activation, deactivation, or enhanced metabolism and elimination. Sulfation of macromolecules has been shown to regulate a number of physiologic and pathophysiological pathways by enhancing binding affinity to regulatory proteins or binding partners. Over the last 25 years, crystal structures of these enzymes have provided a wealth of information on the mechanisms of this process and the specificity of these enzymes. This review will focus on the general commonalities of the sulfotransferases, from enzyme structure to catalytic mechanism as well as providing examples into how structural information is being used to either design drugs that inhibit sulfotransferases or to modify the enzymes to improve drug synthesis. SIGNIFICANCE STATEMENT: This manuscript honors Dr. Masahiko Negishi's contribution to the understanding of sulfotransferase mechanism, specificity, and roles in biology by analyzing the crystal structures that have been solved over the last 25 years.
Collapse
Affiliation(s)
- Lars C Pedersen
- Genome Integrity and Structural Biology Laboratory (L.C.P., L.G.P., A.M.K.) and Reproductive and Developmental Biology Laboratory (M.Y.), National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina; and Department of Chemistry, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (L.G.P.)
| | - MyeongJin Yi
- Genome Integrity and Structural Biology Laboratory (L.C.P., L.G.P., A.M.K.) and Reproductive and Developmental Biology Laboratory (M.Y.), National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina; and Department of Chemistry, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (L.G.P.)
| | - Lee G Pedersen
- Genome Integrity and Structural Biology Laboratory (L.C.P., L.G.P., A.M.K.) and Reproductive and Developmental Biology Laboratory (M.Y.), National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina; and Department of Chemistry, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (L.G.P.)
| | - Andrea M Kaminski
- Genome Integrity and Structural Biology Laboratory (L.C.P., L.G.P., A.M.K.) and Reproductive and Developmental Biology Laboratory (M.Y.), National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina; and Department of Chemistry, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (L.G.P.)
| |
Collapse
|
18
|
Clarke T, Fernandez FE, Dawson PA. Sulfation Pathways During Neurodevelopment. Front Mol Biosci 2022; 9:866196. [PMID: 35495624 PMCID: PMC9047184 DOI: 10.3389/fmolb.2022.866196] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Accepted: 03/24/2022] [Indexed: 01/27/2023] Open
Abstract
Sulfate is an important nutrient that modulates a diverse range of molecular and cellular functions in mammalian physiology. Over the past 2 decades, animal studies have linked numerous sulfate maintenance genes with neurological phenotypes, including seizures, impaired neurodevelopment, and behavioral abnormalities. Despite sulfation pathways being highly conserved between humans and animals, less than one third of all known sulfate maintenance genes are clinically reportable. In this review, we curated the temporal and spatial expression of 91 sulfate maintenance genes in human fetal brain from 4 to 17 weeks post conception using the online Human Developmental Biology Resource Expression. In addition, we performed a systematic search of PubMed and Embase, identifying those sulfate maintenance genes linked to atypical neurological phenotypes in humans and animals. Those findings, together with a search of the Online Mendelian Inheritance in Man database, identified a total of 18 candidate neurological dysfunction genes that are not yet considered in clinical settings. Collectively, this article provides an overview of sulfate biology genes to inform future investigations of perturbed sulfate homeostasis associated with neurological conditions.
Collapse
Affiliation(s)
- Taylor Clarke
- School of Behavioural and Health Sciences, Faculty of Health Sciences, Australian Catholic University, Banyo, QLD, Australia
| | - Francesca E. Fernandez
- School of Behavioural and Health Sciences, Faculty of Health Sciences, Australian Catholic University, Banyo, QLD, Australia
| | - Paul A. Dawson
- Mater Research Institute, University of Queensland, Brisbane, QLD, Australia
- *Correspondence: Paul A. Dawson,
| |
Collapse
|
19
|
Kaczor-Kamińska M, Kamiński K, Wróbel M. Heparan Sulfate, Mucopolysaccharidosis IIIB and Sulfur Metabolism Disorders. Antioxidants (Basel) 2022; 11:antiox11040678. [PMID: 35453363 PMCID: PMC9026333 DOI: 10.3390/antiox11040678] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 03/24/2022] [Accepted: 03/28/2022] [Indexed: 02/01/2023] Open
Abstract
Mucopolysaccharidosis, type IIIB (MPS IIIB) is a rare disease caused by mutations in the N-alpha-acetylglucosaminidase (NAGLU) gene resulting in decreased or absent enzyme activity. On the cellular level, the disorder is characterized by the massive lysosomal storage of heparan sulfate (HS)—one species of glycosaminoglycans. HS is a sulfur-rich macromolecule, and its accumulation should affect the turnover of total sulfur in cells; according to the studies presented here, it, indeed, does. The lysosomal degradation of HS in cells produces monosaccharides and inorganic sulfate (SO42−). Sulfate is a product of L-cysteine metabolism, and any disruption of its levels affects the entire L-cysteine catabolism pathway, which was first reported in 2019. It is known that L-cysteine level is elevated in cells with the Naglu−/− gene mutation and in selected tissues of individuals with MPS IIIB. The level of glutathione and the Naglu−/− cells’ antioxidant potential are significantly reduced, as well as the activity of 3-mercaptopyruvate sulfurtransferase (MPST, EC 2.8.1.2) and the level of sulfane sulfur-containing compounds. The direct reason is not yet known. This paper attempts to identify some of cause-and-effect correlations that may lead to this condition and identifies research directions that should be explored.
Collapse
Affiliation(s)
- Marta Kaczor-Kamińska
- Chair of Medical Biochemistry, Faculty of Medicine, Jagiellonian University Medical College, 7 Kopernika St., 31-034 Krakow, Poland;
- Correspondence: ; Tel.: +48-12-422-7400
| | - Kamil Kamiński
- Department of Physical Chemistry, Faculty of Chemistry, Jagiellonian University, 2 Gronostajowa St., 30-387 Krakow, Poland;
| | - Maria Wróbel
- Chair of Medical Biochemistry, Faculty of Medicine, Jagiellonian University Medical College, 7 Kopernika St., 31-034 Krakow, Poland;
| |
Collapse
|
20
|
Igreja C, Sommer RJ. The Role of Sulfation in Nematode Development and Phenotypic Plasticity. Front Mol Biosci 2022; 9:838148. [PMID: 35223994 PMCID: PMC8869759 DOI: 10.3389/fmolb.2022.838148] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 01/24/2022] [Indexed: 12/25/2022] Open
Abstract
Sulfation is poorly understood in most invertebrates and a potential role of sulfation in the regulation of developmental and physiological processes of these organisms remains unclear. Also, animal model system approaches did not identify many sulfation-associated mechanisms, whereas phosphorylation and ubiquitination are regularly found in unbiased genetic and pharmacological studies. However, recent work in the two nematodes Caenorhabditis elegans and Pristionchus pacificus found a role of sulfatases and sulfotransferases in the regulation of development and phenotypic plasticity. Here, we summarize the current knowledge about the role of sulfation in nematodes and highlight future research opportunities made possible by the advanced experimental toolkit available in these organisms.
Collapse
Affiliation(s)
- Catia Igreja
- *Correspondence: Catia Igreja, ; Ralf J. Sommer,
| | | |
Collapse
|
21
|
Bjornsdottir G, Stefansdottir L, Thorleifsson G, Sulem P, Norland K, Ferkingstad E, Oddsson A, Zink F, Lund SH, Nawaz MS, Bragi Walters G, Skuladottir AT, Gudjonsson SA, Einarsson G, Halldorsson GH, Bjarnadottir V, Sveinbjornsson G, Helgadottir A, Styrkarsdottir U, Gudmundsson LJ, Pedersen OB, Hansen TF, Werge T, Banasik K, Troelsen A, Skou ST, Thørner LW, Erikstrup C, Nielsen KR, Mikkelsen S, Jonsdottir I, Bjornsson A, Olafsson IH, Ulfarsson E, Blondal J, Vikingsson A, Brunak S, Ostrowski SR, Ullum H, Thorsteinsdottir U, Stefansson H, Gudbjartsson DF, Thorgeirsson TE, Stefansson K. Rare SLC13A1 variants associate with intervertebral disc disorder highlighting role of sulfate in disc pathology. Nat Commun 2022; 13:634. [PMID: 35110524 PMCID: PMC8810832 DOI: 10.1038/s41467-022-28167-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Accepted: 01/12/2022] [Indexed: 12/12/2022] Open
Abstract
Back pain is a common and debilitating disorder with largely unknown underlying biology. Here we report a genome-wide association study of back pain using diagnoses assigned in clinical practice; dorsalgia (119,100 cases, 909,847 controls) and intervertebral disc disorder (IDD) (58,854 cases, 922,958 controls). We identify 41 variants at 33 loci. The most significant association (ORIDD = 0.92, P = 1.6 × 10-39; ORdorsalgia = 0.92, P = 7.2 × 10-15) is with a 3'UTR variant (rs1871452-T) in CHST3, encoding a sulfotransferase enzyme expressed in intervertebral discs. The largest effects on IDD are conferred by rare (MAF = 0.07 - 0.32%) loss-of-function (LoF) variants in SLC13A1, encoding a sodium-sulfate co-transporter (LoF burden OR = 1.44, P = 3.1 × 10-11); variants that also associate with reduced serum sulfate. Genes implicated by this study are involved in cartilage and bone biology, as well as neurological and inflammatory processes.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Muhammad S Nawaz
- deCODE Genetics/Amgen, Inc., Reykjavik, Iceland
- Faculty of Medicine, School of Health Sciences, University of Iceland, Reykjavik, Iceland
| | - G Bragi Walters
- deCODE Genetics/Amgen, Inc., Reykjavik, Iceland
- Faculty of Medicine, School of Health Sciences, University of Iceland, Reykjavik, Iceland
| | | | | | | | - Gisli H Halldorsson
- deCODE Genetics/Amgen, Inc., Reykjavik, Iceland
- School of Engineering and Natural Sciences, University of Iceland, Reykjavik, Iceland
| | | | | | | | | | | | - Ole B Pedersen
- Department of Clinical Immunology, Zealand University Hospital, Køge, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Thomas Folkmann Hansen
- Danish Headache Center, Dept. Neurology, Rigshospitalet-Glostrup, Glostrup, Denmark
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Thomas Werge
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Institute of Biological Psychiatry, Mental Health Services, Copenhagen University Hospital, Copenhagen, Denmark
- Lundbeck Foundation for GeoGenetics, GLOBE Institute, University of Copenhagen, Copenhagen, Denmark
| | - Karina Banasik
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Anders Troelsen
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Orthopaedic Surgery, CAG ROAD-Research OsteoArthritis Denmark, Copenhagen University Hospital, Hvidovre, Denmark
| | - Soren T Skou
- Research Unit for Musculoskeletal Function and Physiotherapy, Department of Sports Science and Clinical Biomechanics, University of Southern Denmark, Odense, Denmark
- The Research Unit PROgrez, Department of Physiotherapy and Occupational Therapy, Næstved-Slagelse-Ringsted Hospitals, Næstved, Denmark
| | - Lise Wegner Thørner
- Department of Clinical Immunology, Copenhagen University Hospital, Copenhagen, Denmark
| | - Christian Erikstrup
- Department of Clinical Immunology, Aarhus University Hospital, Aarhus, Denmark
| | - Kaspar Rene Nielsen
- Department of Clinical Immunology, Aalborg University Hospital, Aalborg, Denmark
| | - Susan Mikkelsen
- Department of Clinical Immunology, Aarhus University Hospital, Aarhus, Denmark
| | - Ingileif Jonsdottir
- deCODE Genetics/Amgen, Inc., Reykjavik, Iceland
- Faculty of Medicine, School of Health Sciences, University of Iceland, Reykjavik, Iceland
| | - Aron Bjornsson
- Department of Neurosurgery, Landspitali University Hospital, Reykjavik, Iceland
| | - Ingvar H Olafsson
- Department of Neurosurgery, Landspitali University Hospital, Reykjavik, Iceland
| | - Elfar Ulfarsson
- Department of Neurosurgery, Landspitali University Hospital, Reykjavik, Iceland
| | - Josep Blondal
- Health Care Institution of West Iceland, Stykkisholmur, Iceland
| | | | - Soren Brunak
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Sisse R Ostrowski
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Clinical Immunology, Copenhagen University Hospital, Copenhagen, Denmark
| | - Henrik Ullum
- Statens Serum Institut, Copenhagen, Copenhagen, Denmark
| | - Unnur Thorsteinsdottir
- deCODE Genetics/Amgen, Inc., Reykjavik, Iceland
- Faculty of Medicine, School of Health Sciences, University of Iceland, Reykjavik, Iceland
| | | | - Daniel F Gudbjartsson
- deCODE Genetics/Amgen, Inc., Reykjavik, Iceland
- School of Engineering and Natural Sciences, University of Iceland, Reykjavik, Iceland
| | | | - Kari Stefansson
- deCODE Genetics/Amgen, Inc., Reykjavik, Iceland.
- Faculty of Medicine, School of Health Sciences, University of Iceland, Reykjavik, Iceland.
| |
Collapse
|
22
|
Lane JDE, Greenwood WJH, Day VW, Jolliffe KA, Bowman-James K, Adriaenssens L. Bis[squaramido]ferrocenes as electrochemical sulfate receptors. NEW J CHEM 2022. [DOI: 10.1039/d2nj03951f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The bis[squaramido]ferrocene scaffold is introduced and shown to selectively bind and electrochemically report sulfate in competitive water/DMSO mixtures.
Collapse
Affiliation(s)
- Jakob D. E. Lane
- School of Chemistry, The University of Sydney, NSW, 2006, Australia
| | | | - Victor W. Day
- Department of Chemistry, University of Kansas, Lawrence, Kansas, 66045, USA
| | - Katrina A. Jolliffe
- School of Chemistry, The University of Sydney, NSW, 2006, Australia
- The University of Sydney Nano Institute (Sydney Nano), The University of Sydney, NSW, 2006, Australia
| | | | | |
Collapse
|
23
|
Sun L, Konstantinidi A, Ye Z, Nason R, Zhang Y, Büll C, Kahl-Knutson B, Hansen L, Leffler H, Vakhrushev SY, Yang Z, Clausen H, Narimatsu Y. Installation of O-glycan sulfation capacities in human HEK293 cells for display of sulfated mucins. J Biol Chem 2021; 298:101382. [PMID: 34954141 PMCID: PMC8789585 DOI: 10.1016/j.jbc.2021.101382] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 10/30/2021] [Accepted: 11/02/2021] [Indexed: 11/25/2022] Open
Abstract
The human genome contains at least 35 genes that encode Golgi sulfotransferases that function in the secretory pathway, where they are involved in decorating glycosaminoglycans, glycolipids, and glycoproteins with sulfate groups. Although a number of important interactions by proteins such as selectins, galectins, and sialic acid–binding immunoglobulin-like lectins are thought to mainly rely on sulfated O-glycans, our insight into the sulfotransferases that modify these glycoproteins, and in particular GalNAc-type O-glycoproteins, is limited. Moreover, sulfated mucins appear to accumulate in respiratory diseases, arthritis, and cancer. To explore further the genetic and biosynthetic regulation of sulfated O-glycans, here we expanded a cell-based glycan array in the human embryonic kidney 293 (HEK293) cell line with sulfation capacities. We stably engineered O-glycan sulfation capacities in HEK293 cells by site-directed knockin of sulfotransferase genes in combination with knockout of genes to eliminate endogenous O-glycan branching (core2 synthase gene GCNT1) and/or sialylation capacities in order to provide simplified substrates (core1 Galβ1–3GalNAcα1–O-Ser/Thr) for the introduced sulfotransferases. Expression of the galactose 3-O-sulfotransferase 2 in HEK293 cells resulted in sulfation of core1 and core2 O-glycans, whereas expression of galactose 3-O-sulfotransferase 4 resulted in sulfation of core1 only. We used the engineered cell library to dissect the binding specificity of galectin-4 and confirmed binding to the 3-O-sulfo-core1 O-glycan. This is a first step toward expanding the emerging cell-based glycan arrays with the important sulfation modification for display and production of glycoconjugates with sulfated O-glycans.
Collapse
Affiliation(s)
- Lingbo Sun
- Copenhagen Center for Glycomics, Departments of Cellular and Molecular Medicine and School of Dentistry, Faculty of Health Sciences, University of Copenhagen, Blegdamsvej 3, DK-2200 Copenhagen N, Denmark; Medical College of Yan'an University, Yan'an University, Yan'an, 716000, Shaanxi Province, China
| | - Andriana Konstantinidi
- Copenhagen Center for Glycomics, Departments of Cellular and Molecular Medicine and School of Dentistry, Faculty of Health Sciences, University of Copenhagen, Blegdamsvej 3, DK-2200 Copenhagen N, Denmark
| | - Zilu Ye
- Copenhagen Center for Glycomics, Departments of Cellular and Molecular Medicine and School of Dentistry, Faculty of Health Sciences, University of Copenhagen, Blegdamsvej 3, DK-2200 Copenhagen N, Denmark
| | - Rebecca Nason
- Copenhagen Center for Glycomics, Departments of Cellular and Molecular Medicine and School of Dentistry, Faculty of Health Sciences, University of Copenhagen, Blegdamsvej 3, DK-2200 Copenhagen N, Denmark
| | - Yuecheng Zhang
- Department of Biomedical Sciences, Faculty of Health and Society, Malmö University, Jan Waldenströms gata 25, 205 06 Malmö, Sweden
| | - Christian Büll
- Copenhagen Center for Glycomics, Departments of Cellular and Molecular Medicine and School of Dentistry, Faculty of Health Sciences, University of Copenhagen, Blegdamsvej 3, DK-2200 Copenhagen N, Denmark
| | - Barbro Kahl-Knutson
- Department of Laboratory Medicine, Section MIG, Lund University BMC-C1228b, Klinikgatan28, 221 84 Lund, Sweden
| | - Lars Hansen
- Copenhagen Center for Glycomics, Departments of Cellular and Molecular Medicine and School of Dentistry, Faculty of Health Sciences, University of Copenhagen, Blegdamsvej 3, DK-2200 Copenhagen N, Denmark
| | - Hakon Leffler
- Department of Laboratory Medicine, Section MIG, Lund University BMC-C1228b, Klinikgatan28, 221 84 Lund, Sweden
| | - Sergey Y Vakhrushev
- Copenhagen Center for Glycomics, Departments of Cellular and Molecular Medicine and School of Dentistry, Faculty of Health Sciences, University of Copenhagen, Blegdamsvej 3, DK-2200 Copenhagen N, Denmark
| | - Zhang Yang
- Copenhagen Center for Glycomics, Departments of Cellular and Molecular Medicine and School of Dentistry, Faculty of Health Sciences, University of Copenhagen, Blegdamsvej 3, DK-2200 Copenhagen N, Denmark
| | - Henrik Clausen
- Copenhagen Center for Glycomics, Departments of Cellular and Molecular Medicine and School of Dentistry, Faculty of Health Sciences, University of Copenhagen, Blegdamsvej 3, DK-2200 Copenhagen N, Denmark.
| | - Yoshiki Narimatsu
- Copenhagen Center for Glycomics, Departments of Cellular and Molecular Medicine and School of Dentistry, Faculty of Health Sciences, University of Copenhagen, Blegdamsvej 3, DK-2200 Copenhagen N, Denmark.
| |
Collapse
|
24
|
Eisele BS, Luka Z, Wu AJ, Yang F, Hale AT, York JD. Sulfation of glycosaminoglycans depends on the catalytic activity of lithium-inhibited phosphatase BPNT2 in vitro. J Biol Chem 2021; 297:101293. [PMID: 34634304 PMCID: PMC8551643 DOI: 10.1016/j.jbc.2021.101293] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 09/29/2021] [Accepted: 10/06/2021] [Indexed: 01/02/2023] Open
Abstract
Golgi-resident bisphosphate nucleotidase 2 (BPNT2) is a member of a family of magnesium-dependent, lithium-inhibited phosphatases that share a three-dimensional structural motif that directly coordinates metal binding to effect phosphate hydrolysis. BPNT2 catalyzes the breakdown of 3'-phosphoadenosine-5'-phosphate, a by-product of glycosaminoglycan (GAG) sulfation. KO of BPNT2 in mice leads to skeletal abnormalities because of impaired GAG sulfation, especially chondroitin-4-sulfation, which is critical for proper extracellular matrix development. Mutations in BPNT2 have also been found to underlie a chondrodysplastic disorder in humans. The precise mechanism by which the loss of BPNT2 impairs sulfation remains unclear. Here, we used mouse embryonic fibroblasts (MEFs) to test the hypothesis that the catalytic activity of BPNT2 is required for GAG sulfation in vitro. We show that a catalytic-dead Bpnt2 construct (D108A) does not rescue impairments in intracellular or secreted sulfated GAGs, including decreased chondroitin-4-sulfate, present in Bpnt2-KO MEFs. We also demonstrate that missense mutations in Bpnt2 adjacent to the catalytic site, which are known to cause chondrodysplasia in humans, recapitulate defects in overall GAG sulfation and chondroitin-4-sulfation in MEF cultures. We further show that treatment of MEFs with lithium (a common psychotropic medication) inhibits GAG sulfation and that this effect depends on the presence of BPNT2. Taken together, this work demonstrates that the catalytic activity of an enzyme potently inhibited by lithium can modulate GAG sulfation and therefore extracellular matrix composition, revealing new insights into lithium pharmacology.
Collapse
Affiliation(s)
- Brynna S Eisele
- Department of Biochemistry, Vanderbilt University, Nashville, Tennessee, USA; Department of Pharmacology, Vanderbilt University, Nashville, Tennessee, USA
| | - Zigmund Luka
- Department of Biochemistry, Vanderbilt University, Nashville, Tennessee, USA
| | - Alice J Wu
- Department of Biochemistry, Vanderbilt University, Nashville, Tennessee, USA
| | - Fei Yang
- Department of Biochemistry, Vanderbilt University, Nashville, Tennessee, USA
| | - Andrew T Hale
- Department of Biochemistry, Vanderbilt University, Nashville, Tennessee, USA
| | - John D York
- Department of Biochemistry, Vanderbilt University, Nashville, Tennessee, USA.
| |
Collapse
|
25
|
Thomsson KA, Vitiazeva V, Mateoiu C, Jin C, Liu J, Holgersson J, Weijdegård B, Sundfeldt K, Karlsson NG. Sulfation of O-glycans on Mucin-type Proteins From Serous Ovarian Epithelial Tumors. Mol Cell Proteomics 2021; 20:100150. [PMID: 34555499 PMCID: PMC8527052 DOI: 10.1016/j.mcpro.2021.100150] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 08/19/2021] [Accepted: 09/15/2021] [Indexed: 11/19/2022] Open
Abstract
Despite sulfated O-linked glycans being abundant on ovarian cancer (OC) glycoproteins, their regulation during cancer development and involvement in cancer pathogenesis remain unexplored. We characterized O-glycans carrying sulfation on galactose residues and compared their expression with defined sulfotransferases regulated during OC development. Desialylated sulfated oligosaccharides were released from acidic glycoproteins in the cyst fluid from one patient with a benign serous cyst and one patient with serous OC. Oligosaccharides characterized by LC-MSn were identified as core 1 and core 2 O-glycans up to the size of decamers and with 1 to 4 sulfates linked to GlcNAc residues and to C-3 and/or C-6 of Gal. To study the specificity of the potential ovarian sulfotransferases involved, Gal3ST2 (Gal-3S)-, Gal3ST4 (Gal-3S)-, and CHST1 (Gal-6S)-encoding expression plasmids were transfected individually into CHO cells also expressing the P-selectin glycoprotein ligand-1/mouse immunoglobulin G2b (PSGL-1/mIg G2b) fusion protein and the human core 2 transferase (GCNT1). Characterization of the PSGL-1/mIg G2b O-glycans showed that Gal3ST2 preferentially sulfated Gal on the C-6 branch of core 2 structures and Gal3ST4 preferred Gal on the C-3 branch independently if core-1 or -2. CHST1 sulfated Gal residues on both the C-3 (core 1/2) and C-6 branches of core 2 structures. Using serous ovarian tissue micro array, Gal3ST2 was found to be decreased in tissue classified as malignant compared with tissues classified as benign or borderline, with the lowest expression in poorly differentiated malignant tissue. Neither Gal3ST4 nor CHST1 was differentially expressed in benign, borderline, or malignant tissue, and there was no correlation between expression level and differentiation stage. The data displays a complex sulfation pattern of O-glycans on OC glycoproteins and that aggressiveness of the cancer is associated with a decreased expression of the Gal3ST2 transferase. Ovarian cancer tissue contains highly sulfated O-glycoproteins. Sulfation occurs on GlcNAc (6-position) and Gal (3- and 6-position). Sulfation of Gal can be mimicked recombinantly with selected sulfotransferase. The Gal3ST2 sulfotransferase level is lower in malignant cancer compared with benign.
Collapse
Affiliation(s)
- Kristina A Thomsson
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Varvara Vitiazeva
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Constantina Mateoiu
- Department of Clinical Pathology, Sahlgrenska University Hostpital, Gotenburg, Sweden
| | - Chunsheng Jin
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Jining Liu
- Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Jan Holgersson
- Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Birgitta Weijdegård
- Department of Obstetrics and Gynecology, Institute of Clinical Sciences, Sahlgrenska Center for Cancer Research, University of Gothenburg, Gothenburg, Sweden
| | - Karin Sundfeldt
- Department of Obstetrics and Gynecology, Institute of Clinical Sciences, Sahlgrenska Center for Cancer Research, University of Gothenburg, Gothenburg, Sweden
| | - Niclas G Karlsson
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden; Department of Life Sciences and Health, Faculty of Health Sciences, Oslo Metropolitan University, Oslo, Norway.
| |
Collapse
|
26
|
Zhou S, Xu X, Ma N, Jung F, Lendlein A. Influence of sterilization conditions on sulfate-functionalized polyGGE. Clin Hemorheol Microcirc 2021; 79:597-608. [PMID: 34420943 DOI: 10.3233/ch-211241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Sulfated biomolecules are known to influence numerous biological processes in all living organisms. Particularly, they contribute to prevent and inhibit the hypercoagulation condition. The failure of polymeric implants and blood contacting devices is often related to hypercoagulation and microbial contamination. Here, bioactive sulfated biomacromolecules are mimicked by sulfation of poly(glycerol glycidyl ether) (polyGGE) films. Autoclaving, gamma-ray irradiation and ethylene oxide (EtO) gas sterilization techniques were applied to functionalized materials. The sulfate group density and hydrophilicity of sulfated polymers were decreased while chain mobility and thermal degradation were enhanced post autoclaving when compared to those after EtO sterilization. These results suggest that a quality control after sterilization is mandatory to ensure the amount and functionality of functionalized groups are retained.
Collapse
Affiliation(s)
- Shuo Zhou
- Institute of Active Polymers, Helmholtz-Zentrum Hereon, Teltow, Germany.,Institute of Chemistry, University of Potsdam, Potsdam, Germany
| | - Xun Xu
- Institute of Active Polymers, Helmholtz-Zentrum Hereon, Teltow, Germany.,Berlin-Brandenburg Centre for Regenerative Therapies, Helmholtz-Zentrum Hereon, Teltow, Germany
| | - Nan Ma
- Institute of Active Polymers, Helmholtz-Zentrum Hereon, Teltow, Germany.,Berlin-Brandenburg Centre for Regenerative Therapies, Helmholtz-Zentrum Hereon, Teltow, Germany.,Institute of Chemistry and Biochemistry, Freie Universität Berlin, Berlin, Germany
| | - Friedrich Jung
- Institute of Active Polymers, Helmholtz-Zentrum Hereon, Teltow, Germany.,Berlin-Brandenburg Centre for Regenerative Therapies, Helmholtz-Zentrum Hereon, Teltow, Germany
| | - Andreas Lendlein
- Institute of Active Polymers, Helmholtz-Zentrum Hereon, Teltow, Germany.,Institute of Chemistry, University of Potsdam, Potsdam, Germany.,Berlin-Brandenburg Centre for Regenerative Therapies, Helmholtz-Zentrum Hereon, Teltow, Germany.,Institute of Chemistry and Biochemistry, Freie Universität Berlin, Berlin, Germany
| |
Collapse
|
27
|
Ren S, Wei Y, Niu M, Li R, Wang R, Wei S, Wen J, Wang D, Yang T, Chen X, Wu S, Tong Y, Jing M, Li H, Wang M, Zhao Y. Mechanism of rutaecarpine on ethanol-induced acute gastric ulcer using integrated metabolomics and network pharmacology. Biomed Pharmacother 2021; 138:111490. [PMID: 33773465 DOI: 10.1016/j.biopha.2021.111490] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 02/22/2021] [Accepted: 03/09/2021] [Indexed: 11/15/2022] Open
Abstract
This study was aimed to explore the mechanism of rutaecarpine (RUT) on ethanol-induced gastric ulcer (GU) in mice by integrated approaches. At first, the efficacy was determined through the macroscopic and microscopic state of stomach tissue and the expression levels of GU-related factors. Then, the serum metabolomics method based on UPLC-Q-TOF/MS was used to explore the specific metabolites and metabolic pathways. Finally, the upstream key protein targets of these specific metabolites were analyzed by network pharmacology and verified by PCR to explore the potential mechanism. RUT alleviated the histological and pathological damage of gastric tissue caused by ethanol, and could remarkably ameliorate the level of GU-related factors. Subsequently, a total of 7 potential metabolites involved in 9 metabolic pathways were identified by metabolomics analysis. Then, a 'component-targets-metabolites' interaction network was constructed, and therefore 4 key target proteins (PLA2G1B, PDE5A, MIF and SRC) that may regulate the specific metabolites were obtained. This case was further verified by the results of PCR. ALL the above results strongly demonstrated that RUT exerted a gastroprotective effect against GU. And it is the first time to combine metabolomics combined with network pharmacology to elucidate the mechanism of RUT on GU, which may be related to the regulation of energy metabolism, oxidative stress, and inflammation, and these pathways may be regulated through the upstream protein PLA2G1B, PDE5A, MIF and SRC.
Collapse
Affiliation(s)
- Sichen Ren
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; Department of Pharmacy, the Fifth Medical Center of Chinese PLA General Hospital, Beijing 100039, China
| | - Ying Wei
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; Department of Pharmacy, the Fifth Medical Center of Chinese PLA General Hospital, Beijing 100039, China
| | - Ming Niu
- Department of China Military Institute of Chinese Materia, the Fifth Medical Center of Chinese PLA General Hospital, Beijing 100039, China
| | - Ruisheng Li
- Research Center for Clinical and Translational Medicine, the Fifth Medical Center of Chinese PLA General Hospital, Beijing 100039, China
| | - Ruilin Wang
- Integrative Medical Center, the Fifth Medical Center of Chinese PLA General Hospital, Beijing 100039, China
| | - Shizhang Wei
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; Department of Pharmacy, the Fifth Medical Center of Chinese PLA General Hospital, Beijing 100039, China
| | - Jianxia Wen
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; Department of Pharmacy, the Fifth Medical Center of Chinese PLA General Hospital, Beijing 100039, China
| | - Dan Wang
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; Department of Pharmacy, the Fifth Medical Center of Chinese PLA General Hospital, Beijing 100039, China
| | - Tao Yang
- Department of Pharmacy, the Fifth Medical Center of Chinese PLA General Hospital, Beijing 100039, China; College of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China
| | - Xing Chen
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; Department of Pharmacy, the Fifth Medical Center of Chinese PLA General Hospital, Beijing 100039, China
| | - Shihua Wu
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; Department of Pharmacy, the Fifth Medical Center of Chinese PLA General Hospital, Beijing 100039, China
| | - Yuling Tong
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; Department of Pharmacy, the Fifth Medical Center of Chinese PLA General Hospital, Beijing 100039, China
| | - Manyi Jing
- Department of Pharmacy, the Fifth Medical Center of Chinese PLA General Hospital, Beijing 100039, China
| | - Haotian Li
- Department of Pharmacy, the Fifth Medical Center of Chinese PLA General Hospital, Beijing 100039, China
| | - Min Wang
- Department of Pharmacy, the Fifth Medical Center of Chinese PLA General Hospital, Beijing 100039, China
| | - Yanling Zhao
- Department of Pharmacy, the Fifth Medical Center of Chinese PLA General Hospital, Beijing 100039, China.
| |
Collapse
|
28
|
Baglia RA, Mills KR, Mitra K, Tutol JN, Ball D, Page KM, Kallu J, Gottipolu S, D'Arcy S, Nielsen SO, Dodani SC. An activity-based fluorescent sensor for the detection of the phenol sulfotransferase SULT1A1 in living cells. RSC Chem Biol 2021; 2:830-834. [PMID: 34212150 PMCID: PMC8190907 DOI: 10.1039/d0cb00231c] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Accepted: 03/04/2021] [Indexed: 11/21/2022] Open
Abstract
Human phenol sulfotransferases mediate the transfer of a sulfuryl moiety from the activated sulfate donor PAPS to hydroxy-containing substrates, altering substrate solubility and charge to affect phase II metabolism and cell signaling. Here, we present the development, computational modeling, in vitro enzymology, and biological application of STS-3, an activity-based fluorescent sensor for the SULT1A1 isoform.
Collapse
Affiliation(s)
- Regina A Baglia
- Department of Chemistry and Biochemistry, The University of Texas at Dallas Richardson TX 75080 USA
| | - Kira R Mills
- Department of Chemistry and Biochemistry, The University of Texas at Dallas Richardson TX 75080 USA
| | - Koushambi Mitra
- Department of Chemistry and Biochemistry, The University of Texas at Dallas Richardson TX 75080 USA
| | - Jasmine N Tutol
- Department of Chemistry and Biochemistry, The University of Texas at Dallas Richardson TX 75080 USA
| | - Darby Ball
- Department of Chemistry and Biochemistry, The University of Texas at Dallas Richardson TX 75080 USA
| | - Kierstin M Page
- Department of Chemistry and Biochemistry, The University of Texas at Dallas Richardson TX 75080 USA
| | - Jyothi Kallu
- Department of Chemistry and Biochemistry, The University of Texas at Dallas Richardson TX 75080 USA
| | - Sriharika Gottipolu
- Department of Chemistry and Biochemistry, The University of Texas at Dallas Richardson TX 75080 USA
| | - Sheena D'Arcy
- Department of Chemistry and Biochemistry, The University of Texas at Dallas Richardson TX 75080 USA
| | - Steven O Nielsen
- Department of Chemistry and Biochemistry, The University of Texas at Dallas Richardson TX 75080 USA
| | - Sheel C Dodani
- Department of Chemistry and Biochemistry, The University of Texas at Dallas Richardson TX 75080 USA
| |
Collapse
|
29
|
Paganini C, Gramegna Tota C, Superti-Furga A, Rossi A. Skeletal Dysplasias Caused by Sulfation Defects. Int J Mol Sci 2020; 21:ijms21082710. [PMID: 32295296 PMCID: PMC7216085 DOI: 10.3390/ijms21082710] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 04/09/2020] [Accepted: 04/10/2020] [Indexed: 12/18/2022] Open
Abstract
Proteoglycans (PGs) are macromolecules present on the cell surface and in the extracellular matrix that confer specific mechanical, biochemical, and physical properties to tissues. Sulfate groups present on glycosaminoglycans, linear polysaccharide chains attached to PG core proteins, are fundamental for correct PG functions. Indeed, through the negative charge of sulfate groups, PGs interact with extracellular matrix molecules and bind growth factors regulating tissue structure and cell behavior. The maintenance of correct sulfate metabolism is important in tissue development and function, particularly in cartilage where PGs are fundamental and abundant components of the extracellular matrix. In chondrocytes, the main sulfate source is the extracellular space, then sulfate is taken up and activated in the cytosol to the universal sulfate donor to be used in sulfotransferase reactions. Alteration in each step of sulfate metabolism can affect macromolecular sulfation, leading to the onset of diseases that affect mainly cartilage and bone. This review presents a panoramic view of skeletal dysplasias caused by mutations in genes encoding for transporters or enzymes involved in macromolecular sulfation. Future research in this field will contribute to the understanding of the disease pathogenesis, allowing the development of targeted therapies aimed at alleviating, preventing, or modifying the disease progression.
Collapse
Affiliation(s)
- Chiara Paganini
- Department of Molecular Medicine, Unit of Biochemistry, University of Pavia, 27100 Pavia, Italy; (C.P.); (C.G.T.)
| | - Chiara Gramegna Tota
- Department of Molecular Medicine, Unit of Biochemistry, University of Pavia, 27100 Pavia, Italy; (C.P.); (C.G.T.)
| | - Andrea Superti-Furga
- Division of Genetic Medicine, Lausanne University Hospital, University of Lausanne, 1011 Lausanne, Switzerland;
| | - Antonio Rossi
- Department of Molecular Medicine, Unit of Biochemistry, University of Pavia, 27100 Pavia, Italy; (C.P.); (C.G.T.)
- Correspondence:
| |
Collapse
|
30
|
Sebastiani F, Verde AV, Heyden M, Schwaab G, Havenith M. Cooperativity and ion pairing in magnesium sulfate aqueous solutions from the dilute regime to the solubility limit. Phys Chem Chem Phys 2020; 22:12140-12153. [DOI: 10.1039/c9cp06845g] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
A combined THz and simulation study on MgSO4 find no contact ion pairs in highly concentrated solutions.
Collapse
Affiliation(s)
| | - Ana Vila Verde
- Department of Theory & Bio-systems
- Max Planck Institute for Colloids and Interfaces
- Potsdam
- Germany
| | - Matthias Heyden
- School of Molecular Sciences
- Arizona State University
- Tempe
- USA
| | - Gerhard Schwaab
- Department of Physical Chemistry II
- Ruhr-University Bochum
- Bochum
- Germany
| | - Martina Havenith
- Department of Physical Chemistry II
- Ruhr-University Bochum
- Bochum
- Germany
| |
Collapse
|
31
|
Chen J, Wu JS, Mize T, Moreno M, Hamid M, Servin F, Bashy B, Zhao Z, Jia P, Tsuang MT, Kendler KS, Xiong M, Chen X. A Frameshift Variant in the CHST9 Gene Identified by Family-Based Whole Genome Sequencing Is Associated with Schizophrenia in Chinese Population. Sci Rep 2019; 9:12717. [PMID: 31481703 PMCID: PMC6722128 DOI: 10.1038/s41598-019-49052-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Accepted: 08/19/2019] [Indexed: 12/30/2022] Open
Abstract
Recent studies imply that rare variants contribute to the risk of schizophrenia, however, the exact variants or genes responsible for this condition are largely unknown. In this study, we conducted whole genome sequencing (WGS) of 20 Chinese families. Each family consisted of at least two affected siblings diagnosed with schizophrenia and at least one unaffected sibling. We examined functional variants that were found in affected sibling(s) but not in unaffected sibling(s) within a family. Matching this criterion, a frameshift heterozygous deletion of CA (–/CA) at chromosome 18:24722722, also referred to as rs752084147, in the Carbohydrate Sulfotransferase 9 (CHST9) gene, was detected in two families. This deletion was confirmed by PCR-based Sanger sequencing. With the observed frequency of 0.00076 in Han Chinese population, we performed both case-control and family-based analyses to evaluate its association with schizophrenia. In the case-control analyses, Chi-square test P-value was 6.80e-12 and the P-value was 0.0008 after one million simulations. In family-based segregation analyses, segregation P-value was 7.72e-7 and simulated P-value was 5.70e-6. For both the case-control and family-based analyses, the CA deletion was significantly associated with schizophrenia in the Chinese population. Further investigation of this gene is warranted in the development of schizophrenia by utilizing larger and more ethnically diverse samples.
Collapse
Affiliation(s)
- Jingchun Chen
- Nevada Institute of Personalized Medicine, University of Nevada, Las Vegas, 4505 S, Maryland Parkway, Las Vegas, NV, 89154-4009, USA.
| | - Jain-Shing Wu
- Nevada Institute of Personalized Medicine, University of Nevada, Las Vegas, 4505 S, Maryland Parkway, Las Vegas, NV, 89154-4009, USA
| | - Travis Mize
- Nevada Institute of Personalized Medicine, University of Nevada, Las Vegas, 4505 S, Maryland Parkway, Las Vegas, NV, 89154-4009, USA.,Department of Psychology, University of Nevada, Las Vegas, 4505 S, Maryland Parkway, Las Vegas, NV, 89154-4009, USA
| | - Marvi Moreno
- Nevada Institute of Personalized Medicine, University of Nevada, Las Vegas, 4505 S, Maryland Parkway, Las Vegas, NV, 89154-4009, USA
| | - Mahtab Hamid
- Nevada Institute of Personalized Medicine, University of Nevada, Las Vegas, 4505 S, Maryland Parkway, Las Vegas, NV, 89154-4009, USA
| | - Francisco Servin
- Nevada Institute of Personalized Medicine, University of Nevada, Las Vegas, 4505 S, Maryland Parkway, Las Vegas, NV, 89154-4009, USA
| | - Bita Bashy
- Nevada Institute of Personalized Medicine, University of Nevada, Las Vegas, 4505 S, Maryland Parkway, Las Vegas, NV, 89154-4009, USA
| | - Zhongming Zhao
- Center for Precision Health, School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX, 77030, USA.,Department of Psychiatry and Behavioral Sciences, The University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Peilin Jia
- Center for Precision Health, School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Ming T Tsuang
- Department of Psychiatry, University of California at San Diego, San Diego, CA, 92093, USA
| | - Kenneth S Kendler
- Virginia Institute of Psychiatric and Behavioral Genetics, Medical College of Virginia and Virginia Commonwealth University, Richmond, VA, 23298, USA
| | - Momiao Xiong
- Department of Biostatistics and Data Science, Human Genetics Center, University of Texas School of Public Health, Houston, TX, 77030, USA
| | | |
Collapse
|
32
|
Abstract
Sulfur is present in the amino acids cysteine and methionine and in a large range of essential coenzymes and cofactors and is therefore essential for all organisms. It is also a constituent of sulfate esters in proteins, carbohydrates, and numerous cellular metabolites. The sulfation and desulfation reactions modifying a variety of different substrates are commonly known as sulfation pathways. Although relatively little is known about the function of most sulfated metabolites, the synthesis of activated sulfate used in sulfation pathways is essential in both animal and plant kingdoms. In humans, mutations in the genes encoding the sulfation pathway enzymes underlie a number of developmental aberrations, and in flies and worms, their loss-of-function is fatal. In plants, a lower capacity for synthesizing activated sulfate for sulfation reactions results in dwarfism, and a complete loss of activated sulfate synthesis is also lethal. Here, we review the similarities and differences in sulfation pathways and associated processes in animals and plants, and we point out how they diverge from bacteria and yeast. We highlight the open questions concerning localization, regulation, and importance of sulfation pathways in both kingdoms and the ways in which findings from these "red" and "green" experimental systems may help reciprocally address questions specific to each of the systems.
Collapse
Affiliation(s)
- Süleyman Günal
- Botanical Institute, Cluster of Excellence on Plant Sciences (CEPLAS), University of Cologne, Cologne 50674, Germany
| | - Rebecca Hardman
- Institute of Metabolism and Systems Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, United Kingdom
| | - Stanislav Kopriva
- Botanical Institute, Cluster of Excellence on Plant Sciences (CEPLAS), University of Cologne, Cologne 50674, Germany.
| | - Jonathan Wolf Mueller
- Institute of Metabolism and Systems Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, United Kingdom; Centre for Endocrinology, Diabetes and Metabolism (CEDAM), Birmingham Health Partners, Birmingham B15 2TH, United Kingdom.
| |
Collapse
|
33
|
Lee S, Temple FT, Dawson PA. Kidney microRNA profile in pregnant mice reveals molecular insights into kidney adaptation to pregnancy: A pilot study. Mol Genet Metab Rep 2019; 20:100486. [PMID: 31249785 PMCID: PMC6587019 DOI: 10.1016/j.ymgmr.2019.100486] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Revised: 06/12/2019] [Indexed: 02/06/2023] Open
Abstract
The maternal kidneys undergo numerous physiological changes during pregnancy to maintain a healthy pregnancy for mother and child. Over the past decade, interest in microRNAs (miRNAs) for regulating gene expression during pregnancy has expanded. However, the role of miRNAs in modulating kidney physiology during pregnancy has not been extensively investigated. In this study, miRNome profiling suggested differential expression of 163 miRNAs (of 887 miRNAs detected) in the kidneys from pregnant mice at 6.5 days gestation when compared to non-pregnant female mice, of which 35 and 128 miRNAs were potentially down- and up-regulated, respectively. We performed network and pathway analyses of the >1700 potential mRNA targets of the differentially expressed miRNAs using MiRNet, Gene Ontology, Reactome and KEGG analyses. The mRNA targets were over-represented in numerous cellular signalling pathways, including cellular protective responses. In addition, we explored 13 and 29 potential differentially expressed miRNAs to have putative binding sites in the Slc13a1 and Slc26a1 sulfate transporter mRNAs, respectively, and that decreased levels of mir-466k may potentially explain the increased expression of these sulfate transporters in early mouse gestation. Collectively, this study suggests altered expression levels of miRNAs during mouse gestation, which provides pilot data for future investigations into the molecular events that modulate kidney adaptsation to pregnancy.
Collapse
Affiliation(s)
- Soohyun Lee
- Mater Research Institute, University of Queensland, Brisbane, Queensland, Australia
| | - Fergal Thomas Temple
- Mater Research Institute, University of Queensland, Brisbane, Queensland, Australia
| | - Paul Anthony Dawson
- Mater Research Institute, University of Queensland, Brisbane, Queensland, Australia
| |
Collapse
|
34
|
Swaroop M, Brooks MJ, Gieser L, Swaroop A, Zheng W. Patient iPSC-derived neural stem cells exhibit phenotypes in concordance with the clinical severity of mucopolysaccharidosis I. Hum Mol Genet 2019; 27:3612-3626. [PMID: 30052969 DOI: 10.1093/hmg/ddy259] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Accepted: 07/09/2018] [Indexed: 12/29/2022] Open
Abstract
Mucopolysaccharidosis type I (MPS I) is caused by deficiency of α-l-iduronidase (IDUA), a lysosomal enzyme involved in the breakdown and recycling of glycosaminoglycans (GAGs). Although enzyme replacement therapy is available, the efficacy of the treatment for neuropathic manifestations is limited. To facilitate drug discovery and model disease pathophysiology, we generated neural stem cells (NSCs) from MPS I patient-derived induced pluripotent stem cells (iPSCs). The NSCs exhibited characteristic disease phenotypes with deficiency of IDUA, accumulation of GAGs and enlargement of lysosomes, in agreement with the severity of clinical subgroups of MPS I. Transcriptome profiling of NSCs revealed 429 genes that demonstrated a more extensive change in expression in the most severe Hurler syndrome subgroup compared to the intermediate Hurler-Scheie or the least severe Scheie syndrome subgroups. Clustering and pathway analysis revealed high concordance of the severity of neurological defects with marked dysregulation of GAG biosynthesis, GAG degradation, lysosomal function and autophagy. Gene ontology (GO) analysis identified a dramatic upregulation of the autophagy pathway, especially in the Hurler syndrome subgroup. We conclude that GAG accumulation in the patient-derived cells disrupts lysosomal homeostasis, affecting multiple related cellular pathways in response to IDUA deficiency. These dysregulated processes likely lead to enhanced autophagy and progressively severe disease states. Our study provides potentially useful targets for clinical biomarker development, disease diagnosis and prognosis, and drug discovery.
Collapse
Affiliation(s)
- Manju Swaroop
- National Therapeutics for Rare and Neglected Diseases, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, USA
| | - Matthew J Brooks
- Neurobiology-Neurodegeneration and Repair Laboratory, National Eye Institute, National Institutes of Health, Bethesda, MD, USA
| | - Linn Gieser
- Neurobiology-Neurodegeneration and Repair Laboratory, National Eye Institute, National Institutes of Health, Bethesda, MD, USA
| | - Anand Swaroop
- Neurobiology-Neurodegeneration and Repair Laboratory, National Eye Institute, National Institutes of Health, Bethesda, MD, USA
| | - Wei Zheng
- National Therapeutics for Rare and Neglected Diseases, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, USA
| |
Collapse
|
35
|
Whittamore JM, Stephens CE, Hatch M. Absence of the sulfate transporter SAT-1 has no impact on oxalate handling by mouse intestine and does not cause hyperoxaluria or hyperoxalemia. Am J Physiol Gastrointest Liver Physiol 2019; 316:G82-G94. [PMID: 30383413 PMCID: PMC6383384 DOI: 10.1152/ajpgi.00299.2018] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
The anion exchanger SAT-1 [sulfate anion transporter 1 (Slc26a1)] is considered an important regulator of oxalate and sulfate homeostasis, but the mechanistic basis of these critical roles remain undetermined. Previously, characterization of the SAT-1-knockout (KO) mouse suggested that the loss of SAT-1-mediated oxalate secretion by the intestine was responsible for the hyperoxaluria, hyperoxalemia, and calcium oxalate urolithiasis reportedly displayed by this model. To test this hypothesis, we compared the transepithelial fluxes of 14C-oxalate, 35SO42- , and 36Cl- across isolated, short-circuited segments of the distal ileum, cecum, and distal colon from wild-type (WT) and SAT-1-KO mice. The absence of SAT-1 did not impact the transport of these anions by any part of the intestine examined. Additionally, SAT-1-KO mice were neither hyperoxaluric nor hyperoxalemic. Instead, 24-h urinary oxalate excretion was almost 50% lower than in WT mice. With no contribution from the intestine, we suggest that this may reflect the loss of SAT-1-mediated oxalate efflux from the liver. SAT-1-KO mice were, however, profoundly hyposulfatemic, even though there were no changes to intestinal sulfate handling, and the renal clearances of sulfate and creatinine indicated diminished rates of sulfate reabsorption by the proximal tubule. Aside from this distinct sulfate phenotype, we were unable to reproduce the hyperoxaluria, hyperoxalemia, and urolithiasis of the original SAT-1-KO model. In conclusion, oxalate and sulfate transport by the intestine were not dependent on SAT-1, and we found no evidence supporting the long-standing hypothesis that intestinal SAT-1 contributes to oxalate and sulfate homeostasis. NEW & NOTEWORTHY SAT-1 is a membrane-bound transport protein expressed in the intestine, liver, and kidney, where it is widely considered essential for the excretion of oxalate, a potentially toxic waste metabolite. Previously, calcium oxalate kidney stone formation by the SAT-1-knockout mouse generated the hypothesis that SAT-1 has a major role in oxalate excretion via the intestine. We definitively tested this proposal and found no evidence for SAT-1 as an intestinal anion transporter contributing to oxalate homeostasis.
Collapse
Affiliation(s)
- Jonathan M. Whittamore
- Department of Pathology, Immunology, and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, Florida
| | - Christine E. Stephens
- Department of Pathology, Immunology, and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, Florida
| | - Marguerite Hatch
- Department of Pathology, Immunology, and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, Florida
| |
Collapse
|
36
|
Balyan R, Cai M, Zhao W, Dai Z, Zhai Y, Chen G. Repeated restraint stress upregulates rat sulfotransferase 1A1. J Basic Clin Physiol Pharmacol 2018; 30:265-273. [PMID: 30864418 DOI: 10.1515/jbcpp-2016-0038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2016] [Accepted: 11/15/2018] [Indexed: 11/15/2022]
Abstract
BackgroundSulfotransferases (SULTs) are phase II drug-metabolizing enzymes. SULTs also regulate the biological activities of biological signaling molecules, such as various hormones, bile acids, and monoamine neurotransmitters; therefore, they play critical roles in the endocrine and nervous systems. People are subject to various kinds of physical, chemical, toxicological, physiological, and psychological stresses at one time or another. The study of the effects produced by stress may lead to finding novel remedies for many disease conditions. The effect of repeated restraint stress on rat SULT expression has not been studied. MethodsThis study involves the effect of repeated restraint stress on SULT1A1 expressions. Male Sprague-Dawley rats (n=4) were subjected to repeated restraint stress 2 h/day for 7 days. Protein and RNA expression of SULT1A1 were analyzed by western blot and quantitative real time reverse transcription polymerase chain reaction, respectively, in important tissues. ResultsWe observed that repeated restraint stress increased the expression of SULT1A1 in the liver, adrenal glands, cerebellum, hypothalamus, and cerebral cortex in male rats. Patterns of enhanced expression were observed at both mRNA and protein level, indicating that repeated restraint stress stimulates enzyme expression at the transcriptional level. ConclusionsChanges of SULT1A1 expression in important tissues caused by repeated restraint stress will have a significant effect on drug metabolism and xenobiotics detoxification. The significant changes in endocrine glands and brain sections may also cause disturbances in hormone homeostasis, therefore leading to disease conditions. This report provides clues for the understanding of the effect of stresses on health.
Collapse
Affiliation(s)
- Rajiv Balyan
- Department of Physiological Sciences, Center for Veterinary Health Sciences, Oklahoma State University, Stillwater, OK, USA
| | - Ma Cai
- College of Life Science, Zhongkai University of Agriculture and Engineering, Guangzhou, Guangdong Province, China
| | - Wenhong Zhao
- College of Light Industry and Food Science, Zhongkai University of Agriculture and Engineering, Guangzhou, Guangdong Province, China
| | - Zhao Dai
- School of Environmental and Chemical Engineering, Tianjin Polytechnic University, Tianjin, China
| | - Yujia Zhai
- Department of Anesthesiology, The Third Affiliated Hospital, Shenzhen University, Shenzhen, China
| | - Guangping Chen
- Department of Physiological Sciences, Center for Veterinary Health Sciences, Oklahoma State University, Stillwater, OK 74074, USA, Phone: +405-744-2349
| |
Collapse
|
37
|
Mamatkulov SI, Rinne KF, Buchner R, Netz RR, Bonthuis DJ. Water-separated ion pairs cause the slow dielectric mode of magnesium sulfate solutions. J Chem Phys 2018; 148:222812. [DOI: 10.1063/1.5000385] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Affiliation(s)
- Shavkat I. Mamatkulov
- Fachbereich Physik, Freie Universität Berlin, 14195 Berlin, Germany
- Ion-Plasma and Laser Technologies Institute of the Uzbekistan AS, Tashkent, Uzbekistan
| | - Klaus F. Rinne
- Fachbereich Physik, Freie Universität Berlin, 14195 Berlin, Germany
| | - Richard Buchner
- Institut für Physikalische und Theoretische Chemie, Universität Regensburg, D-93040 Regensburg, Germany
| | - Roland R. Netz
- Fachbereich Physik, Freie Universität Berlin, 14195 Berlin, Germany
| | | |
Collapse
|
38
|
Sorrell JM, Somoza RA, Caplan AI. Human mesenchymal stem cells induced to differentiate as chondrocytes follow a biphasic pattern of extracellular matrix production. J Orthop Res 2018; 36:1757-1766. [PMID: 29194731 PMCID: PMC5976510 DOI: 10.1002/jor.23820] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Accepted: 11/27/2017] [Indexed: 02/04/2023]
Abstract
Regenerative medicine and tissue engineering studies are actively developing novel means to repair adult articular cartilage defects using biological approaches. One such approach is the harnessing of adult human therapeutic cells such as those referred to as mesenchymal stem cells. Upon exposure to chondrogenic signals, these cells differentiate and initiate the production of a complex and voluminous cartilaginous matrix that is crucial to both the structure and function of cartilage. Furthermore, this complexity requires the time-sensitive activation of a large number of genes to produce the components of this matrix. The current study analyzed the kinetics of matrix production in an aggregate culture model where adult human mesenchymal stem cells were induced to differentiate as chondrocytes. The results indicate the existence of a biphasic mode of differentiation and maturation during which matrix genes and molecules are differentially activated and secreted. These results have important implications for developing novel approaches for the creation of tissue engineered articular cartilage. © 2017 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 36:1757-1766, 2018.
Collapse
Affiliation(s)
- J. Michael Sorrell
- Department of Biology, Skeletal Research Center; Case Western Reserve University; Cleveland Ohio 44106
| | - Rodrigo A. Somoza
- Department of Biology, Skeletal Research Center; Case Western Reserve University; Cleveland Ohio 44106
| | - Arnold I. Caplan
- Department of Biology, Skeletal Research Center; Case Western Reserve University; Cleveland Ohio 44106
| |
Collapse
|
39
|
Gallo C, Nuzzo G, d'Ippolito G, Manzo E, Sardo A, Fontana A. Sterol Sulfates and Sulfotransferases in Marine Diatoms. Methods Enzymol 2018; 605:101-138. [PMID: 29909823 DOI: 10.1016/bs.mie.2018.03.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Sterol sulfates are widely occurring molecules in marine organisms. Their importance has been so far underestimated although many of these compounds are crucial mediators of physiological and ecological functions in other organisms. Biosynthesis of sterol sulfates is controlled by cytosolic sulfotransferases (SULTs), a varied family of enzymes that catalyze the transfer of a sulfo residue (-SO3H) from the universal donor 3'-phosphoadenosine-5'-phosphosulfate to the hydroxyl function at C-3 of the steroid skeleton. The absence of molecular tools has been the main impediment to the development of a biosynthetic study of this class of compounds in marine organisms. In fact, there is very limited information about these enzymes in marine environments. SULT activity has, however, been reported in several marine species, and, recently, the production of sterol sulfates has been linked to the control of growth in marine diatoms. In this chapter, we describe methods for the study of sterol sulfates in this lineage of marine microalgae. The main aim is to provide the tools useful to deal with the biosynthesis and regulation of these compounds and to circumvent the bottleneck of the lack of molecular information. The protocols have been designed for marine diatoms, but most of the procedures can be used for other marine organisms.
Collapse
Affiliation(s)
- Carmela Gallo
- National Research Council of Italy, Institute of Biomolecular Chemistry, Pozzuoli, Naples, Italy
| | - Genoveffa Nuzzo
- National Research Council of Italy, Institute of Biomolecular Chemistry, Pozzuoli, Naples, Italy
| | - Giuliana d'Ippolito
- National Research Council of Italy, Institute of Biomolecular Chemistry, Pozzuoli, Naples, Italy.
| | - Emiliano Manzo
- National Research Council of Italy, Institute of Biomolecular Chemistry, Pozzuoli, Naples, Italy
| | - Angela Sardo
- National Research Council of Italy, Institute of Biomolecular Chemistry, Pozzuoli, Naples, Italy
| | - Angelo Fontana
- National Research Council of Italy, Institute of Biomolecular Chemistry, Pozzuoli, Naples, Italy.
| |
Collapse
|
40
|
Small Intestinal Absorption of Methylsulfonylmethane (MSM) and Accumulation of the Sulfur Moiety in Selected Tissues of Mice. Nutrients 2017; 10:nu10010019. [PMID: 29295596 PMCID: PMC5793247 DOI: 10.3390/nu10010019] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Revised: 12/05/2017] [Accepted: 12/20/2017] [Indexed: 12/14/2022] Open
Abstract
The principal dietary sources of sulfur, the amino acids methionine and cysteine, may not always be consumed in adequate amounts to meet sulfur requirements. The naturally occurring organosulfur compound, methylsulfonylmethane (MSM), is available as a dietary supplement and has been associated with multiple health benefits. Absorption of MSM by the small intestine and accumulation of the associated sulfur moiety in selected tissues with chronic (8 days) administration were evaluated using juvenile male mice. Intestinal absorption was not saturated at 50 mmol, appeared passive and carrier-independent, with a high capacity (at least 2 g/d-mouse). The 35S associated with MSM did not increase in serum or tissue homogenates between days 2 and 8, indicating a stable equilibrium between intake and elimination was established. In contrast, proteins isolated from the preparations using gel electrophoresis revealed increasing incorporation of 35S in the protein fraction of serum, cellular elements of blood, liver, and small intestine but not skeletal muscle. The potential contributions of protein synthesis using labeled sulfur amino acids synthesized by the gut bacteria and posttranslational sulfation of proteins by incorporation of the labeled sulfate of MSM in 3'-phosphoadenosine 5'-phosphosulfate (PAPS) and subsequent transfer by sulfotransferases are discussed.
Collapse
|
41
|
Dawson PA, Richard K, Perkins A, Zhang Z, Simmons DG. Review: Nutrient sulfate supply from mother to fetus: Placental adaptive responses during human and animal gestation. Placenta 2017; 54:45-51. [PMID: 28089504 DOI: 10.1016/j.placenta.2017.01.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2016] [Revised: 12/23/2016] [Accepted: 01/04/2017] [Indexed: 01/20/2023]
Abstract
Nutrient sulfate has numerous roles in mammalian physiology and is essential for healthy fetal growth and development. The fetus has limited capacity to generate sulfate and relies on sulfate supplied from the maternal circulation via placental sulfate transporters. The placenta also has a high sulfate requirement for numerous molecular and cellular functions, including sulfate conjugation (sulfonation) to estrogen and thyroid hormone which leads to their inactivation. Accordingly, the ratio of sulfonated (inactive) to unconjugated (active) hormones modulates endocrine function in fetal, placental and maternal tissues. During pregnancy, there is a marked increase in the expression of genes involved in transport and generation of sulfate in the mouse placenta, in line with increasing fetal and placental demands for sulfate. The maternal circulation also provides a vital reservoir of sulfate for the placenta and fetus, with maternal circulating sulfate levels increasing by 2-fold from mid-gestation. However, despite evidence from animal studies showing the requirement of maternal sulfate supply for placental and fetal physiology, there are no routine clinical measurements of sulfate or consideration of dietary sulfate intake in pregnant women. This is also relevant to certain xenobiotics or pharmacological drugs which when taken by the mother use significant quantities of circulating sulfate for detoxification and clearance, and thereby have the potential to decrease sulfonation capacity in the placenta and fetus. This article will review the physiological adaptations of the placenta for maintaining sulfate homeostasis in the fetus and placenta, with a focus on pathophysiological outcomes in animal models of disturbed sulfate homeostasis.
Collapse
Affiliation(s)
- P A Dawson
- Mater Research Institute, The University of Queensland, Woolloongabba, Australia; School of Biomedical Sciences, The University of Queensland, St. Lucia, Australia.
| | - K Richard
- Conjoint Endocrine Laboratory, Chemical Pathology, Pathology Queensland, Queensland Health, Herston, Australia
| | - A Perkins
- School of Medical Science, Menzies Health Institute Queensland, Griffith University, Gold Coast Campus, Australia
| | - Z Zhang
- Mater Research Institute, The University of Queensland, Woolloongabba, Australia; School of Biomedical Sciences, The University of Queensland, St. Lucia, Australia
| | - D G Simmons
- Mater Research Institute, The University of Queensland, Woolloongabba, Australia; School of Biomedical Sciences, The University of Queensland, St. Lucia, Australia
| |
Collapse
|