1
|
Kim N, Li Y, Yu R, Kwon HS, Song A, Jun MH, Jeong JY, Lee JH, Lim HH, Kim MJ, Kim JW, Oh WJ. Repulsive Sema3E-Plexin-D1 signaling coordinates both axonal extension and steering via activating an autoregulatory factor, Mtss1. eLife 2024; 13:e96891. [PMID: 38526535 PMCID: PMC11001299 DOI: 10.7554/elife.96891] [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: 02/07/2024] [Accepted: 03/14/2024] [Indexed: 03/26/2024] Open
Abstract
Axon guidance molecules are critical for neuronal pathfinding because they regulate directionality and growth pace during nervous system development. However, the molecular mechanisms coordinating proper axonal extension and turning are poorly understood. Here, metastasis suppressor 1 (Mtss1), a membrane protrusion protein, ensured axonal extension while sensitizing axons to the Semaphorin 3E (Sema3E)-Plexin-D1 repulsive cue. Sema3E-Plexin-D1 signaling enhanced Mtss1 expression in projecting striatonigral neurons. Mtss1 localized to the neurite axonal side and regulated neurite outgrowth in cultured neurons. Mtss1 also aided Plexin-D1 trafficking to the growth cone, where it signaled a repulsive cue to Sema3E. Mtss1 ablation reduced neurite extension and growth cone collapse in cultured neurons. Mtss1-knockout mice exhibited fewer striatonigral projections and irregular axonal routes, and these defects were recapitulated in Plxnd1- or Sema3e-knockout mice. These findings demonstrate that repulsive axon guidance activates an exquisite autoregulatory program coordinating both axonal extension and steering during neuronal pathfinding.
Collapse
Affiliation(s)
- Namsuk Kim
- Neurovascular Unit Research Group, Korea Brain Research InstituteDaeguRepublic of Korea
| | - Yan Li
- Neurovascular Unit Research Group, Korea Brain Research InstituteDaeguRepublic of Korea
| | - Ri Yu
- Neurovascular Unit Research Group, Korea Brain Research InstituteDaeguRepublic of Korea
| | - Hyo-Shin Kwon
- Neurovascular Unit Research Group, Korea Brain Research InstituteDaeguRepublic of Korea
| | - Anji Song
- Neurovascular Unit Research Group, Korea Brain Research InstituteDaeguRepublic of Korea
| | - Mi-Hee Jun
- Neurovascular Unit Research Group, Korea Brain Research InstituteDaeguRepublic of Korea
| | - Jin-Young Jeong
- Neurovascular Unit Research Group, Korea Brain Research InstituteDaeguRepublic of Korea
- Department of Brain and Cognitive Sciences, Daegu Gyeongbuk Institute of Science and TechnologyDaeguRepublic of Korea
| | - Ji Hyun Lee
- Neurovascular Unit Research Group, Korea Brain Research InstituteDaeguRepublic of Korea
| | - Hyun-Ho Lim
- Neurovascular Unit Research Group, Korea Brain Research InstituteDaeguRepublic of Korea
| | - Mi-Jin Kim
- Department of Life Sciences, Chung-Ang UniversitySeoulRepublic of Korea
| | - Jung-Woong Kim
- Department of Life Sciences, Chung-Ang UniversitySeoulRepublic of Korea
| | - Won-Jong Oh
- Neurovascular Unit Research Group, Korea Brain Research InstituteDaeguRepublic of Korea
| |
Collapse
|
2
|
Chen M, Dong Y, Tian L, Zhou J, Zhu E, Yuan H, Li X, Wang B. Metastasis suppressor 1 interacts with protein tyrosine phosphatase receptor-δ to regulate adipogenesis. FASEB J 2023; 37:e22857. [PMID: 36906292 DOI: 10.1096/fj.202201322r] [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: 08/13/2022] [Revised: 02/07/2023] [Accepted: 02/22/2023] [Indexed: 03/13/2023]
Abstract
Adipogenesis is a finely controlled process and its dysfunction may contribute to metabolic disorders such as obesity. Metastasis suppressor 1 (MTSS1) is a player in tumorigenesis and metastasis of various types of cancers. To date, it is not known whether and how MTSS1 plays a role in adipocyte differentiation. In the current study, we found that MTSS1 was upregulated during adipogenic differentiation of established mesenchymal cell lines and primary cultured bone marrow stromal cells. Gain-of-function and loss-of-function experiments uncovered that MTSS1 facilitated adipocyte differentiation from mesenchymal progenitor cells. Mechanistic explorations revealed that MTSS1 bound and interacted with FYN, a member of Src family of tyrosine kinases (SFKs), and protein tyrosine phosphatase receptor-δ (PTPRD). We demonstrated that PTPRD was capable of inducing the differentiation of adipocytes. Overexpression of PTPRD attenuated the impaired adipogenesis induced by the siRNA targeting MTSS1. Both MTSS1 and PTPRD activated SFKs by suppressing the phosphorylation of SFKs at Tyr530 and inducing the phosphorylation of FYN at Tyr419. Further investigation showed that MTSS1 and PTPRD were able to activate FYN. Collectively, our study has for the first time unraveled that MTSS1 plays a role in adipocyte differentiation in vitro through interacting with PTPRD and thereby activating SFKs such as FYN tyrosine kinase.
Collapse
Affiliation(s)
- Meng Chen
- NHC Key Lab of Hormones and Development, Tianjin Key Lab of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Institute of Endocrinology, Tianjin Medical University, Tianjin, China
| | - Yuan Dong
- College of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Lijie Tian
- NHC Key Lab of Hormones and Development, Tianjin Key Lab of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Institute of Endocrinology, Tianjin Medical University, Tianjin, China
| | - Jie Zhou
- NHC Key Lab of Hormones and Development, Tianjin Key Lab of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Institute of Endocrinology, Tianjin Medical University, Tianjin, China
| | - Endong Zhu
- NHC Key Lab of Hormones and Development, Tianjin Key Lab of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Institute of Endocrinology, Tianjin Medical University, Tianjin, China
| | - Hairui Yuan
- NHC Key Lab of Hormones and Development, Tianjin Key Lab of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Institute of Endocrinology, Tianjin Medical University, Tianjin, China
| | - Xiaoxia Li
- College of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Baoli Wang
- NHC Key Lab of Hormones and Development, Tianjin Key Lab of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Institute of Endocrinology, Tianjin Medical University, Tianjin, China
| |
Collapse
|
3
|
Gao H, Tripathi U, Trushin S, Okromelidze L, Pichurin NP, Wei L, Zhuang Y, Wang L, Trushina E. A genome-wide association study in human lymphoblastoid cells supports safety of mitochondrial complex I inhibitor. Mitochondrion 2021; 58:83-94. [PMID: 33610756 PMCID: PMC8743030 DOI: 10.1016/j.mito.2021.02.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Accepted: 02/08/2021] [Indexed: 01/12/2023]
Abstract
Novel therapeutic strategies for Alzheimer's disease (AD) are of the greatest priority given the consistent failure of recent clinical trials focused on Aβ or pTau. Earlier, we demonstrated that mild mitochondrial complex I inhibitor CP2 blocks neurodegeneration and cognitive decline in multiple mouse models of AD. To evaluate the safety of CP2 in humans, we performed a genome-wide association study (GWAS) using 196 lymphoblastoid cell lines and identified 11 SNP loci and 64 mRNA expression probe sets that potentially associate with CP2 susceptibility. Using primary mouse neurons and pharmacokinetic study, we show that CP2 is generally safe at a therapeutic dose.
Collapse
Affiliation(s)
- Huanyao Gao
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, 200 First St. SW, Rochester, MN, 55905, USA
| | - Utkarsh Tripathi
- Department of Neurology, Mayo Clinic, 200 First St. SW, Rochester, MN, 55905, USA
| | - Sergey Trushin
- Department of Neurology, Mayo Clinic, 200 First St. SW, Rochester, MN, 55905, USA
| | - Lela Okromelidze
- Department of Neurology, Mayo Clinic, 200 First St. SW, Rochester, MN, 55905, USA
| | - Nicholas P Pichurin
- Department of Neurology, Mayo Clinic, 200 First St. SW, Rochester, MN, 55905, USA
| | - Lixuan Wei
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, 200 First St. SW, Rochester, MN, 55905, USA
| | - Yongxian Zhuang
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, 200 First St. SW, Rochester, MN, 55905, USA
| | - Liewei Wang
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, 200 First St. SW, Rochester, MN, 55905, USA
| | - Eugenia Trushina
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, 200 First St. SW, Rochester, MN, 55905, USA; Department of Neurology, Mayo Clinic, 200 First St. SW, Rochester, MN, 55905, USA.
| |
Collapse
|
4
|
Di Paolo A, Eastman G, Mesquita-Ribeiro R, Farias J, Macklin A, Kislinger T, Colburn N, Munroe D, Sotelo Sosa JR, Dajas-Bailador F, Sotelo-Silveira JR. PDCD4 regulates axonal growth by translational repression of neurite growth-related genes and is modulated during nerve injury responses. RNA (NEW YORK, N.Y.) 2020; 26:1637-1653. [PMID: 32747606 PMCID: PMC7566564 DOI: 10.1261/rna.075424.120] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2020] [Accepted: 07/20/2020] [Indexed: 05/07/2023]
Abstract
Programmed cell death 4 (PDCD4) protein is a tumor suppressor that inhibits translation through the mTOR-dependent initiation factor EIF4A, but its functional role and mRNA targets in neurons remain largely unknown. Our work identified that PDCD4 is highly expressed in axons and dendrites of CNS and PNS neurons. Using loss- and gain-of-function experiments in cortical and dorsal root ganglia primary neurons, we demonstrated the capacity of PDCD4 to negatively control axonal growth. To explore PDCD4 transcriptome and translatome targets, we used Ribo-seq and uncovered a list of potential targets with known functions as axon/neurite outgrowth regulators. In addition, we observed that PDCD4 can be locally synthesized in adult axons in vivo, and its levels decrease at the site of peripheral nerve injury and before nerve regeneration. Overall, our findings demonstrate that PDCD4 can act as a new regulator of axonal growth via the selective control of translation, providing a target mechanism for axon regeneration and neuronal plasticity processes in neurons.
Collapse
Affiliation(s)
- Andrés Di Paolo
- Departamento de Proteínas y Ácidos Nucleicos, Instituto de Investigaciones Biológicas Clemente Estable, Montevideo 11600, Uruguay
| | - Guillermo Eastman
- Departamento de Genómica, Instituto de Investigaciones Biológicas Clemente Estable, Montevideo 11600, Uruguay
| | | | - Joaquina Farias
- Departamento de Genómica, Instituto de Investigaciones Biológicas Clemente Estable, Montevideo 11600, Uruguay
| | - Andrew Macklin
- Princess Margaret Cancer Centre, University Health Network, Toronto M5G 1L7, Canada
| | - Thomas Kislinger
- Princess Margaret Cancer Centre, University Health Network, Toronto M5G 1L7, Canada
- University of Toronto, Department of Medical Biophysics, Toronto M5S 1A1, Canada
| | - Nancy Colburn
- Former Chief of Laboratory of Cancer Prevention at the National Cancer Institute-NIH at Frederick, Maryland 21702, USA
| | - David Munroe
- Former Laboratory of Molecular Technologies, LEIDOS at Frederick National Laboratory for Cancer Research, Frederick, Maryland 21702, USA
| | - José R Sotelo Sosa
- Departamento de Proteínas y Ácidos Nucleicos, Instituto de Investigaciones Biológicas Clemente Estable, Montevideo 11600, Uruguay
| | | | - José R Sotelo-Silveira
- Departamento de Genómica, Instituto de Investigaciones Biológicas Clemente Estable, Montevideo 11600, Uruguay
- Departamento de Biología Celular y Molecular, Facultad de Ciencias UdelaR, Montevideo 11400, Uruguay
| |
Collapse
|
5
|
Harvey PD, Sun N, Bigdeli TB, Fanous AH, Aslan M, Malhotra AK, Lu Q, Hu Y, Li B, Chen Q, Mane S, Miller P, Rajeevan N, Sayward F, Cheung KH, Li Y, Greenwood TA, Gur RE, Braff DL, Brophy M, Pyarajan S, O'Leary TJ, Gleason T, Przygodszki R, Muralidhar S, Gaziano JM, Concato J, Zhao H, Siever LJ. Genome-wide association study of cognitive performance in U.S. veterans with schizophrenia or bipolar disorder. Am J Med Genet B Neuropsychiatr Genet 2020; 183:181-194. [PMID: 31872970 DOI: 10.1002/ajmg.b.32775] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Revised: 11/22/2019] [Accepted: 12/09/2019] [Indexed: 12/25/2022]
Abstract
Cognitive impairment is a frequent and serious problem in patients with various forms of severe mental illnesses (SMI), including schizophrenia (SZ) and bipolar disorder (BP). Recent research suggests genetic links to several cognitive phenotypes in both SMI and in the general population. Our goal in this study was to identify potential genomic signatures of cognitive functioning in veterans with severe mental illness and compare them to previous findings for cognition across different populations. Veterans Affairs (VA) Cooperative Studies Program (CSP) Study #572 evaluated cognitive and functional capacity measures among SZ and BP patients. In conjunction with the VA Million Veteran Program, 3,959 European American (1,095 SZ, 2,864 BP) and 2,601 African American (1,095 SZ, 2,864 BP) patients were genotyped using a custom Affymetrix Axiom Biobank array. We performed a genome-wide association study of global cognitive functioning, constructed polygenic scores for SZ and cognition in the general population, and examined genetic correlations with 2,626 UK Biobank traits. Although no single locus attained genome-wide significance, observed allelic effects were strongly consistent with previous studies. We observed robust associations between global cognitive functioning and polygenic scores for cognitive performance, intelligence, and SZ risk. We also identified significant genetic correlations with several cognition-related traits in UK Biobank. In a diverse cohort of U.S. veterans with SZ or BP, we demonstrate broad overlap of common genetic effects on cognition in the general population, and find that greater polygenic loading for SZ risk is associated with poorer cognitive performance.
Collapse
Affiliation(s)
- Philip D Harvey
- Research Service, Bruce W. Carter Miami Veterans Affairs (VA) Medical Center, Miami, Florida.,Department of Psychiatry and Behavioral Sciences, University of Miami School of Medicine, Miami, Florida
| | - Ning Sun
- Clinical Epidemiology Research Center (CERC), VA Connecticut Healthcare System, West Haven, Connecticut.,Yale University School of Medicine, New Haven, Connecticut
| | - Tim B Bigdeli
- Department of Psychiatry, VA New York Harbor Healthcare System, Brooklyn, New York.,Department of Psychiatry and Behavioral Sciences, SUNY Downstate Medical Center, Brooklyn, New York
| | - Ayman H Fanous
- Department of Psychiatry, VA New York Harbor Healthcare System, Brooklyn, New York.,Department of Psychiatry and Behavioral Sciences, SUNY Downstate Medical Center, Brooklyn, New York
| | - Mihaela Aslan
- Clinical Epidemiology Research Center (CERC), VA Connecticut Healthcare System, West Haven, Connecticut.,Yale University School of Medicine, New Haven, Connecticut
| | - Anil K Malhotra
- Center for Psychiatric Neuroscience, Feinstein Institute for Medical Research, Manhasset, New York.,Division of Psychiatry Research, The Zucker Hillside Hospital, Northwell Health, Glen Oaks, New York.,Department of Psychiatry, Hofstra Northwell School of Medicine, Hempstead, New York
| | - Qiongshi Lu
- Yale University School of Medicine, New Haven, Connecticut.,Department of Biostatistics & Medical Informatics, University of Wisconsin-Madison, Madison, Wisconsin
| | - Yiming Hu
- Yale University School of Medicine, New Haven, Connecticut
| | - Boyang Li
- Yale University School of Medicine, New Haven, Connecticut
| | - Quan Chen
- Clinical Epidemiology Research Center (CERC), VA Connecticut Healthcare System, West Haven, Connecticut.,Yale University School of Medicine, New Haven, Connecticut
| | - Shrikant Mane
- Yale University School of Medicine, New Haven, Connecticut
| | - Perry Miller
- Clinical Epidemiology Research Center (CERC), VA Connecticut Healthcare System, West Haven, Connecticut.,Yale University School of Medicine, New Haven, Connecticut
| | - Nallakkandi Rajeevan
- Clinical Epidemiology Research Center (CERC), VA Connecticut Healthcare System, West Haven, Connecticut.,Yale University School of Medicine, New Haven, Connecticut
| | - Frederick Sayward
- Clinical Epidemiology Research Center (CERC), VA Connecticut Healthcare System, West Haven, Connecticut.,Yale University School of Medicine, New Haven, Connecticut
| | - Kei-Hoi Cheung
- Clinical Epidemiology Research Center (CERC), VA Connecticut Healthcare System, West Haven, Connecticut.,Yale University School of Medicine, New Haven, Connecticut
| | - Yuli Li
- Clinical Epidemiology Research Center (CERC), VA Connecticut Healthcare System, West Haven, Connecticut.,Yale University School of Medicine, New Haven, Connecticut
| | | | - Raquel E Gur
- Department of Psychiatry, University of Pennsylvania Perelman School of Medicine and Children's Hospital of Philadelphia, Philadelphia, Pennsylvania.,Department of Child & Adolescent Psychiatry and Lifespan Brain Institute, University of Pennsylvania Perelman School of Medicine and Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - David L Braff
- Department of Psychiatry, University of California, San Diego, California.,VISN-22 Mental Illness, Research, Education and Clinical Center (MIRECC), VA San Diego Healthcare System, San Diego, California
| | | | - Mary Brophy
- Massachusetts Area Veterans Epidemiology Research, and Information Center (MAVERIC), Jamaica Plain, Massachusetts.,Boston University School of Medicine, Boston, Massachusetts
| | - Saiju Pyarajan
- Massachusetts Area Veterans Epidemiology Research, and Information Center (MAVERIC), Jamaica Plain, Massachusetts
| | - Timothy J O'Leary
- Office of Research and Development, Veterans Health Administration, Washington, District of Columbia
| | - Theresa Gleason
- Office of Research and Development, Veterans Health Administration, Washington, District of Columbia
| | - Ronald Przygodszki
- Office of Research and Development, Veterans Health Administration, Washington, District of Columbia
| | - Sumitra Muralidhar
- Office of Research and Development, Veterans Health Administration, Washington, District of Columbia
| | - J Michael Gaziano
- Massachusetts Area Veterans Epidemiology Research, and Information Center (MAVERIC), Jamaica Plain, Massachusetts.,Department of Medicine, Harvard University, Boston, Massachusetts
| | - John Concato
- Clinical Epidemiology Research Center (CERC), VA Connecticut Healthcare System, West Haven, Connecticut.,Yale University School of Medicine, New Haven, Connecticut
| | - Hongyu Zhao
- Clinical Epidemiology Research Center (CERC), VA Connecticut Healthcare System, West Haven, Connecticut.,Yale University School of Medicine, New Haven, Connecticut
| | - Larry J Siever
- James J. Peters Veterans Affairs Medical Center, Bronx, New York.,Department of Psychiatry, Mount Sinai School of Medicine, New York, New York
| |
Collapse
|
6
|
Uhl GR, Martinez MJ. PTPRD: neurobiology, genetics, and initial pharmacology of a pleiotropic contributor to brain phenotypes. Ann N Y Acad Sci 2019; 1451:112-129. [PMID: 30648269 PMCID: PMC6629525 DOI: 10.1111/nyas.14002] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Revised: 11/12/2018] [Accepted: 12/19/2018] [Indexed: 12/12/2022]
Abstract
Receptor-type protein tyrosine phosphatase, receptor type D (PTPRD) has likely roles as a neuronal cell adhesion molecule and synaptic specifier. Interest in its neurobiology and genomics has been stimulated by results from human genetics and mouse models for phenotypes related to addiction, restless leg syndrome, neurofibrillary pathology in Alzheimer's disease, cognitive impairment/intellectual disability, mood lability, and obsessive-compulsive disorder. We review PTPRD's discovery, gene family, candidate homomeric and heteromeric binding partners, phosphatase activities, brain distribution, human genetic associations with nervous system phenotypes, and mouse model data relevant to these phenotypes. We discuss the recently reported discovery of the first small molecule inhibitor of PTPRD phosphatase, the identification of its addiction-related effects, and the implications of these findings for the PTPRD-associated brain phenotypes. In assembling PTPRD neurobiology, human genetics, and mouse genetic and pharmacological datasets, we provide a compelling picture of the roles played by PTPRD, its variation, and its potential as a target for novel therapeutics.
Collapse
Affiliation(s)
- George R Uhl
- Neurology and Research Services, New Mexico VA Healthcare System, Albuquerque, New Mexico.,Departments of Neurology, Neuroscience, Molecular Genetics and Microbiology, University of New Mexico, Albuquerque, New Mexico.,Biomedical Research Institute of New Mexico, Albuquerque, New Mexico.,Departments of Neurology, Neuroscience and Mental Health, Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - Maria J Martinez
- Neurology and Research Services, New Mexico VA Healthcare System, Albuquerque, New Mexico.,Biomedical Research Institute of New Mexico, Albuquerque, New Mexico
| |
Collapse
|
7
|
Wang M, Chen Y, Guo Z, Yang C, Qi J, Fu Y, Chen Z, Chen P, Wang Y. Changes in the mitochondrial proteome in human hepatocytes in response to alpha-amanitin hepatotoxicity. Toxicon 2018; 156:34-40. [PMID: 30399359 DOI: 10.1016/j.toxicon.2018.11.002] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Revised: 10/20/2018] [Accepted: 11/01/2018] [Indexed: 01/16/2023]
Abstract
Amanitin-induced apoptosis is proposed to have a significant effect on the pathogenesis of liver damage. However, few reports have focused on proteome changes induced by α-amanitin (α-AMA). Here, we evaluated changes in mitochondrial proteins of hepatocytes in response to 2 μM α-AMA, a concentration at which α-AMA-induced cell damage could be rescued at cellular level by common clinical drugs. We found 56 proteins were differentially expressed in an α-AMA-treated group. Among them, 38 proteins were downregulated and 18 were upregulated. Downregulated functional proteins included importer TOMM40, respiratory chain component cytochrome C, and metabolic enzymes of citrate acid cycle such as malate dehydrogenase, which localize on the mitochondrial outer membrane, inner membrane and matrix respectively. Immunoblot analysis showed that α-AMA decreased mitochondrial import receptor subunit TOMM40 and cytochrome c accompanied by an increase in the cytosol although their total protein levels were not affected significantly. The mitochondrial membrane potential was also destroyed by α-AMA and was restored by the clinical drug silibinin. Immunofluorescence suggested that mitochondrial morphology did not change. Taken together, our results provide further insights into the toxic mechanism of α-AMA on hepatocytes.
Collapse
Affiliation(s)
- Mei Wang
- The Key Laboratory of Protein Chemistry and Developmental Biology of Ministry of Education, College of Life Sciences, Hunan Normal University, Changsha 410081, PR China
| | - Yu Chen
- The Key Laboratory of Protein Chemistry and Developmental Biology of Ministry of Education, College of Life Sciences, Hunan Normal University, Changsha 410081, PR China
| | - Zhen Guo
- The Key Laboratory of Protein Chemistry and Developmental Biology of Ministry of Education, College of Life Sciences, Hunan Normal University, Changsha 410081, PR China
| | - Changcheng Yang
- The Key Laboratory of Protein Chemistry and Developmental Biology of Ministry of Education, College of Life Sciences, Hunan Normal University, Changsha 410081, PR China
| | - Jiaomei Qi
- The Key Laboratory of Protein Chemistry and Developmental Biology of Ministry of Education, College of Life Sciences, Hunan Normal University, Changsha 410081, PR China
| | - Yujuan Fu
- The Key Laboratory of Protein Chemistry and Developmental Biology of Ministry of Education, College of Life Sciences, Hunan Normal University, Changsha 410081, PR China
| | - Zuohong Chen
- The Key Laboratory of Protein Chemistry and Developmental Biology of Ministry of Education, College of Life Sciences, Hunan Normal University, Changsha 410081, PR China
| | - Ping Chen
- The Key Laboratory of Protein Chemistry and Developmental Biology of Ministry of Education, College of Life Sciences, Hunan Normal University, Changsha 410081, PR China.
| | - Ying Wang
- The Key Laboratory of Protein Chemistry and Developmental Biology of Ministry of Education, College of Life Sciences, Hunan Normal University, Changsha 410081, PR China.
| |
Collapse
|
8
|
Tang D, Xiao Z, Xu Y, Zeng J, Peng D, Liang S, Tang C, Liu Z. The peptide toxin δ-hexatoxin-MrIX inhibits fast inactivation of Na Vs in mouse cerebellar granule cells. Peptides 2018; 102:47-53. [PMID: 29501398 DOI: 10.1016/j.peptides.2018.02.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Revised: 02/27/2018] [Accepted: 02/27/2018] [Indexed: 12/19/2022]
Abstract
Spider venom is rich in peptide toxins that could be used to explore the structure and function of voltage-gated sodium channels (NaVs). This study has characterized a 44-amino acid peptide toxin, δ-hexatoxin-MrIX (δ-HXTX-MrIX), from the venom of the spider Macrothele raveni. δ-hexatoxin-MrIX potently inhibited the fast inactivation of NaVs in mouse cerebellar granule cells (CGCs) with an EC50 of 35.3 ± 5.9 nM. The toxin shifted both the steady-state activation and the steady-state inactivation curves of CGC NaVs to the hyperpolarized direction. δ-hexatoxin-MrIX also acted on NaV1.3 and NaV1.4 channels heterologously expressed in HEK293T cells, as well as on NaVs in acutely isolated cockroach DUM neurons. However, the NaV1.5, NaV1.7 and NaV1.8 channels were resistant to δ-hexatoxin-MrIX. The toxin inhibited the fast inactivation of NaV1.3 and NaV1.4 with high affinity (EC50 values of 82.0 ± 3.0 nM and 24.0 ± 4.7 nM, respectively), but the saturating dose of toxin showed distinct efficacy on these two types of channels. δ-hexatoxin-MrIX is a peptide toxin acting on CGC NaVs and could be used as a pharmacological tool to explore the role of NaVs in granule cell maturation during cerebellum development.
Collapse
Affiliation(s)
- Dongfang Tang
- The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, 410081, China
| | - Zhen Xiao
- The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, 410081, China
| | - Yan Xu
- The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, 410081, China
| | - Jiao Zeng
- The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, 410081, China
| | - Dezheng Peng
- The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, 410081, China
| | - Songping Liang
- The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, 410081, China
| | - Cheng Tang
- The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, 410081, China.
| | - Zhonghua Liu
- The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, 410081, China.
| |
Collapse
|