1
|
Irisarri A, Corral A, Perez-Salvador N, Bellver-Sanchis A, Ribalta-Vilella M, Bentanachs R, Alegret M, Laguna JC, Barroso E, Palomer X, Ortuño-Sahagún D, Vázquez-Carrera M, Pallàs M, Herrero L, Griñán-Ferré C. FTO inhibition mitigates high-fat diet-induced metabolic disturbances and cognitive decline in SAMP8 mice. Mol Med 2025; 31:73. [PMID: 39984825 PMCID: PMC11843768 DOI: 10.1186/s10020-025-01126-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Accepted: 02/12/2025] [Indexed: 02/23/2025] Open
Abstract
This study investigated the effects of fat mass and obesity-associated (FTO) inhibition on cognitive function and metabolic parameters of senescence-accelerated mouse prone 8 (SAMP8) mice fed a high-fat diet (HFD). SAMP8 mice fed an HFD exhibited increased body weight, impaired glucose tolerance, and elevated serum leptin levels. In epididymal white adipose tissue (eWAT), pharmacological treatment with FB23, a well-established FTO inhibitor, increased leptin production and modulated genes involved in lipid metabolism (Cpt1a, Atgl, Hsl, Fas), oxidative stress (OS) (Bip, Edem), and inflammation (Mcp1, Tnfα). Expression of hepatic genes related to lipid metabolism (Cpt1a, Atgl, Mgl, Dgat2, Srebp, Plin2) and OS (catalase, Edem) were modulated by FB23, although hepatic steatosis remained unchanged. Remarkably, FB23 treatment increased m6A RNA methylation in the brain, accompanied by changes in N6-methyladenosine (m6A)-regulatory enzymes and modulation of neuroinflammatory markers (Il6, Mcp1, iNOS). FTO inhibition reduced the activity of matrix metalloproteases (Mmp2, Mmp9) and altered IGF1 signaling (Igf1, Pten). Notably, enhanced leptin signaling was observed through increased expression of immediate early genes (Arc, Fos) and the transcription factor Stat3. Improved synaptic plasticity was evident, as shown by increased levels of neurotrophic factors (Bdnf, Ngf) and restored neurite length and spine density. Consistent with these findings, behavioral tests demonstrated that FB23 treatment effectively rescued cognitive impairments in SAMP8 HFD mice. The novel object recognition test (NORT) and object location test (OLT) revealed that treated mice exhibited enhanced short- and long-term memory and spatial memory compared to the HFD control group. Additionally, the open field test showed a reduction in anxiety-like behavior after treatment with FB23. In conclusion, pharmacological FTO inhibition ameliorated HFD-induced metabolic disturbances and cognitive decline in SAMP8 mice. These results suggest that targeting FTO may be a promising therapeutic approach to counteract obesity-induced cognitive impairment and age-related neurodegeneration.
Collapse
Grants
- PID2021-122116OB-100 Ministerio de Economía, Industria Economía, Industria y Competitividad (Agencia Estatal de Investigación, AEI) and European Union NextGenerationEU/PRTR
- PDC2022-133441-I00 Ministerio de Economía, Industria Economía, Industria y Competitividad (Agencia Estatal de Investigación, AEI) and European Union NextGenerationEU/PRTR
- PID2020-114953RB-C21 Ministerio de Economía, Industria Economía, Industria y Competitividad (Agencia Estatal de Investigación, AEI) and European Union NextGenerationEU/PRTR
- PID2022-139016OA-I00 Ministerio de Economía, Industria Economía, Industria y Competitividad (Agencia Estatal de Investigación, AEI) and European Union NextGenerationEU/PRTR
- CIBERDEM CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM)
- 2021 SGR 00357 the Government of Catalonia
- CB06/03/0001 CIBEROBN
- 2021SGR00367 Carlos III Health Institute project and the Government of Catalonia
- Producte 0092 Departament d'Empresa i Coneixement de la Generalitat de Catalunya 2023
- Llavor 005 and 007 Departament d'Empresa i Coneixement de la Generalitat de Catalunya 2023
- Departament d’Empresa i Coneixement de la Generalitat de Catalunya 2023
Collapse
Affiliation(s)
- Alba Irisarri
- pHD Program in Biotechnology, Faculty of Pharmacy and Food Sciences, University of Barcelona, Avda. Joan XXIII 27, 08028, Barcelona, Spain
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, University of Barcelona, Avda. Joan XXIII 27, 08028, Barcelona, Spain
- Institute of Neurosciences of the University of Barcelona, University of Barcelona, 08035, Barcelona, Spain
| | - Ana Corral
- Department of Biochemistry and Physiology, School of Pharmacy and Food Sciences, University of Barcelona, Avda. Joan XXIII 27, 08028, Barcelona, Spain
- Institute of Biomedicine of the University of Barcelona (IBUB), University of Barcelona, Avda. Joan XXIII 27, 08028, Barcelona, Spain
| | - Núria Perez-Salvador
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, University of Barcelona, Avda. Joan XXIII 27, 08028, Barcelona, Spain
| | - Aina Bellver-Sanchis
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, University of Barcelona, Avda. Joan XXIII 27, 08028, Barcelona, Spain
- Institute of Neurosciences of the University of Barcelona, University of Barcelona, 08035, Barcelona, Spain
| | - Marta Ribalta-Vilella
- pHD Program in Biotechnology, Faculty of Pharmacy and Food Sciences, University of Barcelona, Avda. Joan XXIII 27, 08028, Barcelona, Spain
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, University of Barcelona, Avda. Joan XXIII 27, 08028, Barcelona, Spain
- Institute of Neurosciences of the University of Barcelona, University of Barcelona, 08035, Barcelona, Spain
| | - Roger Bentanachs
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, University of Barcelona, Avda. Joan XXIII 27, 08028, Barcelona, Spain
- Institute of Biomedicine of the University of Barcelona (IBUB), University of Barcelona, Avda. Joan XXIII 27, 08028, Barcelona, Spain
| | - Marta Alegret
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, University of Barcelona, Avda. Joan XXIII 27, 08028, Barcelona, Spain
- Institute of Biomedicine of the University of Barcelona (IBUB), University of Barcelona, Avda. Joan XXIII 27, 08028, Barcelona, Spain
- Centro de Investigación Biomédica en Red (CIBER) de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, 28029, Madrid, Spain
| | - Juan Carlos Laguna
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, University of Barcelona, Avda. Joan XXIII 27, 08028, Barcelona, Spain
- Institute of Biomedicine of the University of Barcelona (IBUB), University of Barcelona, Avda. Joan XXIII 27, 08028, Barcelona, Spain
- Centro de Investigación Biomédica en Red (CIBER) de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, 28029, Madrid, Spain
| | - Emma Barroso
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, University of Barcelona, Avda. Joan XXIII 27, 08028, Barcelona, Spain
- Institute of Biomedicine of the University of Barcelona (IBUB), University of Barcelona, Avda. Joan XXIII 27, 08028, Barcelona, Spain
- Spanish Biomedical Research Center in Diabetes and Associated Metabolic Diseases (CIBERDEM) - National Institute of Health Carlos III, 28029, Madrid, Spain
- Pediatric Research Institute-Hospital Sant Joan de Déu, 08950, Esplugues de Llobregat, Barcelona, Spain
| | - Xavier Palomer
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, University of Barcelona, Avda. Joan XXIII 27, 08028, Barcelona, Spain
- Institute of Biomedicine of the University of Barcelona (IBUB), University of Barcelona, Avda. Joan XXIII 27, 08028, Barcelona, Spain
- Spanish Biomedical Research Center in Diabetes and Associated Metabolic Diseases (CIBERDEM) - National Institute of Health Carlos III, 28029, Madrid, Spain
- Pediatric Research Institute-Hospital Sant Joan de Déu, 08950, Esplugues de Llobregat, Barcelona, Spain
| | - Daniel Ortuño-Sahagún
- Laboratorio de Neuroinmunología Molecular, Instituto de Investigación de Ciencias Biomédicas (IICB) CUCS, Universidad de Guadalajara, 44340, Guadalajara, Jalisco, Mexico
| | - Manuel Vázquez-Carrera
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, University of Barcelona, Avda. Joan XXIII 27, 08028, Barcelona, Spain
- Institute of Biomedicine of the University of Barcelona (IBUB), University of Barcelona, Avda. Joan XXIII 27, 08028, Barcelona, Spain
- Spanish Biomedical Research Center in Diabetes and Associated Metabolic Diseases (CIBERDEM) - National Institute of Health Carlos III, 28029, Madrid, Spain
- Pediatric Research Institute-Hospital Sant Joan de Déu, 08950, Esplugues de Llobregat, Barcelona, Spain
| | - Mercè Pallàs
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, University of Barcelona, Avda. Joan XXIII 27, 08028, Barcelona, Spain
- Institute of Neurosciences of the University of Barcelona, University of Barcelona, 08035, Barcelona, Spain
- Centro de Investigación en Red, Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Laura Herrero
- Department of Biochemistry and Physiology, School of Pharmacy and Food Sciences, University of Barcelona, Avda. Joan XXIII 27, 08028, Barcelona, Spain
- Institute of Biomedicine of the University of Barcelona (IBUB), University of Barcelona, Avda. Joan XXIII 27, 08028, Barcelona, Spain
- Centro de Investigación Biomédica en Red (CIBER) de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, 28029, Madrid, Spain
| | - Christian Griñán-Ferré
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, University of Barcelona, Avda. Joan XXIII 27, 08028, Barcelona, Spain.
- Institute of Neurosciences of the University of Barcelona, University of Barcelona, 08035, Barcelona, Spain.
- Centro de Investigación en Red, Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain.
| |
Collapse
|
2
|
Pinto AP, Muñoz VR, Tavares MEA, Neto IVDS, Dos Santos JR, Rodrigues GS, Carolino ROG, Alberici LC, Simabuco FM, Teixeira GR, Pauli JR, de Moura LP, Cintra DE, Ropelle ER, Freitas EC, Rivas DA, da Silva ASR. Short-term exercise counteracts accelerated ageing impacts on physical performance and liver health in mice. Clin Exp Pharmacol Physiol 2024; 51:e70001. [PMID: 39477552 DOI: 10.1111/1440-1681.70001] [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: 07/02/2024] [Revised: 09/15/2024] [Accepted: 10/01/2024] [Indexed: 01/06/2025]
Abstract
Senescence impairs liver physiology, mitochondrial function and circadian regulation, resulting in systemic metabolic dysregulation. Given the limited research on the effects of combined exercise on an ageing liver, this study aimed to evaluate its impact on liver metabolism, circadian rhythms and mitochondrial function in senescence-accelerated mouse-prone 8 (SAMP8) and senescence-accelerated mouse-resistant 1 (SAMR1) mice. Histological, reverse transcription quantitative polymerase chain reaction (RT-qPCR) and immunoblotting analyses were conducted, supplemented by transcriptomic data sets and AML12 hepatocyte studies. Sedentary SAMP8 mice exhibited decreased muscle strength, reduced mitochondrial complex I levels and increased lipid droplet accumulation. In contrast, combined exercise mitigated muscle strength loss, upregulated proteins involved in mitochondrial complexes (CIII, CIV, CV) and increased Bmal1 messenger RNA (mRNA) expression in the liver. These molecular adaptations are associated with healthier liver phenotypes and may influence metabolic function and cellular longevity. Notably, elevated lipid content in aged mice was reduced post-exercise, indicating liver benefits even after a relatively short intervention. The combined exercise regimen did not improve aerobic capacity, likely due to the low volume and brief duration of running. Moreover, no significant effects were observed in SAMR1 mice, possibly because the training intensity was insufficient for younger, healthier animals. These findings underscore the potential of combined strength and endurance exercise to attenuate age-related liver dysfunction, particularly in ageing populations.
Collapse
Affiliation(s)
- Ana P Pinto
- School of Physical Education and Sport of Ribeirão Preto, University of São Paulo (USP), São Paulo, Brazil
| | - Vitor R Muñoz
- School of Physical Education and Sport of Ribeirão Preto, University of São Paulo (USP), São Paulo, Brazil
| | - Maria Eduarda A Tavares
- Department of Physical Education, State University of São Paulo (UNESP), São Paulo, Brazil
- Multicentric Program of Postgraduate in Physiological Sciences, School of Dentistry of Araçatuba, São Paulo State University (UNESP), São Paulo, Brazil
| | - Ivo V de Sousa Neto
- School of Physical Education and Sport of Ribeirão Preto, University of São Paulo (USP), São Paulo, Brazil
| | - Jonathas R Dos Santos
- Department of Biomolecular Sciences, School of Pharmaceutical Sciences of Ribeirao Preto, University of São Paulo-FCFRP USP, São Paulo, Brazil
| | - Guilherme S Rodrigues
- Department of Internal Medicine, Ribeirão Preto Medical School, University of São Paulo (USP), São Paulo, Brazil
| | - Ruither O Gomes Carolino
- School of Physical Education and Sport of Ribeirão Preto, University of São Paulo (USP), São Paulo, Brazil
| | - Luciane C Alberici
- Department of Biomolecular Sciences, School of Pharmaceutical Sciences of Ribeirao Preto, University of São Paulo-FCFRP USP, São Paulo, Brazil
| | - Fernando M Simabuco
- Laboratory of Molecular Biology of Exercise (LaBMEx), School of Applied Sciences, University of Campinas (UNICAMP), São Paulo, Brazil
- Department of Biochemistry, Federal University of São Paulo (UNIFESP), São Paulo, Brazil
| | - Giovana R Teixeira
- Department of Physical Education, State University of São Paulo (UNESP), São Paulo, Brazil
- Multicentric Program of Postgraduate in Physiological Sciences, School of Dentistry of Araçatuba, São Paulo State University (UNESP), São Paulo, Brazil
| | - José R Pauli
- Laboratory of Molecular Biology of Exercise (LaBMEx), School of Applied Sciences, University of Campinas (UNICAMP), São Paulo, Brazil
- Nutrigenomics and Lipids Research Center, CELN, School of Applied Sciences, UNICAMP, São Paulo, Brazil
| | - Leandro P de Moura
- Laboratory of Molecular Biology of Exercise (LaBMEx), School of Applied Sciences, University of Campinas (UNICAMP), São Paulo, Brazil
| | - Dennys E Cintra
- Laboratory of Molecular Biology of Exercise (LaBMEx), School of Applied Sciences, University of Campinas (UNICAMP), São Paulo, Brazil
- Nutrigenomics and Lipids Research Center, CELN, School of Applied Sciences, UNICAMP, São Paulo, Brazil
| | - Eduardo R Ropelle
- Laboratory of Molecular Biology of Exercise (LaBMEx), School of Applied Sciences, University of Campinas (UNICAMP), São Paulo, Brazil
- Nutrigenomics and Lipids Research Center, CELN, School of Applied Sciences, UNICAMP, São Paulo, Brazil
| | - Ellen C Freitas
- School of Physical Education and Sport of Ribeirão Preto, University of São Paulo (USP), São Paulo, Brazil
- Department of Health Sciences, Ribeirão Preto Medical School, University of São Paulo (USP), São Paulo, Brazil
| | - Donato A Rivas
- Center for Exercise Medicine Research, Fralin Biomedical Research Institute, Virginia Tech Carilion, Roanoke, Virginia, USA
| | - Adelino S R da Silva
- School of Physical Education and Sport of Ribeirão Preto, University of São Paulo (USP), São Paulo, Brazil
- Postgraduate Program in Rehabilitation and Functional Performance, Ribeirão Preto Medical School, University of São Paulo (USP), São Paulo, Brazil
| |
Collapse
|
3
|
Liu Q, Liu M, Yang T, Wang X, Cheng P, Zhou H. What can we do to optimize mitochondrial transplantation therapy for myocardial ischemia-reperfusion injury? Mitochondrion 2023; 72:72-83. [PMID: 37549815 DOI: 10.1016/j.mito.2023.08.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 05/20/2023] [Accepted: 08/04/2023] [Indexed: 08/09/2023]
Abstract
Mitochondrial transplantation is a promising solution for the heart following ischemia-reperfusion injury due to its capacity to replace damaged mitochondria and restore cardiac function. However, many barriers (such as inadequate mitochondrial internalization, poor survival of transplanted mitochondria, few mitochondria colocalized with cardiac cells) compromise the replacement of injured mitochondria with transplanted mitochondria. Therefore, it is necessary to optimize mitochondrial transplantation therapy to improve clinical effectiveness. By analogy, myocardial ischemia-reperfusion injury is like a withered flower, it needs to absorb enough nutrients to recover and bloom. In this review, we present a comprehensive overview of "nutrients" (source of exogenous mitochondria and different techniques for mitochondrial isolation), "absorption" (mitochondrial transplantation approaches, mitochondrial transplantation dose and internalization mechanism), and "flowering" (the mechanism of mitochondrial transplantation in cardioprotection) for myocardial ischemia-reperfusion injury.
Collapse
Affiliation(s)
- Qian Liu
- Institute of Cardiovascular Disease of Integrated Traditional Chinese Medicine and Western Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Meng Liu
- Comprehensive treatment area of Traditional Chinese Medicine, Guanghua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Tianshu Yang
- Institute of Cardiovascular Disease of Integrated Traditional Chinese Medicine and Western Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xinting Wang
- Institute of Cardiovascular Disease of Integrated Traditional Chinese Medicine and Western Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Peipei Cheng
- Institute of Cardiovascular Disease of Integrated Traditional Chinese Medicine and Western Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Hua Zhou
- Institute of Cardiovascular Disease of Integrated Traditional Chinese Medicine and Western Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| |
Collapse
|
4
|
Kepchia D, Huang L, Currais A, Liang Z, Fischer W, Maher P. The Alzheimer's disease drug candidate J147 decreases blood plasma fatty acid levels via modulation of AMPK/ACC1 signaling in the liver. Biomed Pharmacother 2022; 147:112648. [PMID: 35051863 PMCID: PMC8854339 DOI: 10.1016/j.biopha.2022.112648] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 01/07/2022] [Accepted: 01/12/2022] [Indexed: 12/30/2022] Open
Abstract
J147 is a novel drug candidate developed to treat neurological dysfunction. Numerous studies have demonstrated the beneficial effects of J147 in cellular and animal models of disease which has led to the transitioning of the compound into human clinical trials. However, no biomarkers for its target engagement have been identified. Here, we determined if specific metabolites in the plasma could be indicative of J147's activity in vivo. Plasma lipidomics data from three independent rodent studies were assessed along with liver lipidomics data from one of the studies. J147 consistently reduced plasma free fatty acid (FFA) levels across the independent studies. Decreased FFA levels were also found in the livers of J147-treated mice that correlated well with those in the plasma. These changes in the liver were associated with activation of the AMP-activated protein kinase/acetyl-CoA carboxylase 1 signaling pathway. A reduction in FFA levels by J147 was confirmed in HepG2 cells, where activation of the AMPK/ACC1 pathway was seen along with increases in acetyl-CoA and ATP levels which correlated with enhanced cellular bioenergetics. Our data show that J147 targets liver cells to activate the AMPK/ACC1 signaling pathway and preserve energy at the expense of inhibiting FFA synthesis.
Collapse
Affiliation(s)
- Devin Kepchia
- Cellular Neurobiology Laboratory, The Salk Institute for Biological Studies, 10010 N. Torrey Pines Rd., La Jolla, CA 92037, USA.
| | - Ling Huang
- The Razavi Newman Integrative Genomics and Bioinformatics
Core, The Salk Institute for Biological Studies, 10010 N. Torrey Pines Rd. La Jolla,
CA 92037, USA
| | - Antonio Currais
- Cellular Neurobiology Laboratory, The Salk Institute for
Biological Studies, 10010 N. Torrey Pines Rd. La Jolla, CA 92037, USA
| | - Zhibin Liang
- Cellular Neurobiology Laboratory, The Salk Institute for
Biological Studies, 10010 N. Torrey Pines Rd. La Jolla, CA 92037, USA
| | - Wolfgang Fischer
- Cellular Neurobiology Laboratory, The Salk Institute for
Biological Studies, 10010 N. Torrey Pines Rd. La Jolla, CA 92037, USA
| | - Pamela Maher
- Cellular Neurobiology Laboratory, The Salk Institute for Biological Studies, 10010 N. Torrey Pines Rd., La Jolla, CA 92037, USA.
| |
Collapse
|
5
|
Li R, Li Y, Tian M, Zhang H, Lou L, Liu K, Zhang J, Zhao Y, Zhang J, Le S, Fu X, Zhou Y, Li W, Gao X, Nie Y. Comparative proteomic profiling reveals a pathogenic role for the O-GlcNAcylated AIMP2-PARP1 complex in aging-related hepatic steatosis in mice. FEBS Lett 2022; 596:128-145. [PMID: 34817071 DOI: 10.1002/1873-3468.14242] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2021] [Revised: 10/29/2021] [Accepted: 11/15/2021] [Indexed: 11/07/2022]
Abstract
The prevalence of non-alcoholic fatty liver disease (NAFLD) increases with aging. However, the mechanism of aging-related NAFLD remains unclear. Herein, we constructed an aging-related hepatic steatosis model and analyzed the differentially expressed proteins (DEPs) in livers from young and old mice using liquid chromatography-mass spectrometry. Five hundred and eighty-eight aging-related DEPs and novel pathways were identified. Aminoacyl tRNA synthetase complex-interacting multifunctional protein 2 (AIMP2), the most significantly upregulated protein, promoted poly(ADP-ribose) polymerase 1 (PARP1) activation in aging-related hepatic steatosis. Additionally, mice liver-specific O-GlcNAcase knockout promoted AIMP2 and PARP1 expression. O-GlcNAc transferase (OGT) overexpression and O-GlcNAcase inhibition by genetic or pharmaceutical manipulations increased AIMP2 and PARP1 levels in vitro. Mechanistically, O-GlcNAcylation increased AIMP2 protein stability, leading to its aggregation. Our study reveals O-GlcNAcylated AIMP2 as a novel pathogenic regulator of aging-related hepatic steatosis.
Collapse
Affiliation(s)
- Renlong Li
- State key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Xijing Hospital of Digestive Diseases, Air Force Military Medical University, Xi'an, China
| | - Yan Li
- State key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Xijing Hospital of Digestive Diseases, Air Force Military Medical University, Xi'an, China
| | - Miaomiao Tian
- State key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Xijing Hospital of Digestive Diseases, Air Force Military Medical University, Xi'an, China
| | - Haohao Zhang
- State key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Xijing Hospital of Digestive Diseases, Air Force Military Medical University, Xi'an, China
| | - Lijun Lou
- State key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Xijing Hospital of Digestive Diseases, Air Force Military Medical University, Xi'an, China
| | - Kun Liu
- State key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Xijing Hospital of Digestive Diseases, Air Force Military Medical University, Xi'an, China
| | - Jiehao Zhang
- State key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Xijing Hospital of Digestive Diseases, Air Force Military Medical University, Xi'an, China
| | - Yu Zhao
- State key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Xijing Hospital of Digestive Diseases, Air Force Military Medical University, Xi'an, China
| | - Jing Zhang
- State key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Xijing Hospital of Digestive Diseases, Air Force Military Medical University, Xi'an, China
| | - Shuangshuang Le
- State key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Xijing Hospital of Digestive Diseases, Air Force Military Medical University, Xi'an, China
| | - Xin Fu
- State key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Xijing Hospital of Digestive Diseases, Air Force Military Medical University, Xi'an, China
| | - Yao Zhou
- State key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Xijing Hospital of Digestive Diseases, Air Force Military Medical University, Xi'an, China
| | - Wenjiao Li
- State key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Xijing Hospital of Digestive Diseases, Air Force Military Medical University, Xi'an, China
| | - Xianchun Gao
- State key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Xijing Hospital of Digestive Diseases, Air Force Military Medical University, Xi'an, China
| | - Yongzhan Nie
- State key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Xijing Hospital of Digestive Diseases, Air Force Military Medical University, Xi'an, China
| |
Collapse
|
6
|
Abstract
As the powerhouse of the cell, mitochondria, plays a crucial role in many aspects of life, whereby mitochondrial dysfunctions are associated with pathogenesis of many diseases, like neurodegenerative diseases, obesity, cancer, and metabolic as well as cardiovascular disorders. Mitochondria analysis frequently starts with isolation and enrichment procedures, which have become increasingly important in biomedical research. Unfortunately, isolation procedures can easily cause changes in the structural integrity of mitochondria during in vitro handling having impact on their function. This carries the risk that conclusions about isolated mitochondria may be drawn on the basis of experimental artifacts. Here we critically review a commonly used isolation procedure for mitochondria utilizing differential (gradient) centrifugation and depict major challenges to achieve "functional" mitochondria as basis for comprehensive physiological studies.
Collapse
Affiliation(s)
- Stefan Lehr
- Institute for Clinical Biochemistry and Pathobiochemistry, German Diabetes Center at the Heinrich-Heine-University Duesseldorf, Leibniz Center for Diabetes Research, Duesseldorf, Germany.
- German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany.
| | - Sonja Hartwig
- Institute for Clinical Biochemistry and Pathobiochemistry, German Diabetes Center at the Heinrich-Heine-University Duesseldorf, Leibniz Center for Diabetes Research, Duesseldorf, Germany
- German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany
| | - Jorg Kotzka
- Institute for Clinical Biochemistry and Pathobiochemistry, German Diabetes Center at the Heinrich-Heine-University Duesseldorf, Leibniz Center for Diabetes Research, Duesseldorf, Germany
- German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany
| |
Collapse
|
7
|
Kepchia D, Currais A, Dargusch R, Finley K, Schubert D, Maher P. Geroprotective effects of Alzheimer's disease drug candidates. Aging (Albany NY) 2021; 13:3269-3289. [PMID: 33550278 PMCID: PMC7906177 DOI: 10.18632/aging.202631] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2020] [Accepted: 01/14/2021] [Indexed: 04/18/2023]
Abstract
Geroprotectors are compounds that slow the biological aging process in model organisms and may therefore extend healthy lifespan in humans. It is hypothesized that they do so by preserving the more youthful function of multiple organ systems. However, this hypothesis has rarely been tested in any organisms besides C. elegans and D. melanogaster. To determine if two life-extending compounds for Drosophila maintain a more youthful phenotype in old mice, we asked if they had anti-aging effects in both the brain and kidney. We utilized rapidly aging senescence-accelerated SAMP8 mice to investigate age-associated protein level alterations in these organs. The test compounds were two cognition-enhancing Alzheimer's disease drug candidates, J147 and CMS121. Mice were fed the compounds in the last quadrant of their lifespan, when they have cognitive deficits and are beginning to develop CKD. Both compounds improved physiological markers for brain and kidney function. However, these two organs had distinct, tissue-specific protein level alterations that occurred with age, but in both cases, drug treatments restored a more youthful level. These data show that geroprotective AD drug candidates J147 and CMS121 prevent age-associated disease in both brain and kidney, and that their apparent mode of action in each tissue is distinct.
Collapse
Affiliation(s)
- Devin Kepchia
- Cellular Neurobiology Laboratory, The Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Antonio Currais
- Cellular Neurobiology Laboratory, The Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Richard Dargusch
- Cellular Neurobiology Laboratory, The Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Kim Finley
- Donald P. Shiley BioScience Center, San Diego State University, San Diego, CA 92115, USA
| | - David Schubert
- Cellular Neurobiology Laboratory, The Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Pamela Maher
- Cellular Neurobiology Laboratory, The Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| |
Collapse
|
8
|
Gil-Hernández A, Silva-Palacios A. Relevance of endoplasmic reticulum and mitochondria interactions in age-associated diseases. Ageing Res Rev 2020; 64:101193. [PMID: 33069818 DOI: 10.1016/j.arr.2020.101193] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 09/29/2020] [Accepted: 10/05/2020] [Indexed: 02/06/2023]
Abstract
Although the elixir of youth remains in the darkness, medical and scientific advances have succeeded in increasing human longevity; however, the predisposition to disease and its high economic cost are raising. Different strategies (e.g., antioxidants) and signaling pathways (e.g., Nrf2) have been identified to help regulate disease progression, nevertheless, there are still missing links that we need to understand. Contact sites called mitochondria-associated membranes (MAM) allow bi-directional communication between organelles as part of the essential functions in the cell to maintain its homeostasis. Different groups have deeply studied the role of MAM in aging; however, it's necessary to analyze their involvement in the progression of age-related diseases. In this review, we highlight the role of contact sites in these conditions, as well as the morphological and functional changes of mitochondria and ER in aging. We emphasize the intimate relationship between both organelles as a reflection of the biological processes that take place in the cell to try to regulate the deterioration characteristic of the aging process; proposing MAM as a potential target to help limit the disease progression with age.
Collapse
|
9
|
Asokan SM, Wang T, Wang MF, Lin WT. A novel dipeptide from potato protein hydrolysate augments the effects of exercise training against high-fat diet-induced damages in senescence-accelerated mouse-prone 8 by boosting pAMPK / SIRT1/ PGC-1α/ pFOXO3 pathway. Aging (Albany NY) 2020; 12:7334-7349. [PMID: 32335547 PMCID: PMC7202530 DOI: 10.18632/aging.103081] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Accepted: 03/29/2020] [Indexed: 12/13/2022]
Abstract
The pathological effects of obesity are often severe in aging condition. Although exercise training is found to be advantageous, the intensity of exercise performed is limited in aging condition. Therefore in this study we assessed the effect of a combined treatment regimen with a short-peptide IF isolated from alcalase potato-protein hydrolysates and a moderate exercise training for 15 weeks in a 6 month old HFD induced obese senescence accelerated mouse-prone 8 (SAMP8) mice model. Animals were divided into 6 groups (n=6) (C:Control+BSA); (HF:HFD+BSA); (EX:Control+ BSA+Exercise); (HF+IF:HFD+ IF); (HF+EX:HFD+Exercise); (HF+EX+IF:HFD+Exercise+IF). A moderate incremental swimming exercise training was provided for 6 weeks and after 3 weeks of exercise, IF was orally administered (1 mg/kg body Weight). The results show that combined administration of IF and exercise provides a better protection to aging animals by reducing body weight and regulated tissue damage. IF intake and exercise training provided protection against cardiac hypertrophy and maintains the tissue homeostasis in the heart and liver sections. Interestingly, IF and exercise training showed an effective upregulation in pAMPK/ SIRT1/ PGC-1α/ pFOXO3 mechanism of cellular longevity. Therefore, exercise training with IF intake is a possible strategy for anti-obesity benefits and superior cardiac and hepatic protection in aging condition.
Collapse
Affiliation(s)
- Shibu Marthandam Asokan
- Cardiovascular and Mitochondria Related Disease Research Center, Buddhist Tzu Chi Hospital, Hualien, Taiwan.,Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan
| | - Ting Wang
- Department of Hospitality Management, College of Agriculture, Tunghai University, Taichung, Taiwan
| | - Ming-Fu Wang
- Department of Food and Nutrition, Providence University, Taichung, Taiwan
| | - Wan-Teng Lin
- Department of Hospitality Management, College of Agriculture, Tunghai University, Taichung, Taiwan.,Department of Senior Wellness and Sport Science, College of Agriculture, Tunghai University, Taichung, Taiwan
| |
Collapse
|
10
|
Quantitative proteomics to study aging in rabbit liver. Mech Ageing Dev 2020; 187:111227. [PMID: 32126221 DOI: 10.1016/j.mad.2020.111227] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 01/24/2020] [Accepted: 02/27/2020] [Indexed: 12/23/2022]
Abstract
Aging globally effects cellular and organismal metabolism across a range of mammalian species, including humans and rabbits. Rabbits (Oryctolagus cuniculus are an attractive model system of aging due to their genetic similarity with humans and their short lifespans. This model can be used to understand metabolic changes in aging especially in major organs such as liver where we detected pronounced variations in fat metabolism, mitochondrial dysfunction, and protein degradation. Such changes in the liver are consistent across several mammalian species however in rabbits the downstream effects of these changes have not yet been explored. We have applied proteomics to study changes in the liver proteins from young, middle, and old age rabbits using a multiplexing cPILOT strategy. This resulted in the identification of 2,586 liver proteins, among which 45 proteins had significant p < 0.05) changes with aging. Seven proteins were differentially-expressed at all ages and include fatty acid binding protein, aldehyde dehydrogenase, enoyl-CoA hydratase, 3-hydroxyacyl CoA dehydrogenase, apolipoprotein C3, peroxisomal sarcosine oxidase, adhesion G-protein coupled receptor, and glutamate ionotropic receptor kinate. Insights to how alterations in metabolism affect protein expression in liver have been gained and demonstrate the utility of rabbit as a model of aging.
Collapse
|
11
|
Huda N, Liu G, Hong H, Yan S, Khambu B, Yin XM. Hepatic senescence, the good and the bad. World J Gastroenterol 2019; 25:5069-5081. [PMID: 31558857 PMCID: PMC6747293 DOI: 10.3748/wjg.v25.i34.5069] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Revised: 07/25/2019] [Accepted: 08/07/2019] [Indexed: 02/06/2023] Open
Abstract
Gradual alterations of cell’s physiology and functions due to age or exposure to various stresses lead to the conversion of normal cells to senescent cells. Once becoming senescent, the cell stops dividing permanently but remains metabolically active. Cellular senescence does not have a single marker but is characterized mainly by a combination of multiple markers, such as, morphological changes, expression of cell cycle inhibitors, senescence associated β-galactosidase activity, and changes in nuclear membrane. When cells in an organ become senescent, the entire organism can be affected. This may occur through the senescence-associated secretory phenotype (SASP). SASP may exert beneficial or harmful effects on the microenvironment of tissues. Research on senescence has become a very exciting field in cell biology since the link between age-related diseases, including cancer, and senescence has been established. The loss of regenerative and homeostatic capacity of the liver over the age is somehow connected to cellular senescence. The major contributors of senescence properties in the liver are hepatocytes and cholangiocytes. Senescent cells in the liver have been implicated in the etiology of chronic liver diseases including cirrhosis and hepatocellular carcinoma and in the interference of liver regeneration. This review summarizes recently reported findings in the understanding of the molecular mechanisms of senescence and its relationship with liver diseases.
Collapse
Affiliation(s)
- Nazmul Huda
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, United States
| | - Gang Liu
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, United States
| | - Honghai Hong
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, United States
| | - Shengmin Yan
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, United States
| | - Bilon Khambu
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, United States
| | - Xiao-Ming Yin
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, United States
| |
Collapse
|
12
|
Yu HM, Yao XD, Zhang RM, Zhuang HF, Wang PW, Li YZ. Repression of let-7b-5p prevents the development of multifidus muscle dysfunction by promoting vitamin D accumulation via upregulation of electron transfer flavoprotein alpha subunit in a rat model of multifidus muscle injury. J Cell Biochem 2019; 120:7458-7473. [PMID: 30387180 DOI: 10.1002/jcb.28020] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Accepted: 10/15/2018] [Indexed: 01/24/2023]
Abstract
Multifidus muscle dysfunction is associated with the multifidus muscle injury (MMI), which ultimately result in the low-back pain. Increasing evidence shows that microRNAs (miRs) may be involved in multifidus muscle dysfunction. In this study, we tested the hypothesis that downregulation of let-7b-5p may inhibit the multifidus muscle dysfunction development and progression. The target prediction program and luciferase activity determination confirmed electron transfer flavoprotein alpha subunit (ETFA) as a direct target gene of let-7b-5p. To study the mechanisms and functions of let-7b-5p in relation to ETFA in MMI progression, we prepared rats with experimental MMI, and a lentivirus-based packaging system was designed to upregulate expressions of let-7b-5p, and downregulate the expression of ETFA. ETFA was identified as a target gene of let-7b-5p. Older age, a longer duration of pain, and higher visual analog scale and Oswestry disability index scores for the patients with chronic low-back pain were linked to a more severe degree of degenerative muscle atrophy and fatty infiltration. Increased expression of let-7b-5p and decreased expression of ETFA and vitamin D receptor (VDR) were positively correlated with multifidus muscle dysfunction. Downregulated let-7b-5p could inhibit infiltration of collagen fibers, reverse the ultrastructural changes of multifidus muscle, and induce the VDR expression, thereby repair the MMI. The results provided a potential basis for let-7b-5p that could support targeted intervention in multifidus muscle dysfunction. Collectively, this study confirmed that downregulation of let-7b-5p has a potential inhibitory effect on the development of the function of the musculus myocytes by upregulating ETFA.
Collapse
Affiliation(s)
- Hai-Ming Yu
- Department of Orthopaedics, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Xue-Dong Yao
- Department of Orthopaedics, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Rong-Mou Zhang
- Department of Orthopaedics, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Hua-Feng Zhuang
- Department of Orthopaedics, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Pei-Wen Wang
- Department of Orthopaedics, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Yi-Zhong Li
- Department of Orthopaedics, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| |
Collapse
|
13
|
Yin S, Gambe RG, Sun J, Martinez AZ, Cartun ZJ, Regis FFD, Wan Y, Fan J, Brooks AN, Herman SEM, Ten Hacken E, Taylor-Weiner A, Rassenti LZ, Ghia EM, Kipps TJ, Obeng EA, Cibulskis CL, Neuberg D, Campagna DR, Fleming MD, Ebert BL, Wiestner A, Leshchiner I, DeCaprio JA, Getz G, Reed R, Carrasco RD, Wu CJ, Wang L. A Murine Model of Chronic Lymphocytic Leukemia Based on B Cell-Restricted Expression of Sf3b1 Mutation and Atm Deletion. Cancer Cell 2019; 35:283-296.e5. [PMID: 30712845 PMCID: PMC6372356 DOI: 10.1016/j.ccell.2018.12.013] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Revised: 10/24/2018] [Accepted: 12/28/2018] [Indexed: 12/26/2022]
Abstract
SF3B1 is recurrently mutated in chronic lymphocytic leukemia (CLL), but its role in the pathogenesis of CLL remains elusive. Here, we show that conditional expression of Sf3b1-K700E mutation in mouse B cells disrupts pre-mRNA splicing, alters cell development, and induces a state of cellular senescence. Combination with Atm deletion leads to the overcoming of cellular senescence and the development of CLL-like disease in elderly mice. These CLL-like cells show genome instability and dysregulation of multiple CLL-associated cellular processes, including deregulated B cell receptor signaling, which we also identified in human CLL cases. Notably, human CLLs harboring SF3B1 mutations exhibit altered response to BTK inhibition. Our murine model of CLL thus provides insights into human CLL disease mechanisms and treatment.
Collapse
MESH Headings
- Adenine/analogs & derivatives
- Agammaglobulinaemia Tyrosine Kinase/antagonists & inhibitors
- Agammaglobulinaemia Tyrosine Kinase/metabolism
- Alternative Splicing
- Animals
- Antineoplastic Agents/pharmacology
- Ataxia Telangiectasia Mutated Proteins/deficiency
- Ataxia Telangiectasia Mutated Proteins/genetics
- Ataxia Telangiectasia Mutated Proteins/metabolism
- B-Lymphocytes/drug effects
- B-Lymphocytes/immunology
- B-Lymphocytes/metabolism
- Cellular Senescence/drug effects
- DNA Damage
- Gene Deletion
- Genetic Predisposition to Disease
- Genomic Instability
- Humans
- Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy
- Leukemia, Lymphocytic, Chronic, B-Cell/genetics
- Leukemia, Lymphocytic, Chronic, B-Cell/immunology
- Leukemia, Lymphocytic, Chronic, B-Cell/metabolism
- Mice, 129 Strain
- Mice, Inbred C57BL
- Mice, Knockout
- Mice, Mutant Strains
- Mutation
- Neoplasms, Experimental/drug therapy
- Neoplasms, Experimental/genetics
- Neoplasms, Experimental/immunology
- Neoplasms, Experimental/metabolism
- Phenotype
- Phosphoproteins/genetics
- Phosphoproteins/metabolism
- Piperidines
- Protein Kinase Inhibitors/pharmacology
- Pyrazoles/pharmacology
- Pyrimidines/pharmacology
- RNA Splicing Factors/genetics
- RNA Splicing Factors/metabolism
- Receptors, Antigen, B-Cell/immunology
- Receptors, Antigen, B-Cell/metabolism
- Signal Transduction
- Tumor Cells, Cultured
Collapse
Affiliation(s)
- Shanye Yin
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA; Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Rutendo G Gambe
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Jing Sun
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | | | - Zachary J Cartun
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Fara Faye D Regis
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Youzhong Wan
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Jean Fan
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
| | | | - Sarah E M Herman
- Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Elisa Ten Hacken
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | | | - Laura Z Rassenti
- Moores Cancer Center, University of California, San Diego, La Jolla, CA 92093, USA
| | - Emanuela M Ghia
- Moores Cancer Center, University of California, San Diego, La Jolla, CA 92093, USA
| | - Thomas J Kipps
- Moores Cancer Center, University of California, San Diego, La Jolla, CA 92093, USA
| | | | | | - Donna Neuberg
- Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Dean R Campagna
- Department of Pathology, Boston Children's Hospital, Boston, MA, USA
| | - Mark D Fleming
- Department of Pathology, Boston Children's Hospital, Boston, MA, USA
| | - Benjamin L Ebert
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA; Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Adrian Wiestner
- Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | | | - James A DeCaprio
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Gad Getz
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Robin Reed
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Ruben D Carrasco
- Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA; Department of Oncologic Pathology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Catherine J Wu
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA; Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA.
| | - Lili Wang
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA; Department of Systems Biology, Beckman Research Institute, City of Hope, Monrovia, CA, USA.
| |
Collapse
|
14
|
Mitoproteomics: Tackling Mitochondrial Dysfunction in Human Disease. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:1435934. [PMID: 30533169 PMCID: PMC6250043 DOI: 10.1155/2018/1435934] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Accepted: 08/29/2018] [Indexed: 12/11/2022]
Abstract
Mitochondria are highly dynamic and regulated organelles that historically have been defined based on their crucial role in cell metabolism. However, they are implicated in a variety of other important functions, making mitochondrial dysfunction an important axis in several pathological contexts. Despite that conventional biochemical and molecular biology approaches have provided significant insight into mitochondrial functionality, innovative techniques that provide a global view of the mitochondrion are still necessary. Proteomics fulfils this need by enabling accurate, systems-wide quantitative analysis of protein abundance. More importantly, redox proteomics approaches offer unique opportunities to tackle oxidative stress, a phenomenon that is intimately linked to aging, cardiovascular disease, and cancer. In addition, cutting-edge proteomics approaches reveal how proteins exert their functions in complex interaction networks where even subtle alterations stemming from early pathological states can be monitored. Here, we describe the proteomics approaches that will help to deepen the role of mitochondria in health and disease by assessing not only changes to mitochondrial protein composition but also alterations to their redox state and how protein interaction networks regulate mitochondrial function and dynamics. This review is aimed at showing the reader how the application of proteomics approaches during the last 20 years has revealed crucial mitochondrial roles in the context of aging, neurodegenerative disorders, metabolic disease, and cancer.
Collapse
|
15
|
Li P, Yang H, Liu G, Ma W, Li C, Huo H, He J, Liu L. PpSARK Regulates Moss Senescence and Salt Tolerance through ABA Related Pathway. Int J Mol Sci 2018; 19:E2609. [PMID: 30177627 PMCID: PMC6163601 DOI: 10.3390/ijms19092609] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Revised: 08/30/2018] [Accepted: 08/30/2018] [Indexed: 11/18/2022] Open
Abstract
Senescence-associated receptor-like kinase (SARK) family members in Arabidopsis, soybean, and rice are known to be positive regulators of leaf senescence. In the meantime, SARKs are extensively involved in stress response. However, their function and underlying molecular mechanism in stress responses in moss are not well known. Here, we investigated functional roles of SARK isolated from Physcomitrella patens (PpSARK) in salt stress response and senescence. PpSARK transcripts significantly accumulated under NaCl and abscisic acid (ABA) treatments, with higher expression in the moss gametophyte stage. Insertional gain-of-function mutants of PpSARK (PpSARKg) were more tolerant to salt stress and ABA than wild type (WT), whereas senescence of mutants was delayed during the protonema stage. Expression of stress-responsive genes in the ABA related pathway, such as PpABI3, PpABI5, PpPP2C, and PpLEA were significantly higher in PpSARKg and WT under salt stress conditions, suggesting that PpSARK might positively regulate salt tolerance via an ABA-related pathway. Endogenous ABA contents also increased 3-fold under salt stress conditions. These results indicate that PpSARK functions as a positive regulator in salt stress responses, while possibly functioning as a negative regulator in senescence in moss.
Collapse
Affiliation(s)
- Ping Li
- Key Laboratory of Economic Plants and Biotechnology, Germplasm Bank of Wild Species, Kunming Institute of Botany, Chinese Academy of Sciences, Yunnan Key Laboratory for Wild Plant Resources, Kunming 650201, China.
- University of Chinese Academy of Sciences, Beijing 100049, China.
| | - Hong Yang
- Key Laboratory of Economic Plants and Biotechnology, Germplasm Bank of Wild Species, Kunming Institute of Botany, Chinese Academy of Sciences, Yunnan Key Laboratory for Wild Plant Resources, Kunming 650201, China.
- University of Chinese Academy of Sciences, Beijing 100049, China.
| | - Gaojing Liu
- Key Laboratory of Economic Plants and Biotechnology, Germplasm Bank of Wild Species, Kunming Institute of Botany, Chinese Academy of Sciences, Yunnan Key Laboratory for Wild Plant Resources, Kunming 650201, China.
| | - Wenzhang Ma
- Key Laboratory of Economic Plants and Biotechnology, Germplasm Bank of Wild Species, Kunming Institute of Botany, Chinese Academy of Sciences, Yunnan Key Laboratory for Wild Plant Resources, Kunming 650201, China.
| | - Chuanhong Li
- National Key Laboratory of Crop Genetic Improvement and National Centre of Plant Gene Research, Huazhong Agricultural University, Wuhan 430070, China.
| | - Heqiang Huo
- Mid-Florida Research and Education Center, Department of Environmental Horticulture, University of Florida, Apopka, FL 32703, USA.
| | - Jianfang He
- Key Laboratory of Economic Plants and Biotechnology, Germplasm Bank of Wild Species, Kunming Institute of Botany, Chinese Academy of Sciences, Yunnan Key Laboratory for Wild Plant Resources, Kunming 650201, China.
- University of Chinese Academy of Sciences, Beijing 100049, China.
| | - Li Liu
- Key Laboratory of Economic Plants and Biotechnology, Germplasm Bank of Wild Species, Kunming Institute of Botany, Chinese Academy of Sciences, Yunnan Key Laboratory for Wild Plant Resources, Kunming 650201, China.
| |
Collapse
|
16
|
Bloomer SA, Wellen KE, Henderson GC. Sexual dimorphism in the hepatic protein response to a moderate trans fat diet in senescence-accelerated mice. Lipids Health Dis 2017; 16:243. [PMID: 29237473 PMCID: PMC5729490 DOI: 10.1186/s12944-017-0639-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Accepted: 12/05/2017] [Indexed: 01/15/2023] Open
Abstract
BACKGROUND Aging is characterized by increases in inflammation and oxidative stress, conditions that are exacerbated by environmental factors such as diet. In this study, we investigated the effects of a trans-fatty acid (TFA) diet on the liver in adult (25 wk) and old (60 wk) senescence-accelerated mice (SAMP8 strain) of both sexes. Our goal was to assess the effects of the diet on protein markers of inflammation and oxidative stress in the liver. METHODS Male and female mice were placed on life-long diets containing similar amounts of total fat (17%), with differing amounts of TFA: 2% (moderate TFA group) or 0.2% of total energy from TFA (control diet group). At the indicated ages, livers were harvested and evaluated for markers of inflammation and oxidative stress, as well as for enzymes of fat metabolism via immunoblotting. Relative densities of protein bands were determined and compared via a three-factor ANOVA. RESULTS Compared to males, females demonstrated significantly lower inflammatory protein expression (ICAM-1, MCP-1, COX-2), along with lower expression of the DNA damage marker, Gadd153, and the oxidative stress marker, HO-1. Female mice demonstrated higher expression of antioxidant enzymes (SOD-1, SOD-2, and Ref-1) and lipogenic enzymes (FASN, ACLY) compared to male mice. While HO-1 was elevated in the female mice fed the TFA diet compared to controls, the diet did not affect other markers of oxidative stress or inflammation. However, the diet was associated with significant increases in FASN and ACLY in adult (25 wk) male mice. CONCLUSIONS Our results suggest sexually dimorphic protein expression in the liver, with female mice demonstrating lower inflammation and increased oxidative stress defenses. Additionally, considering that FASN and ACLY contribute to hepatic lipogenesis, our results suggest a potential mechanism for the dyslipidemia in adult male mice that is associated with TFA diets.
Collapse
Affiliation(s)
- Steven A Bloomer
- Division of Science and Engineering, Penn State University, Abington College, 1600 Woodland Rd, Abington, PA, 19001, USA.
| | - Kathryn E Wellen
- Department of Cancer Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA.,Abramson Family Cancer Research Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | | |
Collapse
|
17
|
Liu HW, Chan YC, Wei CC, Chen YA, Wang MF, Chang SJ. An alternative model for studying age-associated metabolic complications: Senescence-accelerated mouse prone 8. Exp Gerontol 2017; 99:61-68. [PMID: 28843510 DOI: 10.1016/j.exger.2017.08.023] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Revised: 08/13/2017] [Accepted: 08/16/2017] [Indexed: 12/14/2022]
Abstract
Rodent animal models take at least 18months to develop aging phenotypes for researchers to investigate the mechanism of age-related metabolic complications. Senescence-accelerated mouse prone 8 (SAMP8) shortens the process of aging and may facilitate an alternative model for studying age-related insulin resistance. The short-lived strain SAMP8 and two long-lived strains SAM resistant 1 (SAMR1) mice and C57BL/6 mice at 12 (young) and 40weeks old (old) were used in the present study. Glucose tolerance test, histology and signaling pathways involved in lipid metabolism in adipose tissue and liver and key components of insulin signaling pathway in the skeletal muscle were determined in these three strains. We found that short-lived SAMP8 mice developed symptoms of insulin resistance including hyperglycemia, hyperinsulinemia, and impaired glucose tolerance in association with adipocyte hypertrophy and ectopic lipid accumulation in liver and muscle at 40-wk.-old. Significantly increased serum IL-6, leptin, and resistin levels and adipogenic transcription factor PPARγ and macrophage marker F4/80 mRNA expression in adipose tissues were observed in old SAMP8 mice, compared with that in young SAMP8 mice. Marked increases in SREBP1 and PPARγ and a decrease in PPARα at mRNA level in accordance with activation of mTOR/Akt pathway were contributed to hepatic lipid accumulation in old SAMP8 mice. Down-regulation of insulin signaling pathway including IRβ, IRS1, and AS160 at protein level in skeletal muscle was observed in old SAMP8 mice. At 40-wk.-old, both long-lived SAMR1 and C57BL/6 mice have not been fully developed age-related metabolic disorders including insulin resistance and visceral fat expansion in line with fewer defects in lipid metabolism and skeletal muscle insulin signaling pathway. In conclusion, our data suggest the suitability of the SAMP8 mice as a model for studying age-related metabolic complications.
Collapse
Affiliation(s)
- Hung-Wen Liu
- Department of Life Sciences, National Cheng Kung University, Tainan, Taiwan; Department of Physical education, National Taiwan Normal University, Taipei, Taiwan
| | - Yin-Ching Chan
- Department of Food and Nutrition, Providence University, Taichung, Taiwan
| | - Chu-Chun Wei
- Department of Life Sciences, National Cheng Kung University, Tainan, Taiwan
| | - Yun-An Chen
- Department of Life Sciences, National Cheng Kung University, Tainan, Taiwan
| | - Ming-Fu Wang
- Department of Food and Nutrition, Providence University, Taichung, Taiwan
| | - Sue-Joan Chang
- Department of Life Sciences, National Cheng Kung University, Tainan, Taiwan.
| |
Collapse
|
18
|
Cortegano I, Rodríguez M, Martín I, Prado MC, Ruíz C, Hortigüela R, Alía M, Vilar M, Mira H, Cano E, Domínguez M, de Andrés B, Gaspar ML. Altered marginal zone and innate-like B cells in aged senescence-accelerated SAMP8 mice with defective IgG1 responses. Cell Death Dis 2017; 8:e3000. [PMID: 28817118 PMCID: PMC5596542 DOI: 10.1038/cddis.2017.351] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Revised: 06/27/2017] [Accepted: 07/02/2017] [Indexed: 12/31/2022]
Abstract
Aging has a strong impact on the activity of the immune system, enhancing susceptibility to pathogens and provoking a predominant pre-inflammatory status, whereas dampening responses to vaccines in humans and mice. Here, we demonstrate a loss of marginal zone B lymphocytes (MZ, CD19+CD45R+CD21++CD23lo) and a decrease of naive B cells (CD19+IgD+), whereas there is an enhancement of a CD19+CD45Rlo innate-like B cell population (B1REL) and the so-called aged B cell compartment (ABC, CD45R+CD21loCD23loCD5-CD11b-) in aged senescence-accelerated (SAMP8) mice but not in aged senescence-resistant (SAMR1) mice. These changes in aged SAMP8 mice were associated with lower IgG isotype levels, displaying low variable gene usage repertoires of the immunoglobulin heavy chain (VH) diversity, with a diminution on IgG1-memory B cells (CD11b-Gr1-CD138-IgM-IgD-CD19+CD38+IgG1+), an increase in T follicular helper (TFH, CD4+CXCR5+PD1+) cell numbers, and an altered MOMA-1 (metallophilic macrophages) band in primary follicles. LPS-mediated IgG1 responses were impaired in the B1REL and ABC cell compartments, both in vitro and in vivo. These data demonstrate the prominent changes to different B cell populations and in structural follicle organization that occur upon aging in SAMP8 mice. These novel results raise new questions regarding the importance of the cellular distribution in the B cell layers, and their effector functions needed to mount a coordinated and effective humoral response.
Collapse
MESH Headings
- Aging/genetics
- Aging/immunology
- Animals
- Antigens, CD/genetics
- Antigens, CD/immunology
- B-Lymphocytes/drug effects
- B-Lymphocytes/immunology
- B-Lymphocytes/pathology
- Cell Death/drug effects
- Cell Proliferation/drug effects
- Gene Expression Regulation, Developmental
- IgG Deficiency/genetics
- IgG Deficiency/metabolism
- IgG Deficiency/pathology
- Immunity, Humoral
- Immunity, Innate
- Immunoglobulin D/genetics
- Immunoglobulin D/metabolism
- Immunoglobulin G/genetics
- Immunoglobulin G/metabolism
- Immunoglobulin Heavy Chains
- Immunoglobulin M/genetics
- Immunoglobulin M/metabolism
- Immunologic Memory
- Lipopolysaccharides/pharmacology
- Mice, Inbred C57BL
- Mice, Transgenic
- Primary Cell Culture
- Signal Transduction
- Spleen/cytology
- Spleen/drug effects
- Spleen/immunology
- T-Lymphocytes, Helper-Inducer/cytology
- T-Lymphocytes, Helper-Inducer/drug effects
- T-Lymphocytes, Helper-Inducer/immunology
Collapse
Affiliation(s)
- Isabel Cortegano
- Centro Nacional de Microbiología, Instituto de Salud Carlos III (ISCIII), Majadahonda, Madrid, Spain
| | - Mercedes Rodríguez
- Centro Nacional de Microbiología, Instituto de Salud Carlos III (ISCIII), Majadahonda, Madrid, Spain
| | - Isabel Martín
- Centro Nacional de Microbiología, Instituto de Salud Carlos III (ISCIII), Majadahonda, Madrid, Spain
| | - Maria Carmen Prado
- Centro Nacional de Microbiología, Instituto de Salud Carlos III (ISCIII), Majadahonda, Madrid, Spain
| | - Carolina Ruíz
- Centro Nacional de Microbiología, Instituto de Salud Carlos III (ISCIII), Majadahonda, Madrid, Spain
| | - Rafael Hortigüela
- Unidad Funcional de Investigación en Enfermedades Crónicas, Instituto de Salud Carlos III (ISCIII), Majadahonda, 28220 Madrid, Spain
| | - Mario Alía
- Centro Nacional de Microbiología, Instituto de Salud Carlos III (ISCIII), Majadahonda, Madrid, Spain
| | - Marçal Vilar
- Instituto de Biomedicina de Valencia, Valencia 46010, Spain
| | - Helena Mira
- Instituto de Biomedicina de Valencia, Valencia 46010, Spain
| | - Eva Cano
- Unidad Funcional de Investigación en Enfermedades Crónicas, Instituto de Salud Carlos III (ISCIII), Majadahonda, 28220 Madrid, Spain
| | - Mercedes Domínguez
- Centro Nacional de Microbiología, Instituto de Salud Carlos III (ISCIII), Majadahonda, Madrid, Spain
| | - Belén de Andrés
- Centro Nacional de Microbiología, Instituto de Salud Carlos III (ISCIII), Majadahonda, Madrid, Spain
| | - María Luisa Gaspar
- Centro Nacional de Microbiología, Instituto de Salud Carlos III (ISCIII), Majadahonda, Madrid, Spain
| |
Collapse
|
19
|
Tang L, Luo JR, Li YL, Ge R, Li QS. Hepatotoxicity and proteomic mechanism of Di-n-butyl-di-(4-chlorobenzohydroxamato)tin(IV) (DBDCT) in vivo. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2017; 51:38-44. [PMID: 28273564 DOI: 10.1016/j.etap.2017.01.012] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/23/2016] [Revised: 01/17/2017] [Accepted: 01/19/2017] [Indexed: 06/06/2023]
Abstract
Di-n-butyl-di-(4-chlorobenzohydroxamato)tin(IV) (DBDCT) is an anti-tumour organotin(IV) compound with hepatotoxicity. To investigate the hepatotoxicity and mechanisms of DBDCT in vivo, proteomic technology 2D gel combined with MALDI-TOF-MS was used in our research. Results indicated that DBDCT increased AST, AKP and ACP activities and decreased ALT activity. Further, sporadic eosinophilic changes and nuclear pyknosis were visible in hepatic pathological observation. Proteomic analysis showed that twenty-two proteins involved in amino acid, nucleic acid, carbohydrate and lipid metabolism, stress response, multicellular organism development and cell apoptosis were differentially expressed and identified. Notably, a considerable amount of the altered proteins, such as OAT, HPPD, M2GD, GSTM2, Glud1, GSTa, HS90β and PDIA3 participated in multi-metabolic pathways and oxidative stress reactions. Our findings indicated that the inhibition of enzyme activity and oxidative stress were the major mechanisms by which DBDCT induced hepatotoxicity, and the altered proteins could be potential drug targets for the further design of new type of organic tin with high activity and low toxicology.
Collapse
Affiliation(s)
- Li Tang
- School of Pharmaceutical Science, Shanxi Medical University, Taiyuan 030001, PR China
| | - Jie-Ran Luo
- School of Pharmaceutical Science, Shanxi Medical University, Taiyuan 030001, PR China
| | - Yun-Lan Li
- School of Pharmaceutical Science, Shanxi Medical University, Taiyuan 030001, PR China
| | - Rui Ge
- School of Pharmaceutical Science, Shanxi Medical University, Taiyuan 030001, PR China
| | - Qing-Shan Li
- School of Pharmaceutical Science, Shanxi Medical University, Taiyuan 030001, PR China; Shanxi University of Traditional Chinese Medicine, Taiyuan 030024, PR China.
| |
Collapse
|
20
|
Zhang L, Hatzakis E, Nichols RG, Hao R, Correll J, Smith PB, Chiaro CR, Perdew GH, Patterson AD. Metabolomics Reveals that Aryl Hydrocarbon Receptor Activation by Environmental Chemicals Induces Systemic Metabolic Dysfunction in Mice. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2015; 49:8067-77. [PMID: 26023891 PMCID: PMC4890155 DOI: 10.1021/acs.est.5b01389] [Citation(s) in RCA: 73] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/17/2023]
Abstract
Environmental exposure to dioxins and dioxin-like compounds poses a significant health risk for human health. Developing a better understanding of the mechanisms of toxicity through activation of the aryl hydrocarbon receptor (AHR) is likely to improve the reliability of risk assessment. In this study, the AHR-dependent metabolic response of mice exposed to 2,3,7,8-tetrachlorodibenzofuran (TCDF) was assessed using global (1)H nuclear magnetic resonance (NMR)-based metabolomics and targeted metabolite profiling of extracts obtained from serum and liver. (1)H NMR analyses revealed that TCDF exposure suppressed gluconeogenesis and glycogenolysis, stimulated lipogenesis, and triggered inflammatory gene expression in an Ahr-dependent manner. Targeted analyses using gas chromatography coupled with mass spectrometry showed TCDF treatment altered the ratio of unsaturated/saturated fatty acids. Consistent with this observation, an increase in hepatic expression of stearoyl coenzyme A desaturase 1 was observed. In addition, TCDF exposure resulted in inhibition of de novo fatty acid biosynthesis manifested by down-regulation of acetyl-CoA, malonyl-CoA, and palmitoyl-CoA metabolites and related mRNA levels. In contrast, no significant changes in the levels of glucose and lipid were observed in serum and liver obtained from Ahr-null mice following TCDF treatment, thus strongly supporting the important role of the AHR in mediating the metabolic effects seen following TCDF exposure.
Collapse
Affiliation(s)
- Limin Zhang
- Center for Molecular Toxicology and Carcinogenesis, Department of Veterinary and Biomedical Sciences, The Pennsylvania State University, University Park, Pennsylvania, 16802, USA
- CAS Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Wuhan Centre for Magnetic Resonance, Wuhan Institute of Physics and Mathematics, Chinese Academy of Sciences (CAS), Wuhan 430071, China
| | - Emmanuel Hatzakis
- Department of Chemistry, The Pennsylvania State University, University Park, Pennsylvania, 16802, USA
| | - Robert G. Nichols
- Center for Molecular Toxicology and Carcinogenesis, Department of Veterinary and Biomedical Sciences, The Pennsylvania State University, University Park, Pennsylvania, 16802, USA
| | - Ruixin Hao
- Center for Molecular Toxicology and Carcinogenesis, Department of Veterinary and Biomedical Sciences, The Pennsylvania State University, University Park, Pennsylvania, 16802, USA
| | - Jared Correll
- Center for Molecular Toxicology and Carcinogenesis, Department of Veterinary and Biomedical Sciences, The Pennsylvania State University, University Park, Pennsylvania, 16802, USA
| | - Philip B. Smith
- Metabolomics Facility, The Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, Pennsylvania, 16802, USA
| | - Christopher R. Chiaro
- Center for Molecular Toxicology and Carcinogenesis, Department of Veterinary and Biomedical Sciences, The Pennsylvania State University, University Park, Pennsylvania, 16802, USA
| | - Gary H. Perdew
- Center for Molecular Toxicology and Carcinogenesis, Department of Veterinary and Biomedical Sciences, The Pennsylvania State University, University Park, Pennsylvania, 16802, USA
| | - Andrew D. Patterson
- Center for Molecular Toxicology and Carcinogenesis, Department of Veterinary and Biomedical Sciences, The Pennsylvania State University, University Park, Pennsylvania, 16802, USA
| |
Collapse
|
21
|
He J, Hao S, Zhang H, Guo F, Huang L, Xiao X, He D. Chronological protein synthesis in regenerating rat liver. Electrophoresis 2015; 36:1622-32. [DOI: 10.1002/elps.201500019] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2014] [Revised: 03/05/2015] [Accepted: 04/02/2015] [Indexed: 01/20/2023]
Affiliation(s)
- Jinjun He
- Key Laboratory of Cell Proliferation and Regulation Biology Ministry of Education; Universities of the Confederated Institute for Proteomics, Beijing Normal University; Beijing P. R. China
| | - Shuai Hao
- Key Laboratory of Cell Proliferation and Regulation Biology Ministry of Education; Universities of the Confederated Institute for Proteomics, Beijing Normal University; Beijing P. R. China
| | - Hao Zhang
- Key Laboratory of Cell Proliferation and Regulation Biology Ministry of Education; Universities of the Confederated Institute for Proteomics, Beijing Normal University; Beijing P. R. China
| | - Fuzheng Guo
- Key Laboratory of Cell Proliferation and Regulation Biology Ministry of Education; Universities of the Confederated Institute for Proteomics, Beijing Normal University; Beijing P. R. China
| | - Lingyun Huang
- Key Laboratory of Cell Proliferation and Regulation Biology Ministry of Education; Universities of the Confederated Institute for Proteomics, Beijing Normal University; Beijing P. R. China
| | - Xueyuan Xiao
- Key Laboratory of Cell Proliferation and Regulation Biology Ministry of Education; Universities of the Confederated Institute for Proteomics, Beijing Normal University; Beijing P. R. China
| | - Dacheng He
- Key Laboratory of Cell Proliferation and Regulation Biology Ministry of Education; Universities of the Confederated Institute for Proteomics, Beijing Normal University; Beijing P. R. China
| |
Collapse
|
22
|
Abstract
As the powerhouse of the cell, mitochondria play a crucial role in many aspects of life, whereby mitochondrial dysfunctions are associated with pathogenesis of many diseases, like neurodegenerative diseases, obesity, cancer, and metabolic as well as cardiovascular disorders. Mitochondria analysis frequently starts with isolation and enrichment procedures potentially affecting mitochondrial morphology having impact on their function. Due to the complex mitochondrial morphology, the major task is to preserve their structural integrity. Here we critically review a commonly used isolation procedure for mitochondria utilizing differential (gradient) centrifugation and depict major challenges to achieve "functional" mitochondria as basis for comprehensive physiological studies.
Collapse
|
23
|
Peinado JR, Diaz-Ruiz A, Frühbeck G, Malagon MM. Mitochondria in metabolic disease: getting clues from proteomic studies. Proteomics 2014; 14:452-66. [PMID: 24339000 DOI: 10.1002/pmic.201300376] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2013] [Revised: 11/08/2013] [Accepted: 11/21/2013] [Indexed: 01/11/2023]
Abstract
Mitochondria play a key role as major regulators of cellular energy homeostasis, but in the context of mitochondrial dysfunction, mitochondria may generate reactive oxidative species and induce cellular apoptosis. Indeed, altered mitochondrial status has been linked to the pathogenesis of several metabolic disorders and specially disorders related to insulin resistance, such as obesity, type 2 diabetes, and other comorbidities comprising the metabolic syndrome. In the present review, we summarize information from various mitochondrial proteomic studies of insulin-sensitive tissues under different metabolic states. To that end, we first focus our attention on the pancreas, as mitochondrial malfunction has been shown to contribute to beta cell failure and impaired insulin release. Furthermore, proteomic studies of mitochondria obtained from liver, muscle, and adipose tissue are summarized, as these tissues constitute the primary insulin target metabolic tissues. Since recent advances in proteomic techniques have exposed the importance of PTMs in the development of metabolic disease, we also present information on specific PTMs that may directly affect mitochondria during the pathogenesis of metabolic disease. Specifically, mitochondrial protein acetylation, phosphorylation, and other PTMs related to oxidative damage, such as nitrosylation and carbonylation, are discussed.
Collapse
Affiliation(s)
- Juan R Peinado
- Department of Medical Sciences, Faculty of Medicine, Ciudad Real, Spain
| | | | | | | |
Collapse
|
24
|
Proteome analysis of hepatic non-parenchymal cells of immune liver fibrosis rats. SCIENCE CHINA-LIFE SCIENCES 2014; 57:303-314. [PMID: 24562544 DOI: 10.1007/s11427-014-4619-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/20/2013] [Accepted: 11/20/2013] [Indexed: 12/31/2022]
Abstract
Elucidation of the mechanisms of liver fibrogenesis is important to treat liver fibrosis. In this study, we established rat models of liver fibrosis with stages from 0-1, 2, and 3-4 to 4 at 2, 4, 6, and 8 weeks, respectively, by injection of pig serum. Liver fibrogenesis was detected by Masson's trichrome staining. Rat non-parenchymal cells (NPCs) were enriched 4-fold by Percoll density gradient centrifugation. Protein extracts from NPCs were prepared at 4 and 8 weeks, separated by two-dimensional electrophoresis, and then stained with Coomassie Blue G-250. At 4 weeks, we identified 18 non-redundant differentially expressed proteins of which protein disulfide-isomerase associated protein 3 (PDIA3) and NDUV showed consistent expression at protein and mRNA levels from 4 to 8 weeks. PDIA3 was found to be down-regulated by Western blotting in the rat model and immunohistochemically in human liver. Our results revealed important aspects of the pathogenesis/progression of liver fibrosis and demonstrated important changes in protein expression levels of NPCs at various stages of liver fibrosis.
Collapse
|
25
|
Chen X, Li J, Hou J, Xie Z, Yang F. Mammalian mitochondrial proteomics: insights into mitochondrial functions and mitochondria-related diseases. Expert Rev Proteomics 2014; 7:333-45. [DOI: 10.1586/epr.10.22] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
26
|
Cheng XR, Zhou WX, Zhang YX. The behavioral, pathological and therapeutic features of the senescence-accelerated mouse prone 8 strain as an Alzheimer's disease animal model. Ageing Res Rev 2014; 13:13-37. [PMID: 24269312 DOI: 10.1016/j.arr.2013.10.002] [Citation(s) in RCA: 104] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2013] [Revised: 10/10/2013] [Accepted: 10/30/2013] [Indexed: 12/22/2022]
Abstract
Alzheimer's disease (AD) is a widespread and devastating progressive neurodegenerative disease. Disease-modifying treatments remain beyond reach, and the etiology of the disease is uncertain. Animal model are essential for identifying disease mechanisms and developing effective therapeutic strategies. Research on AD is currently being carried out in rodent models. The most common transgenic mouse model mimics familial AD, which accounts for a small percentage of cases. The senescence-accelerated mouse prone 8 (SAMP8) strain is a spontaneous animal model of accelerated aging. Many studies indicate that SAMP8 mice harbor the behavioral and histopathological signatures of AD, namely AD-like cognitive and behavioral alterations, neuropathological phenotypes (neuron and dendrite spine loss, spongiosis, gliosis and cholinergic deficits in the forebrain), β-amyloid deposits resembling senile plaques, and aberrant hyperphosphorylation of Tau-like neurofibrillary tangles. SAMP8 mice are useful in the development of novel therapies, and many pharmacological agents and approaches are effective in SAMP8 mice. SAMP8 mice are considered a robust model for exploring the etiopathogenesis of sporadic AD and a plausible experimental model for developing preventative and therapeutic treatments for late-onset/age-related AD, which accounts for the vast majority of cases.
Collapse
Affiliation(s)
- Xiao-rui Cheng
- Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Wen-xia Zhou
- Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China.
| | - Yong-xiang Zhang
- Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China.
| |
Collapse
|
27
|
Hardeland R. Melatonin and the theories of aging: a critical appraisal of melatonin's role in antiaging mechanisms. J Pineal Res 2013; 55:325-56. [PMID: 24112071 DOI: 10.1111/jpi.12090] [Citation(s) in RCA: 195] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2013] [Accepted: 08/23/2013] [Indexed: 02/06/2023]
Abstract
The classic theories of aging such as the free radical theory, including its mitochondria-related versions, have largely focused on a few specific processes of senescence. Meanwhile, numerous interconnections have become apparent between age-dependent changes previously thought to proceed more or less independently. Increased damage by free radicals is not only linked to impairments of mitochondrial function, but also to inflammaging as it occurs during immune remodeling and by release of proinflammatory cytokines from mitotically arrested, DNA-damaged cells that exhibit the senescence-associated secretory phenotype (SASP). Among other effects, SASP can cause mutations in stem cells that reduce the capacity for tissue regeneration or, in worst case, lead to cancer stem cells. Oxidative stress has also been shown to promote telomere attrition. Moreover, damage by free radicals is connected to impaired circadian rhythmicity. Another nexus exists between cellular oscillators and metabolic sensing, in particular to the aging-suppressor SIRT1, which acts as an accessory clock protein. Melatonin, being a highly pleiotropic regulator molecule, interacts directly or indirectly with all the processes mentioned. These influences are critically reviewed, with emphasis on data from aged organisms and senescence-accelerated animals. The sometimes-controversial findings obtained either in a nongerontological context or in comparisons of tumor with nontumor cells are discussed in light of evidence obtained in senescent organisms. Although, in mammals, lifetime extension by melatonin has been rarely documented in a fully conclusive way, a support of healthy aging has been observed in rodents and is highly likely in humans.
Collapse
Affiliation(s)
- Rüdiger Hardeland
- Johann Friedrich Blumenbach Institute of Zoology and Anthropology, University of Göttingen, Göttingen, Germany
| |
Collapse
|
28
|
Bayram B, Nikolai S, Huebbe P, Ozcelik B, Grimm S, Grune T, Frank J, Rimbach G. Biomarkers of oxidative stress, antioxidant defence and inflammation are altered in the senescence-accelerated mouse prone 8. AGE (DORDRECHT, NETHERLANDS) 2013; 35:1205-1217. [PMID: 22767392 PMCID: PMC3705129 DOI: 10.1007/s11357-012-9448-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/28/2011] [Accepted: 06/20/2012] [Indexed: 06/01/2023]
Abstract
In this study we compared biomarkers of oxidative stress, stress response, antioxidant defence and inflammation between mice (n = 10 per group, female, 7 months old) with an accelerated (SAMP8) and a normal ageing phenotype (SAMR1). As compared to SAMR1 mice, SAMP8 mice exhibited higher levels of lipid peroxides and protein carbonyls as well as a lower activity of the proteasomal subunit β-5. Furthermore, heme oxygenase-1 and paraoxonase-1 (PON-1) status was lower in SAMP8 mice indicating impaired stress response. Biomarkers of inflammation such as C-reactive protein and serum amyloid P were elevated in SAMP8 mice. Interestingly, impaired stress response and increased inflammation in SAMP8 mice were associated with elevated concentrations of ascorbic acid and α-tocopherol in the liver. An age-dependent increase in hepatic vitamin E and a decline in PON-1 gene expression were also observed in aged compared to young C57BL/6 mice.
Collapse
Affiliation(s)
- Banu Bayram
- />Institute of Human Nutrition and Food Science, Christian-Albrechts-University, Hermann Rodewald Strasse 6, 24098 Kiel, Germany
- />Department of Food Engineering, Istanbul Technical University, 34469 Maslak, Istanbul, Turkey
| | - Sibylle Nikolai
- />Institute of Human Nutrition and Food Science, Christian-Albrechts-University, Hermann Rodewald Strasse 6, 24098 Kiel, Germany
| | - Patricia Huebbe
- />Institute of Human Nutrition and Food Science, Christian-Albrechts-University, Hermann Rodewald Strasse 6, 24098 Kiel, Germany
| | - Beraat Ozcelik
- />Department of Food Engineering, Istanbul Technical University, 34469 Maslak, Istanbul, Turkey
| | - Stefanie Grimm
- />Institute of Nutrition, Department of Nutritional Toxicology, Friedrich Schiller University, Dornburger Strasse 24, 07743 Jena, Germany
| | - Tilman Grune
- />Institute of Nutrition, Department of Nutritional Toxicology, Friedrich Schiller University, Dornburger Strasse 24, 07743 Jena, Germany
| | - Jan Frank
- />Institute of Biological Chemistry and Nutrition, University of Hohenheim, Garbenstrasse 30, 70599 Stuttgart, Germany
| | - Gerald Rimbach
- />Institute of Human Nutrition and Food Science, Christian-Albrechts-University, Hermann Rodewald Strasse 6, 24098 Kiel, Germany
| |
Collapse
|
29
|
Zhuang H, Gan Z, Jiang W, Zhang X, Hua ZC. Comparative proteomics analysis reveals roles for FADD in the regulation of energy metabolism and proteolysis pathway in mouse embryonic fibroblast. Proteomics 2013; 13:2398-413. [PMID: 23744592 DOI: 10.1002/pmic.201300017] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2013] [Revised: 05/01/2013] [Accepted: 05/16/2013] [Indexed: 12/19/2022]
Abstract
Fas-associated death domain-containing protein (FADD) is a classical apoptotic pathway adaptor. Further studies revealed that it also plays essential roles in nonapoptotic processes, which is assumed to be regulated by its phosphorylation. However, the exact mechanisms are still poorly understood. To study the nonapoptotic effects of FADD, a comprehensive strategy of proteomics identification combined with bioinformatic analysis was undertaken to identify proteins differentially expressed in three cell lines containing FADD and its mutant, FADD-A and FADD-D. The cell lines were thought to bear wild-type FADD, unphosphorylated FADD mimic and constitutive phosphorylated FADD mimic, respectively. A total of 47 proteins were identified to be significantly changed due to FADD phosphorylation. Network analysis using MetaCore™ identified a number of changed proteins that were involved in cellular metabolic process, including lipid metabolism, fatty acid metabolism, glycolysis, and oxidative phosphorylation. The finding that FADD-D cell line showed an increase in fatty acid oxidation argues that it could contribute to the leaner phenotype of FADD-D mice as reported previously. In addition, six proteins related to the ubiquitin-proteasome pathway were also specifically overexpressed in FADD-D cell line. Finally, the c-Myc gene represents a convergent hub lying at the center of dysregulated pathways, and was upregulated in FADD-D cells. Taken together, these studies allowed us to conclude that impaired mitochondrial function and proteolysis might play pivotal roles in the dysfunction associated with FADD phosphorylation-induced disorders.
Collapse
Affiliation(s)
- Hongqin Zhuang
- The State Key Laboratory of Pharmaceutical Biotechnology, College of Life Science and School of Stomatology, Affiliated Stomatological Hospital, Nanjing University, Nanjing, PR China
| | | | | | | | | |
Collapse
|
30
|
Guo SJ, Qi CH, Zhou WX, Zhang YX, Zhang XM, Wang J, Wang HX. Proteomic data show an increase in autoantibodies and alpha-fetoprotein and a decrease in apolipoprotein A-II with time in sera from senescence-accelerated mice. Braz J Med Biol Res 2013; 46:417-25. [PMID: 23588375 PMCID: PMC3854399 DOI: 10.1590/1414-431x20132663] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2012] [Accepted: 01/04/2013] [Indexed: 11/22/2022] Open
Abstract
We evaluated changes in levels by comparing serum proteins in
senescence-accelerated mouse-prone 8 (SAMP8) mice at 2, 6, 12, and 15 months of
age (SAMP8-2 m, -6 m, -12 m, -15 m) to age-matched
SAM-resistant 1 (SAMR1) mice. Mice were sacrificed, and blood was analyzed by
2-dimensional electrophoresis combined with mass spectrometry. Five protein
spots were present in all SAMP8 serum samples, but only appeared in SAMR1
samples at 15 months of age except for spot 3, which also showed a slight
expression in SAMR1-12 m sera. Two proteins decreased in the sera from
SAMP8-2 m, -6 m, and -12 m mice, and divided into 2 spots
each in SAMP8-15 m sera. Thus, the total number of altered spots in SAMP8
sera was 7; of these, 4 were identified as Ig kappa chain V region (M-T413),
chain A of an activity suppressing Fab fragment to cytochrome P450 aromatase
(32C2_A), alpha-fetoprotein, and apolipoprotein A-II. M-T413 is a monoclonal CD4
antibody, which inhibits T cell proliferation. We found that M-T413 RNA level
was significantly enhanced in splenocytes from SAMP8-2 m mice. This
agreed with serum M-T413 protein alterations and a strikingly lower blood
CD4+ T cell count in SAMP8 mice when compared to the
age-matched SAMR1 mice, with the latter negatively correlating with serum M-T413
protein volume. Age-related changes in serum proteins favored an increase in
autoantibodies and alpha-fetoprotein and a decrease of apolipoprotein A-II,
which occurred in SAMP8 mice at 2 months of age and onwards. These proteins may
serve as candidate biomarkers for early aging.
Collapse
Affiliation(s)
- S J Guo
- Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | | | | | | | | | | | | |
Collapse
|
31
|
He J, Liu Y, Wang Q, Ji J. Purification and proteomic analysis of liver membrane skeletons. Methods Mol Biol 2012; 909:97-111. [PMID: 22903711 DOI: 10.1007/978-1-61779-959-4_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/13/2023]
Abstract
The detergent-resistant membrane skeletons play a critical role in cell shaping and signaling. The focus of the methods described in this chapter is first on the preparation of membrane skeletons from liver by multistep sucrose density gradient centrifugation, and then on the analysis of the protein components of membrane skeletons using proteomics techniques. Two proteomic analysis strategies are described. In the first strategy, membrane skeleton proteins are separated by two-dimensional gel electrophoresis and identified by matrix-assisted laser desorption/ionization tandem time-of-flight mass spectrometry. In the other strategy, proteins are separated by SDS-PAGE and identified by liquid chromatography-electrospray ionization Fourier transform ion cyclotron resonance mass spectrometry. The methods facilitate the understanding of the structure of membrane skeletons.
Collapse
Affiliation(s)
- Jintang He
- State Key Laboratory of Protein and Plant Gene Research, College of Life Sciences, Peking University, Beijing, People's Republic of China
| | | | | | | |
Collapse
|
32
|
Melanogenesis stimulation in B16-F10 melanoma cells induces cell cycle alterations, increased ROS levels and a differential expression of proteins as revealed by proteomic analysis. Exp Cell Res 2012; 318:1913-25. [DOI: 10.1016/j.yexcr.2012.05.019] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2012] [Revised: 04/26/2012] [Accepted: 05/16/2012] [Indexed: 12/29/2022]
|
33
|
Rauniyar N, Prokai L. Isotope-coded dimethyl tagging for differential quantification of posttranslational protein carbonylation by 4-hydroxy-2-nonenal, an end-product of lipid peroxidation. JOURNAL OF MASS SPECTROMETRY : JMS 2011; 46:976-85. [PMID: 22012663 PMCID: PMC3197809 DOI: 10.1002/jms.1978] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
Peroxidation of cellular membrane lipids, rich in polyunsaturated fatty acids, generates electrophilic, α, β-unsaturated aldehydes such as 4-hydroxy-2-nonenal (HNE). HNE is a highly reactive and cytotoxic molecule that can react with the nucleophilic sites in proteins causing posttranslational modification. The identification of protein targets is an important first step; however, quantitative profiling of site-specific modifications is necessary to understand the biological impact of HNE-induced carbonylation. We report a method that uses light (H(12)CHO) and heavy (D(13)CDO) isotopic variant of formaldehyde to differentially label primary amines (N-termini and ε-amino group of lysines) in peptides through reductive methylation and, combined with selective enrichment of modified peptides, permits comparison of the extent of carbonylation in two samples after mixing for simultaneous liquid chromatography-mass spectrometry. Specifically, dimethyl-labeled peptide carbonyls were fractionated from unmodified peptides using solid-phase hydrazide chemistry to immobilize them to porous glass beads and, after removing the unmodified peptides by thoroughly washing the beads, subsequently recover them by acid-catalyzed hydrolysis. The method was developed using HNE-modified synthetic peptides and also showing enrichment from a complex matrix of digested human plasma proteins. Applicability was confirmed using apomyoglobin as an analyte, implicating thereby its potential value to proteome-wide identification and relative quantification of posttranslational protein carbonylation with residue-specific information. Because HNE attachment may not necessarily cause change in protein abundance, this modification-focused quantification should facilitate the characterization of accompanied changes in protein function and, also, provide important insights into molecular signaling mechanisms and a better understanding of cellular processes associated with oxidative stress.
Collapse
Affiliation(s)
| | - Laszlo Prokai
- Correspondence to: Laszlo Prokai, Department of Molecular Biology & Immunology, University of North Texas Health Science Center, 3500 Camp Bowie Boulevard, Fort Worth, Texas 76107
| |
Collapse
|
34
|
Lin X, Wang Q, Cheng Y, Ji J, Yu LC. Changes of protein expression profiles in the amygdala during the process of morphine-induced conditioned place preference in rats. Behav Brain Res 2011; 221:197-206. [DOI: 10.1016/j.bbr.2011.03.002] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2010] [Revised: 02/23/2011] [Accepted: 03/01/2011] [Indexed: 11/17/2022]
|
35
|
Zhang L, Peng X, Zhang Z, Feng Y, Jia X, Shi Y, Yang H, Zhang Z, Zhang X, Liu L, Yin L, Yuan Z. Subcellular proteome analysis unraveled annexin A2 related to immune liver fibrosis. J Cell Biochem 2010; 110:219-28. [PMID: 20225235 DOI: 10.1002/jcb.22529] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
It is important to study the mechanism of liver fibrogenesis, and find new non-invasive biomarkers. In this study, we used subcellular proteomic technology to study the plasma membrane (PM) proteins related to immune liver fibrosis and search for new non-invasive biomarkers. A rat liver fibrosis model was induced by pig serum injection. The liver fibrogenesis from stage (S) S0-1, S2, S3-4, and S4 was detected by Masson staining and HE staining in this rat model after 2, 4, 6, and 8 weeks of treatment. The liver PM was enriched and analyzed using subcellular proteomic technology. The differentially expressed proteins were verified by Western blotting, immunohistochemistry, and ELISA. PM with 149-fold purification was obtained and 22 differentially expressed proteins were identified. Of which, annexin A2 (ANXA2) was detected to be increased obviously in S4 compared with S0-1, and verified by Western blotting of rat liver tissue and immunohistochemistry of rat and human liver tissue. The expression of ANXA2 in human plasma with S1-2 was also found to be up-regulated for 1.4-fold than that in S0. Furthermore, ANXA2 was detected to translocate from nuclear membrane and cytosol to PM as HBV stimulation through immunocytochemical analysis in vitro. This study identified 22 differentially expressed proteins related to liver fibrosis, and verified a potential biomarker (ANXA2) for non-invasive diagnosis of immune liver fibrosis. To our knowledge, it was the first time to dynamically study the proteins related to liver fibrosis and select biomarkers for liver fibrosis diagnosis through PM proteome research.
Collapse
Affiliation(s)
- Lijun Zhang
- Shanghai Public Health Clinical Center, Shanghai 201508, China.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Lin X, Wang Q, Ji J, Yu LC. Role of MEK-ERK pathway in morphine-induced conditioned place preference in ventral tegmental area of rats. J Neurosci Res 2010; 88:1595-604. [PMID: 20091775 DOI: 10.1002/jnr.22326] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
A major goal of research on drug addiction is to develop the effective treatments to deal with the long-term behavioral disorders especially reinstatement induced by the addictive drugs such as opiates, cocaine, and cannabinoid. The molecular mechanisms underlying these substance-related disorders remain unclear so far. Here we used the model of morphine-induced conditioned place preference (CPP) in rats to mimic the progress of drug-taking, withdrawal and relapse in human. The tissue of ventral tegmental area (VTA), one of the most important brain structures associated with abused drug-related disorders, was taken and two-dimensional electrophoresis (2-DE) was performed to analyze and compare the changes of protein expression patterns during the different stages of morphine-induced CPP. First, we found that there were 80 proteins identified to be changed in the process of morphine-induced CPP. Furthermore, as the mitogen-activated protein kinase kinase 1 (MAPKK1) was increased significantly in the stages of establishment and reinstatement, we confirmed the change of activated extracellular signal-regulated kinase (ERK) by Western blotting in VTA tissue and cultured cell. The results demonstrated that the activated MEK-ERK pathway by chronic morphine treatment in VTA was involved in morphine-induced reinstatement. Moreover, inhibition of MEK-ERK pathway by infusion the MEK inhibitor U0126 in VTA blocked the establishment of morphine-induced CPP. The present study found significant changes in a group of protein expressions in VTA during morphine-induced CPP and further confirmed the role of MEK-ERK cell signaling pathway of VTA in morphine addiction.
Collapse
Affiliation(s)
- XiaoJing Lin
- The National Laboratory of Protein Engineering and Plant Genetic Engineering, College of Life Sciences, Peking University, Beijing, China
| | | | | | | |
Collapse
|
37
|
Cai XW, Shedden K, Ao X, Davis M, Fu XL, Lawrence TS, Lubman DM, Kong FMS. Plasma proteomic analysis may identify new markers for radiation-induced lung toxicity in patients with non-small-cell lung cancer. Int J Radiat Oncol Biol Phys 2010; 77:867-76. [PMID: 20510197 DOI: 10.1016/j.ijrobp.2010.01.038] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2009] [Revised: 01/19/2010] [Accepted: 01/25/2010] [Indexed: 01/08/2023]
Abstract
PURPOSE To study whether radiation induces differential changes in plasma proteomics in patients with and without radiation-induced lung toxicity (RILT) of Grade >/=2 (RILT2). METHODS AND MATERIALS A total of 57 patients with NSCLC received radiation therapy (RT) were eligible. Twenty patients, 6 with RILT2 with tumor stage matched to 14 without RILT2, were enrolled for this analysis. Platelet-poor plasma was obtained before RT, at 2, 4, 6 weeks during RT, and 1 and 3 months after RT. Plasma proteomes were compared using a multiplexed quantitative proteomics approach involving ExacTag labeling, reverse-phase high-performance liquid chromatography and nano-LC electrospray tandem mass spectrometry. Variance components models were used to identify the differential protein expression between patients with and without RILT2. RESULTS More than 100 proteins were identified and quantified. After excluding proteins for which there were not at least 2 subjects with data for at least two time points, 76 proteins remained for this preliminary analysis. C4b-binding protein alpha chain, Complement C3, and Vitronectin had significantly higher expression levels in patients with RILT2 compared with patients without RILT2, based on both the data sets of RT start to 3 months post-RT (p < 0.01) and RT start to the end of RT (p < 0.01). The expression ratios of patients with RILT2 vs. without RILT2 were 1.60, 1.36, 1.46, and 1.66, 1.34, 1.46, for the above three proteins, respectively. CONCLUSIONS This proteomic approach identified new plasma protein markers for future studies on RILT prediction.
Collapse
Affiliation(s)
- Xu-Wei Cai
- Department of Radiation Oncology, University of Michigan Medical Center, Ann Arbor, MI 48108, USA
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Pang CY, Wang H, Pang Y, Xu C, Jiao Y, Qin YM, Western TL, Yu SX, Zhu YX. Comparative proteomics indicates that biosynthesis of pectic precursors is important for cotton fiber and Arabidopsis root hair elongation. Mol Cell Proteomics 2010; 9:2019-33. [PMID: 20525998 DOI: 10.1074/mcp.m110.000349] [Citation(s) in RCA: 136] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
The quality of cotton fiber is determined by its final length and strength, which is a function of primary and secondary cell wall deposition. Using a comparative proteomics approach, we identified 104 proteins from cotton ovules 10 days postanthesis with 93 preferentially accumulated in the wild type and 11 accumulated in the fuzzless-lintless mutant. Bioinformatics analysis indicated that nucleotide sugar metabolism was the most significantly up-regulated biochemical process during fiber elongation. Seven protein spots potentially involved in pectic cell wall polysaccharide biosynthesis were specifically accumulated in wild-type samples at both the protein and transcript levels. Protein and mRNA expression of these genes increased when either ethylene or lignoceric acid (C24:0) was added to the culture medium, suggesting that these compounds may promote fiber elongation by modulating the production of cell wall polymers. Quantitative analysis revealed that fiber primary cell walls contained significantly higher amounts of pectin, whereas more hemicellulose was found in ovule samples. Significant fiber growth was observed when UDP-L-rhamnose, UDP-D-galacturonic acid, or UDP-D-glucuronic acid, all of which were readily incorporated into the pectin fraction of cell wall preparations, was added to the ovule culture medium. The short root hairs of Arabidopsis uer1-1 and gae6-1 mutants were complemented either by genetic transformation of the respective cotton cDNA or by adding a specific pectin precursor to the growth medium. When two pectin precursors, produced by either UDP-4-keto-6-deoxy-D-glucose 3,5-epimerase 4-reductase or by UDP-D-glucose dehydrogenase and UDP-D-glucuronic acid 4-epimerase successively, were used in the chemical complementation assay, wild-type root hair lengths were observed in both cut1 and ein2-5 Arabidopsis seedlings, which showed defects in C24:0 biosynthesis or ethylene signaling, respectively. Our results suggest that ethylene and C24:0 may promote cotton fiber and Arabidopsis root hair growth by activating the pectin biosynthesis network, especially UDP-L-rhamnose and UDP-D-galacturonic acid synthesis.
Collapse
Affiliation(s)
- Chao-You Pang
- The National Laboratory of Protein Engineering and Plant Genetic Engineering, College of Life Sciences, Peking University, Beijing, China
| | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Liu Y, He J, Li C, Benitez R, Fu S, Marrero J, Lubman DM. Identification and confirmation of biomarkers using an integrated platform for quantitative analysis of glycoproteins and their glycosylations. J Proteome Res 2010; 9:798-805. [PMID: 19961239 DOI: 10.1021/pr900715p] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Hepatocellular carcinoma (HCC) is the most common primary malignant tumor of the liver. However, accurate diagnosis can be difficult as most of the patients who develop this tumor have symptoms similar to those caused by longstanding liver disease. Herein we developed an integrated platform to discover the glycoprotein biomarkers in early HCC. At first, lectin arrays were applied to investigate the differences in glycan structures on serum glycoproteins from HCC and cirrhosis patients. The intensity for AAL and LCA was significantly higher in HCC, indicating an elevation of fucosylation level. Then serum from 10 HCC samples and 10 cirrhosis samples were used to screen the altered fucosylated proteins by a combination of Exactag labeling, lectin extraction and LC-MS/MS. Finally, 27 HCC and 27 cirrhosis serum samples were used for lectin-antibody arrays to confirm the change of these fucosylated proteins. C3, CE, HRG, CD14 and HGF were found to be biomarker candidates for distinguishing early HCC from cirrhosis, with a sensitivity of 72% and specificity of 79%. Our work gives insight to the detection of early HCC, and the application of this comprehensive strategy has the potential to facilitate biomarker discovery on a large scale.
Collapse
Affiliation(s)
- Yashu Liu
- Department of Surgery, University of Michigan Medical Center, Ann Arbor, Michigan 48109-0656, USA
| | | | | | | | | | | | | |
Collapse
|
40
|
Wang Q, He J, Meng L, Liu Y, Pu H, Ji J. A proteomics analysis of rat liver membrane skeletons: the investigation of actin- and cytokeratin-based protein components. J Proteome Res 2010; 9:22-9. [PMID: 19354234 DOI: 10.1021/pr900102n] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Membrane skeletons, which are defined for their resistance to Triton extraction of cell membrane, play a pivotal role in cell shape and signal transduction. In the present work, we applied a complementary proteomics strategy: 2-DE combined with MALDI-TOF MS and 1D-PAGE coupled with LC-ESI-FTICR MS to analyze a membrane skeleton fraction isolated from Sprague-Dawley (SD) rat livers. We report confident identification of 104 proteins (39 membrane skeleton proteins) using 2-DE and MALDI-TOF MS approach and 402 proteins (87 membrane skeleton proteins) using 1D-PAGE LC-MS/MS analysis. In total, 100 membrane skeleton proteins were identified using the two complementary proteomics means. To the best of our knowledge, this is the largest data set of membrane skeleton proteins to date. Noteworthily, almost all of these membrane skeleton proteins were associated with actin or cytokeratin, and more than half of them were involved in various cell junctions. Our results offer insights into the protein components of the actin- and cytokeratin-based membrane skeletons in rat livers, which would improve our understanding of their biological roles.
Collapse
Affiliation(s)
- Qingsong Wang
- The National Laboratory of Protein Engineering and Plant Genetic Engineering, College of Life Sciences, Peking University, Beijing 100871, China
| | | | | | | | | | | |
Collapse
|
41
|
Gao X, Zhang X, Zheng J, He F. Proteomics in China: Ready for prime time. SCIENCE CHINA-LIFE SCIENCES 2010; 53:22-33. [DOI: 10.1007/s11427-010-0027-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/20/2009] [Accepted: 12/28/2009] [Indexed: 12/27/2022]
|
42
|
Hardeland R. Neuroprotection by radical avoidance: search for suitable agents. Molecules 2009; 14:5054-102. [PMID: 20032877 PMCID: PMC6255388 DOI: 10.3390/molecules14125054] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2009] [Revised: 11/30/2009] [Accepted: 12/04/2009] [Indexed: 02/07/2023] Open
Abstract
Neurodegeneration is frequently associated with damage by free radicals. However, increases in reactive oxygen and nitrogen species, which may ultimately lead to neuronal cell death, do not necessarily reflect its primary cause, but can be a consequence of otherwise induced cellular dysfunction. Detrimental processes which promote free radical formation are initiated, e.g., by disturbances in calcium homeostasis, mitochondrial malfunction, and an age-related decline in the circadian oscillator system. Free radicals generated at high rates under pathophysiological conditions are insufficiently detoxified by scavengers. Interventions at the primary causes of dysfunction, which avoid secondary rises in radical formation, may be more efficient. The aim of such approaches should be to prevent calcium overload, to reduce mitochondrial electron dissipation, to support electron transport capacity, and to avoid circadian perturbations. L-theanine and several amphiphilic nitrones are capable of counteracting excitotoxicity and/or mitochondrial radical formation. Resveratrol seems to promote mitochondrial biogenesis. Mitochondrial effects of leptin include attenuation of electron leakage. Melatonin combines all the requirements mentioned, additionally regulates anti- and pro-oxidant enzymes and is, with few exceptions, very well tolerated. In this review, the perspectives, problems and limits of drugs are compared which may be suitable for reducing the formation of free radicals.
Collapse
Affiliation(s)
- Rüdiger Hardeland
- Johann Friedrich Blumenbach Institute of Zoology and Anthropology, University of Göttingen, Berliner str. 28, D-37073 Göttingen, Germany.
| |
Collapse
|
43
|
Wu J, An Y, Pu H, Shan Y, Ren X, An M, Wang Q, Wei S, Ji J. Enrichment of serum low-molecular-weight proteins using C18 absorbent under urea/dithiothreitol denatured environment. Anal Biochem 2009; 398:34-44. [PMID: 19891953 DOI: 10.1016/j.ab.2009.10.047] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2009] [Revised: 10/19/2009] [Accepted: 10/30/2009] [Indexed: 01/26/2023]
Abstract
Serum low-molecular-weight proteins (LMWPs, molecular weight<30kDa) are closely related to the body physiological and pathological situations, whereas many difficulties are encountered when enriching and fractionating them. Using C(18) absorbent (100 A) enrichment and fractionation under urea/dithiothreitol (DTT) denatured environment followed by 60% acetonitrile (ACN) elution, serum LMWPs could be enriched more than 100-fold and were evaluated by sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE), two-dimensional gel electrophoresis (2-DE), and isotope-coded affinity tag (ICAT) labeling quantification. Proteins existing in human serum at low nanograms/milliliter (ng/ml) levels, such as myeloid-related proteins (MRPs), could be identified directly from 2-DE coupled with matrix-assisted laser desorption/ionization tandem time-of-flight mass spectrometry (MALDI-TOF/TOF MS) and LTQ-Orbitrap MS. Sixteen proteins were confidentially identified and quantified using ICAT labeling and liquid chromatography-tandem mass spectrometry (LC-MS/MS). By virtue of its easy operation and high reproducibility to process large quantity complex serum samples, this method has potential uses in enriching LMWPs either in serum or in cell and tissue samples.
Collapse
Affiliation(s)
- Jing Wu
- National Laboratory of Protein Engineering and Plant Genetic Engineering, College of Life Sciences, Peking University, Beijing 100871, People's Republic of China
| | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Chaiyarit S, Thongboonkerd V. Comparative analyses of cell disruption methods for mitochondrial isolation in high-throughput proteomics study. Anal Biochem 2009; 394:249-58. [DOI: 10.1016/j.ab.2009.07.026] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2009] [Revised: 07/14/2009] [Accepted: 07/17/2009] [Indexed: 01/30/2023]
|
45
|
Hartwig S, Feckler C, Lehr S, Wallbrecht K, Wolgast H, Müller-Wieland D, Kotzka J. A critical comparison between two classical and a kit-based method for mitochondria isolation. Proteomics 2009; 9:3209-14. [PMID: 19415664 DOI: 10.1002/pmic.200800344] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Numerous protocols for isolation of mitochondria are available. Here, three methods for the isolation of intact mitochondria from mouse liver tissues are compared with regard to yield, purity and activity. Mitochondria were isolated by sucrose density gradient ultracentrifugation, free-flow electrophoresis or a commercially available kit-based method. Our analyses show that the sophisticated (and most expensive) free-flow electrophoresis method enables isolation of intact mitochondria with an enrichment of approximately 70%. Using the classical density centrifugation method is very laborious and time-consuming, but delivers about 57% intact mitochondria. Using standard laboratory equipment in a quick and simple procedure, the kit provides approximately 50% intact mitochondria, suitable for most standard investigations.
Collapse
Affiliation(s)
- Sonja Hartwig
- Institute of Clinical Biochemistry and Pathobiochemistry, German Diabetes Center at the Heinrich-Heine-University Düsseldorf, Leibniz Center for Diabetes Research, Düsseldorf, Germany
| | | | | | | | | | | | | |
Collapse
|
46
|
Wang Q, Zhao X, He S, Liu Y, An M, Ji J. Differential proteomics analysis of specific carbonylated proteins in the temporal cortex of aged rats: the deterioration of antioxidant system. Neurochem Res 2009; 35:13-21. [PMID: 19562484 DOI: 10.1007/s11064-009-0023-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2009] [Accepted: 06/15/2009] [Indexed: 01/02/2023]
Abstract
Oxidative stress plays a pivotal role in normal brain aging and various neurodegenerative diseases, including Alzheimer's disease (AD). Irreversible protein carbonylation, a widely used marker for oxidative stress, rises during aging. The temporal cortex is essential for learning and memory and particularly susceptible to oxidative stress during aging and in AD patients. In this study, we used 2-DE, MALDI-TOF/TOF MS, and Western blotting to analyze the differentially carbonylated proteins in the rat temporal cortex between 1-month-old and 24-month-old. We showed that the carbonyl levels of ten protein spots corresponding to six gene products: SOD1, SOD2, peroxiredoxin 1, peptidylprolyl isomerase A, cofilin 1, and adenylate kinase 1, significantly increased in the temporal cortex of aged rats. These proteins are associated with antioxidant defense, the cytoskeleton, and energy metabolism. Several oxidized proteins identified in aged rat brain are known to be involved in neurodegenerative disorders as well. Our findings indicate that these carbonylated proteins may be implicated in the decline of normal brain aging process and provide insights into the mechanisms underlying age-associated dysfunction of temporal cortex.
Collapse
Affiliation(s)
- Qingsong Wang
- The National Laboratory of Protein Engineering and Plant Genetic Engineering, College of Life Sciences, Peking University, 100871 Beijing, People's Republic of China.
| | | | | | | | | | | |
Collapse
|
47
|
Scaloni A, Codarin E, Di Maso V, Arena S, Renzone G, Tiribelli C, Quadrifoglio F, Tell G. Modern strategies to identify new molecular targets for the treatment of liver diseases: The promising role of Proteomics and Redox Proteomics investigations. Proteomics Clin Appl 2009; 3:242-62. [DOI: 10.1002/prca.200800169] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2008] [Indexed: 12/16/2022]
|
48
|
KASHIWAGI A, KUROSAKI H, LUO H, YAMAMOTO H, OSHIMURA M, SHIBAHARA T. Effects of Tritrichomonas muris on the Mouse Intestine: A Proteomic Analysis. Exp Anim 2009; 58:537-42. [PMID: 19897938 DOI: 10.1538/expanim.58.537] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Affiliation(s)
- Akiko KASHIWAGI
- Division of Laboratory Animal Science, Research Center for Bioscience and Technology, Tottori University
| | - Hajime KUROSAKI
- Department of Biomedical Science, Institute of Regenerative Medicine and Biofunction, Graduate School of Medical Science, Tottori University
| | - Hong LUO
- Graduate School of Medicine, Faculty of Medicine, Tottori University
| | - Hiroshi YAMAMOTO
- Division of Animal Resources and Development, Life Science Research Center, University of Toyama
| | - Mitsuo OSHIMURA
- Department of Biomedical Science, Institute of Regenerative Medicine and Biofunction, Graduate School of Medical Science, Tottori University
| | - Toshiyuki SHIBAHARA
- Comparative Animal Science Laboratory, Department of Animal Pharmaceutical Science, Faculty of Pharmacy, Chiba Institute of Science
| |
Collapse
|
49
|
Ruiz-Romero C, Blanco FJ. Mitochondrial proteomics and its application in biomedical research. MOLECULAR BIOSYSTEMS 2009; 5:1130-42. [DOI: 10.1039/b906296n] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
|
50
|
Pocsfalvi G. Chapter 5 Selective Enrichment in Phosphopeptides for the Identification of Phosphorylated Mitochondrial Proteins. Methods Enzymol 2009; 457:81-96. [DOI: 10.1016/s0076-6879(09)05005-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
|