1
|
Rickard BP, Overchuk M, Chappell VA, Kemal Ruhi M, Sinawang PD, Nguyen Hoang TT, Akin D, Demirci U, Franco W, Fenton SE, Santos JH, Rizvi I. Methods to Evaluate Changes in Mitochondrial Structure and Function in Cancer. Cancers (Basel) 2023; 15:2564. [PMID: 37174030 PMCID: PMC10177605 DOI: 10.3390/cancers15092564] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 04/25/2023] [Accepted: 04/26/2023] [Indexed: 05/15/2023] Open
Abstract
Mitochondria are regulators of key cellular processes, including energy production and redox homeostasis. Mitochondrial dysfunction is associated with various human diseases, including cancer. Importantly, both structural and functional changes can alter mitochondrial function. Morphologic and quantifiable changes in mitochondria can affect their function and contribute to disease. Structural mitochondrial changes include alterations in cristae morphology, mitochondrial DNA integrity and quantity, and dynamics, such as fission and fusion. Functional parameters related to mitochondrial biology include the production of reactive oxygen species, bioenergetic capacity, calcium retention, and membrane potential. Although these parameters can occur independently of one another, changes in mitochondrial structure and function are often interrelated. Thus, evaluating changes in both mitochondrial structure and function is crucial to understanding the molecular events involved in disease onset and progression. This review focuses on the relationship between alterations in mitochondrial structure and function and cancer, with a particular emphasis on gynecologic malignancies. Selecting methods with tractable parameters may be critical to identifying and targeting mitochondria-related therapeutic options. Methods to measure changes in mitochondrial structure and function, with the associated benefits and limitations, are summarized.
Collapse
Affiliation(s)
- Brittany P. Rickard
- Curriculum in Toxicology & Environmental Medicine, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Marta Overchuk
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, Chapel Hill, NC, and North Carolina State University, Raleigh, NC 27695, USA
| | - Vesna A. Chappell
- Mechanistic Toxicology Branch, Division of Translational Toxicology, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA
| | - Mustafa Kemal Ruhi
- Institute of Biomedical Engineering, Boğaziçi University, Istanbul 34684, Turkey
| | - Prima Dewi Sinawang
- Canary Center at Stanford for Cancer Early Detection, Department of Radiology, School of Medicine, Palo Alto, CA 94304, USA
- Department of Chemical Engineering, Stanford University, Stanford, CA 94305, USA
| | - Tina Thuy Nguyen Hoang
- Department of Biomedical Engineering, University of Massachusetts Lowell, Lowell, MA 01854, USA
| | - Demir Akin
- Canary Center at Stanford for Cancer Early Detection, Department of Radiology, School of Medicine, Palo Alto, CA 94304, USA
- Center for Cancer Nanotechnology Excellence for Translational Diagnostics (CCNE-TD), School of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Utkan Demirci
- Canary Center at Stanford for Cancer Early Detection, Department of Radiology, School of Medicine, Palo Alto, CA 94304, USA
| | - Walfre Franco
- Department of Biomedical Engineering, University of Massachusetts Lowell, Lowell, MA 01854, USA
| | - Suzanne E. Fenton
- Mechanistic Toxicology Branch, Division of Translational Toxicology, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA
| | - Janine H. Santos
- Mechanistic Toxicology Branch, Division of Translational Toxicology, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA
| | - Imran Rizvi
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, Chapel Hill, NC, and North Carolina State University, Raleigh, NC 27695, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
- Center for Environmental Health and Susceptibility, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| |
Collapse
|
2
|
Simsek B, Yanar K, Çakatay U. Proatherogenic Importance of Carbamylation-induced Protein Damage and Type 2 Diabetes Mellitus: A Systematic Review. Curr Diabetes Rev 2020; 16:608-618. [PMID: 31914914 DOI: 10.2174/1573399816666200107102918] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2019] [Revised: 10/26/2019] [Accepted: 12/12/2019] [Indexed: 02/06/2023]
Abstract
INTRODUCTION & BACKGROUND Protein carbamylation is a non-enzymatic and irreversible posttranslational process. It affects functions of numerous enzymes, hormones and receptors playing several roles in diabetes pathogenesis by changing their native structures. Detrimental consequences of oxidative protein damage comprise, but are not limited to glyoxidation, lipoxidation and carbonylation reactions. Since the carbamylated plasma proteins are strongly related to the glycemic control parameters of diabetes, they may have an additive value and emerge as potential biomarkers for the follow up, prognosis and treatment of diabetes mellitus. METHODS & RESULTS To conduct our systematic review, we used PubMed and Semantic Scholar, and used 'Protein carbamylation and diabetes' and 'Protein carbamylation and atherosclerosis' as keywords and looked into about five hundred manuscripts. Manuscripts that are not in English were excluded as well as manuscripts that did not mention carbamylation to maintain the focus of the present article. Similar to glycation, carbamylation is able to alter functions of plasma proteins and their interactions with endothelial cells and has been shown to be involved in the development of atherosclerosis. CONCLUSION At this stage, it seems clear that protein carbamylation leads to worse clinical outcomes. To improve patient care, but maybe more importantly to improve healthcare-prevention, we believe the next stage involves understanding how exactly protein carbamylation leads to worse outcomes and when and in what group of people anti-carbamylation therapies must be employed.
Collapse
Affiliation(s)
- Bahadir Simsek
- Cerrahpasa, Cerrahpasa Medical School, Medical Program, 34096, Istanbul, Turkey
| | - Karolin Yanar
- Department of Medical Biochemistry, Cerrahpasa Medical School , Istanbul University, 34096, Istanbul, Turkey
| | - Ufuk Çakatay
- Department of Medical Biochemistry, Cerrahpasa Medical School , Istanbul University, 34096, Istanbul, Turkey
| |
Collapse
|
3
|
Bavaresco DV, Schwalm MT, Farias BMD, Ceretta LB, Rosa MID, Valvassori SS. INCREASED OXIDATIVE STRESS IN THE BLOOD OF OSTOMY PATIENTS. ARQUIVOS DE GASTROENTEROLOGIA 2018; 55:164-169. [PMID: 30043867 DOI: 10.1590/s0004-2803.201800000-29] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/22/2017] [Accepted: 01/09/2018] [Indexed: 01/14/2023]
Abstract
BACKGROUND Ostomy is a surgical procedure that creates a stoma that aims to construct a new path for the output of feces or urine. The relationship of oxidative stress (OxS) markers in patients with ostomy is still poorly described. OBJECTIVE The present study was aimed at investigating the changes in oxidative stress parameters in peripheral blood collected from ostomy patients when compared with a healthy control group. METHODS It was evaluated 29 ostomy patients and 30 healthy control patients. The oxidative stress parameters evaluated were: lipid peroxidation [lipid hydroperoxide (LPO), 8-isoprostane (8-ISO) and 4-hydroxynonenal (4-HNE)], protein oxidation and nitration [carbonyl and 3-nitrotyrosine (3-NT)] and DNA oxidation [8-hydroxy-2'-deoxyguanosine (8-OHDG)] in serum from ostomy patients compared to health controls. RESULTS The data showed an increase of LPO, 8-ISO, 4-HNE, 3-NT and 8-OHDG in serum collected from ostomy patients when compared to healthy controls. CONCLUSION The findings support the hypothesis that ostomy triggers the oxidative stress observed in the blood collected from these patients.
Collapse
Affiliation(s)
- Daniela V Bavaresco
- Universidade do Extremo Sul Catarinense, Unidade Acadêmica de Ciências da Saúde, Programa de Pós-Graduação em Ciências da Saúde, Laboratório de Sinalização Neural e Psi-co-farmacologia, Criciúma, SC, Brasil.,Universidade do Extremo Sul Catarinense: Laboratório de Epidemiologia; Programa de Pós-Graduação em Saúde Coletiva; Programa de Pós-Graduação em Ciências da Saúde, Criciúma, SC, Brasil
| | - Mágada T Schwalm
- Universidade do Extremo Sul Catarinense, Unidade Acadêmica de Ciências da Saúde, Curso de Enfermagem, Criciúma, SC, Brasil
| | - Beatriz M de Farias
- Universidade do Extremo Sul Catarinense, Unidade Acadêmica de Ciências da Saúde, Curso de Enfermagem, Criciúma, SC, Brasil
| | - Luciane B Ceretta
- Universidade do Extremo Sul Catarinense, Unidade Acadêmica de Ciências da Saúde, Programa de Pós-Graduação em Saúde Coletiva, Laboratório de Saúde Coletiva, Criciúma, SC, Brasil
| | - Maria Inês da Rosa
- Universidade do Extremo Sul Catarinense: Laboratório de Epidemiologia; Programa de Pós-Graduação em Saúde Coletiva; Programa de Pós-Graduação em Ciências da Saúde, Criciúma, SC, Brasil
| | - Samira S Valvassori
- Universidade do Extremo Sul Catarinense, Unidade Acadêmica de Ciências da Saúde, Programa de Pós-Graduação em Ciências da Saúde, Laboratório de Sinalização Neural e Psi-co-farmacologia, Criciúma, SC, Brasil
| |
Collapse
|
4
|
Valvassori SS, Bavaresco DV, Feier G, Cechinel-Recco K, Steckert AV, Varela RB, Borges C, Carvalho-Silva M, Gomes LM, Streck EL, Quevedo J. Increased oxidative stress in the mitochondria isolated from lymphocytes of bipolar disorder patients during depressive episodes. Psychiatry Res 2018; 264:192-201. [PMID: 29653348 DOI: 10.1016/j.psychres.2018.03.089] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Revised: 02/19/2018] [Accepted: 03/31/2018] [Indexed: 10/25/2022]
Abstract
The present study aims to investigate the oxidative stress parameters in isolated mitochondria, as well as looking at mitochondrial complex activity in patients with Bipolar Disorder (BD) during depressive or euthymic episodes. This study evaluated the levels of mitochondrial complex (I, II, II-III and IV) activity in lymphocytes from BD patients. We evaluated the following oxidative stress parameters: superoxide, thiobarbituric acid reactive species (TBARS) and carbonyl levels in submitochondrial particles of lymphocytes from bipolar patients. 51 bipolar patients were recruited into this study: 34 in the euthymic phase, and 17 in the depressive phase. Our results indicated that the depressive phase could increase the levels of mitochondrial superoxide, carbonyl and TBARS, and superoxide dismutase, and could decrease the levels of mitochondrial complex II activity in the lymphocytes of bipolar patients. It was also observed that there was a negative correlation between the Hamilton Depression Rating Scale (HDRS) and complex II activity in the lymphocytes of depressive bipolar patients. In addition, there was a positive correlation between HDRS and superoxide, superoxide dismutase, TBARS and carbonyl. Additionally, there was a negative correlation between complex II activity and oxidative stress parameters. In conclusion, our results suggest that mitochondrial oxidative stress and mitochondrial complex II dysfunction play important roles in the depressive phase of BD.
Collapse
Affiliation(s)
- Samira S Valvassori
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil.
| | - Daniela V Bavaresco
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
| | - Gustavo Feier
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
| | - Kelen Cechinel-Recco
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
| | - Amanda V Steckert
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
| | - Roger B Varela
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
| | - Cenita Borges
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
| | - Milena Carvalho-Silva
- Experimental Neurology Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
| | - Lara M Gomes
- Experimental Neurology Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
| | - Emílio L Streck
- Experimental Neurology Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
| | - João Quevedo
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil; Center of Excellence on Mood Disorders, Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA; Neuroscience Graduate Program, The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA; Translational Psychiatry Program, Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA
| |
Collapse
|
5
|
Khatibi S, Babon J, Wagner J, Manton JH, Tan CW, Zhu HJ, Wormald S, Burgess AW. TGF-β and IL-6 family signalling crosstalk: an integrated model. Growth Factors 2017; 35:100-124. [PMID: 28948853 DOI: 10.1080/08977194.2017.1363746] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Mathematical models for TGF-β and IL-6 signalling have been linked, providing a platform for analyzing the crosstalk between the systems. An integrated IL-6:TGF-β model was developed via a reduced set of reaction equations which incorporate both feedback loops and appropriate time-delays for transcription and translation processes. The model simulates stable, robust and realistic responses to both ligands. Pulsatile (multiple pulses) inputs for both TGF-β and IL-6 have been simulated to investigate the effects of each ligand on the sensitivity, equilibrium and dynamic responses of the integrated signalling system. In our simulations the crosstalk between constant IL-6 and TGF-β signalling via SMAD7 does not appear to be sufficient to render the cells resistant to TGF-β inhibition. However, the simulations predict that pulsatile IL-6 stimulation would increase SMAD7 levels substantially and consequentially, lead to resistance to TGF-β. The model also allows the prediction of the integrated signalling pathway responses to the mutation of key components, e.g. Gp130 F/F.
Collapse
Affiliation(s)
- Shabnam Khatibi
- a Department of Electrical and Electronic Engineering , University of Melbourne , Parkville , VIC , Australia
- b Structural Biology Division, The Walter and Eliza Hall Institute of Medical Research (WEHI) , Parkville , VIC , Australia
| | - Jeff Babon
- b Structural Biology Division, The Walter and Eliza Hall Institute of Medical Research (WEHI) , Parkville , VIC , Australia
| | - John Wagner
- a Department of Electrical and Electronic Engineering , University of Melbourne , Parkville , VIC , Australia
- c IBM Researchtreetience , Carlton , Australia
- d Department of Medical Biology , University of Melbourne , Parkville , VIC , Australia
| | - Jonathan H Manton
- a Department of Electrical and Electronic Engineering , University of Melbourne , Parkville , VIC , Australia
| | - Chin Wee Tan
- b Structural Biology Division, The Walter and Eliza Hall Institute of Medical Research (WEHI) , Parkville , VIC , Australia
- e IBM Research Collaboratory for Life Sciences Research , Victorian Life Sciences Computation Initiative , Carlton , VIC , Australia
| | - Hong-Jian Zhu
- f Department of Surgery (RMH) , University of Melbourne , Parkville , VIC , Australia
| | - Sam Wormald
- g Division of Cancer and Haematology , The Walter and Eliza Hall Institute of Medical Research (WEHI) , Parkville , VIC , Australia
| | - Antony W Burgess
- b Structural Biology Division, The Walter and Eliza Hall Institute of Medical Research (WEHI) , Parkville , VIC , Australia
- e IBM Research Collaboratory for Life Sciences Research , Victorian Life Sciences Computation Initiative , Carlton , VIC , Australia
| |
Collapse
|
6
|
Mazhar F, Malhi SM, Simjee SU. Comparative studies on the effects of clinically used anticonvulsants on the oxidative stress biomarkers in pentylenetetrazole-induced kindling model of epileptogenesis in mice. J Basic Clin Physiol Pharmacol 2017; 28:31-42. [PMID: 27658141 DOI: 10.1515/jbcpp-2016-0034] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Accepted: 07/19/2016] [Indexed: 12/22/2022]
Abstract
BACKGROUND Oxidative stress plays a key role in the pathogenesis of epilepsy and contributes in underlying epileptogenesis process. Anticonvulsant drugs targeting the oxidative stress domain of epileptogenesis may provide better control of seizure. The present study was carried out to investigate the effect of clinically used anti-epileptic drugs (AEDs) on the course of pentylenetetrazole (PTZ)-induced kindling and oxidative stress markers in mice. METHODS Six mechanistically heterogeneous anticonvulsants: phenobarbital, phenytoin, levetiracetam, pregabalin, topiramate, and felbamate were selected and their redox profiles were determined. Diazepam was used as a drug control for comparison. Kindling was induced by repeated injections of a sub-convulsive dose of PTZ (50 mg/kg, s.c.) on alternate days until seizure score 5 was evoked in the control kindled group. Anticonvulsants were administered daily. Following PTZ kindling, oxidative stress biomarkers were assessed in homogenized whole brain samples and estimated for the levels of nitric oxide, peroxide, malondialdehyde, protein carbonyl, reduced glutathione, and activities of nitric oxide synthase and superoxide dismutase. RESULTS Biochemical analysis revealed a significant increase in the levels of reactive oxygen species with a parallel decrease in endogenous anti-oxidants in PTZ-kindled control animals. Daily treatment with levetiracetam and felbamate significantly decreased the PTZ-induced seizure score as well as the levels of nitric oxide (p<0.001), nitric oxide synthase activity (p<0.05), peroxide levels (p<0.05), and malondialdehyde (p<0.05). Levetiracetam and felbamate significantly decreased lipid and protein peroxidation whereas topiramate was found to reduce lipid peroxidation only. CONCLUSIONS An AED that produces anticonvulsant effect by the diversified mechanism of action such as levetiracetam, felbamate, and topiramate exhibited superior anti-oxidative stress activity in addition to their anticonvulsant activity.
Collapse
|
7
|
Di Domenico F, Barone E, Perluigi M, Butterfield DA. The Triangle of Death in Alzheimer's Disease Brain: The Aberrant Cross-Talk Among Energy Metabolism, Mammalian Target of Rapamycin Signaling, and Protein Homeostasis Revealed by Redox Proteomics. Antioxid Redox Signal 2017; 26:364-387. [PMID: 27626216 DOI: 10.1089/ars.2016.6759] [Citation(s) in RCA: 88] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
SIGNIFICANCE Alzheimer's disease (AD) is a multifactorial neurodegenerative disorder and represents one of the most disabling conditions. AD shares many features in common with systemic insulin resistance diseases, suggesting that it can be considered as a metabolic disease, characterized by reduced insulin-stimulated growth and survival signaling, increased oxidative stress (OS), proinflammatory cytokine activation, mitochondrial dysfunction, impaired energy metabolism, and altered protein homeostasis. Recent Advances: Reduced glucose utilization and energy metabolism in AD have been associated with the buildup of amyloid-β peptide and hyperphosphorylated tau, increased OS, and the accumulation of unfolded/misfolded proteins. Mammalian target of rapamycin (mTOR), which is aberrantly activated in AD since early stages, plays a key role during AD neurodegeneration by, on one side, inhibiting insulin signaling as a negative feedback mechanism and, on the other side, regulating protein homeostasis (synthesis/clearance). CRITICAL ISSUES It is likely that the concomitant and mutual alterations of energy metabolism-mTOR signaling-protein homeostasis might represent a self-sustaining triangle of harmful events that trigger the degeneration and death of neurons and the development and progression of AD. Intriguingly, the altered cross-talk between the components of such a triangle of death, beyond altering the redox homeostasis of the neuron, is further exacerbated by increased levels of OS that target and impair key components of the pathways involved. Redox proteomic studies in human samples and animal models of AD-like dementia led to identification of oxidatively modified components of the pathways composing the triangle of death, therefore revealing the crucial role of OS in fueling this aberrant vicious cycle. FUTURE DIRECTIONS The identification of compounds able to restore the function of the pathways targeted by oxidative damage might represent a valuable therapeutic approach to slow or delay AD. Antioxid. Redox Signal. 26, 364-387.
Collapse
Affiliation(s)
- Fabio Di Domenico
- 1 Department of Biochemical Sciences, Sapienza University of Rome , Rome, Italy
| | - Eugenio Barone
- 1 Department of Biochemical Sciences, Sapienza University of Rome , Rome, Italy .,2 Facultad de Salud, Instituto de Ciencias Biomédicas, Universidad Autónoma de Chile , Santiago, Chile
| | - Marzia Perluigi
- 1 Department of Biochemical Sciences, Sapienza University of Rome , Rome, Italy
| | - D Allan Butterfield
- 3 Department of Chemistry, Sanders-Brown Center of Aging, University of Kentucky , Lexington, Kentucky
| |
Collapse
|
8
|
Luna C, Alique M, Navalmoral E, Noci MV, Bohorquez-Magro L, Carracedo J, Ramírez R. Aging-associated oxidized albumin promotes cellular senescence and endothelial damage. Clin Interv Aging 2016; 11:225-36. [PMID: 27042026 PMCID: PMC4780186 DOI: 10.2147/cia.s91453] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Increased levels of oxidized proteins with aging have been considered a cardiovascular risk factor. However, it is unclear whether oxidized albumin, which is the most abundant serum protein, induces endothelial damage. The results of this study indicated that with aging processes, the levels of oxidized proteins as well as endothelial microparticles release increased, a novel marker of endothelial damage. Among these, oxidized albumin seems to play a principal role. Through in vitro studies, endothelial cells cultured with oxidized albumin exhibited an increment of endothelial damage markers such as adhesion molecules and apoptosis levels. In addition, albumin oxidation increased the amount of endothelial microparticles that were released. Moreover, endothelial cells with increased oxidative stress undergo senescence. In addition, endothelial cells cultured with oxidized albumin shown a reduction in endothelial cell migration measured by wound healing. As a result, we provide the first evidence that oxidized albumin induces endothelial injury which then contributes to the increase of cardiovascular disease in the elderly subjects.
Collapse
Affiliation(s)
- Carlos Luna
- Nephrology Unit, Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Reina Sofía University Hospital, Córdoba, Spain
| | - Matilde Alique
- Department of Systems Biology, Physiology Unit, Universidad de Alcalá, Madrid, Spain
| | - Estefanía Navalmoral
- Department of Systems Biology, Physiology Unit, Universidad de Alcalá, Madrid, Spain
| | | | | | - Julia Carracedo
- Nephrology Unit, Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Reina Sofía University Hospital, Córdoba, Spain
| | - Rafael Ramírez
- Department of Systems Biology, Physiology Unit, Universidad de Alcalá, Madrid, Spain
| |
Collapse
|
9
|
It Is All about (U)biquitin: Role of Altered Ubiquitin-Proteasome System and UCHL1 in Alzheimer Disease. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2016; 2016:2756068. [PMID: 26881020 PMCID: PMC4736377 DOI: 10.1155/2016/2756068] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Accepted: 11/26/2015] [Indexed: 02/07/2023]
Abstract
Free radical-mediated damage to macromolecules and the resulting oxidative modification of different cellular components are a common feature of aging, and this process becomes much more pronounced in age-associated pathologies, including Alzheimer disease (AD). In particular, proteins are particularly sensitive to oxidative stress-induced damage and these irreversible modifications lead to the alteration of protein structure and function. In order to maintain cell homeostasis, these oxidized/damaged proteins have to be removed in order to prevent their toxic accumulation. It is generally accepted that the age-related accumulation of “aberrant” proteins results from both the increased occurrence of damage and the decreased efficiency of degradative systems. One of the most important cellular proteolytic systems responsible for the removal of oxidized proteins in the cytosol and in the nucleus is the proteasomal system. Several studies have demonstrated the impairment of the proteasome in AD thus suggesting a direct link between accumulation of oxidized/misfolded proteins and reduction of this clearance system. In this review we discuss the impairment of the proteasome system as a consequence of oxidative stress and how this contributes to AD neuropathology. Further, we focus the attention on the oxidative modifications of a key component of the ubiquitin-proteasome pathway, UCHL1, which lead to the impairment of its activity.
Collapse
|
10
|
Mehrabi S, Wallace L, Cohen S, Yao X, Aikhionbare FO. Differential Measurements of Oxidatively Modified Proteins in Colorectal Adenopolyps. ACTA ACUST UNITED AC 2015; 6:288-299. [PMID: 26069854 PMCID: PMC4461072 DOI: 10.4236/ijcm.2015.64037] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Introduction Adenopolyps patients have a three-fold higher risk of colon cancer over the general population, which increases to six-fold if the polyps are multiple and with lower survival among African American population. Currently, 6% of CRC can be ascribed to mutations in particular genes. Moreover, the optimal management of patients with colorectal adenopolyps depends on the accuracy of appropriate staging strategies because patients with similar colorectal adenocarcinoma architecture display heterogeneity in the course and outcome of the disease. Oxidative stress, due to an imbalance between reactive oxygen species (ROS) and antioxidant capacities as well as a disruption of redox signaling, causes a wide range of damage to DNA, proteins, and lipids which promote tumor formation. Objective/Method This study applied spectrophotometric, dinitrophenylhydrazone (DNPH) assay, two-dimensional gel electrophoresis, and western blot analyses to assess the levels of oxidatively modified proteins in 41 pairs of primary colorectal tissues including normal/surrounding, adenopolyps (tubular, tubulovillous, villous, polypvillous) and carcinoma. Analysis of variance (ANOVA) and Student’s t-tests were utilized for the resulting data set. Results Our data showed that the levels of reactive protein carbonyl groups significantly increased as colorectal adenopolyps progresses to malignancy. No significant differences were found in the levels of carbonyl proteins between gender samples analyzed. For African American patients, there were, relative to Caucasians, 10% higher levels of reactive carbonyls in proteins of tubulovillous tissue samples (P < 0.05) and over 36% higher in levels in adenocarcinomas (P < 0.05). In normal tissues and tubular, there were no significant differences between the two groups in levels of protein carbonyls. Differences in the levels of protein carbonyl expression within individual patient samples with different number of tumor cells were notably evident. Conclusion Results suggested that oxidative stress could be involved in the modification of oxidatively carbonyl proteins in the precancer stages, leading to increased aggressiveness of colorectal polyps.
Collapse
Affiliation(s)
- Sharifeh Mehrabi
- Department of Medicine, Morehouse School of Medicine, Atlanta, USA
| | | | - Shakeria Cohen
- Department of Medicine, Morehouse School of Medicine, Atlanta, USA
| | - Xuebiao Yao
- Department of Physiology, Morehouse School of Medicine, Atlanta, USA
| | | |
Collapse
|
11
|
Oxidatively modified proteins in the serous subtype of ovarian carcinoma. BIOMED RESEARCH INTERNATIONAL 2014; 2014:585083. [PMID: 24795885 PMCID: PMC3985143 DOI: 10.1155/2014/585083] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/25/2013] [Revised: 02/01/2014] [Accepted: 02/24/2014] [Indexed: 01/17/2023]
Abstract
Serous subtype of ovarian cancer is considered to originate from fallopian epithelium mucosa that has been exposed to physiological changes resulting from ovulation. Ovulation influences an increased in inflammation of epithelial ovarian cells as results of constant exposure of cells to ROS. The imbalance between ROS and antioxidant capacities, as well as a disruption of redox signaling, causes a wide range of damage to DNA, proteins, and lipids. This study applied spectrophotometric, dinitrophenylhydrazone (DNPH) assay, two-dimensional gel electrophoresis, and Western blot analyses to assess the levels of oxidatively modified proteins in 100 primary serous epithelial ovarian carcinoma and normal/surrounding tissues. These samples were obtained from 56 Caucasian and 44 African-American patients within the age range of 61 ± 10 years. Analyses showed that the levels of reactive protein carbonyl groups increased as stages progressed to malignancy. Additionally, the levels of protein carbonyls in serous ovarian carcinoma among African Americans are 40% (P < 0.05) higher relative to Caucasian at similar advanced stages. Results suggest that oxidative stress is involved in the modification of carbonyl protein groups, leading to increased aggressiveness of epithelial ovarian tumors and may contribute to the disease's invasiveness among African Americans.
Collapse
|
12
|
Umstead TM, Phelps DS, Wang G, Floros J, Tarkington BK. In vitro exposure of proteins to ozone. Toxicol Mech Methods 2012; 12:1-16. [PMID: 20597812 DOI: 10.1080/15376510209167932] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
An in vitro system has been developed to expose proteins to ozone. The system is designed to deliver consistent and accurate levels of ozone over a range of concentrations (between 0.1 and >/=10 ppm) with extended exposure times (24 h or longer) in a humidified environment (100%). In the experiment presented in this article, ozone concentrations between 0.1 and 2.0 ppm were used. Ozone was generated by an electrical discharge ozonizer to ensure stability; it was continually monitored by an ultraviolet ozone analyzer and was precisely controlled by mass flow controllers, which gave reproducible results between runs. Humidity was closely regulated in the system to allow small amounts of protein solutions (50 muL or less) to be exposed without significant changes (<0.2%) in sample volume. The degree of surfactant protein-A (SP-A) oxidation by ozone was measured between runs to demonstrate the reproducibility of the system. A detailed description of the system is given, and protein oxidation detection methods and their limitations are discussed. Using these methods, we were able to assess oxidation of SP-A that apparently occurred prior to its isolation from the lung by bronchoalveolar lavage. This in vitro system allowed us to expose small amounts of protein to ozone in a simple, highly controlled, and reproducible manner.
Collapse
Affiliation(s)
- Todd M Umstead
- Department of Pediatrics, The Pennsylvania State University College of Medicine, Hershey, PennsylvaniaUSA
| | | | | | | | | |
Collapse
|
13
|
Huang X, Zhang J, Liu J, Sun L, Zhao H, Lu Y, Wang J, Li J. C-reactive protein promotes adhesion of monocytes to endothelial cells via NADPH oxidase-mediated oxidative stress. J Cell Biochem 2012; 113:857-67. [PMID: 22020763 DOI: 10.1002/jcb.23415] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Enhanced monocyte adhesion to endothelial cells is an early event in atherogenesis. It has been shown that C-reactive protein (CRP) plays a key role in atherogenesis. Here, we investigated the effects of CRP on monocyte-endothelial cell adhesion and tested the hypothesis that NADPH oxidase (NOX)-mediated oxidative stress might play a key role in CRP-induced monocyte-endothelial cell adhesion. Firstly, 36 patients with carotid intima-media thickness (IMT) incrassation and 34 controls were enrolled in this study. The levels of glucose, lipids, CRP, monocyte chemotractant protein (MCP-1), malondialdehyde (MDA), and protein carbonylation were analyzed. The results showed that carotid IMT was associated with abnormal lipid metabolism, including elevated CRP, triglycerides (TG) (P < 0.01) and decreased high density lipoprotein (HDL) level (P < 0.05). The levels of CRP and MCP-1 in patients with carotid IMT incrassation were increased compared with the controls (P < 0.01). Moreover, patients with carotid IMT incrassation displayed enhanced MDA and protein carbonylation levels (P < 0.01), accompanied by activation and up-regulation of NOX in monocytes (P < 0.05) compared with the controls. The monocytes isolated from five healthy donors were used for in vitro experiments. Reactive oxygen species (ROS) production and NOX expression in monocytes were examined. The results also indicated that CRP could promote the adhesion of monocyte-endothelial cell by up-regulation of MCP-1 expression (P < 0.05). Importantly, NFκ B and p38 MAPK signaling pathways, which were activated by NOX-derived ROS, were involved in CRP-induced monocyte-endothelial cell adhesion and up-regulation of MCP-1 expression. These data suggested that CRP could promote the adhesion of monocytes to endothelial cells via NOX-mediated oxidative stress. J. Cell. Biochem. 113: 857-867, 2012. © 2011 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Xiuqing Huang
- The Key Laboratory of Geriatrics, Beijing Hospital &Beijing Institute of Geriatrics, Ministry of Health, Beijing 100730, PR China
| | | | | | | | | | | | | | | |
Collapse
|
14
|
Andreazza AC, Andersen ML, Alvarenga TA, de-Oliveira MR, Armani F, Ruiz FS, Giglio L, Moreira JCF, Kapczinski F, Tufik S. Impairment of the mitochondrial electron transport chain due to sleep deprivation in mice. J Psychiatr Res 2010; 44:775-80. [PMID: 20176368 DOI: 10.1016/j.jpsychires.2010.01.015] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
It has been demonstrated that sleep deprivation is associated with altered expression of genes related to metabolic processes, response to stress and inflammation, circadian sleep/wake cycles, regulation of cell proliferation and various signaling pathways. However, the molecular mechanisms underlying these changes remain poorly understood. Thus, the present study aims to characterize the function of the mitochondrial electron transport chain in the brain using an animal model of paradoxical sleep deprivation (PSD). The question of whether sleep recovery (rebound) can reverse changes found after PSD is also addressed. Adult male inbred C57BL/6J mice were randomly distributed into three groups: home-cage control, PSD and sleep rebound groups. The PSD and rebound groups were subjected to PSD for 72 h. After this sleep deprivation period, the rebound group was returned to its home cage and allowed to sleep in an undisturbed and spontaneous fashion for 24h. The mitochondrial complex I-III, complex II, succinate dehydrogenase and complex II-III activities were then measured by spectrophotometric methods in sub-mitochondrial particles extracted from the prefrontal cortex, hippocampus, striatum and hypothalamus. Our results showed a significant decrease in the activity of complex I-III in the PSD and rebound groups as compared to the control group. The complex II and II-III activity were particularly decreased in the hypothalamus of the sleep rebound group. These results are consistent with the involvement of sleep in energy metabolism and corroborate previous experiments demonstrating the importance of the hypothalamus in sleep regulation.
Collapse
Affiliation(s)
- Ana C Andreazza
- Bipolar Disorders Program and Laboratory of Molecular Psychiatry, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Bacterial stressors in minimally processed food. Int J Mol Sci 2009; 10:3076-3105. [PMID: 19742126 PMCID: PMC2738913 DOI: 10.3390/ijms10073076] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2009] [Revised: 06/29/2009] [Accepted: 06/29/2009] [Indexed: 11/17/2022] Open
Abstract
Stress responses are of particular importance to microorganisms, because their habitats are subjected to continual changes in temperature, osmotic pressure, and nutrients availability. Stressors (and stress factors), may be of chemical, physical, or biological nature. While stress to microorganisms is frequently caused by the surrounding environment, the growth of microbial cells on its own may also result in induction of some kinds of stress such as starvation and acidity. During production of fresh-cut produce, cumulative mild processing steps are employed, to control the growth of microorganisms. Pathogens on plant surfaces are already stressed and stress may be increased during the multiple mild processing steps, potentially leading to very hardy bacteria geared towards enhanced survival. Cross-protection can occur because the overlapping stress responses enable bacteria exposed to one stress to become resistant to another stress. A number of stresses have been shown to induce cross protection, including heat, cold, acid and osmotic stress. Among other factors, adaptation to heat stress appears to provide bacterial cells with more pronounced cross protection against several other stresses. Understanding how pathogens sense and respond to mild stresses is essential in order to design safe and effective minimal processing regimes.
Collapse
|
16
|
Thybert D, Avner S, Lucchetti-Miganeh C, Chéron A, Barloy-Hubler F. OxyGene: an innovative platform for investigating oxidative-response genes in whole prokaryotic genomes. BMC Genomics 2008; 9:637. [PMID: 19117520 PMCID: PMC2631583 DOI: 10.1186/1471-2164-9-637] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2008] [Accepted: 12/31/2008] [Indexed: 11/25/2022] Open
Abstract
Background Oxidative stress is a common stress encountered by living organisms and is due to an imbalance between intracellular reactive oxygen and nitrogen species (ROS, RNS) and cellular antioxidant defence. To defend themselves against ROS/RNS, bacteria possess a subsystem of detoxification enzymes, which are classified with regard to their substrates. To identify such enzymes in prokaryotic genomes, different approaches based on similarity, enzyme profiles or patterns exist. Unfortunately, several problems persist in the annotation, classification and naming of these enzymes due mainly to some erroneous entries in databases, mistake propagation, absence of updating and disparity in function description. Description In order to improve the current annotation of oxidative stress subsystems, an innovative platform named OxyGene has been developed. It integrates an original database called OxyDB, holding thoroughly tested anchor-based signatures associated to subfamilies of oxidative stress enzymes, and a new anchor-driven annotator, for ab initio detection of ROS/RNS response genes. All complete Bacterial and Archaeal genomes have been re-annotated, and the results stored in the OxyGene repository can be interrogated via a Graphical User Interface. Conclusion OxyGene enables the exploration and comparative analysis of enzymes belonging to 37 detoxification subclasses in 664 microbial genomes. It proposes a new classification that improves both the ontology and the annotation of the detoxification subsystems in prokaryotic whole genomes, while discovering new ORFs and attributing precise function to hypothetical annotated proteins. OxyGene is freely available at:
Collapse
Affiliation(s)
- David Thybert
- CNRS UMR 6026, Interactions Cellulaires et Moléculaires, Equipe B@SIC, Université de Rennes 1, IFR140 GFAS, Campus de Beaulieu, Av. du Général Leclerc, 35042 Rennes, France.
| | | | | | | | | |
Collapse
|
17
|
Sultana R, Butterfield DA. Redox proteomics studies of in vivo amyloid beta-peptide animal models of Alzheimer's disease: Insight into the role of oxidative stress. Proteomics Clin Appl 2008; 2:685-96. [PMID: 21136866 DOI: 10.1002/prca.200780024] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2007] [Indexed: 12/21/2022]
Abstract
Alzheimer's disease (AD) is an age-related neurodegenerative disease. AD is characterized by the presence of senile plaques, neurofibrillary tangles, and synaptic loss. Amyloid β-peptide (Aβ), a component of senile plaques, has been proposed to play an important role in oxidative stress in AD brain and could be one of the key factors in the pathogenesis of AD. In the present review, we discuss some of the AD animal models that express Aβ, and compare the proteomics-identified oxidatively modified proteins between AD brain and those of Aβ models. Such a comparison would allow better understanding of the role of Aβ in AD pathogenesis thereby helping in developing potential therapeutics to treat or delay AD.
Collapse
Affiliation(s)
- Rukhsana Sultana
- Department of Chemistry, University of Kentucky, Lexington, KY, USA; Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, USA
| | | |
Collapse
|
18
|
Butterfield DA, Perluigi M, Sultana R. Oxidative stress in Alzheimer's disease brain: New insights from redox proteomics. Eur J Pharmacol 2006; 545:39-50. [PMID: 16860790 DOI: 10.1016/j.ejphar.2006.06.026] [Citation(s) in RCA: 253] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2005] [Revised: 11/28/2005] [Accepted: 06/13/2006] [Indexed: 12/20/2022]
Abstract
Alzheimer's disease, an age-related neurodegenerative disorder, is characterized clinically by a progressive loss of memory and cognitive functions. Neuropathologically, Alzheimer's disease is defined by the accumulation of extracellular amyloid protein deposited senile plaques and intracellular neurofibrillary tangles made of abnormal and hyperphosphorylated tau protein, regionalized neuronal death, and loss of synaptic connections within selective brain regions. Evidence has suggested a critical role for amyloid-beta peptide metabolism and oxidative stress in Alzheimer's disease pathogenesis and progression. Among the other indices of oxidative stress in Alzheimer's disease brain are protein carbonyls and 3-nitrotyrosine, which are the markers of protein oxidation. Thus, in this review, we discuss the application of redox proteomics for the identification of oxidatively modified proteins in Alzheimer's disease brain and also discuss the functions associated with the identified oxidized proteins in relation to Alzheimer's disease pathology. The information obtained from proteomics may be helpful in understanding the molecular mechanisms involved in the development and progression of Alzheimer's disease as well as of other neurodegenerative disorders. Further, redox proteomics may provide potential targets for drug therapy in Alzheimer's disease.
Collapse
|
19
|
|
20
|
Stadtman ER, Arai H, Berlett BS. Protein oxidation by the cytochrome P450 mixed-function oxidation system. Biochem Biophys Res Commun 2005; 338:432-6. [PMID: 16140263 DOI: 10.1016/j.bbrc.2005.07.203] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2005] [Accepted: 07/20/2005] [Indexed: 10/25/2022]
Abstract
This mini-review summarizes results of studies on the oxidation of proteins and low-density lipoprotein (LDL) by various mixed-function oxidation (MFO) systems. Oxidation of LDL by the O2/FeCl3/H2O2/ascorbate MFO system is dependent on all four components and is much greater when reactions are carried out in the presence of a physiological bicarbonate/CO2 buffer system as compared to phosphate buffer. However, FeCl3 in this system could be replaced by hemin or the heme-containing protein, hemoglobin, or cytochrome c. Oxidation of LDL by the O2/cytochrome P450 cytochrome c reductase/NADPH/FeCl3 MFO system is only slightly higher (25%) in the bicarbonate/CO2 buffer as compared to phosphate buffer, but is dependent on all components except FeCl3. Omission of FeCl3 led to a 60% loss of activity. These results suggest that peroxymonobicarbonate and/or free radical derivatives of bicarbonate ion and/or CO2 might contribute to LDL oxidation by these MFO systems.
Collapse
Affiliation(s)
- Earl R Stadtman
- Laboratory of Biochemistry, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892-8012, USA.
| | | | | |
Collapse
|
21
|
Khan MAS, Chock PB, Stadtman ER. Knockout of caspase-like gene, YCA1, abrogates apoptosis and elevates oxidized proteins in Saccharomyces cerevisiae. Proc Natl Acad Sci U S A 2005; 102:17326-31. [PMID: 16301538 PMCID: PMC1287485 DOI: 10.1073/pnas.0508120102] [Citation(s) in RCA: 88] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
In our previous study, we established that inhibition of apoptosis by the general caspase inhibitor is associated with an increase in the level of oxidized proteins in a multicellular eukaryotic system. To gain further insight into a potential link between oxidative stress and apoptosis, we carried out studies with Saccharomyces cerevisiae, which contains a gene (YCA1) that encodes synthesis of metacaspase, a homologue of the mammalian caspase, and is known to play a crucial role in the regulation of yeast apoptosis. We show that upon exposure to H(2)O(2), the accumulation of protein carbonyls is much greater in a Delta yca1 strain lacking the YCA1 gene than in the wild type and that apoptosis was abrogated in the Delta yca1 strain, whereas wild type underwent apoptosis as measured by externalization of phosphatidylserine and the display of TUNEL-positive nuclei. We also show that H(2)O(2)-mediated stress leads to up-regulation of the 20S proteasome and suppression of ubiquitinylation activities. These findings suggest that deletion of the apoptotic-related caspase-like gene leads to a large H(2)O(2)-dependent accumulation of oxidized proteins and up-regulation of 20S proteasome activity.
Collapse
Affiliation(s)
- Mohammed A S Khan
- Laboratory of Biochemistry, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892-8012, USA
| | | | | |
Collapse
|
22
|
Papin JA, Hunter T, Palsson BO, Subramaniam S. Reconstruction of cellular signalling networks and analysis of their properties. Nat Rev Mol Cell Biol 2005; 6:99-111. [PMID: 15654321 DOI: 10.1038/nrm1570] [Citation(s) in RCA: 321] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
The study of cellular signalling over the past 20 years and the advent of high-throughput technologies are enabling the reconstruction of large-scale signalling networks. After careful reconstruction of signalling networks, their properties must be described within an integrative framework that accounts for the complexity of the cellular signalling network and that is amenable to quantitative modelling.
Collapse
Affiliation(s)
- Jason A Papin
- Department of Bioengineering, 9500 Gilman Drive, Mail Code 0412, University of California, San Diego, La Jolla, California 92093-0412, USA
| | | | | | | |
Collapse
|
23
|
Papin JA, Palsson BO. The JAK-STAT signaling network in the human B-cell: an extreme signaling pathway analysis. Biophys J 2005; 87:37-46. [PMID: 15240442 PMCID: PMC1304358 DOI: 10.1529/biophysj.103.029884] [Citation(s) in RCA: 86] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Large-scale models of signaling networks are beginning to be reconstructed and corresponding analysis frameworks are being developed. Herein, a reconstruction of the JAK-STAT signaling system in the human B-cell is described and a scalable framework for its network analysis is presented. This approach is called extreme signaling pathway analysis and involves the description of network properties with systemically independent basis vectors called extreme pathways. From the extreme signaling pathways, emergent systems properties of the JAK-STAT signaling network have been characterized, including 1), a mathematical definition of network crosstalk; 2), an analysis of redundancy in signaling inputs and outputs; 3), a study of reaction participation in the network; and 4), a delineation of 85 correlated reaction sets, or systemic signaling modules. This study is the first such analysis of an actual biological signaling system. Extreme signaling pathway analysis is a topologically based approach and assumes a balanced use of the signaling network. As large-scale reconstructions of signaling networks emerge, such scalable analyses will lead to a description of the fundamental systems properties of signal transduction networks.
Collapse
Affiliation(s)
- Jason A Papin
- Department of Bioengineering, University of California, San Diego, La Jolla 92093, USA
| | | |
Collapse
|
24
|
Jeong J, Rouault TA, Levine RL. Identification of a Heme-sensing Domain in Iron Regulatory Protein 2. J Biol Chem 2004; 279:45450-4. [PMID: 15316013 DOI: 10.1074/jbc.m407562200] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Iron regulatory protein 2 coordinates the cellular regulation of iron metabolism by binding to iron-responsive elements in mRNA. The protein is synthesized constitutively but is rapidly degraded when iron stores are replete. The mechanisms that prevent degradation during iron deficiency or promote degradation during iron sufficiency are not delineated. Iron regulatory protein 2 contains a domain not present in the closely related iron regulatory protein 1, and we found that this domain binds heme with high affinity. A cysteine within the domain is axially liganded to the heme, as occurs in cytochrome P450. The protein-bound heme reacts with molecular oxygen to mediate the oxidation of cysteine, including beta-elimination of the sulfur to yield alanine. This covalent modification may thus mark the protein molecule for degradation by the proteasome system, providing another mechanism by which heme can regulate the level of iron regulatory protein 2.
Collapse
Affiliation(s)
- Jinsook Jeong
- Laboratory of Biochemistry, NHLBI, National Institutes of Health, Bethesda, MD 20892, USA
| | | | | |
Collapse
|
25
|
Mostertz J, Hecker M. Patterns of protein carbonylation following oxidative stress in wild-type and sigB Bacillus subtilis cells. Mol Genet Genomics 2003; 269:640-8. [PMID: 12845527 DOI: 10.1007/s00438-003-0877-4] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2003] [Accepted: 06/01/2003] [Indexed: 10/26/2022]
Abstract
Oxidative stress causes damage to nucleic acids, membrane lipids and proteins. One striking effect is the metal-catalyzed, site-specific carbonylation of proteins. In the gram-positive soil bacterium Bacillus subtilis, the PerR-dependent specific stress response and the sigmaB-dependent general stress response act together to make cells more resistant to oxidative stress. In this study, we analyzed the carbonylation of cytoplasmic proteins in response to hydrogen peroxide stress in B. subtilis. Furthermore, we asked whether the sigmaB-dependent response to oxidative stress also confers protection against protein carbonylation. To monitor the amount and specificity of protein damage, carbonyls were derivatized with 2,4-dinitrophenylhydrazine, and the resulting stable hydrazones were detected by immunoanalysis of proteins separated by one- or two-dimensional gel electrophoresis. The overall level of protein carbonylation increased strongly in cells treated with hydrogen peroxide. Several proteins, including the elongation factors EF-G, TufA and EF-Ts, were found to be highly carbonylated. Induction of the peroxide specific stress response by treatment with sub-lethal peroxide concentrations, prior to exposure to otherwise lethal levels of peroxide, markedly reduced the degree of protein carbonylation. Cells starved for glucose also showed only minor amounts of peroxide-mediated protein carbonylation compared to exponentially growing cells. We could not detect any differences between wild-type and deltasigB cells starved for glucose or preadapted by heat treatment with respect to the amount or specificity of protein damage incurred upon subsequent exposure to peroxide stress. However, artificial preloading with proteins that are normally induced by sigmaB-dependent mechanisms resulted in a lower level of protein carbonylation when cells were later subjected to oxidative stress.
Collapse
Affiliation(s)
- J Mostertz
- Institut für Mikrobiologie, Ernst-Moritz-Arndt-Universität Greifswald, F.-L.-Jahnstr. 15, 17489 Greifswald, Germany
| | | |
Collapse
|
26
|
Abstract
Mathematical modeling is one of the key methodologies of metabolic engineering. Based on a given metabolic model different computational tools for the simulation, data evaluation, systems analysis, prediction, design and optimization of metabolic systems have been developed. The currently used metabolic modeling approaches can be subdivided into structural models, stoichiometric models, carbon flux models, stationary and nonstationary mechanistic models and models with gene regulation. However, the power of a model strongly depends on its basic modeling assumptions, the simplifications made and the data sources used. Model validation turns out to be particularly difficult for metabolic systems. The different modeling approaches are critically reviewed with respect to their potential and benefits for the metabolic engineering cycle. Several tools that have emerged from the different modeling approaches including structural pathway synthesis, stoichiometric pathway analysis, metabolic flux analysis, metabolic control analysis, optimization of regulatory architectures and the evaluation of rapid sampling experiments are discussed.
Collapse
Affiliation(s)
- Wolfgang Wiechert
- Department of Simulation and Computer Science, Institute of Mechanical and Control Engineering, University of Siegen, Paul-Bonatz-Str. 9-11, D-57068 Siegen, Germany.
| |
Collapse
|
27
|
Covert MW, Schilling CH, Palsson B. Regulation of gene expression in flux balance models of metabolism. J Theor Biol 2001; 213:73-88. [PMID: 11708855 DOI: 10.1006/jtbi.2001.2405] [Citation(s) in RCA: 271] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Genome-scale metabolic networks can now be reconstructed based on annotated genomic data augmented with biochemical and physiological information about the organism. Mathematical analysis can be performed to assess the capabilities of these reconstructed networks. The constraints-based framework, with flux balance analysis (FBA), has been used successfully to predict time course of growth and by-product secretion, effects of mutation and knock-outs, and gene expression profiles. However, FBA leads to incorrect predictions in situations where regulatory effects are a dominant influence on the behavior of the organism. Thus, there is a need to include regulatory events within FBA to broaden its scope and predictive capabilities. Here we represent transcriptional regulatory events as time-dependent constraints on the capabilities of a reconstructed metabolic network to further constrain the space of possible network functions. Using a simplified metabolic/regulatory network, growth is simulated under various conditions to illustrate systemic effects such as catabolite repression, the aerobic/anaerobic diauxic shift and amino acid biosynthesis pathway repression. The incorporation of transcriptional regulatory events in FBA enables us to interpret, analyse and predict the effects of transcriptional regulation on cellular metabolism at the systemic level.
Collapse
Affiliation(s)
- M W Covert
- Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093-0412, USA
| | | | | |
Collapse
|
28
|
Abstract
Although different theories have been proposed to explain the aging process, it is generally agreed that there is a correlation between aging and the accumulation of oxidatively damaged proteins, lipids, and nucleic acids. Oxidatively modified proteins have been shown to increase as a function of age. Studies reveal an age-related increase in the level of protein carbonyl content, oxidized methionine, protein hydrophobicity, and cross-linked and glycated proteins as well as the accumulation of less active enzymes that are more susceptible to heat inactivation and proteolytic degredation. Factors that decelerate protein oxidation also increase the life span of animals and vice versa. Furthermore, a number of age-related diseases have been shown to be associated with elevated levels of oxidatively modified proteins. The chemistry of reactive oxygen species-mediated protein modification will be discussed. The accumulation of oxidatively modified proteins may reflect deficiencies in one or more parameters of a complex function that maintains a delicate balance between the presence of a multiplicity of prooxidants, antioxidants, and repair, replacement, or elimination of biologically damaged proteins.
Collapse
Affiliation(s)
- E R Stadtman
- Laboratory of Biochemistry, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland 20892-0342, USA.
| |
Collapse
|
29
|
Abstract
The oxidative modification of proteins by reactive species, especially reactive oxygen species, is implicated in the etiology or progression of a panoply of disorders and diseases. These reactive species form through a large number of physiological and non-physiological reactions. An increase in the rate of their production or a decrease in their rate of scavenging will increase the oxidative modification of cellular molecules, including proteins. For the most part, oxidatively modified proteins are not repaired and must be removed by proteolytic degradation, and a decrease in the efficiency of proteolysis will cause an increase in the cellular content of oxidatively modified proteins. The level of these modified molecules can be quantitated by measurement of the protein carbonyl content, which has been shown to increase in a variety of diseases and processes, most notably during aging. Accumulation of modified proteins disrupts cellular function either by loss of catalytic and structural integrity or by interruption of regulatory pathways.
Collapse
Affiliation(s)
- E R Stadtman
- Laboratory of Biochemistry, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland 20892-0320, USA.
| | | |
Collapse
|
30
|
Moskovitz J, Poston JM, Berlett BS, Nosworthy NJ, Szczepanowski R, Stadtman ER. Identification and characterization of a putative active site for peptide methionine sulfoxide reductase (MsrA) and its substrate stereospecificity. J Biol Chem 2000; 275:14167-72. [PMID: 10799493 DOI: 10.1074/jbc.275.19.14167] [Citation(s) in RCA: 148] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Peptide methionine sulfoxide reductases (MsrA) from many different organisms share a consensus amino acid sequence (GCFWG) that could play an important role in their active site. Site-directed single substitution of each of these amino acids except glycines in the yeast MsrA resulted in total loss of enzyme activity. Nevertheless, all the recombinant MsrA mutants and native proteins had a very similar circular dichroism spectrum. The demonstration that either treatment with iodoacetamide or replacement of the motif cysteine with serine leads to inactivation of the enzyme underscores the singular importance of cysteine residues in the activity of MsrA. The recombinant yeast MsrA was used for general characterization of the enzyme. Its K(m) value was similar to the bovine MsrA and appreciably lower than the K(m) of the bacterial enzyme. Also, it was shown that the enzymatic activity increased dramatically with increasing ionic strength. The recombinant yeast MsrA activity and the reduction activity of free methionine sulfoxide(s) were stereoselective toward the L-methionine S-sulfoxide and S-methyl p-tolyl sulfoxide. It was established that a methionine auxotroph yeast strain could grow on either form of L-methionine sulfoxide.
Collapse
Affiliation(s)
- J Moskovitz
- Laboratory of Biochemistry and Laboratory of Biochemical Genetics, NHLBI, National Institutes of Health, Bethesda, Maryland 20982-0320, USA.
| | | | | | | | | | | |
Collapse
|
31
|
Figueiredo-Pereira ME, Cohen G. The ubiquitin/proteasome pathway: friend or foe in zinc-, cadmium-, and H2O2-induced neuronal oxidative stress. Mol Biol Rep 1999; 26:65-9. [PMID: 10363649 DOI: 10.1023/a:1006909918866] [Citation(s) in RCA: 49] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
One of the hallmarks of neurodegeneration is the accumulation of ubiquitinated proteins in intraneuronal inclusions in the cytosol, endosomes/lysosomes and nuclei of affected cells. The relationship between inclusion production and cell viability is not well understood. On the one hand inclusions may be beneficial and result from an attempt of the cell to isolate a subclass of ubiquitinated proteins that are not effectively degraded. On the other hand, the inclusions may impede normal cell function contributing to cell death. To address this issue we treated mouse neuronal HT4 cells with three toxic agents cadmium, zinc and H2O2, and investigated their effects on glutathione homeostasis, on accumulation of ubiquitinated proteins and on cell viability. The three treatments induce oxidative stress manifested by decreases in glutathione (GSH) and/or increases in protein mixed disulfides (PrSSG). After an overnight recovery period in the absence of treatment, GSH and PrSSG were restored to almost normal levels. However, the levels of ubiquitinated proteins were significantly increased, and cell viability was sharply reduced. These results suggest that the ubiquitin-proteasome pathway is recruited for removal of proteins that are oxidatively modified. However, if the ubiquitinated proteins are not efficiently degraded, they accumulate in the cell and contribute to a decrease in cell viability.
Collapse
Affiliation(s)
- M E Figueiredo-Pereira
- Department of Biological Sciences, Hunter College of City University of New York, NY 10021, USA
| | | |
Collapse
|
32
|
Figueiredo-Pereira ME, Yakushin S, Cohen G. Disruption of the intracellular sulfhydryl homeostasis by cadmium-induced oxidative stress leads to protein thiolation and ubiquitination in neuronal cells. J Biol Chem 1998; 273:12703-9. [PMID: 9582293 DOI: 10.1074/jbc.273.21.12703] [Citation(s) in RCA: 152] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Cadmium is a potent cell poison known to cause oxidative stress by increasing lipid peroxidation and/or by changing intracellular glutathione levels and to affect the ubiquitin/ATP-dependent proteolytic pathway. However, the cellular mechanisms involved in cadmium toxicity are still not well understood, especially in neuronal cells. To investigate the relationship between cadmium-induced oxidative stress and the ubiquitin/ATP-dependent pathway, we treated cultures of neuronal cells with different concentrations of the metal ion. In addition to decreases in glutathione levels, we observed marked increases in protein-mixed disulfides (Pr-SSGs) after exposure of HT4 cells (a mouse neuronal cell line) or rat primary mesencephalic cultures to Cd2+. The increases in intracellular levels of Pr-SSGs were concurrent with increases in the levels of ubiquitinated proteins (Ub proteins) when the HT4 cells were subjected to lower (25 microM or less) concentrations of cadmium. However, higher concentrations of cadmium (50 microM), which were toxic, led to increases in Pr-SSGs but inhibited ubiquitination, probably reflecting inhibition of ubiquitinating enzymes. The cadmium-induced changes in Pr-SSGs and Ub proteins were not affected when more than 85% of intracellular glutathione was removed from the cells by the glutathione synthetase inhibitor L-buthionine-(S,R)-sulfoximine. However, the reducing agent dithiothreitol, which prevented the build up of Pr-SSGs in the cell, also blocked the accumulation of Ub proteins induced by cadmium. In addition, dithiothreitol blocked the effects of the higher toxic (50 microM) concentrations of cadmium on cytotoxicity and on glutathione, Pr-SSGs, and Ub proteins. Together, these results strongly suggest that changes in the levels of intracellular Pr-SSGs and ubiquitin-protein conjugates in neuronal cells are responses closely associated with the disruption of intracellular sulfhydryl homeostasis caused by cadmium-mediated oxidative stress.
Collapse
Affiliation(s)
- M E Figueiredo-Pereira
- Department of Biological Sciences, Hunter College of City University of New York, New York 10021, USA.
| | | | | |
Collapse
|
33
|
Abstract
Highly reactive oxygen species that are formed during normal metabolism and under conditions of oxidative stress are able to oxidize proteins or convert lipid and carbohydrate derivatives to compounds that react with functional groups on proteins. Among other changes, these ROS-mediated reactions lead to the formation of protein carbonyl derivatives, which serves as a marker of ROS-mediated protein damage. On the basis of this marker, it is established that oxidatively damaged protein is associated with aging and some diseases. The accumulation of oxidatively damaged protein reflects the balance among a myriad of factors that govern the rates of ROS generation and the rate at which damaged protein is degraded. Peroxynitrite, which is formed under normal physiological conditions, is able to oxidize methionine residues in proteins and to nitrate tyrosine residues; however, its ability to do so is dependent on the availability of CO2, which stimulates the nitration of tyrosine residues but inhibits the oxidation of methionine residues. Nitration of tyrosine residues may contribute to peroxynitrite toxicity, as nitration precludes the phosphorylation or nucleotidylation of tyrosine residues and thereby seriously compromises one of the most important mechanisms of cellular regulation and signal transduction.
Collapse
Affiliation(s)
- E R Stadtman
- Laboratory of Biochemistry, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland 20892-0342, USA
| | | |
Collapse
|
34
|
Abstract
It is well established that periods of increased contractile activity result in significant changes in muscle structure and function. Such morphological changes as sarcomeric Z-line disruption and sarcoplasmic reticulum vacuolization are characteristic of exercise-induced muscle injury. While the precise mechanism(s) underlying the perturbations to muscle following exercise remains to be elucidated, it is clear that disturbances in Ca2+ homeostasis and changes in the rate of protein degradation occur. The resulting elevation in intracellular [Ca2+] activates the non-lysosomal cysteine protease, calpain. Because calpain cleaves a variety of protein substrates including cytoskeletal and myofibrillar proteins, calpain-mediated degradation is thought to contribute to the changes in muscle structure and function that occur immediately following exercise. In addition, calpain activation may trigger the adaptation response to muscle injury. The purpose of this paper is to: (i) review the chemistry of the calpain-calpastatin system; (ii) provide evidence for the involvement of the non-lysosomal, calcium-activated neutral protease (calpain) in the response of skeletal muscle protein breakdown to exercise (calpain hypothesis); and (iii) describe the possible involvement of calpain in the inflammatory and regeneration response to exercise.
Collapse
Affiliation(s)
- A N Belcastro
- School of Rehabilitation Sciences, Faculty of Medicine, University of British Columbia, Vancouver, Canada
| | | | | |
Collapse
|
35
|
Affiliation(s)
- B S Berlett
- Laboratory of Biochemistry, NHLBI, National Institutes of Health, Bethesda, Maryland 20892, USA
| | | |
Collapse
|
36
|
|
37
|
Reinbothe C, Apel K, Reinbothe S. A light-induced protease from barley plastids degrades NADPH:protochlorophyllide oxidoreductase complexed with chlorophyllide. Mol Cell Biol 1995; 15:6206-12. [PMID: 7565773 PMCID: PMC230872 DOI: 10.1128/mcb.15.11.6206] [Citation(s) in RCA: 52] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
The NADPH:protochlorophyllide oxidoreductase precursor protein (pPorA) of barley (Hordeum vulgare L. cv. Carina), synthesized from a full-length cDNA clone by coupling in vitro transcription and translation, is a catalytically active protein. It converts protochlorophyllide to chlorophyllide in a light- and NADPH-dependent manner. At least the pigment product of catalysis remains tightly bound to the precursor protein. The chlorophyllide-pPorA complex differs markedly from the protochlorophyllide-pPorA complex with respect to sensitivity to attack by a light-induced, nucleus-encoded, and energy-dependent protease activity of barley plastids. The pPorA-chlorophyllide complex is rapidly degraded, in contrast to pPorA-protochlorophyllide complexes containing or lacking NADPH, which are both resistant to protease treatment. Unexpectedly, pPorA devoid of its substrates or products was less sensitive to proteolysis than the pPorA-chlorophyllide complex, suggesting that both substrate binding and product formation during catalysis had caused differential changes in protein conformation.
Collapse
Affiliation(s)
- C Reinbothe
- Department of Genetics, Swiss Federal Institute of Technology, Zurich, Switzerland
| | | | | |
Collapse
|
38
|
Affiliation(s)
- E R Stadtman
- NHLBI, Laboratory of Biochemistry, National Institutes of Health, Bethesda, Maryland 20892, USA
| |
Collapse
|
39
|
Modification of glucose-6-phosphate dehydrogenase by 4-hydroxy-2-nonenal. Formation of cross-linked protein that inhibits the multicatalytic protease. J Biol Chem 1994. [DOI: 10.1016/s0021-9258(17)31853-7] [Citation(s) in RCA: 240] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
|
40
|
Belcastro AN, Gilchrist JS, Scrubb JA, Arthur G. Calcium-supported calpain degradation rates for cardiac myofibrils in diabetes. Sulfhydryl and hydrophobic interactions. Mol Cell Biochem 1994; 135:51-60. [PMID: 7816056 DOI: 10.1007/bf00925960] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
OBJECTIVE The purpose was to investigate the calcium required for calpain-mediated degradation of selected cardiac myofibril proteins modified by diabetes, sulfhydryl (SH) and hydrophobic reagents. METHODS After 20 weeks of streptozotocin-induced (55 mg.kg-1) diabetes, calcium sensitive calpain (1.5 U.ml-1) degradation rates of purified cardiac myofibrillar proteins (1 mg.ml-1) were measured, in vitro, and compared to degradation rates for N-ethylmaleimide (NEM) and 2-p-toluidinylnapthalene-6-sulfonate (TNS) treated samples. RESULTS Diabetes (blood glucose of 550 +/- 32 mg.dl-1) reduced the yield of purified myofibrillar protein with minimal change in fibril protein composition. Total SH group reactivities (nmol.mg-1.30min) were 220 +/- 21, 163 +/- 17 and 156 +/- 24 for control, diabetic and NEM-treated (0.5 mM) myofibrils (p < or = 0.05). Calpain degradation rates were faster for all diabetic and SH modified myofibrillar proteins (p < or = 0.05), with a 45 and 35% reduction in the pCa50 for a 37 kDa protein of diabetic and NEM-treated fibril complexes. For control myofibrils, both 100 and 200 uM TNS, reduced calpain degradation rates to a similar extent for all substrate proteins. In contrast, diabetic and NEM-treated samples showed a further reduction in calpain degradation rates with increasing TNS from 100 to 200 uM. CONCLUSION Our results support the hypothesis that in diabetes the calcium requirements for calpain degradation rates are reduced and dependent upon sulfhydryl group status and Ca(2+)-induced hydrophobic interactions, implicating a 37 kDa myofbillar-complexed protein.
Collapse
Affiliation(s)
- A N Belcastro
- Laboratory of Cell Physiology and Exercise, University of British Columbia, Vancouver, Canada
| | | | | | | |
Collapse
|
41
|
Mukhopadhyay C, Chatterjee I. NADPH-initiated cytochrome P450-mediated free metal ion-independent oxidative damage of microsomal proteins. Exclusive prevention by ascorbic acid. J Biol Chem 1994. [DOI: 10.1016/s0021-9258(17)36845-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
|
42
|
Pell EJ, Eckardt NA, Glick RE. Biochemical and molecular basis for impairment of photosynthetic potential. PHOTOSYNTHESIS RESEARCH 1994; 39:453-462. [PMID: 24311136 DOI: 10.1007/bf00014598] [Citation(s) in RCA: 26] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/1993] [Accepted: 11/08/1993] [Indexed: 06/02/2023]
Abstract
Ozone induces reductions in net photosynthesis in a large number of plant species. A primary mechanism by which photosynthesis is reduced is through impact on carbon dioxide fixation. Ozone induces loss in Rubisco activity associated with loss in concentration of the protein. Evidence is presented that ozone may induce oxidative modification of Rubisco leading to subsequent proteolysis. In addition, plants exposed to ozone sustain reduction in rbcS, the mRNA for the small subunit of Rubisco. This loss in rbcS mRNA may lead to a reduced potential for synthesis of the protein. The regulation of O3-induced loss of Rubisco, and implications of the decline in this protein in relation to accelerated senescence are discussed.
Collapse
Affiliation(s)
- E J Pell
- Department of Plant Pathology, the Pennsylvania State University, 16802, University Park, PA, USA
| | | | | |
Collapse
|
43
|
Schwerdt G, Huth W. Turnover and transformation of mitochondrial acetyl-CoA acetyltransferase into CoA-modified forms. Biochem J 1993; 292 ( Pt 3):915-9. [PMID: 8100417 PMCID: PMC1134201 DOI: 10.1042/bj2920915] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Rat liver mitochondrial acetyl-CoA acetyltransferase (acetoacetyl-CoA thiolase, EC 2.3.1.9) exists additionally in the CoA-modified forms A1 and A2. After a pulse of radioactivity using [35S]methionine in hepatocytes, the highest radioactivity was obtained in the unmodified enzyme. Over the chase time, the radioactivity in the unmodified enzyme decreased, but simultaneously increased in both CoA-modified forms, thus proving that the fully active unmodified enzyme exists before the partially active modified forms A1 and A2. Also, the specific radioactivity (ratio % radioactivity/% immunoreactive area) of A1 > A2 demonstrates a sequential CoA modification of form A1 to form A2. Acetyl-CoA acetyltransferase was degraded with an apparent half-life of 38.0 h: the modified forms A1 and A2 have half-lives of 24.5 and 7.2 h. The physiological meaning of the CoA modification of acetyl-CoA acetyltransferase is not yet understood.
Collapse
Affiliation(s)
- G Schwerdt
- Institut für Biochemie und Molekulare Zellbiologie, Georg-August-Universität Göttingen, Federal Republic of Germany
| | | |
Collapse
|
44
|
Abstract
By analogy to some pathologies (such as demyelinating diseases, arthritis and inflammatory processes) where the loss of cellular integrity is the starting point of tissue oxidative damage, it is proposed that some dementia types could be derived from a similar mechanism. The following oxidative events are proposed: (a) different agents could alter capillary or neuron integrity with the subsequent leakage of oxidases, proteases and transition metals from cellular compartments; (b) the persistence of the damaging agent, possible depletion of antioxidative defenses and concomitant loss of neuron function; (c) alteration of adjacent cells in the same manner; and (d) finally localized brain necrosis and progression of the dementia.
Collapse
Affiliation(s)
- E Roche
- Department de Biochimie Clinique, CMU, Genève, Switzerland
| | | |
Collapse
|
45
|
Sanllorenti PM, Tardivo DB, Conde RD. Dietary level of protein regulates glyceraldehyde-3-phosphate dehydrogenase content and synthesis rate in mouse liver cytosol. Mol Cell Biochem 1992; 115:117-28. [PMID: 1448056 DOI: 10.1007/bf00230321] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
The content of liver cytosolic proteins was studied in mice subjected to protein depletion followed by refeeding with a normal diet. Depletion elicited either the accumulation or the decrease of several polypeptides, being the early increase of a M(r) 36,000 polypeptide the most pronounced change observed. The refeeding with a normal diet for 2 days caused a return of the cytosol protein composition to that of normally fed animals. The M(r) 36,000 polypeptide was identified as glyceraldehyde-3-phosphate dehydrogenase (GAPDH). Its molecular weight, the sequence of its first twenty amino acid residues, its amino acid composition and its antigenic properties were found to be similar with those of GAPDH from different mammalian cells. During the first 2 days of protein depletion, both the GAPDH polypeptide content and activity increased. Thereafter, the enzymatic activity of GAPDH decreased, whereas GAPDH protein mass decreased in a lesser extent. The accumulation of GAPDH and other particular polypeptides in the cytosols of protein depleted mice was associated with an increased synthesis. The refeeding with a normal diet caused an immediate return to the synthesis pattern of normal livers.
Collapse
Affiliation(s)
- P M Sanllorenti
- Instituto de Investigaciones Biologicas, Facultad de Ciencias Exactas y Naturales, Universidad Nacional de Mar del Plata, Agentina
| | | | | |
Collapse
|
46
|
Santini SA, Mordente A, Meucci E, Miggiano GA, Martorana GE. Conformational stability of bovine alpha-crystallin. Evidence for a destabilizing effect of ascorbate. Biochem J 1992; 287 ( Pt 1):107-12. [PMID: 1417762 PMCID: PMC1133130 DOI: 10.1042/bj2870107] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Short-term incubation of bovine alpha-crystallin with ascorbate alters the protein conformational stability. The denaturation curves with urea and guanidinium-chloride show different patterns, suggesting a deviation from a two-state mechanism owing to the presence of one or more intermediates in the unfolding of ascorbate-modified alpha-crystallin. Furthermore, the latter protein profiles are shifted to lower denaturant concentrations indicating a destabilizing action of ascorbate, which is capable of facilitating protein dissociation into subunits as demonstrated by gel filtration with 1.5 M-urea. The decrease in conformational stability cannot be ascribed to any major structural alteration, but rather to localized changes in the protein molecule. In fact, no difference between native and ascorbate-treated alpha-crystallin can be detected by amino acid analysis but perturbation of the tryptophan and tyrosine environment is indicated by alterations in intrinsic fluorescence. Furthermore, turbidity and light-scattering measurements suggest an involvement of the lysine side chains, since aggregability patterns with acetylsalicylic acid are significantly altered. The ascorbate-destabilizing effect on the conformational stability of alpha-crystallin, probably exerted through oxidative modification of amino acid residues and/or the formation of covalent adducts, provokes unfavourable steric interactions between residues along the polypeptide chains, thus favouring aggregation and insolubilization of crystallins which can lead to cataract formation, as also demonstrated by proteolytic digestion patterns which show a lower rate of degradation of the ascorbate-modified alpha-crystallin.
Collapse
Affiliation(s)
- S A Santini
- Istituto di Chimica Biologica, Università Cattolica del S. Cuore, Facoltà di Medicina e Chirurgia, Agostino Gemelli, Roma, Italy
| | | | | | | | | |
Collapse
|
47
|
Simpson JA, Narita S, Gieseg S, Gebicki S, Gebicki JM, Dean RT. Long-lived reactive species on free-radical-damaged proteins. Biochem J 1992; 282 ( Pt 3):621-4. [PMID: 1554345 PMCID: PMC1130831 DOI: 10.1042/bj2820621] [Citation(s) in RCA: 151] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
We have demonstrated two novel reactive species on radical-modified proteins which are relatively long-lived, one oxidizing and one reducing. The two species are reactive with critical biological components, and so may be of physiological and pathological importance. The oxidizing species, which have been identified as protein hydroperoxides, can consume key cellular reductants, such as ascorbate and glutathione. The reducing species can act on both free and metalloprotein forms of copper and iron ions, which participate in radical generation. These findings suggest that proteins may act as traps for the chemical energy released by free radicals, with the capacity to pass it on to other molecules. The long-lived nature of both the reactive moieties indicates that they may be able to diffuse and transfer damaging reactions to distant sites.
Collapse
Affiliation(s)
- J A Simpson
- Heart Research Institute, Camperdown, Sydney, N.S.W., Australia
| | | | | | | | | | | |
Collapse
|
48
|
Tierney DJ, Haas AL, Koop DR. Degradation of cytochrome P450 2E1: selective loss after labilization of the enzyme. Arch Biochem Biophys 1992; 293:9-16. [PMID: 1309987 DOI: 10.1016/0003-9861(92)90358-4] [Citation(s) in RCA: 114] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Mechanism-based inactivation of cytochrome P450 can result in the chemical modification of the heme, the protein, or both as a result of covalent binding of modified heme to the protein. In the present study we took advantage of different modes of inactivation of P450 2E1 by CCl4, 1-aminobenzotriazole, or 3-amino-1,2,4-triazole to investigate parameters which target P450 2E1 for proteolysis from the microsomal membrane. Treatment of mice with CCl4 at the point of maximal induction of P450 2E1 after a single oral dose of acetone resulted in the complete loss of P450 2E1-dependent p-nitrophenol hydroxylation and a 75% loss of immunochemically detectable protein within 1 h of administration. Treatment with 1-aminobenzotriazole at the point of maximal induction caused a complete loss of P450 2E1-dependent p-nitrophenol hydroxylation but only a 12% loss of immunochemically detectable protein 1 h after administration. Treatment of mice with 3-amino-1,2,4-triazole caused a rapid loss of both catalytic activity and microsomal p-nitrophenol hydroxylase activity. However, unlike CCl4 treatment, the activity and enzyme level rebounded 5 and 9 h after treatment. The P450 2E1 ligand, 4-methylpyrazole, administered at the point of maximal induction maintained the acetone-induced catalytic and immunochemical level of P450 2E1. These results suggest that differentially modified forms of P450 2E1 show a characteristic susceptibility to degradation. While there are many potential pathways for protein degradation, the loss of P450 2E1 was associated with increased formation of high molecular weight microsomal ubiquitin conjugates. The formation of ubiquitin-conjugated microsomal protein which correlates with P450 2E1 loss suggests that ubiquitination may represent a proteolytic signal for the rapid and selective proteolysis of certain labilized conformations of P450 2E1 from the endoplasmic reticulum.
Collapse
Affiliation(s)
- D J Tierney
- Department of Environmental Health Sciences, Case Western Reserve University School of Medicine, Cleveland, Ohio 97201
| | | | | |
Collapse
|
49
|
Abstract
Cytochrome P450IIE1 is involved in the metabolic activation of many xenobiotics involved with human toxicity. In particular, cellular concentrations of P450IIE1 are significantly induced by the most widely abused drug in our society today, alcohol. As a result, the synthesis and degradation of this form of P450 has significant health consequences. The regulation of the steady-state concentration of P450IIE1 is an extremely complex process. The enzyme is regulated by transcriptional activation, mRNA stabilization, increased mRNA translatability and decreased protein degradation. The principal mechanism which controls the induction process depends on the chemical nature of the inducer, the age, and the nutritional and hormonal status of the animal. There also appear to be significant sex differences in the expression of P450IIE1. It is entirely possible that the regulation of the enzyme concentration under any given set of conditions will involve all of the mechanisms to different extents.
Collapse
Affiliation(s)
- D R Koop
- Department of Environmental Health Sciences, Case Western Reserve University, School of Medicine, Cleveland, OH 44106
| | | |
Collapse
|
50
|
O'Hara MB, Hageman JH. Energy and calcium ion dependence of proteolysis during sporulation of Bacillus subtilis cells. J Bacteriol 1990; 172:4161-70. [PMID: 2115863 PMCID: PMC213238 DOI: 10.1128/jb.172.8.4161-4170.1990] [Citation(s) in RCA: 31] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Bacterial cells degrade intracellular proteins at elevated rates during starvation and can selectively degrade proteins by energy-dependent processes. Sporulating bacteria can degrade protein with apparent first-order rate constants of over 0.20 h-1. We have shown, with an optimized [14C]leucine-labeling and chasing procedure, in a chemically defined sporulation medium, that intracellular protein degradation in sporulating cells of Bacillus subtilis 168 (trpC2) is apparently energy dependent. Sodium arsenate, sodium azide, carbonyl cyanide m-chlorophenylhydrozone, and N,N'-dicyclohexylcarbodiimide, at levels which did not induce appreciable lysis (less than or equal to 10%) over 10-h periods of sporulation, inhibited intracellular proteolysis by 13 to 93%. Exponentially growing cells acquired arsenate resistance. In contrast to earlier reports, we found that chloramphenicol (100 micrograms/ml) strongly inhibited proteolysis (68%) even when added 6 h into the sporulation process. Restricting the calcium ion concentration (less than 2 microM) in the medium had no effect on rates or extent of vegetative growth, strongly inhibited sporulation (98%), and inhibited rates of proteolysis by 60% or more. Inhibitors of energy metabolism, at the same levels which inhibited proteolysis, did not affect the rate or degree of uptake of Ca2+ by cells, which suggested that the Ca2+ and metabolic energy requirements of proteolysis were independent. Restricting the Ca2+ concentration in the medium reduced by threefold the specific activity in cells of the major intracellular serine proteinase after 12 h of sporulation. Finally, cells of a mutant of B. subtilis bearing an insertionally inactivated gene for the Ca2(+)-dependent intracellular proteinase-1 degraded protein in chemically defined sporulation medium at a rate indistinguishable from that of the wild-type cells for periods of 8 h.
Collapse
Affiliation(s)
- M B O'Hara
- Department of Chemistry, New Mexico State University, Las Cruces 88003
| | | |
Collapse
|