1
|
Cobley JN, Chatzinikolaou PN, Schmidt CA. The nonlinear cysteine redox dynamics in the i-space: A proteoform-centric theory of redox regulation. Redox Biol 2025; 81:103523. [PMID: 39929052 PMCID: PMC11849597 DOI: 10.1016/j.redox.2025.103523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 01/27/2025] [Accepted: 01/29/2025] [Indexed: 02/27/2025] Open
Abstract
The post-translational redox regulation of protein function by cysteine oxidation controls diverse biological processes, from cell division to death. However, most current site-centric paradigms fail to capture the nonlinear and emergent nature of redox regulation in proteins with multiple cysteines. Here, we present a proteoform-centric theory of redox regulation grounded in the i-space. The i-space encapsulates the theoretical landscape of all possible cysteine proteoforms. Using computational approaches, we quantify the vast size of the abstract i-space, revealing its scale-free architecture-elucidating the disproportionate influence of cysteine-rich proteins. We define mathematical rules governing cysteine proteoform dynamics. Their dynamics are inherently nonlinear, context-dependent, and fundamentally constrained by protein copy numbers. Monte Carlo simulations of the human protein PTP1B reveal extensive i-space sampling beyond site-centric models, supporting the "oxiform conjecture". This conjecture posits that highly oxidised proteoforms, molecules bearing multiple oxidised cysteines, are central to redox regulation. In support, even 90%-reduced proteomes can house vast numbers of unique, potentially functioanlly diverse, oxiforms. This framework offers a transformative lens for understanding the redox biology of proteoforms.
Collapse
|
2
|
Chartier CA, Woods VA, Xu Y, van Vlimmeren AE, Johns AC, Jovanovic M, McDermott AE, Keedy DA, Shah NH. Allosteric regulation of the tyrosine phosphatase PTP1B by a protein-protein interaction. Protein Sci 2025; 34:e70016. [PMID: 39723820 DOI: 10.1002/pro.70016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 11/11/2024] [Accepted: 12/13/2024] [Indexed: 12/28/2024]
Abstract
The rapid identification of protein-protein interactions has been significantly enabled by mass spectrometry (MS) proteomics-based methods, including affinity purification-MS, crosslinking-MS, and proximity-labeling proteomics. While these methods can reveal networks of interacting proteins, they cannot reveal how specific protein-protein interactions alter protein function or cell signaling. For instance, when two proteins interact, there can be emergent signaling processes driven purely by the individual activities of those proteins being co-localized. Alternatively, protein-protein interactions can allosterically regulate function, enhancing or suppressing activity in response to binding. In this work, we investigate the interaction between the tyrosine phosphatase PTP1B and the adaptor protein Grb2, which have been annotated as binding partners in a number of proteomics studies. This interaction has been postulated to co-localize PTP1B with its substrate IRS-1 by forming a ternary complex, thereby enhancing the dephosphorylation of IRS-1 to suppress insulin signaling. Here, we report that Grb2 binding to PTP1B also allosterically enhances PTP1B catalytic activity. We show that this interaction is dependent on the proline-rich region of PTP1B, which interacts with the C-terminal SH3 domain of Grb2. Using NMR spectroscopy and hydrogen-deuterium exchange mass spectrometry (HDX-MS) we show that Grb2 binding alters PTP1B structure and/or dynamics. Finally, we use MS proteomics to identify other interactors of the PTP1B proline-rich region that may also regulate PTP1B function similarly to Grb2. This work presents one of the first examples of a protein allosterically regulating the enzymatic activity of PTP1B and lays the foundation for discovering new mechanisms of PTP1B regulation in cell signaling.
Collapse
Affiliation(s)
| | - Virgil A Woods
- Structural Biology Initiative, CUNY Advanced Science Research Center, New York, New York, USA
- PhD Program in Biochemistry, CUNY Graduate Center, New York, New York, USA
| | - Yunyao Xu
- Department of Chemistry, Columbia University, New York, New York, USA
| | - Anne E van Vlimmeren
- Department of Chemistry, Columbia University, New York, New York, USA
- Department of Biological Sciences, Columbia University, New York, New York, USA
| | - Andrew C Johns
- Department of Chemistry, Columbia University, New York, New York, USA
| | - Marko Jovanovic
- Department of Biological Sciences, Columbia University, New York, New York, USA
| | - Ann E McDermott
- Department of Chemistry, Columbia University, New York, New York, USA
| | - Daniel A Keedy
- Structural Biology Initiative, CUNY Advanced Science Research Center, New York, New York, USA
- Department of Chemistry and Biochemistry, City College of New York, New York, New York, USA
- PhD Programs in Biochemistry, Biology, & Chemistry, CUNY Graduate Center, New York, New York, USA
| | - Neel H Shah
- Department of Chemistry, Columbia University, New York, New York, USA
| |
Collapse
|
3
|
Mackova V, Raudenska M, Polanska HH, Jakubek M, Masarik M. Navigating the redox landscape: reactive oxygen species in regulation of cell cycle. Redox Rep 2024; 29:2371173. [PMID: 38972297 PMCID: PMC11637001 DOI: 10.1080/13510002.2024.2371173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/09/2024] Open
Abstract
Objectives: To advance our knowledge of disease mechanisms and therapeutic options, understanding cell cycle regulation is critical. Recent research has highlighted the importance of reactive oxygen species (ROS) in cell cycle regulation. Although excessive ROS levels can lead to age-related pathologies, ROS also play an essential role in normal cellular functions. Many cell cycle regulatory proteins are affected by their redox status, but the precise mechanisms and conditions under which ROS promote or inhibit cell proliferation are not fully understood.Methods: This review presents data from the scientific literature and publicly available databases on changes in redox state during the cell cycle and their effects on key regulatory proteins.Results: We identified redox-sensitive targets within the cell cycle machinery and analysed different effects of ROS (type, concentration, duration of exposure) on cell cycle phases. For example, moderate levels of ROS can promote cell proliferation by activating signalling pathways involved in cell cycle progression, whereas excessive ROS levels can induce DNA damage and trigger cell cycle arrest or cell death.Discussion: Our findings encourage future research focused on identifying redox-sensitive targets in the cell cycle machinery, potentially leading to new treatments for diseases with dysregulated cell proliferation.
Collapse
Affiliation(s)
- Viktoria Mackova
- Department of Pathological Physiology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Martina Raudenska
- Department of Pathological Physiology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
- Department of Physiology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Hana Holcova Polanska
- Department of Pathological Physiology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
- Department of Physiology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Milan Jakubek
- BIOCEV, First Faculty of Medicine, Charles University, Vestec, Czech Republic
| | - Michal Masarik
- Department of Pathological Physiology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
- Department of Physiology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
- BIOCEV, First Faculty of Medicine, Charles University, Vestec, Czech Republic
- Institute of Pathophysiology, First Faculty of Medicine, Charles University, Prague, Czech Republic
| |
Collapse
|
4
|
Gallero S, Persson KW, Henríquez-Olguín C. Unresolved questions in the regulation of skeletal muscle insulin action by reactive oxygen species. FEBS Lett 2024; 598:2145-2159. [PMID: 38803005 DOI: 10.1002/1873-3468.14937] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 04/10/2024] [Accepted: 04/22/2024] [Indexed: 05/29/2024]
Abstract
Reactive oxygen species (ROS) are well-established signaling molecules implicated in a wide range of cellular processes, including both oxidative stress and intracellular redox signaling. In the context of insulin action within its target tissues, ROS have been reported to exert both positive and negative regulatory effects. However, the precise molecular mechanisms underlying this duality remain unclear. This Review examines the complex role of ROS in insulin action, with a particular focus on skeletal muscle. We aim to address three critical aspects: (a) the proposed intracellular pro-oxidative redox shift elicited by insulin, (b) the evidence supporting that redox-sensitive cysteine modifications impact insulin signaling and action, and (c) cellular mechanisms underlying how ROS can paradoxically act as both enhancers and inhibitors of insulin action. This Review underscores the urgent need for more systematic research to identify specific reactive species, redox targets, and the physiological significance of redox signaling in maintaining insulin action and metabolic health, with a particular emphasis on human skeletal muscle.
Collapse
Affiliation(s)
- Samantha Gallero
- The August Krogh Section for Molecular Physiology, Department of Nutrition, Exercise and Sports, University of Copenhagen, Denmark
| | - Kaspar W Persson
- The August Krogh Section for Molecular Physiology, Department of Nutrition, Exercise and Sports, University of Copenhagen, Denmark
| | - Carlos Henríquez-Olguín
- The August Krogh Section for Molecular Physiology, Department of Nutrition, Exercise and Sports, University of Copenhagen, Denmark
- Exercise Science Laboratory, Faculty of Medicine, Universidad Finis Terrae, Santiago, Chile
| |
Collapse
|
5
|
Coronell-Tovar A, Pardo JP, Rodríguez-Romero A, Sosa-Peinado A, Vásquez-Bochm L, Cano-Sánchez P, Álvarez-Añorve LI, González-Andrade M. Protein tyrosine phosphatase 1B (PTP1B) function, structure, and inhibition strategies to develop antidiabetic drugs. FEBS Lett 2024; 598:1811-1838. [PMID: 38724486 DOI: 10.1002/1873-3468.14901] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 04/08/2024] [Accepted: 04/09/2024] [Indexed: 08/13/2024]
Abstract
Tyrosine protein phosphatase non-receptor type 1 (PTP1B; also known as protein tyrosine phosphatase 1B) is a member of the protein tyrosine phosphatase (PTP) family and is a soluble enzyme that plays an essential role in different physiological processes, including the regulation of metabolism, specifically in insulin and leptin sensitivity. PTP1B is crucial in the pathogenesis of type 2 diabetes mellitus and obesity. These biological functions have made PTP1B validated as an antidiabetic and anti-obesity, and potentially anticancer, molecular target. Four main approaches aim to inhibit PTP1B: orthosteric, allosteric, bidentate inhibition, and PTPN1 gene silencing. Developing a potent and selective PTP1B inhibitor is still challenging due to the enzyme's ubiquitous expression, subcellular location, and structural properties. This article reviews the main advances in the study of PTP1B since it was first isolated in 1988, as well as recent contextual information related to the PTP family to which this protein belongs. Furthermore, we offer an overview of the role of PTP1B in diabetes and obesity, and the challenges to developing selective, effective, potent, bioavailable, and cell-permeable compounds that can inhibit the enzyme.
Collapse
Affiliation(s)
- Andrea Coronell-Tovar
- Laboratorio de Biosensores y Modelaje molecular, Departamento de Bioquímica, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Juan P Pardo
- Laboratorio de Biosensores y Modelaje molecular, Departamento de Bioquímica, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | | | - Alejandro Sosa-Peinado
- Laboratorio de Biosensores y Modelaje molecular, Departamento de Bioquímica, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Luz Vásquez-Bochm
- Laboratorio de Biosensores y Modelaje molecular, Departamento de Bioquímica, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Patricia Cano-Sánchez
- Instituto de Química, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Laura Iliana Álvarez-Añorve
- Laboratorio de Biosensores y Modelaje molecular, Departamento de Bioquímica, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Martin González-Andrade
- Laboratorio de Biosensores y Modelaje molecular, Departamento de Bioquímica, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| |
Collapse
|
6
|
Delibegović M, Dall'Angelo S, Dekeryte R. Protein tyrosine phosphatase 1B in metabolic diseases and drug development. Nat Rev Endocrinol 2024; 20:366-378. [PMID: 38519567 DOI: 10.1038/s41574-024-00965-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/16/2024] [Indexed: 03/25/2024]
Abstract
Protein tyrosine phosphatase 1B (PTP1B), a non-transmembrane phosphatase, has a major role in a variety of signalling pathways, including direct negative regulation of classic insulin and leptin signalling pathways, and is implicated in the pathogenesis of several cardiometabolic diseases and cancers. As such, PTP1B has been a therapeutic target for over two decades, with PTP1B inhibitors identified either from natural sources or developed throughout the years. Some of these inhibitors have reached phase I and/or II clinical trials in humans for the treatment of type 2 diabetes mellitus, obesity and/or metastatic breast cancer. In this Review, we summarize the cellular processes and regulation of PTP1B, discuss evidence from in vivo preclinical and human studies of the association between PTP1B and different disorders, and discuss outcomes of clinical trials. We outline challenges associated with the targeting of this phosphatase (which was, until the past few years, viewed as difficult to target), the current state of the field of PTP1B inhibitors (and dual phosphatase inhibitors) and future directions for manipulating the activity of this key metabolic enzyme.
Collapse
Affiliation(s)
- Mirela Delibegović
- Aberdeen Cardiovascular and Diabetes Centre, University of Aberdeen, Institute of Medical Sciences, Aberdeen, UK.
| | - Sergio Dall'Angelo
- Aberdeen Cardiovascular and Diabetes Centre, University of Aberdeen, Institute of Medical Sciences, Aberdeen, UK
| | - Ruta Dekeryte
- Aberdeen Cardiovascular and Diabetes Centre, University of Aberdeen, Institute of Medical Sciences, Aberdeen, UK
| |
Collapse
|
7
|
Welsh CL, Madan LK. Protein Tyrosine Phosphatase regulation by Reactive Oxygen Species. Adv Cancer Res 2024; 162:45-74. [PMID: 39069369 DOI: 10.1016/bs.acr.2024.05.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/30/2024]
Abstract
Protein Tyrosine Phosphatases (PTPs) help to maintain the balance of protein phosphorylation signals that drive cell division, proliferation, and differentiation. These enzymes are also well-suited to redox-dependent signaling and oxidative stress response due to their cysteine-based catalytic mechanism, which requires a deprotonated thiol group at the active site. This review focuses on PTP structural characteristics, active site chemical properties, and vulnerability to change by reactive oxygen species (ROS). PTPs can be oxidized and inactivated by H2O2 through three non-exclusive mechanisms. These pathways are dependent on the coordinated actions of other H2O2-sensitive proteins, such as peroxidases like Peroxiredoxins (Prx) and Thioredoxins (Trx). PTPs undergo reversible oxidation by converting their active site cysteine from thiol to sulfenic acid. This sulfenic acid can then react with adjacent cysteines to form disulfide bonds or with nearby amides to form sulfenyl-amide linkages. Further oxidation of the sulfenic acid form to the sulfonic or sulfinic acid forms causes irreversible deactivation. Understanding the structural changes involved in both reversible and irreversible PTP oxidation can help with their chemical manipulation for therapeutic intervention. Nonetheless, more information remains unidentified than is presently known about the precise dynamics of proteins participating in oxidation events, as well as the specific oxidation states that can be targeted for PTPs. This review summarizes current information on PTP-specific oxidation patterns and explains how ROS-mediated signal transmission interacts with phosphorylation-based signaling machinery controlled by growth factor receptors and PTPs.
Collapse
Affiliation(s)
- Colin L Welsh
- Department of Cell and Molecular Pharmacology & Experimental Therapeutics, College of Medicine, Medical University of South Carolina, Charleston, SC, United States
| | - Lalima K Madan
- Department of Cell and Molecular Pharmacology & Experimental Therapeutics, College of Medicine, Medical University of South Carolina, Charleston, SC, United States; Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, United States.
| |
Collapse
|
8
|
Trinh VH, Choi JM, Nguyen Huu T, Sah DK, Yoon HJ, Park SC, Jung YS, Ahn YK, Lee KH, Lee SR. Redox Regulation of Phosphatase and Tensin Homolog by Bicarbonate and Hydrogen Peroxide: Implication of Peroxymonocarbonate in Cell Signaling. Antioxidants (Basel) 2024; 13:473. [PMID: 38671920 PMCID: PMC11047460 DOI: 10.3390/antiox13040473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Revised: 04/05/2024] [Accepted: 04/15/2024] [Indexed: 04/28/2024] Open
Abstract
Phosphatase and tensin homolog (PTEN) is a negative regulator of the phosphoinositide 3-kinases/protein kinase B (PI3K/AKT) signaling pathway. Notably, its active site contains a cysteine residue that is susceptible to oxidation by hydrogen peroxide (H2O2). This oxidation inhibits the phosphatase function of PTEN, critically contributing to the activation of the PI3K/AKT pathway. Upon the stimulation of cell surface receptors, the activity of NADPH oxidase (NOX) generates a transient amount of H2O2, serving as a mediator in this pathway by oxidizing PTEN. The mechanism underlying this oxidation, occurring despite the presence of highly efficient and abundant cellular oxidant-protecting and reducing systems, continues to pose a perplexing conundrum. Here, we demonstrate that the presence of bicarbonate (HCO3-) promoted the rate of H2O2-mediated PTEN oxidation, probably through the formation of peroxymonocarbonate (HCO4-), and consequently potentiated the phosphorylation of AKT. Acetazolamide (ATZ), a carbonic anhydrase (CA) inhibitor, was shown to diminish the oxidation of PTEN. Thus, CA can also be considered as a modulator in this context. In essence, our findings consolidate the crucial role of HCO3- in the redox regulation of PTEN by H2O2, leading to the presumption that HCO4- is a signaling molecule during cellular physiological processes.
Collapse
Affiliation(s)
- Vu Hoang Trinh
- Department of Biochemistry, Department of Biomedical Sciences, Chonnam National University Medical School, Gwangju 501190, Republic of Korea; (V.H.T.); (T.N.H.); (D.K.S.); (H.-J.Y.)
- Department of Oncology, Department of Medical Sciences, Pham Ngoc Thach University of Medicine, Ho Chi Minh City 700000, Vietnam
| | - Jin-Myung Choi
- Luxanima Inc., Room 102, 12-55, Sandan-gil, Hwasun-eup, Hwasun-gun 58128, Republic of Korea;
| | - Thang Nguyen Huu
- Department of Biochemistry, Department of Biomedical Sciences, Chonnam National University Medical School, Gwangju 501190, Republic of Korea; (V.H.T.); (T.N.H.); (D.K.S.); (H.-J.Y.)
| | - Dhiraj Kumar Sah
- Department of Biochemistry, Department of Biomedical Sciences, Chonnam National University Medical School, Gwangju 501190, Republic of Korea; (V.H.T.); (T.N.H.); (D.K.S.); (H.-J.Y.)
| | - Hyun-Joong Yoon
- Department of Biochemistry, Department of Biomedical Sciences, Chonnam National University Medical School, Gwangju 501190, Republic of Korea; (V.H.T.); (T.N.H.); (D.K.S.); (H.-J.Y.)
| | - Sang-Chul Park
- The Future Life & Society Research Center, Advanced Institute of Aging Science, Chonnam National University, Gwangju 61469, Republic of Korea;
| | - Yu-Seok Jung
- Chonnam National University Medical School, Gwangju 501190, Republic of Korea;
| | - Young-Keun Ahn
- Department of Cardiology, Chonnam National University Hospital, Gwangju 61469, Republic of Korea;
| | - Kun-Ho Lee
- Department of Biomedical Science, Chosun University, Gwangju 61452, Republic of Korea;
- Department of Neural Development and Disease, Korea Brain Research Institute, Daegu 41062, Republic of Korea
| | - Seung-Rock Lee
- Department of Biochemistry, Department of Biomedical Sciences, Chonnam National University Medical School, Gwangju 501190, Republic of Korea; (V.H.T.); (T.N.H.); (D.K.S.); (H.-J.Y.)
| |
Collapse
|
9
|
Szablewski L. Changes in Cells Associated with Insulin Resistance. Int J Mol Sci 2024; 25:2397. [PMID: 38397072 PMCID: PMC10889819 DOI: 10.3390/ijms25042397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2024] [Revised: 02/10/2024] [Accepted: 02/14/2024] [Indexed: 02/25/2024] Open
Abstract
Insulin is a polypeptide hormone synthesized and secreted by pancreatic β-cells. It plays an important role as a metabolic hormone. Insulin influences the metabolism of glucose, regulating plasma glucose levels and stimulating glucose storage in organs such as the liver, muscles and adipose tissue. It is involved in fat metabolism, increasing the storage of triglycerides and decreasing lipolysis. Ketone body metabolism also depends on insulin action, as insulin reduces ketone body concentrations and influences protein metabolism. It increases nitrogen retention, facilitates the transport of amino acids into cells and increases the synthesis of proteins. Insulin also inhibits protein breakdown and is involved in cellular growth and proliferation. On the other hand, defects in the intracellular signaling pathways of insulin may cause several disturbances in human metabolism, resulting in several chronic diseases. Insulin resistance, also known as impaired insulin sensitivity, is due to the decreased reaction of insulin signaling for glucose levels, seen when glucose use in response to an adequate concentration of insulin is impaired. Insulin resistance may cause, for example, increased plasma insulin levels. That state, called hyperinsulinemia, impairs metabolic processes and is observed in patients with type 2 diabetes mellitus and obesity. Hyperinsulinemia may increase the risk of initiation, progression and metastasis of several cancers and may cause poor cancer outcomes. Insulin resistance is a health problem worldwide; therefore, mechanisms of insulin resistance, causes and types of insulin resistance and strategies against insulin resistance are described in this review. Attention is also paid to factors that are associated with the development of insulin resistance, the main and characteristic symptoms of particular syndromes, plus other aspects of severe insulin resistance. This review mainly focuses on the description and analysis of changes in cells due to insulin resistance.
Collapse
Affiliation(s)
- Leszek Szablewski
- Chair and Department of General Biology and Parasitology, Medical University of Warsaw, Chałubińskiego Str. 5, 02-004 Warsaw, Poland
| |
Collapse
|
10
|
Franco C, Canzoniero LMT. Zinc homeostasis and redox alterations in obesity. Front Endocrinol (Lausanne) 2024; 14:1273177. [PMID: 38260166 PMCID: PMC10800374 DOI: 10.3389/fendo.2023.1273177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2023] [Accepted: 12/18/2023] [Indexed: 01/24/2024] Open
Abstract
Impairment of both cellular zinc and redox homeostasis is a feature of several chronic diseases, including obesity. A significant two-way interaction exists between redox metabolism and the relatively redox-inert zinc ion. Redox metabolism critically influences zinc homeostasis and controls its cellular availability for various cellular functions by regulating zinc exchange from/to zinc-binding proteins. Zinc can regulate redox metabolism and exhibits multiple pro-antioxidant properties. On the other hand, even minor disturbances in zinc status and zinc homeostasis affect systemic and cellular redox homeostasis. At the cellular level, zinc homeostasis is regulated by a multi-layered machinery consisting of zinc-binding molecules, zinc sensors, and two selective families of zinc transporters, the Zinc Transporter (ZnT) and Zrt, Irt-like protein (ZIP). In the present review, we summarize the current state of knowledge on the role of the mutual interaction between zinc and redox homeostasis in physiology and pathophysiology, pointing to the role of zinc in the alterations responsible for redox stress in obesity. Since zinc transporters primarily control zinc homeostasis, we describe how changes in the expression and activity of these zinc-regulating proteins are associated with obesity.
Collapse
|
11
|
Ando M, Suzuki K, Kitamoto R, Nakayama A, Watanabe N, Kawahara M. Differences in serum selenoprotein P profile between C57BL/6 and BALB/c mice fed high-fat diet. J Trace Elem Med Biol 2024; 81:127340. [PMID: 37984217 DOI: 10.1016/j.jtemb.2023.127340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 11/01/2023] [Accepted: 11/07/2023] [Indexed: 11/22/2023]
Abstract
BACKGROUND C57BL/6 mice generally show hyperglycaemia and insulin resistance when fed a high-fat diet (HFD) compared to those of BALB/c mice. However, whether these strains also show different expression profiles of selenoprotein P, a diabetes-related hepatokine, after HFD feeding is unclear. We investigated the effects of HFD on body weight, glucose metabolism, and plasma selenoprotein P levels in C57BL/6 and BALB/c mice. METHODS Male C57BL/6 and BALB/c mice aged seven weeks were divided into normal diet (ND) and HFD groups. Fasting body weights and blood sugar levels were measured weekly. Blood specimens were collected after 16 h of fasting (in weeks 7, 9, and 11) and after 24 h of subsequent refeeding (in weeks 9 and 11) to analyse plasma selenoprotein P and insulin levels. RESULTS The mean body weight of the HFD group was consistently higher than that of the ND group for both strains. However, a significant elevation in fasting plasma glucose levels from the early stage was observed only in the HFD group of C57BL/6 mice. In BALB/c mice, a difference in fasting glucose levels between the HFD and ND groups was observed after nine weeks. After seven, nine, and eleven weeks, the fasting plasma insulin levels were higher in the HFD group than in the ND group for both strains. During this period, plasma selenoprotein P levels in the HFD group were significantly higher than those in the ND group of C57BL/6 mice. However, BALB/c mice did not show a significant difference in plasma levels of selenoprotein P between the ND and HFD groups. After refeeding, the plasma insulin and selenoprotein P levels increased compared to those observed during fasting in the ND group for both strains. Elevation of insulin levels, but not of selenoprotein P levels, after refeeding was noticed in the HFD group for both strains. Plasma selenoprotein P level after refeeding was significantly lower than that during fasting in the HFD group of C57BL/6 mice. CONCLUSION Unlike C57BL/6 mice, BALB/c mice did not show elevated fasting plasma selenoprotein P levels despite HFD feeding. Additionally, the pattern of selenoprotein P levels in the plasma after refeeding differed between C57BL/6 and BALB/c mice. These differences in selenoprotein P expression among strains may be related to different susceptibilities of individuals to diabetes.
Collapse
Affiliation(s)
- Motozumi Ando
- Laboratory of Clinical Pharmacy, School of Pharmacy, Aichi Gakuin University, 1-100 Kusumoto-cho Chikusa-ku, Nagoya, Aichi 464-8650, Japan.
| | - Keiko Suzuki
- Laboratory of Clinical Pharmacy, School of Pharmacy, Aichi Gakuin University, 1-100 Kusumoto-cho Chikusa-ku, Nagoya, Aichi 464-8650, Japan.
| | - Riko Kitamoto
- Laboratory of Clinical Pharmacy, School of Pharmacy, Aichi Gakuin University, 1-100 Kusumoto-cho Chikusa-ku, Nagoya, Aichi 464-8650, Japan.
| | - Ayako Nakayama
- Laboratory of Clinical Pharmacy, School of Pharmacy, Aichi Gakuin University, 1-100 Kusumoto-cho Chikusa-ku, Nagoya, Aichi 464-8650, Japan.
| | - Norio Watanabe
- Laboratory of Clinical Pharmacy, School of Pharmacy, Aichi Gakuin University, 1-100 Kusumoto-cho Chikusa-ku, Nagoya, Aichi 464-8650, Japan.
| | - Masami Kawahara
- Laboratory of Clinical Pharmacy, School of Pharmacy, Aichi Gakuin University, 1-100 Kusumoto-cho Chikusa-ku, Nagoya, Aichi 464-8650, Japan.
| |
Collapse
|
12
|
Mazumdar D, Singh S. Diabetic Encephalopathy: Role of Oxidative and Nitrosative Factors in Type 2 Diabetes. Indian J Clin Biochem 2024; 39:3-17. [PMID: 38223005 PMCID: PMC10784252 DOI: 10.1007/s12291-022-01107-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Accepted: 12/07/2022] [Indexed: 12/23/2022]
Abstract
Diabetes mellitus is a set of complex metabolic disorders characterized by chronic hyperglycaemic condition due to defective insulin secretion (Type 1) and action (Type 2), which leads to serious micro and macro-vascular damage, inflammation, oxidative and nitrosative stress and a deranged energy homeostasis due to imbalance in the glucose and lipid metabolism. Moreover, patient with diabetes mellitus often showed the nervous system disorders known as diabetic encephalopathy. The precise pathological mechanism of diabetic encephalopathy by which it effects the central nervous system directly or indirectly causing the cognitive and motor impairment, is not completely understood. However, it has been speculated that like other extracerebellar tissues, oxidative and nitrosative stress may play significant role in the pathogenesis of diabetic encephalopathy. Therefore, the present review aimed to explain the possible association of the oxidative and nitrosative stress caused by the chronic hyperglycaemic condition with the central nervous system complications of the type 2 diabetes mellitus induced diabetic encephalopathy.
Collapse
Affiliation(s)
- Debashree Mazumdar
- Biochemistry and Molecular Biology Laboratory, Department of Zoology, Guru Ghasidas Vishwavidyalaya (A Central University), Bilaspur, Chhattisgarh 495009 India
| | - Santosh Singh
- Biochemistry and Molecular Biology Laboratory, Department of Zoology, Guru Ghasidas Vishwavidyalaya (A Central University), Bilaspur, Chhattisgarh 495009 India
| |
Collapse
|
13
|
Machado IF, Miranda RG, Dorta DJ, Rolo AP, Palmeira CM. Targeting Oxidative Stress with Polyphenols to Fight Liver Diseases. Antioxidants (Basel) 2023; 12:1212. [PMID: 37371941 DOI: 10.3390/antiox12061212] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 05/26/2023] [Accepted: 05/31/2023] [Indexed: 06/29/2023] Open
Abstract
Reactive oxygen species (ROS) are important second messengers in many metabolic processes and signaling pathways. Disruption of the balance between ROS generation and antioxidant defenses results in the overproduction of ROS and subsequent oxidative damage to biomolecules and cellular components that disturb cellular function. Oxidative stress contributes to the initiation and progression of many liver pathologies such as ischemia-reperfusion injury (LIRI), non-alcoholic fatty liver disease (NAFLD), and hepatocellular carcinoma (HCC). Therefore, controlling ROS production is an attractive therapeutic strategy in relation to their treatment. In recent years, increasing evidence has supported the therapeutic effects of polyphenols on liver injury via the regulation of ROS levels. In the current review, we summarize the effects of polyphenols, such as quercetin, resveratrol, and curcumin, on oxidative damage during conditions that induce liver injury, such as LIRI, NAFLD, and HCC.
Collapse
Affiliation(s)
- Ivo F Machado
- CNC-Center for Neuroscience and Cell Biology, CIBB-Center for Innovative Biomedicine and Biotechnology, University of Coimbra, 3000 Coimbra, Portugal
- IIIUC-Institute of Interdisciplinary Research, University of Coimbra, 3000 Coimbra, Portugal
| | - Raul G Miranda
- School of Pharmaceutical Science of Ribeirão Preto, University of São Paulo, São Paulo 14040, Brazil
| | - Daniel J Dorta
- Department of Chemistry, Faculty of Philosophy, Sciences and Letters of Ribeirão Preto, University of São Paulo, Ribeirão Preto 14040, Brazil
| | - Anabela P Rolo
- CNC-Center for Neuroscience and Cell Biology, CIBB-Center for Innovative Biomedicine and Biotechnology, University of Coimbra, 3000 Coimbra, Portugal
- Department of Life Sciences, University of Coimbra, 3000 Coimbra, Portugal
| | - Carlos M Palmeira
- CNC-Center for Neuroscience and Cell Biology, CIBB-Center for Innovative Biomedicine and Biotechnology, University of Coimbra, 3000 Coimbra, Portugal
- Department of Life Sciences, University of Coimbra, 3000 Coimbra, Portugal
| |
Collapse
|
14
|
Argaev-Frenkel L, Rosenzweig T. Redox Balance in Type 2 Diabetes: Therapeutic Potential and the Challenge of Antioxidant-Based Therapy. Antioxidants (Basel) 2023; 12:antiox12050994. [PMID: 37237860 DOI: 10.3390/antiox12050994] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 04/19/2023] [Accepted: 04/24/2023] [Indexed: 05/28/2023] Open
Abstract
Oxidative stress is an important factor in the development of type 2 diabetes (T2D) and associated complications. Unfortunately, most clinical studies have failed to provide sufficient evidence regarding the benefits of antioxidants (AOXs) in treating this disease. Based on the known complexity of reactive oxygen species (ROS) functions in both the physiology and pathophysiology of glucose homeostasis, it is suggested that inappropriate dosing leads to the failure of AOXs in T2D treatment. To support this hypothesis, the role of oxidative stress in the pathophysiology of T2D is described, together with a summary of the evidence for the failure of AOXs in the management of diabetes. A comparison of preclinical and clinical studies indicates that suboptimal dosing of AOXs might explain the lack of benefits of AOXs. Conversely, the possibility that glycemic control might be adversely affected by excess AOXs is also considered, based on the role of ROS in insulin signaling. We suggest that AOX therapy should be given in a personalized manner according to the need, which is the presence and severity of oxidative stress. With the development of gold-standard biomarkers for oxidative stress, optimization of AOX therapy may be achieved to maximize the therapeutic potential of these agents.
Collapse
Affiliation(s)
| | - Tovit Rosenzweig
- Department of Molecular Biology, Ariel University, Ariel 4070000, Israel
- Adison School of Medicine, Ariel University, Ariel 4070000, Israel
| |
Collapse
|
15
|
Thomas HJ, Ang T, Morrison DJ, Keske MA, Parker L. Acute exercise and high-glucose ingestion elicit dynamic and individualized responses in systemic markers of redox homeostasis. Front Immunol 2023; 14:1127088. [PMID: 37063903 PMCID: PMC10102861 DOI: 10.3389/fimmu.2023.1127088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Accepted: 03/14/2023] [Indexed: 03/31/2023] Open
Abstract
BackgroundBiomarkers of oxidation-reduction (redox) homeostasis are commonly measured in human blood to assess whether certain stimuli (e.g., high-glucose ingestion or acute exercise) lead to a state of oxidative distress (detrimental to health) or oxidative eustress (beneficial to health). Emerging research indicates that redox responses are likely to be highly individualized, yet few studies report individual responses. Furthermore, the effects of complex redox stimuli (e.g., high-glucose-ingestion after exercise) on redox homeostasis remains unclear. We investigated the effect of acute exercise (oxidative eustress), high-glucose ingestion (oxidative distress), and high-glucose ingestion after exercise (both oxidative eu/distress), on commonly measured redox biomarkers in serum/plasma.MethodsIn a randomized crossover fashion, eight healthy men (age: 28 ± 4 years; BMI: 24.5 ± 1.5 kg/m2 [mean ± SD]) completed two separate testing conditions; 1) consumption of a high-glucose mixed-nutrient meal (45% carbohydrate [1.1 g glucose.kg-1], 20% protein, and 35% fat) at rest (control trial), and 2) consumption of the same meal 3 h and 24 h after 1 h of moderate-intensity cycling exercise (exercise trial). Plasma and serum were analyzed for an array of commonly studied redox biomarkers.ResultsOxidative stress and antioxidant defense markers (hydrogen peroxide, 8-isoprostanes, catalase, superoxide dismutase, and nitrate levels) increased immediately after exercise (p < 0.05), whereas nitric oxide activity and thiobarbituric acid reactive substances (TBARS) remained similar to baseline (p > 0.118). Nitric oxide activity and nitrate levels decreased at 3 h post-exercise compared to pre-exercise baseline levels. Depending on when the high-glucose mixed nutrient meal was ingested and the postprandial timepoint investigated, oxidative stress and antioxidant defense biomarkers either increased (hydrogen peroxide, TBARS, and superoxide dismutase), decreased (hydrogen peroxide, 8-isoprostanes, superoxide dismutase, nitric oxide activity, nitrate, and nitrite), or remained similar to pre-meal baseline levels (hydrogen peroxide, 8-isoprostanes, TBARS, catalase, superoxide dismutase and nitrite). Redox responses exhibited large inter-individual variability in the magnitude and/or direction of responses.ConclusionFindings highlight the necessity to interpret redox biomarkers in the context of the individual, biomarker measured, and stimuli observed. Individual redox responsiveness may be of physiological relevance and should be explored as a potential means to inform personalized redox intervention.
Collapse
Affiliation(s)
- Hannah J. Thomas
- Institute for Physical Activity and Nutrition (IPAN), School of Exercise and Nutrition Sciences, Deakin University, Geelong, VIC, Australia
| | - Teddy Ang
- Institute for Physical Activity and Nutrition (IPAN), School of Exercise and Nutrition Sciences, Deakin University, Geelong, VIC, Australia
| | - Dale J. Morrison
- Institute for Physical Activity and Nutrition (IPAN), School of Exercise and Nutrition Sciences, Deakin University, Geelong, VIC, Australia
- Department of Medicine, University of Melbourne, Melbourne, VIC, Australia
| | - Michelle A. Keske
- Institute for Physical Activity and Nutrition (IPAN), School of Exercise and Nutrition Sciences, Deakin University, Geelong, VIC, Australia
| | - Lewan Parker
- Institute for Physical Activity and Nutrition (IPAN), School of Exercise and Nutrition Sciences, Deakin University, Geelong, VIC, Australia
- *Correspondence: Lewan Parker,
| |
Collapse
|
16
|
The PTP1B selective inhibitor MSI-1436 mitigates Tunicamycin-induced ER stress in human hepatocarcinoma cell line through XBP1 splicing modulation. PLoS One 2023; 18:e0278566. [PMID: 36649358 PMCID: PMC9844924 DOI: 10.1371/journal.pone.0278566] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Accepted: 11/18/2022] [Indexed: 01/18/2023] Open
Abstract
Protein tyrosine phosphatase PTP1B is considered as a key metabolic enzyme that has been reported to be associated with insulin resistance onset, and underlying cellular metabolic malfunctions, including ER stress and mitochondrial failure. In this study, effects of selective PTP1B inhibition using MSI-1436 on cellular apoptosis, oxidative stress, mitochondrial dysfunction and ER stress have been assessed using an in vitro model of Tunicamycin induced ER stress in HepG2 cell line. Inhibition of PTP1B using MSI-1436 significantly increased cell viability and reduced the number of apoptotic cells as well as the expression of key apoptosis initiators and effectors. MSI-1436 further mitigated ER stress, by downregulating the expression of IRE1, ATF6 and PERK transcripts, all being key ER stress sensors. Interestingly, MSI-1436 inhibited the XBP1 splicing, and thus its UPR-associated transcriptional activity. PTP1B inhibition further enabled to restore proper mitochondrial biogenesis, by improving transmembrane potential, and diminishing intracellular ROS while restoring of endogenous antioxidant enzymes genes expression. PTP1B inhibition using MSI-1436 could improve cellular apoptosis and metabolic integrity through the mitigation of ER and mitochondrial stress signalling pathways, and excessive ROS accumulation. This strategy may be useful for the treatment of metabolic disorders including IR, NAFLD and diabetes.
Collapse
|
17
|
Mason SA, Parker L, van der Pligt P, Wadley GD. Vitamin C supplementation for diabetes management: A comprehensive narrative review. Free Radic Biol Med 2023; 194:255-283. [PMID: 36526243 DOI: 10.1016/j.freeradbiomed.2022.12.003] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 12/01/2022] [Accepted: 12/05/2022] [Indexed: 12/15/2022]
Abstract
Growing evidence suggests that vitamin C supplementation may be an effective adjunct therapy in the management of people with diabetes. This paper critically reviews the current evidence on effects of vitamin C supplementation and its potential mechanisms in diabetes management. Evidence from meta-analyses of randomized controlled trials (RCTs) show favourable effects of vitamin C on glycaemic control and blood pressure that may be clinically meaningful, and mixed effects on blood lipids and endothelial function. However, evidence is mostly of low evidence certainty. Emerging evidence is promising for effects of vitamin C supplementation on some diabetes complications, particularly diabetic foot ulcers. However, there is a notable lack of robust and well-designed studies exploring effects of vitamin C as a single compound supplement on diabetes prevention and patient-important outcomes (i.e. prevention and amelioration of diabetes complications). RCTs are also required to investigate potential preventative or ameliorative effects of vitamin C on gestational diabetes outcomes. Oral vitamin C doses of 500-1000 mg per day are potentially effective, safe, and affordable for many individuals with diabetes. However, personalisation of supplementation regimens that consider factors such as vitamin C status, disease status, current glycaemic control, vitamin C intake, redox status, and genotype is important to optimize vitamin C's therapeutic effects safely. Finally, given a high prevalence of vitamin C deficiency in patients with complications, it is recommended that plasma vitamin C concentration be measured and monitored in the clinic setting.
Collapse
Affiliation(s)
- Shaun A Mason
- Institute for Physical Activity and Nutrition, School of Exercise and Nutrition Sciences, Deakin University, Geelong, Australia.
| | - Lewan Parker
- Institute for Physical Activity and Nutrition, School of Exercise and Nutrition Sciences, Deakin University, Geelong, Australia
| | - Paige van der Pligt
- Institute for Physical Activity and Nutrition, School of Exercise and Nutrition Sciences, Deakin University, Geelong, Australia; Department of Nutrition and Dietetics, Western Health, Footscray, Australia
| | - Glenn D Wadley
- Institute for Physical Activity and Nutrition, School of Exercise and Nutrition Sciences, Deakin University, Geelong, Australia
| |
Collapse
|
18
|
Netto LES, Machado LESF. Preferential redox regulation of cysteine‐based protein tyrosine phosphatases: structural and biochemical diversity. FEBS J 2022; 289:5480-5504. [DOI: 10.1111/febs.16466] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 03/20/2022] [Accepted: 04/28/2022] [Indexed: 12/30/2022]
Affiliation(s)
- Luís Eduardo S. Netto
- Departamento de Genética e Biologia Evolutiva Instituto de Biociências Universidade de São Paulo Brazil
| | | |
Collapse
|
19
|
Rios-Lugo MJ, Palos-Lucio AG, Victoria-Campos CI, Lugo-Trampe A, Trujillo-Murillo KDC, López-García MA, Espinoza-Ruiz M, Romero-Guzmán ET, Hernández-Mendoza H, Chang-Rueda C. Sex-Specific Association between Fasting Plasma Glucose and Serum Selenium Levels in Adults from Southern Mexico. Healthcare (Basel) 2022; 10:1665. [PMID: 36141277 PMCID: PMC9498661 DOI: 10.3390/healthcare10091665] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Revised: 08/25/2022] [Accepted: 08/27/2022] [Indexed: 11/16/2022] Open
Abstract
Selenium (Se) is an essential trace element that by its antioxidant properties has been studied to elucidate its participation in the development of obesity and type 2 diabetes. We evaluated the association between cardiometabolic traits and serum Se levels in a sample of adults from southern Mexico. In 96 nondiabetic individuals, anthropometric data and clinical biochemistry measurements were analyzed. Serum total Se levels were measured with inductively coupled plasma mass spectrometry (ICP-MS). Serum Se level in the whole sample was 10.309 ± 3.031 μg mL-1 and no difference between the women and men was observed (p = 0.09). Additionally, fasting plasma glucose (FPG) was significantly associated with serum Se level (β = -0.07 ± 0.03, p = 0.02, analysis adjusted for age, sex and BMI). Furthermore, sex shows significant interaction with FPG on the serum Se levels (p = 0.01). A follow-up analysis revealed the particular association between FPG and Se levels in women (β = -0.10 ± 0.04, p = 0.01). In conclusion, our data evidenced a women-specific association between FPG and serum Se levels in a sample of adults from southern Mexico.
Collapse
Affiliation(s)
- María Judith Rios-Lugo
- Sección de Medicina Molecular y Traslacional, Centro de Investigación en Ciencias de la Salud y Biomedicina (CICSaB), Universidad Autónoma de San Luis Potosí, Avda Sierra Leona 550, San Luis 78210, San Luis Potosí, Mexico
- Unidad de Posgrado, Facultad de Enfermería y Nutrición, Universidad Autónoma de San Luis Potosí, Avda. Niño Artillero 130, San Luis Potosí 78210, San Luis Potosí, Mexico
| | - Ana Gabriela Palos-Lucio
- Unidad de Posgrado, Facultad de Enfermería y Nutrición, Universidad Autónoma de San Luis Potosí, Avda. Niño Artillero 130, San Luis Potosí 78210, San Luis Potosí, Mexico
| | - Claudia Inés Victoria-Campos
- Sección de Medicina Molecular y Traslacional, Centro de Investigación en Ciencias de la Salud y Biomedicina (CICSaB), Universidad Autónoma de San Luis Potosí, Avda Sierra Leona 550, San Luis 78210, San Luis Potosí, Mexico
- Unidad de Posgrado, Facultad de Enfermería y Nutrición, Universidad Autónoma de San Luis Potosí, Avda. Niño Artillero 130, San Luis Potosí 78210, San Luis Potosí, Mexico
| | - Angel Lugo-Trampe
- Facultad de Medicina Humana, Campus IV, Universidad Autónoma de Chiapas, Carretera a Puerto Madero Km 1.5, Tapachula 30580, Chiapas, Mexico
| | - Karina Del Carmen Trujillo-Murillo
- Facultad de Medicina Humana, Campus IV, Universidad Autónoma de Chiapas, Carretera a Puerto Madero Km 1.5, Tapachula 30580, Chiapas, Mexico
| | - Maximiliano Arahon López-García
- Facultad de Medicina Humana, Campus IV, Universidad Autónoma de Chiapas, Carretera a Puerto Madero Km 1.5, Tapachula 30580, Chiapas, Mexico
| | - Marisol Espinoza-Ruiz
- Facultad de Ciencias Químicas, Campus IV, Universidad Autónoma de Chiapas, Carretera a Puerto Madero Km 1.5, Tapachula 30580, Chiapas, Mexico
| | - Elizabeth Teresita Romero-Guzmán
- Departamento de Química, Gerencia de Ciencias Básicas, Dirección de Investigación Científica, Instituto Nacional de Investigaciones Nucleares, Carretera Mexico-Toluca s/n, La Marquesa, Ocoyoacác 52750, State of Mexico, Mexico
| | - Héctor Hernández-Mendoza
- Instituto de Investigación de Zonas Desérticas, Universidad Autónoma de San Luis Potosí, Altair 200, San Luis 78377, San Luis Potosí, Mexico
- Universidad del Centro de Mexico, Capitán Caldera 75, San Luis 78250, San Luis Potosí, Mexico
- Hospital General de Soledad de Graciano Sánchez, Secretaría de Salud, Valentín Amador 1112, Soledad de Graciano Sánchez 78435, San Luis Potosí, Mexico
| | - Consuelo Chang-Rueda
- Facultad de Ciencias Químicas, Campus IV, Universidad Autónoma de Chiapas, Carretera a Puerto Madero Km 1.5, Tapachula 30580, Chiapas, Mexico
| |
Collapse
|
20
|
Obesity Hinders the Protective Effect of Selenite Supplementation on Insulin Signaling. Antioxidants (Basel) 2022; 11:antiox11050862. [PMID: 35624726 PMCID: PMC9138114 DOI: 10.3390/antiox11050862] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 04/22/2022] [Accepted: 04/26/2022] [Indexed: 12/11/2022] Open
Abstract
The intake of high-fat diets (HFDs) containing large amounts of saturated long-chain fatty acids leads to obesity, oxidative stress, inflammation, and insulin resistance. The trace element selenium, as a crucial part of antioxidative selenoproteins, can protect against the development of diet-induced insulin resistance in white adipose tissue (WAT) by increasing glutathione peroxidase 3 (GPx3) and insulin receptor (IR) expression. Whether selenite (Se) can attenuate insulin resistance in established lipotoxic and obese conditions is unclear. We confirm that GPX3 mRNA expression in adipose tissue correlates with BMI in humans. Cultivating 3T3-L1 pre-adipocytes in palmitate-containing medium followed by Se treatment attenuates insulin resistance with enhanced GPx3 and IR expression and adipocyte differentiation. However, feeding obese mice a selenium-enriched high-fat diet (SRHFD) only resulted in a modest increase in overall selenoprotein gene expression in WAT in mice with unaltered body weight development, glucose tolerance, and insulin resistance. While Se supplementation improved adipocyte morphology, it did not alter WAT insulin sensitivity. However, mice fed a SRHFD exhibited increased insulin content in the pancreas. Overall, while selenite protects against palmitate-induced insulin resistance in vitro, obesity impedes the effect of selenite on insulin action and adipose tissue metabolism in vivo.
Collapse
|
21
|
Mehan S, Bhalla S, Siddiqui EM, Sharma N, Shandilya A, Khan A. Potential Roles of Glucagon-Like Peptide-1 and Its Analogues in Dementia Targeting Impaired Insulin Secretion and Neurodegeneration. Degener Neurol Neuromuscul Dis 2022; 12:31-59. [PMID: 35300067 PMCID: PMC8921673 DOI: 10.2147/dnnd.s247153] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Accepted: 02/16/2022] [Indexed: 12/20/2022] Open
Abstract
Dementia is a chronic, irreversible condition marked by memory loss, cognitive decline, and mental instability. It is clinically related to various progressive neurological diseases, including Parkinson’s disease, Alzheimer’s disease, and Huntington’s. The primary cause of neurological disorders is insulin desensitization, demyelination, oxidative stress, and neuroinflammation accompanied by various aberrant proteins such as amyloid-β deposits, Lewy bodies accumulation, tau formation leading to neurofibrillary tangles. Impaired insulin signaling is directly associated with amyloid-β and α-synuclein deposition, as well as specific signaling cascades involved in neurodegenerative diseases. Insulin dysfunction may initiate various intracellular signaling cascades, including phosphoinositide 3-kinase (PI3K), c-Jun N-terminal kinases (JNK), and mitogen-activated protein kinase (MAPK). Neuronal death, inflammation, neuronal excitation, mitochondrial malfunction, and protein deposition are all influenced by insulin. Recent research has focused on GLP-1 receptor agonists as a potential therapeutic target. They increase glucose-dependent insulin secretion and are beneficial in neurodegenerative diseases by reducing oxidative stress and cytokine production. They reduce the deposition of abnormal proteins by crossing the blood-brain barrier. The purpose of this article is to discuss the role of insulin dysfunction in the pathogenesis of neurological diseases, specifically dementia. Additionally, we reviewed the therapeutic target (GLP-1) and its receptor activators as a possible treatment of dementia.
Collapse
Affiliation(s)
- Sidharth Mehan
- Neuropharmacology Division, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, India
- Correspondence: Sidharth Mehan, Neuropharmacology Division, Department of Pharmacology, ISF College of Pharmacy, Moga, 142001, Punjab, India, Tel +91 8059889909; +91 9461322911, Email ;
| | - Sonalika Bhalla
- Neuropharmacology Division, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, India
| | - Ehraz Mehmood Siddiqui
- Neuropharmacology Division, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, India
| | - Nidhi Sharma
- Neuropharmacology Division, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, India
| | - Ambika Shandilya
- Neuropharmacology Division, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, India
| | - Andleeb Khan
- Department of Pharmacology & Toxicology, College of Pharmacy, Jazan University, Jazan, Kingdom of Saudi Arabia
| |
Collapse
|
22
|
Nicolas-Francès V, Rossi J, Rosnoblet C, Pichereaux C, Hichami S, Astier J, Klinguer A, Wendehenne D, Besson-Bard A. S-Nitrosation of Arabidopsis thaliana Protein Tyrosine Phosphatase 1 Prevents Its Irreversible Oxidation by Hydrogen Peroxide. FRONTIERS IN PLANT SCIENCE 2022; 13:807249. [PMID: 35222471 PMCID: PMC8867174 DOI: 10.3389/fpls.2022.807249] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Accepted: 01/19/2022] [Indexed: 06/01/2023]
Abstract
Tyrosine-specific protein tyrosine phosphatases (Tyr-specific PTPases) are key signaling enzymes catalyzing the removal of the phosphate group from phosphorylated tyrosine residues on target proteins. This post-translational modification notably allows the regulation of mitogen-activated protein kinase (MAPK) cascades during defense reactions. Arabidopsis thaliana protein tyrosine phosphatase 1 (AtPTP1), the only Tyr-specific PTPase present in this plant, acts as a repressor of H2O2 production and regulates the activity of MPK3/MPK6 MAPKs by direct dephosphorylation. Here, we report that recombinant histidine (His)-AtPTP1 protein activity is directly inhibited by H2O2 and nitric oxide (NO) exogenous treatments. The effects of NO are exerted by S-nitrosation, i.e., the formation of a covalent bond between NO and a reduced cysteine residue. This post-translational modification targets the catalytic cysteine C265 and could protect the AtPTP1 protein from its irreversible oxidation by H2O2. This mechanism of protection could be a conserved mechanism in plant PTPases.
Collapse
Affiliation(s)
- Valérie Nicolas-Francès
- Agroécologie, CNRS, INRAE, Institut Agro, Université de Bourgogne, Université Bourgogne Franche-Comté, Dijon, France
| | - Jordan Rossi
- Agroécologie, CNRS, INRAE, Institut Agro, Université de Bourgogne, Université Bourgogne Franche-Comté, Dijon, France
| | - Claire Rosnoblet
- Agroécologie, CNRS, INRAE, Institut Agro, Université de Bourgogne, Université Bourgogne Franche-Comté, Dijon, France
| | - Carole Pichereaux
- Fédération de Recherche (FR3450), Agrobiosciences, Interactions et Biodiversité (FRAIB), CNRS, Toulouse, France
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse UPS, CNRS, Toulouse, France
| | - Siham Hichami
- Agroécologie, CNRS, INRAE, Institut Agro, Université de Bourgogne, Université Bourgogne Franche-Comté, Dijon, France
| | - Jeremy Astier
- Agroécologie, CNRS, INRAE, Institut Agro, Université de Bourgogne, Université Bourgogne Franche-Comté, Dijon, France
| | - Agnès Klinguer
- Agroécologie, CNRS, INRAE, Institut Agro, Université de Bourgogne, Université Bourgogne Franche-Comté, Dijon, France
| | - David Wendehenne
- Agroécologie, CNRS, INRAE, Institut Agro, Université de Bourgogne, Université Bourgogne Franche-Comté, Dijon, France
| | - Angélique Besson-Bard
- Agroécologie, CNRS, INRAE, Institut Agro, Université de Bourgogne, Université Bourgogne Franche-Comté, Dijon, France
| |
Collapse
|
23
|
Viswambharan H, Yuldasheva NY, Imrie H, Bridge K, Haywood NJ, Skromna A, Hemmings KE, Clark ER, Gatenby VK, Cordell P, Simmons KJ, Makava N, Abudushalamu Y, Endesh N, Brown J, Walker AMN, Futers ST, Porter KE, Cubbon RM, Naseem K, Shah AM, Beech DJ, Wheatcroft SB, Kearney MT, Sukumar P. Novel Paracrine Action of Endothelium Enhances Glucose Uptake in Muscle and Fat. Circ Res 2021; 129:720-734. [PMID: 34420367 PMCID: PMC8448413 DOI: 10.1161/circresaha.121.319517] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Hema Viswambharan
- Leeds Institute for Cardiovascular and Metabolic Medicine, University of Leeds, United Kingdom (H.V., N.Y.Y., H.I., K.B., N.J.H., A.S., K.E.H., E.R.C., V.K.G., P.C., K.J.S., N.M., Y.A., N.E., J.B., A.M.N.W., S.T.F., K.E.P., R.M.C., K.N., D.J.B., S.B.W., M.T.K., P.S.)
| | - Nadira Y Yuldasheva
- Leeds Institute for Cardiovascular and Metabolic Medicine, University of Leeds, United Kingdom (H.V., N.Y.Y., H.I., K.B., N.J.H., A.S., K.E.H., E.R.C., V.K.G., P.C., K.J.S., N.M., Y.A., N.E., J.B., A.M.N.W., S.T.F., K.E.P., R.M.C., K.N., D.J.B., S.B.W., M.T.K., P.S.)
| | - Helen Imrie
- Leeds Institute for Cardiovascular and Metabolic Medicine, University of Leeds, United Kingdom (H.V., N.Y.Y., H.I., K.B., N.J.H., A.S., K.E.H., E.R.C., V.K.G., P.C., K.J.S., N.M., Y.A., N.E., J.B., A.M.N.W., S.T.F., K.E.P., R.M.C., K.N., D.J.B., S.B.W., M.T.K., P.S.)
| | - Katherine Bridge
- Leeds Institute for Cardiovascular and Metabolic Medicine, University of Leeds, United Kingdom (H.V., N.Y.Y., H.I., K.B., N.J.H., A.S., K.E.H., E.R.C., V.K.G., P.C., K.J.S., N.M., Y.A., N.E., J.B., A.M.N.W., S.T.F., K.E.P., R.M.C., K.N., D.J.B., S.B.W., M.T.K., P.S.)
| | - Natalie J Haywood
- Leeds Institute for Cardiovascular and Metabolic Medicine, University of Leeds, United Kingdom (H.V., N.Y.Y., H.I., K.B., N.J.H., A.S., K.E.H., E.R.C., V.K.G., P.C., K.J.S., N.M., Y.A., N.E., J.B., A.M.N.W., S.T.F., K.E.P., R.M.C., K.N., D.J.B., S.B.W., M.T.K., P.S.)
| | - Anna Skromna
- Leeds Institute for Cardiovascular and Metabolic Medicine, University of Leeds, United Kingdom (H.V., N.Y.Y., H.I., K.B., N.J.H., A.S., K.E.H., E.R.C., V.K.G., P.C., K.J.S., N.M., Y.A., N.E., J.B., A.M.N.W., S.T.F., K.E.P., R.M.C., K.N., D.J.B., S.B.W., M.T.K., P.S.)
| | - Karen E Hemmings
- Leeds Institute for Cardiovascular and Metabolic Medicine, University of Leeds, United Kingdom (H.V., N.Y.Y., H.I., K.B., N.J.H., A.S., K.E.H., E.R.C., V.K.G., P.C., K.J.S., N.M., Y.A., N.E., J.B., A.M.N.W., S.T.F., K.E.P., R.M.C., K.N., D.J.B., S.B.W., M.T.K., P.S.)
| | - Emily R Clark
- Leeds Institute for Cardiovascular and Metabolic Medicine, University of Leeds, United Kingdom (H.V., N.Y.Y., H.I., K.B., N.J.H., A.S., K.E.H., E.R.C., V.K.G., P.C., K.J.S., N.M., Y.A., N.E., J.B., A.M.N.W., S.T.F., K.E.P., R.M.C., K.N., D.J.B., S.B.W., M.T.K., P.S.)
| | - V Kate Gatenby
- Leeds Institute for Cardiovascular and Metabolic Medicine, University of Leeds, United Kingdom (H.V., N.Y.Y., H.I., K.B., N.J.H., A.S., K.E.H., E.R.C., V.K.G., P.C., K.J.S., N.M., Y.A., N.E., J.B., A.M.N.W., S.T.F., K.E.P., R.M.C., K.N., D.J.B., S.B.W., M.T.K., P.S.)
| | - Paul Cordell
- Leeds Institute for Cardiovascular and Metabolic Medicine, University of Leeds, United Kingdom (H.V., N.Y.Y., H.I., K.B., N.J.H., A.S., K.E.H., E.R.C., V.K.G., P.C., K.J.S., N.M., Y.A., N.E., J.B., A.M.N.W., S.T.F., K.E.P., R.M.C., K.N., D.J.B., S.B.W., M.T.K., P.S.)
| | - Katie J Simmons
- Leeds Institute for Cardiovascular and Metabolic Medicine, University of Leeds, United Kingdom (H.V., N.Y.Y., H.I., K.B., N.J.H., A.S., K.E.H., E.R.C., V.K.G., P.C., K.J.S., N.M., Y.A., N.E., J.B., A.M.N.W., S.T.F., K.E.P., R.M.C., K.N., D.J.B., S.B.W., M.T.K., P.S.)
| | - Natallia Makava
- Leeds Institute for Cardiovascular and Metabolic Medicine, University of Leeds, United Kingdom (H.V., N.Y.Y., H.I., K.B., N.J.H., A.S., K.E.H., E.R.C., V.K.G., P.C., K.J.S., N.M., Y.A., N.E., J.B., A.M.N.W., S.T.F., K.E.P., R.M.C., K.N., D.J.B., S.B.W., M.T.K., P.S.)
| | - Yilizila Abudushalamu
- Leeds Institute for Cardiovascular and Metabolic Medicine, University of Leeds, United Kingdom (H.V., N.Y.Y., H.I., K.B., N.J.H., A.S., K.E.H., E.R.C., V.K.G., P.C., K.J.S., N.M., Y.A., N.E., J.B., A.M.N.W., S.T.F., K.E.P., R.M.C., K.N., D.J.B., S.B.W., M.T.K., P.S.)
| | - Naima Endesh
- Leeds Institute for Cardiovascular and Metabolic Medicine, University of Leeds, United Kingdom (H.V., N.Y.Y., H.I., K.B., N.J.H., A.S., K.E.H., E.R.C., V.K.G., P.C., K.J.S., N.M., Y.A., N.E., J.B., A.M.N.W., S.T.F., K.E.P., R.M.C., K.N., D.J.B., S.B.W., M.T.K., P.S.)
| | - Jane Brown
- Leeds Institute for Cardiovascular and Metabolic Medicine, University of Leeds, United Kingdom (H.V., N.Y.Y., H.I., K.B., N.J.H., A.S., K.E.H., E.R.C., V.K.G., P.C., K.J.S., N.M., Y.A., N.E., J.B., A.M.N.W., S.T.F., K.E.P., R.M.C., K.N., D.J.B., S.B.W., M.T.K., P.S.)
| | - Andrew M N Walker
- Leeds Institute for Cardiovascular and Metabolic Medicine, University of Leeds, United Kingdom (H.V., N.Y.Y., H.I., K.B., N.J.H., A.S., K.E.H., E.R.C., V.K.G., P.C., K.J.S., N.M., Y.A., N.E., J.B., A.M.N.W., S.T.F., K.E.P., R.M.C., K.N., D.J.B., S.B.W., M.T.K., P.S.)
| | - Simon T Futers
- Leeds Institute for Cardiovascular and Metabolic Medicine, University of Leeds, United Kingdom (H.V., N.Y.Y., H.I., K.B., N.J.H., A.S., K.E.H., E.R.C., V.K.G., P.C., K.J.S., N.M., Y.A., N.E., J.B., A.M.N.W., S.T.F., K.E.P., R.M.C., K.N., D.J.B., S.B.W., M.T.K., P.S.)
| | - Karen E Porter
- Leeds Institute for Cardiovascular and Metabolic Medicine, University of Leeds, United Kingdom (H.V., N.Y.Y., H.I., K.B., N.J.H., A.S., K.E.H., E.R.C., V.K.G., P.C., K.J.S., N.M., Y.A., N.E., J.B., A.M.N.W., S.T.F., K.E.P., R.M.C., K.N., D.J.B., S.B.W., M.T.K., P.S.)
| | - Richard M Cubbon
- Leeds Institute for Cardiovascular and Metabolic Medicine, University of Leeds, United Kingdom (H.V., N.Y.Y., H.I., K.B., N.J.H., A.S., K.E.H., E.R.C., V.K.G., P.C., K.J.S., N.M., Y.A., N.E., J.B., A.M.N.W., S.T.F., K.E.P., R.M.C., K.N., D.J.B., S.B.W., M.T.K., P.S.)
| | - Khalid Naseem
- Leeds Institute for Cardiovascular and Metabolic Medicine, University of Leeds, United Kingdom (H.V., N.Y.Y., H.I., K.B., N.J.H., A.S., K.E.H., E.R.C., V.K.G., P.C., K.J.S., N.M., Y.A., N.E., J.B., A.M.N.W., S.T.F., K.E.P., R.M.C., K.N., D.J.B., S.B.W., M.T.K., P.S.)
| | - Ajay M Shah
- British Heart Foundation Centre of Research Excellence, King's College London (A.M.S.)
| | - David J Beech
- Leeds Institute for Cardiovascular and Metabolic Medicine, University of Leeds, United Kingdom (H.V., N.Y.Y., H.I., K.B., N.J.H., A.S., K.E.H., E.R.C., V.K.G., P.C., K.J.S., N.M., Y.A., N.E., J.B., A.M.N.W., S.T.F., K.E.P., R.M.C., K.N., D.J.B., S.B.W., M.T.K., P.S.)
| | - Stephen B Wheatcroft
- Leeds Institute for Cardiovascular and Metabolic Medicine, University of Leeds, United Kingdom (H.V., N.Y.Y., H.I., K.B., N.J.H., A.S., K.E.H., E.R.C., V.K.G., P.C., K.J.S., N.M., Y.A., N.E., J.B., A.M.N.W., S.T.F., K.E.P., R.M.C., K.N., D.J.B., S.B.W., M.T.K., P.S.)
| | - Mark T Kearney
- Leeds Institute for Cardiovascular and Metabolic Medicine, University of Leeds, United Kingdom (H.V., N.Y.Y., H.I., K.B., N.J.H., A.S., K.E.H., E.R.C., V.K.G., P.C., K.J.S., N.M., Y.A., N.E., J.B., A.M.N.W., S.T.F., K.E.P., R.M.C., K.N., D.J.B., S.B.W., M.T.K., P.S.)
| | - Piruthivi Sukumar
- Leeds Institute for Cardiovascular and Metabolic Medicine, University of Leeds, United Kingdom (H.V., N.Y.Y., H.I., K.B., N.J.H., A.S., K.E.H., E.R.C., V.K.G., P.C., K.J.S., N.M., Y.A., N.E., J.B., A.M.N.W., S.T.F., K.E.P., R.M.C., K.N., D.J.B., S.B.W., M.T.K., P.S.)
| |
Collapse
|
24
|
Bélanger V, Benmoussa A, Napartuk M, Warin A, Laverdière C, Marcoux S, Levy E, Marcil V. The Role of Oxidative Stress and Inflammation in Cardiometabolic Health of Children During Cancer Treatment and Potential Impact of Key Nutrients. Antioxid Redox Signal 2021; 35:293-318. [PMID: 33386063 DOI: 10.1089/ars.2020.8143] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Significance: The 5-year survival rate of childhood cancers is now reaching 84%. However, treatments cause numerous acute and long-term side effects. These include cardiometabolic complications, namely hypertension, dyslipidemia, hyperglycemia, insulin resistance, and increased fat mass. Recent Advances: Many antineoplastic treatments can induce oxidative stress (OxS) and trigger an inflammatory response, which may cause acute and chronic side effects. Critical Issues: Clinical studies have reported a state of heightened OxS and inflammation during cancer treatment in children as the result of treatment cytotoxic action on both cancerous and noncancerous cells. Higher levels of OxS and inflammation are associated with treatment side effects and with the development of cardiometabolic complications. Key nutrients (omega-3 polyunsaturated fatty acids, dietary antioxidants, probiotics, and prebiotics) have the potential to modulate inflammatory and oxidative responses and, therefore, could be considered in the search for adverse complication prevention means as long as antineoplastic treatment efficiency is maintained. Future Directions: There is a need to better understand the relationship between cardiometabolic complications, OxS, inflammation and diet during pediatric cancer treatment, which represents the ultimate goal of this review. Antioxid. Redox Signal. 35, 293-318.
Collapse
Affiliation(s)
- Véronique Bélanger
- Research Centre, CHU Sainte-Justine University Health Centre, Montreal, Canada.,Department of Nutrition, Université de Montréal, Montreal, Canada.,Institute of Nutrition and Functional Foods, Laval University, Quebec City, Canada
| | - Abderrahim Benmoussa
- Research Centre, CHU Sainte-Justine University Health Centre, Montreal, Canada.,Department of Nutrition, Université de Montréal, Montreal, Canada.,Institute of Nutrition and Functional Foods, Laval University, Quebec City, Canada
| | - Mélanie Napartuk
- Research Centre, CHU Sainte-Justine University Health Centre, Montreal, Canada.,Department of Nutrition, Université de Montréal, Montreal, Canada.,Institute of Nutrition and Functional Foods, Laval University, Quebec City, Canada
| | - Alexandre Warin
- Research Centre, CHU Sainte-Justine University Health Centre, Montreal, Canada
| | | | - Sophie Marcoux
- Department of Public Health & Preventive Medicine, Université de Montréal, Montreal, Canada
| | - Emile Levy
- Research Centre, CHU Sainte-Justine University Health Centre, Montreal, Canada.,Department of Nutrition, Université de Montréal, Montreal, Canada.,Institute of Nutrition and Functional Foods, Laval University, Quebec City, Canada.,Department of Pediatrics, Université de Montréal, Montreal, Canada
| | - Valérie Marcil
- Research Centre, CHU Sainte-Justine University Health Centre, Montreal, Canada.,Department of Nutrition, Université de Montréal, Montreal, Canada.,Institute of Nutrition and Functional Foods, Laval University, Quebec City, Canada
| |
Collapse
|
25
|
McKeegan K, Mason SA, Trewin AJ, Keske MA, Wadley GD, Della Gatta PA, Nikolaidis MG, Parker L. Reactive oxygen species in exercise and insulin resistance: Working towards personalized antioxidant treatment. Redox Biol 2021; 44:102005. [PMID: 34049222 PMCID: PMC8167146 DOI: 10.1016/j.redox.2021.102005] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Revised: 04/25/2021] [Accepted: 05/06/2021] [Indexed: 12/11/2022] Open
Abstract
Reactive oxygen species (ROS) are well known for their role in insulin resistance and the development of cardiometabolic disease including type 2 diabetes mellitus (T2D). Conversely, evidence supports the notion that ROS are a necessary component for glucose cell transport and adaptation to physiological stress including exercise and muscle contraction. Although genetic rodent models and cell culture studies indicate antioxidant treatment to be an effective strategy for targeting ROS to promote health, human findings are largely inconsistent. In this review we discuss human research that has investigated antioxidant treatment and glycemic control in the context of health (healthy individuals and during exercise) and disease (insulin resistance and T2D). We have identified key factors that are likely to influence the effectiveness of antioxidant treatment: 1) the context of treatment including whether oxidative distress or eustress is present (e.g., hyperglycemia/lipidaemia or during exercise and muscle contraction); 2) whether specific endogenous antioxidant deficiencies are identified (redox screening); 3) whether antioxidant treatment is specifically designed to target and restore identified deficiencies (antioxidant specificity); 4) and the bioavailability and bioactivity of the antioxidant which are influenced by treatment dose, duration, and method of administration. The majority of human research has failed to account for these factors, limiting their ability to robustly test the effectiveness of antioxidants for health promotion and disease prevention. We propose that a modern "redox screening" and "personalized antioxidant treatment" approach is required to robustly explore redox regulation of human physiology and to elicit more effective antioxidant treatment in humans.
Collapse
Affiliation(s)
- Kathryn McKeegan
- Institute for Physical Activity and Nutrition, School of Exercise and Nutrition Sciences, Deakin University, Geelong, Australia
| | - Shaun A Mason
- Institute for Physical Activity and Nutrition, School of Exercise and Nutrition Sciences, Deakin University, Geelong, Australia
| | - Adam J Trewin
- Institute for Physical Activity and Nutrition, School of Exercise and Nutrition Sciences, Deakin University, Geelong, Australia
| | - Michelle A Keske
- Institute for Physical Activity and Nutrition, School of Exercise and Nutrition Sciences, Deakin University, Geelong, Australia
| | - Glenn D Wadley
- Institute for Physical Activity and Nutrition, School of Exercise and Nutrition Sciences, Deakin University, Geelong, Australia
| | - Paul A Della Gatta
- Institute for Physical Activity and Nutrition, School of Exercise and Nutrition Sciences, Deakin University, Geelong, Australia
| | - Michalis G Nikolaidis
- Department of Physical Education and Sport Science at Serres, Aristotle University of Thessaloniki, Serres, Greece
| | - Lewan Parker
- Institute for Physical Activity and Nutrition, School of Exercise and Nutrition Sciences, Deakin University, Geelong, Australia.
| |
Collapse
|
26
|
Shum M, Shintre CA, Althoff T, Gutierrez V, Segawa M, Saxberg AD, Martinez M, Adamson R, Young MR, Faust B, Gharakhanian R, Su S, Chella Krishnan K, Mahdaviani K, Veliova M, Wolf DM, Ngo J, Nocito L, Stiles L, Abramson J, Lusis AJ, Hevener AL, Zoghbi ME, Carpenter EP, Liesa M. ABCB10 exports mitochondrial biliverdin, driving metabolic maladaptation in obesity. Sci Transl Med 2021; 13:13/594/eabd1869. [PMID: 34011630 DOI: 10.1126/scitranslmed.abd1869] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 01/25/2021] [Accepted: 04/20/2021] [Indexed: 12/12/2022]
Abstract
Although the role of hydrophilic antioxidants in the development of hepatic insulin resistance and nonalcoholic fatty liver disease has been well studied, the role of lipophilic antioxidants remains poorly characterized. A known lipophilic hydrogen peroxide scavenger is bilirubin, which can be oxidized to biliverdin and then reduced back to bilirubin by cytosolic biliverdin reductase. Oxidation of bilirubin to biliverdin inside mitochondria must be followed by the export of biliverdin to the cytosol, where biliverdin is reduced back to bilirubin. Thus, the putative mitochondrial exporter of biliverdin is expected to be a major determinant of bilirubin regeneration and intracellular hydrogen peroxide scavenging. Here, we identified ABCB10 as a mitochondrial biliverdin exporter. ABCB10 reconstituted into liposomes transported biliverdin, and ABCB10 deletion caused accumulation of biliverdin inside mitochondria. Obesity with insulin resistance up-regulated hepatic ABCB10 expression in mice and elevated cytosolic and mitochondrial bilirubin content in an ABCB10-dependent manner. Revealing a maladaptive role of ABCB10-driven bilirubin synthesis, hepatic ABCB10 deletion protected diet-induced obese mice from steatosis and hyperglycemia, improving insulin-mediated suppression of glucose production and decreasing lipogenic SREBP-1c expression. Protection was concurrent with enhanced mitochondrial function and increased inactivation of PTP1B, a phosphatase disrupting insulin signaling and elevating SREBP-1c expression. Restoration of cellular bilirubin content in ABCB10 KO hepatocytes reversed the improvements in mitochondrial function and PTP1B inactivation, demonstrating that bilirubin was the maladaptive effector linked to ABCB10 function. Thus, we identified a fundamental transport process that amplifies intracellular bilirubin redox actions, which can exacerbate insulin resistance and steatosis in obesity.
Collapse
Affiliation(s)
- Michael Shum
- Department of Medicine, Division of Endocrinology, David Geffen School of Medicine at UCLA, 650 Charles E. Young Dr., Los Angeles, CA 90095, USA.,Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, 650 Charles E. Young Dr., Los Angeles, CA 90095, USA.,Molecular Biology Institute at UCLA, 611 Charles E. Young Drive East, Los Angeles, CA 90095-1570, USA
| | - Chitra A Shintre
- Center for Medicines Discovery, University of Oxford, Oxfordshire OX3 7DQ, UK
| | - Thorsten Althoff
- Department of Physiology, University of California, Los Angeles, 650 Charles E. Young Dr., Los Angeles, CA 90095, USA
| | - Vincent Gutierrez
- Department of Medicine, Division of Endocrinology, David Geffen School of Medicine at UCLA, 650 Charles E. Young Dr., Los Angeles, CA 90095, USA.,Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, 650 Charles E. Young Dr., Los Angeles, CA 90095, USA.,Molecular Biology Institute at UCLA, 611 Charles E. Young Drive East, Los Angeles, CA 90095-1570, USA
| | - Mayuko Segawa
- Department of Medicine, Division of Endocrinology, David Geffen School of Medicine at UCLA, 650 Charles E. Young Dr., Los Angeles, CA 90095, USA.,Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, 650 Charles E. Young Dr., Los Angeles, CA 90095, USA.,Molecular Biology Institute at UCLA, 611 Charles E. Young Drive East, Los Angeles, CA 90095-1570, USA
| | - Alexandra D Saxberg
- Department of Molecular Cell Biology, School of Natural Sciences, University of California, Merced, 5200 North Lake Rd., Merced, CA 95343, USA
| | - Melissa Martinez
- Department of Molecular Cell Biology, School of Natural Sciences, University of California, Merced, 5200 North Lake Rd., Merced, CA 95343, USA
| | - Roslin Adamson
- Center for Medicines Discovery, University of Oxford, Oxfordshire OX3 7DQ, UK
| | - Margaret R Young
- Center for Medicines Discovery, University of Oxford, Oxfordshire OX3 7DQ, UK
| | - Belinda Faust
- Center for Medicines Discovery, University of Oxford, Oxfordshire OX3 7DQ, UK
| | - Raffi Gharakhanian
- Department of Medicine, Division of Endocrinology, David Geffen School of Medicine at UCLA, 650 Charles E. Young Dr., Los Angeles, CA 90095, USA.,Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, 650 Charles E. Young Dr., Los Angeles, CA 90095, USA.,Molecular Biology Institute at UCLA, 611 Charles E. Young Drive East, Los Angeles, CA 90095-1570, USA
| | - Shi Su
- Evans Biomedical Research Center, Boston University School of Medicine, 650 Albany St., Boston, MA 02118, USA
| | - Karthickeyan Chella Krishnan
- Department of Human Genetics, David Geffen School of Medicine at UCLA, 650 Charles E. Young Dr., Los Angeles, CA 90095, USA.,Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, 231 Albert Sabin Way, Cincinnati, OH 45267-0575, USA
| | - Kiana Mahdaviani
- Department of Medicine, Division of Endocrinology, David Geffen School of Medicine at UCLA, 650 Charles E. Young Dr., Los Angeles, CA 90095, USA.,Evans Biomedical Research Center, Boston University School of Medicine, 650 Albany St., Boston, MA 02118, USA
| | - Michaela Veliova
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, 650 Charles E. Young Dr., Los Angeles, CA 90095, USA
| | - Dane M Wolf
- Department of Medicine, Division of Endocrinology, David Geffen School of Medicine at UCLA, 650 Charles E. Young Dr., Los Angeles, CA 90095, USA.,Evans Biomedical Research Center, Boston University School of Medicine, 650 Albany St., Boston, MA 02118, USA
| | - Jennifer Ngo
- Department of Medicine, Division of Endocrinology, David Geffen School of Medicine at UCLA, 650 Charles E. Young Dr., Los Angeles, CA 90095, USA.,Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, 650 Charles E. Young Dr., Los Angeles, CA 90095, USA.,Molecular Biology Institute at UCLA, 611 Charles E. Young Drive East, Los Angeles, CA 90095-1570, USA.,Department of Chemistry and Biochemistry, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Laura Nocito
- Evans Biomedical Research Center, Boston University School of Medicine, 650 Albany St., Boston, MA 02118, USA
| | - Linsey Stiles
- Department of Medicine, Division of Endocrinology, David Geffen School of Medicine at UCLA, 650 Charles E. Young Dr., Los Angeles, CA 90095, USA.,Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, 650 Charles E. Young Dr., Los Angeles, CA 90095, USA
| | - Jeff Abramson
- Department of Physiology, University of California, Los Angeles, 650 Charles E. Young Dr., Los Angeles, CA 90095, USA
| | - Aldons J Lusis
- Department of Human Genetics, David Geffen School of Medicine at UCLA, 650 Charles E. Young Dr., Los Angeles, CA 90095, USA.,Department of Medicine, Division of Cardiology, David Geffen School of Medicine at UCLA, 650 Charles E. Young Dr., Los Angeles, CA 90095, USA.,Department of Microbiology, Immunology and Molecular Genetics, David Geffen School of Medicine at UCLA, 650 Charles E. Young Dr., Los Angeles, CA 90095, USA
| | - Andrea L Hevener
- Department of Medicine, Division of Endocrinology, David Geffen School of Medicine at UCLA, 650 Charles E. Young Dr., Los Angeles, CA 90095, USA
| | - Maria E Zoghbi
- Department of Molecular Cell Biology, School of Natural Sciences, University of California, Merced, 5200 North Lake Rd., Merced, CA 95343, USA
| | | | - Marc Liesa
- Department of Medicine, Division of Endocrinology, David Geffen School of Medicine at UCLA, 650 Charles E. Young Dr., Los Angeles, CA 90095, USA. .,Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, 650 Charles E. Young Dr., Los Angeles, CA 90095, USA.,Molecular Biology Institute at UCLA, 611 Charles E. Young Drive East, Los Angeles, CA 90095-1570, USA
| |
Collapse
|
27
|
Abstract
Protein Tyrosine Phosphatases reverse cellular signals initiated by growth factors receptors and other tyrosine kinases by dephosphorylating phosphotyrosine on target proteins. The activity of these enzymes is crucial for maintaining cell homeostasis, yet these enzymes have been often dismissed as humble house-keeping proteins. Understandably, mutations and changes in expression patterns of Protein Tyrosine Phosphatases are implicated in tumorigenesis and various carcinomas. The conserved nature of their catalytic domains makes drug discovery a challenging pursuit. In this review, we focus on describing the various classes of Protein Tyrosine Phosphatases and their catalytic domains. We also summarize their role in cancer and neurodegenerative diseases using specific members as the model system. Finally, we explain the dichotomy in the biological role of catalytically active vs the pseudoenzyme forms of Protein Tyrosine Phosphatases in the context of their membrane bound receptor forms. This chapter aims to provide a current understanding of these proteins, in the background of their foundational past research.
Collapse
Affiliation(s)
- Colin L Welsh
- Department of Cell and Molecular Pharmacology & Experimental Therapeutics, College of Medicine, Medical University of South Carolina, Charleston, SC, United States
| | - Preeti Pandey
- Department of Cell and Molecular Pharmacology & Experimental Therapeutics, College of Medicine, Medical University of South Carolina, Charleston, SC, United States
| | - Lalima G Ahuja
- Department of Cell and Molecular Pharmacology & Experimental Therapeutics, College of Medicine, Medical University of South Carolina, Charleston, SC, United States; Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, United States.
| |
Collapse
|
28
|
Mushtaq U, Bashir M, Nabi S, Khanday FA. Epidermal growth factor receptor and integrins meet redox signaling through P66shc and Rac1. Cytokine 2021; 146:155625. [PMID: 34157521 DOI: 10.1016/j.cyto.2021.155625] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2020] [Revised: 05/23/2021] [Accepted: 06/08/2021] [Indexed: 12/24/2022]
Abstract
This review examines the concerted role of Epidermal Growth Factor Receptor (EGFR) and integrins in regulating Reactive oxygen species (ROS) production through different signaling pathways. ROS as such are not always deleterious to the cells but they also act as signaling molecules, that regulates numerous indespensible physiological fuctions of life. Many adaptor proteins, particularly Shc and Grb2, are involved in mediating the downstream signaling pathways stimulated by EGFR and integrins. Integrin-induced activation of EGFR and subsequent tyrosine phosphorylation of a class of acceptor sites on EGFR leads to alignment and tyrosine phosphorylation of Shc, PLCγ, the p85 subunit of PI-3 K, and Cbl, followed by activation of the downstream targets Erk and Akt/PKB. Functional interactions between these receptors result in the activation of Rac1 via these adaptor proteins, thereby leading to Reactive Oxygen Species. Both GF and integrin activation can produce oxidants independently, however synergistically there is increased ROS generation, suggesting a mutual cooperation between integrins and GFRs for redox signalling. The ROS produced further promotes feed-forward stimulation of redox signaling events such as MAPK activation and gene expression. This relationship has not been reviewed previously. The literature presented here can have multiple implications, ranging from looking at synergistic effects of integrin and EGFR mediated signaling mechanisms of different proteins to possible therapeutic interventions operated by these two receptors. Furthermore, such mutual redox regulation of crosstalk between EGFR and integrins not only add to the established models of pathological oxidative stress, but also can impart new avenues and opportunities for targeted antioxidant based therapeutics.
Collapse
Affiliation(s)
- Umar Mushtaq
- Department of Biotechnology, University of Kashmir, Srinagar, JK 190006, India; Department of Biotechnology, Central University of Kashmir, Ganderbal, JK 191201, India
| | - Muneesa Bashir
- Department of Biotechnology, University of Kashmir, Srinagar, JK 190006, India; Department of Higher Education, Government of Jammu & Kashmir, 190001, India
| | - Sumaiya Nabi
- Department of Biochemistry, University of Kashmir, Srinagar, JK 190006, India
| | - Firdous A Khanday
- Department of Biotechnology, University of Kashmir, Srinagar, JK 190006, India.
| |
Collapse
|
29
|
Pleiotropic and Potentially Beneficial Effects of Reactive Oxygen Species on the Intracellular Signaling Pathways in Endothelial Cells. Antioxidants (Basel) 2021; 10:antiox10060904. [PMID: 34205032 PMCID: PMC8229098 DOI: 10.3390/antiox10060904] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 05/28/2021] [Accepted: 05/31/2021] [Indexed: 02/06/2023] Open
Abstract
Endothelial cells (ECs) are exposed to molecular dioxygen and its derivative reactive oxygen species (ROS). ROS are now well established as important signaling messengers. Excessive production of ROS, however, results in oxidative stress, a significant contributor to the development of numerous diseases. Here, we analyze the experimental data and theoretical concepts concerning positive pro-survival effects of ROS on signaling pathways in endothelial cells (ECs). Our analysis of the available experimental data suggests possible positive roles of ROS in induction of pro-survival pathways, downstream of the Gi-protein-coupled receptors, which mimics insulin signaling and prevention or improvement of the endothelial dysfunction. It is, however, doubtful, whether ROS can contribute to the stabilization of the endothelial barrier.
Collapse
|
30
|
Supasai S, Adamo AM, Mathieu P, Marino RC, Hellmers AC, Cremonini E, Oteiza PI. Gestational zinc deficiency impairs brain astrogliogenesis in rats through multistep alterations of the JAK/STAT3 signaling pathway. Redox Biol 2021; 44:102017. [PMID: 34049221 PMCID: PMC8167189 DOI: 10.1016/j.redox.2021.102017] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2021] [Revised: 05/15/2021] [Accepted: 05/16/2021] [Indexed: 10/27/2022] Open
Abstract
We previously showed that zinc (Zn) deficiency affects the STAT3 signaling pathway in part through redox-regulated mechanisms. Given that STAT3 is central to the process of astrogliogenesis, this study investigated the consequences of maternal marginal Zn deficiency on the developmental timing and key mechanisms of STAT3 activation, and its consequences on astrogliogenesis in the offspring. This work characterized the temporal profile of cortical STAT3 activation from the mid embryonic stage up to young adulthood in the offspring from dams fed a marginal Zn deficient diet (MZD) throughout gestation and until postnatal day (P) 2. All rats were fed a Zn sufficient diet (control) from P2 until P56. Maternal zinc deficiency disrupted cortical STAT3 activation at E19 and P2. This was accompanied by altered activation of JAK2 kinase due to changes in PTP1B phosphatase activity. The underlying mechanisms mediating the adverse impact of a decreased Zn availability on STAT3 activation in the offspring brain include: (i) impaired PTP1B degradation via the ubiquitin/proteasome pathway; (ii) tubulin oxidation, associated decreased interactions with STAT3 and consequent impaired nuclear translocation; and (iii) decreased nuclear STAT3 acetylation. Zn deficiency-associated decreased STAT3 activation adversely impacted astrogliogenesis, leading to a lower astrocyte number in the early postnatal and adult brain cortex. Thus, a decreased availability of Zn during early development can have a major and irreversible adverse effect on astrogliogenesis, in part via multistep alterations in the STAT3 pathway.
Collapse
Affiliation(s)
- Suangsuda Supasai
- Department of Nutrition, University of California, One Shields Avenue, Davis, CA, 95616, USA; Department of Environmental Toxicology, University of California, One Shields Avenue, Davis, CA, 95616, USA; Department of Molecular Tropical Medicine and Genetics, Faculty of Tropical Medicine, Mahidol University, Bangkok, 10400, Thailand
| | - Ana M Adamo
- Department of Biological Chemistry and IQUIFIB (UBA-CONICET), Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Patricia Mathieu
- Department of Biological Chemistry and IQUIFIB (UBA-CONICET), Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Regina C Marino
- Department of Nutrition, University of California, One Shields Avenue, Davis, CA, 95616, USA; Department of Environmental Toxicology, University of California, One Shields Avenue, Davis, CA, 95616, USA
| | - Adelaide C Hellmers
- Department of Nutrition, University of California, One Shields Avenue, Davis, CA, 95616, USA; Department of Environmental Toxicology, University of California, One Shields Avenue, Davis, CA, 95616, USA
| | - Eleonora Cremonini
- Department of Nutrition, University of California, One Shields Avenue, Davis, CA, 95616, USA; Department of Environmental Toxicology, University of California, One Shields Avenue, Davis, CA, 95616, USA
| | - Patricia I Oteiza
- Department of Nutrition, University of California, One Shields Avenue, Davis, CA, 95616, USA; Department of Environmental Toxicology, University of California, One Shields Avenue, Davis, CA, 95616, USA.
| |
Collapse
|
31
|
Londhe AD, Rizvi SHM, Boivin B. In Vitro Activity Assays to Quantitatively Assess the Endogenous Reversible Oxidation State of Protein Tyrosine Phosphatases in Cells. ACTA ACUST UNITED AC 2021; 12:e84. [PMID: 32805074 DOI: 10.1002/cpch.84] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The reversible oxidation of protein tyrosine phosphatases (PTPs) impairs their ability to dephosphorylate substrates in vivo. This transient inactivation of PTPs occurs as their conserved catalytic cysteine residue reacts with cellular oxidants thereby abolishing the ability of this reactive cysteine to attack the phosphate of the target substrate. Hence, in vivo, the inhibition of specific PTPs in response to regulated and localized rises in cellular oxidants enables phospho-dependent signaling. We present assays that measure the endogenous activity of specific PTPs that become transiently inactivated in cells exposed to growth factors. Here, we describe the methods and highlight the pitfalls to avoid post-lysis oxidation of PTPs in order to assess the inactivation and the reactivation of PTPs targeted by cellular oxidants in signal transduction. © 2020 Wiley Periodicals LLC. Basic Protocol 1: Cell transfection (optional) Support Protocol: Preparation of degassed lysis buffers Basic Protocol 2: Cellular extraction in anaerobic conditions Basic Protocol 3: Enrichment and activity assay of specific PTPs Alternate Protocol: Measurement of active PTPs via direct cysteinyl labeling.
Collapse
Affiliation(s)
- Avinash D Londhe
- Department of Nanobioscience, SUNY Polytechnic Institute, Albany, New York
| | - Syed H M Rizvi
- Department of Nanobioscience, SUNY Polytechnic Institute, Albany, New York
| | - Benoit Boivin
- Department of Nanobioscience, SUNY Polytechnic Institute, Albany, New York
| |
Collapse
|
32
|
Insulin Modulates the Bioenergetic and Thermogenic Capacity of Rat Brown Adipocytes In Vivo by Modulating Mitochondrial Mosaicism. Int J Mol Sci 2020; 21:ijms21239204. [PMID: 33287103 PMCID: PMC7730624 DOI: 10.3390/ijms21239204] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 11/09/2020] [Accepted: 11/19/2020] [Indexed: 12/17/2022] Open
Abstract
The effects of insulin on the bioenergetic and thermogenic capacity of brown adipocyte mitochondria were investigated by focusing on key mitochondrial proteins. Two-month-old male Wistar rats were treated acutely or chronically with a low or high dose of insulin. Acute low insulin dose increased expression of all electron transport chain complexes and complex IV activity, whereas high dose increased complex II expression. Chronic low insulin dose decreased complex I and cyt c expression while increasing complex II and IV expression and complex IV activity. Chronic high insulin dose decreased complex II, III, cyt c, and increased complex IV expression. Uncoupling protein (UCP) 1 expression was decreased after acute high insulin but increased following chronic insulin treatment. ATP synthase expression was increased after acute and decreased after chronic insulin treatment. Only a high dose of insulin increased ATP synthase activity in acute and decreased it in chronic treatment. ATPase inhibitory factor protein expression was increased in all treated groups. Confocal microscopy showed that key mitochondrial proteins colocalize differently in different mitochondria within a single brown adipocyte, indicating mitochondrial mosaicism. These results suggest that insulin modulates the bioenergetic and thermogenic capacity of rat brown adipocytes in vivo by modulating mitochondrial mosaicism.
Collapse
|
33
|
Regulation of Metabolic Processes by Hydrogen Peroxide Generated by NADPH Oxidases. Processes (Basel) 2020. [DOI: 10.3390/pr8111424] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Hydrogen peroxide (H2O2) is an important oxidizing molecule that regulates the metabolisms of aerobic organisms. Redox signaling comprises physiological oxidative stress (eustress), while excessive oxidative stress causes damage to molecules. The main enzymatic generators of H2O2 are nicotinamide adenine dinucleotide phosphate oxidases or NADPH oxidases (NOXs) and mitochondrial respiratory chains, as well as various oxidases. The NOX family is constituted of seven enzyme isoforms that produce a superoxide anion (O2−), which can be converted to H2O2 by superoxide dismutase or spontaneously. H2O2 passes through the membranes by some aquaporins (AQPs), known as peroxyporins. It diffuses through cells and tissues to initiate cellular effects, such as proliferation, the recruitment of immune cells, and cell shape changes. Therefore, it has been proposed that H2O2 has the same importance as Ca2+ or adenosine triphosphate (ATP) to act as modulators in signaling and the metabolism. The present overview focuses on the metabolic processes of liver and adipose tissue, regulated by the H2O2 generated by NOXs.
Collapse
|
34
|
Chamorro ME, Maltaneri R, Schiappacasse A, Nesse A, Vittori D. Role of protein tyrosine phosphatase 1B (PTP1B) in the increased sensitivity of endothelial cells to a promigratory effect of erythropoietin in an inflammatory environment. Biol Chem 2020; 401:1167-1180. [PMID: 32386183 DOI: 10.1515/hsz-2020-0136] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Accepted: 05/04/2020] [Indexed: 11/15/2022]
Abstract
The proliferation and migration of endothelial cells are vascular events of inflammation, a process which can also potentiate the effects of promigratory factors. With the aim of investigating possible modifications in the activity of erythropoietin (Epo) in an inflammatory environment, we found that Epo at a non-promigratory concentration was capable of stimulating EA.hy926 endothelial cell migration when TNF-α was present. VCAM-1 and ICAM-1 expression, as well as adhesion of monocytic THP-1 cells to endothelial layers were also increased. Structurally modified Epo (carbamylation or N-homocysteinylation) did not exhibit these effects. The sensitizing effect of TNF-α on Epo activity was mediated by the Epo receptor. Inhibition assays targeting the PI3K/mTOR/NF-κB pathway, shared by Epo and TNF-α, show a cross-talk between both cytokines. As observed in assays using antioxidants, cell migration elicited by TNF-α + Epo depended on TNF-α-generated reactive oxygen species (ROS). ROS-mediated inactivation of protein tyrosine phosphatase 1B (PTP1B), involved in Epo signaling termination, could explain the synergistic effect of these cytokines. Our results suggest that ROS generated by inflammation inactivate PTP1B, causing the Epo signal to last longer. This mechanism, along with the cross-talk between both cytokines, could explain the sensitizing action of TNF-α on the migratory effect of Epo.
Collapse
Affiliation(s)
- María Eugenia Chamorro
- Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas, Instituto del Departamento de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Ciudad Autónoma de Buenos Aires, C1428EHA, Argentina
| | - Romina Maltaneri
- Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas, Instituto del Departamento de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Ciudad Autónoma de Buenos Aires, C1428EHA, Argentina
| | - Agustina Schiappacasse
- Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas, Instituto del Departamento de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Ciudad Autónoma de Buenos Aires, C1428EHA, Argentina
| | - Alcira Nesse
- Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas, Instituto del Departamento de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Ciudad Autónoma de Buenos Aires, C1428EHA, Argentina
| | - Daniela Vittori
- Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas, Instituto del Departamento de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Ciudad Autónoma de Buenos Aires, C1428EHA, Argentina
| |
Collapse
|
35
|
Li N, Zhang S, Xiong F, Eizirik DL, Wang CY. SUMOylation, a multifaceted regulatory mechanism in the pancreatic beta cells. Semin Cell Dev Biol 2020; 103:51-58. [PMID: 32331991 DOI: 10.1016/j.semcdb.2020.03.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2019] [Revised: 01/03/2020] [Accepted: 03/23/2020] [Indexed: 12/19/2022]
Abstract
SUMOylation is an evolutionarily conserved post-translational modification (PTM) that regulates protein subcellular localization, stability, conformation, transcription and enzymatic activity. Recent studies indicate that SUMOylation plays a key role in insulin gene expression, glucose metabolism and insulin exocytosis under physiological conditions in the pancreatic beta cells. Furthermore, SUMOylation is implicated in beta cell survival and recovery following exposure to oxidative stress, ER stress and inflammatory mediators under pathological situations. SUMOylation is closely regulated by the cellular redox status, and it collaborates with other PTMs such as phosphorylation, ubiquitination, and NEDDylation, to maintain beta cellular homeostasis. We hereby provide an update on recent findings regarding the role of SUMOylation in the regulation of pancreatic beta cell viability and function, and discuss its potential implication in beta cell senescence and RNA processing (e.g., pre-mRNA splicing and mRNA methylation). Through which we intend to provide novel insights into this fundamental biological process regarding both maintenance of beta cell viability and functionality, and beta cell dysfunction in diabetes mellitus.
Collapse
Affiliation(s)
- Na Li
- The Center for Biomedical Research, Key Laboratory of Organ Transplantation, Ministry of Education, NHC Key Laboratory of Organ Transplantation, Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Tongji Hospital, Wuhan, China
| | - Shu Zhang
- The Center for Biomedical Research, Key Laboratory of Organ Transplantation, Ministry of Education, NHC Key Laboratory of Organ Transplantation, Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Tongji Hospital, Wuhan, China
| | - Fei Xiong
- The Center for Biomedical Research, Key Laboratory of Organ Transplantation, Ministry of Education, NHC Key Laboratory of Organ Transplantation, Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Tongji Hospital, Wuhan, China
| | - Decio L Eizirik
- ULB Center for Diabetes Research, Université Libre de Bruxelles, 808 Route de Lennik, B-1070, Brussels, Belgium; Indiana Biosciences Research Institute (IBRI), Indianapolis, IN, USA.
| | - Cong-Yi Wang
- The Center for Biomedical Research, Key Laboratory of Organ Transplantation, Ministry of Education, NHC Key Laboratory of Organ Transplantation, Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Tongji Hospital, Wuhan, China.
| |
Collapse
|
36
|
Gong L, Zou Z, Huang L, Guo S, Xing D. Photobiomodulation therapy decreases free fatty acid generation and release in adipocytes to ameliorate insulin resistance in type 2 diabetes. Cell Signal 2020; 67:109491. [DOI: 10.1016/j.cellsig.2019.109491] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2019] [Revised: 12/02/2019] [Accepted: 12/02/2019] [Indexed: 12/17/2022]
|
37
|
Henderson CF, Bica I, Long FT, Irwin DD, Stull CH, Baker BW, Suarez Vega V, Taugher ZM, Fletes ED, Bartleson JM, Humphrey ML, Álvarez L, Akiyama M, Kumagai Y, Fukuto JM, Lin J. Cysteine Trisulfide Protects E. coli from Electrophile-Induced Death through the Generation of Cysteine Hydropersulfide. Chem Res Toxicol 2020; 33:678-686. [PMID: 31977195 DOI: 10.1021/acs.chemrestox.9b00494] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Hydropersulfide and polysulfide species have recently been shown to elicit a wide variety of biological and physiological responses. In this study, we examine the effects of cysteine trisulfide (Cys-SSS-Cys; also known as thiocystine) treatment on E. coli. Previous studies in mammalian cells have shown that Cys-SSS-Cys treatment results in protection from the electrophiles. Here, we show that the protective effect of Cys-SSS-Cys treatment against electrophile-induced cell death is conserved in E. coli. This protection correlates with the rapid generation of cysteine hydropersulfide (Cys-SSH) in the culture media. We go on to demonstrate that an exogenous phosphatase expressed in E. coli, containing only a single catalytic cysteine, is protected from electrophile-induced inactivation in the presence of hydropersulfides. These data together demonstrate that E. coli can utilize Cys-SSS-Cys to generate Cys-SSH and that the Cys-SSH can protect cellular thiols from reactivity with the electrophiles.
Collapse
Affiliation(s)
- Catherine F Henderson
- Department of Biology , Sonoma State University , Rohnert Park , California 94928 , United States
| | - Iris Bica
- Department of Biology , Sonoma State University , Rohnert Park , California 94928 , United States
| | - Faith T Long
- Department of Biology , Sonoma State University , Rohnert Park , California 94928 , United States
| | - Drew D Irwin
- Department of Biology , Sonoma State University , Rohnert Park , California 94928 , United States
| | - Christine H Stull
- Department of Chemistry , Sonoma State University , Rohnert Park , California 94928 , United States
| | - Blaine W Baker
- Department of Chemistry , Sonoma State University , Rohnert Park , California 94928 , United States
| | - Valeria Suarez Vega
- Department of Chemistry , Sonoma State University , Rohnert Park , California 94928 , United States
| | - Zachary M Taugher
- Department of Biology , Sonoma State University , Rohnert Park , California 94928 , United States
| | - Eliza D Fletes
- Department of Biology , Sonoma State University , Rohnert Park , California 94928 , United States
| | - Juliet M Bartleson
- Department of Biology , Sonoma State University , Rohnert Park , California 94928 , United States
| | - Megan L Humphrey
- Department of Biology , Sonoma State University , Rohnert Park , California 94928 , United States
| | - Lucía Álvarez
- Departamento de Química Inorgánica, Analítica y Química Física, Facultad de Ciencias Exactas y Naturales , Universidad de Buenos Aires , INQUIMAE-CONICET, Ciudad Universitaria, C1428EGA Buenos Aires , Argentina
| | - Masahiro Akiyama
- Environmental Biology Section, Faculty of Medicine , University of Tsukuba , Tsukuba , Ibaraki 305-8575 , Japan
| | - Yoshito Kumagai
- Environmental Biology Section, Faculty of Medicine , University of Tsukuba , Tsukuba , Ibaraki 305-8575 , Japan
| | - Jon M Fukuto
- Department of Chemistry , Sonoma State University , Rohnert Park , California 94928 , United States
| | - Joseph Lin
- Department of Biology , Sonoma State University , Rohnert Park , California 94928 , United States
| |
Collapse
|
38
|
AVE0991, a Nonpeptide Angiotensin 1-7 Receptor Agonist, Improves Glucose Metabolism in the Skeletal Muscle of Obese Zucker Rats: Possible Involvement of Prooxidant/Antioxidant Mechanisms. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:6372935. [PMID: 32089774 PMCID: PMC7008284 DOI: 10.1155/2020/6372935] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Revised: 12/22/2019] [Accepted: 12/26/2019] [Indexed: 02/07/2023]
Abstract
Angiotensin 1-7 (Ang 1-7) enhances insulin signaling and glucose transport activity in the skeletal muscle. The aim of our study was to evaluate the effect of AVE0991, a nonpeptide Mas receptor agonist, on the metabolic parameters, expression of RAS components and markers of oxidative stress, and insulin signaling in the skeletal morbidly obese rats. 33-week-old male obese Zucker rats were treated with vehicle and AVE0991 (0.5 mg/kg BW/day) via osmotic minipumps for two weeks. Gene expressions were determined by qPCR and/or Western blot analysis in musculus quadriceps. The enzymatic activities were detected flourometrically (aminopeptidase A) or by colorimetric assay kit (protein tyrosine phosphatase 1B). Administration of AVE0991 enhanced insulin signaling cascade in the skeletal muscle, reflected by improved whole-body glucose tolerance. It has been shown that reactive oxygen species (ROS) have insulin-mimetic action in muscle. The expression of renin receptor, transcription factor PLZF, and prooxidant genes was upregulated by AVE0991 accompanied by elevated expression of genes coding enzymes with antioxidant action. Our results show that AVE0991 administration activates genes involved in both ROS generation and clearance establishing a new prooxidant/antioxidant balance on a higher level, which might contribute to the improved insulin signaling pathway and glucose tolerance of obese Zucker rats.
Collapse
|
39
|
Behring JB, van der Post S, Mooradian AD, Egan MJ, Zimmerman MI, Clements JL, Bowman GR, Held JM. Spatial and temporal alterations in protein structure by EGF regulate cryptic cysteine oxidation. Sci Signal 2020; 13:eaay7315. [PMID: 31964804 PMCID: PMC7263378 DOI: 10.1126/scisignal.aay7315] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Stimulation of plasma membrane receptor tyrosine kinases (RTKs), such as the epidermal growth factor receptor (EGFR), locally increases the abundance of reactive oxygen species (ROS). These ROS then oxidize cysteine residues in proteins to potentiate downstream signaling. Spatial confinement of ROS is an important regulatory mechanism of redox signaling that enables the stimulation of different RTKs to oxidize distinct sets of downstream proteins. To uncover additional mechanisms that specify cysteines that are redox regulated by EGF stimulation, we performed time-resolved quantification of the EGF-dependent oxidation of 4200 cysteine sites in A431 cells. Fifty-one percent of cysteines were statistically significantly oxidized by EGF stimulation. Furthermore, EGF induced three distinct spatiotemporal patterns of cysteine oxidation in functionally organized protein networks, consistent with the spatial confinement model. Unexpectedly, protein crystal structure analysis and molecular dynamics simulations indicated widespread redox regulation of cryptic cysteine residues that are solvent exposed only upon changes in protein conformation. Phosphorylation and increased flux of nucleotide substrates served as two distinct modes by which EGF specified the cryptic cysteine residues that became solvent exposed and redox regulated. Because proteins that are structurally regulated by different RTKs or cellular perturbations are largely unique, these findings suggest that solvent exposure and redox regulation of cryptic cysteine residues contextually delineate redox signaling networks.
Collapse
Affiliation(s)
- Jessica B Behring
- Department of Medicine, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA
| | - Sjoerd van der Post
- Department of Medicine, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA
| | - Arshag D Mooradian
- Department of Medicine, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA
| | - Matthew J Egan
- Department of Medicine, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA
| | - Maxwell I Zimmerman
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA
| | - Jenna L Clements
- Department of Medicine, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA
| | - Gregory R Bowman
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA
| | - Jason M Held
- Department of Medicine, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA.
| |
Collapse
|
40
|
Li Y, Sun S, Tian X, Qiu JG, Jiang B, Wang LJ, Zhang CY. Dephosphorylation-directed tricyclic DNA amplification cascades for sensitive detection of protein tyrosine phosphatase. Chem Commun (Camb) 2020; 56:11581-11584. [DOI: 10.1039/d0cc04714g] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
A new fluorescence method is developed for the sensitive detection of protein tyrosine phosphatase based on dephosphorylation-directed tricyclic DNA amplification cascades.
Collapse
Affiliation(s)
- Yueying Li
- College of Chemistry
- Chemical Engineering and Materials Science
- Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong
- Key Laboratory of Molecular and Nano Probes
- Ministry of Education
| | - Shuli Sun
- College of Chemistry
- Chemical Engineering and Materials Science
- Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong
- Key Laboratory of Molecular and Nano Probes
- Ministry of Education
| | - Xiaorui Tian
- College of Chemistry
- Chemical Engineering and Materials Science
- Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong
- Key Laboratory of Molecular and Nano Probes
- Ministry of Education
| | - Jian-Ge Qiu
- Academy of Medical Sciences
- Zhengzhou University
- Zhengzhou 450000
- China
| | - BingHua Jiang
- Academy of Medical Sciences
- Zhengzhou University
- Zhengzhou 450000
- China
| | - Li-juan Wang
- College of Chemistry
- Chemical Engineering and Materials Science
- Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong
- Key Laboratory of Molecular and Nano Probes
- Ministry of Education
| | - Chun-yang Zhang
- College of Chemistry
- Chemical Engineering and Materials Science
- Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong
- Key Laboratory of Molecular and Nano Probes
- Ministry of Education
| |
Collapse
|
41
|
Henríquez-Olguín C, Boronat S, Cabello-Verrugio C, Jaimovich E, Hidalgo E, Jensen TE. The Emerging Roles of Nicotinamide Adenine Dinucleotide Phosphate Oxidase 2 in Skeletal Muscle Redox Signaling and Metabolism. Antioxid Redox Signal 2019; 31:1371-1410. [PMID: 31588777 PMCID: PMC6859696 DOI: 10.1089/ars.2018.7678] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Significance: Skeletal muscle is a crucial tissue to whole-body locomotion and metabolic health. Reactive oxygen species (ROS) have emerged as intracellular messengers participating in both physiological and pathological adaptations in skeletal muscle. A complex interplay between ROS-producing enzymes and antioxidant networks exists in different subcellular compartments of mature skeletal muscle. Recent evidence suggests that nicotinamide adenine dinucleotide phosphate (NADPH) oxidases (NOXs) are a major source of contraction- and insulin-stimulated oxidants production, but they may paradoxically also contribute to muscle insulin resistance and atrophy. Recent Advances: Pharmacological and molecular biological tools, including redox-sensitive probes and transgenic mouse models, have generated novel insights into compartmentalized redox signaling and suggested that NOX2 contributes to redox control of skeletal muscle metabolism. Critical Issues: Major outstanding questions in skeletal muscle include where NOX2 activation occurs under different conditions in health and disease, how NOX2 activation is regulated, how superoxide/hydrogen peroxide generated by NOX2 reaches the cytosol, what the signaling mediators are downstream of NOX2, and the role of NOX2 for different physiological and pathophysiological processes. Future Directions: Future research should utilize and expand the current redox-signaling toolbox to clarify the NOX2-dependent mechanisms in skeletal muscle and determine whether the proposed functions of NOX2 in cells and animal models are conserved into humans.
Collapse
Affiliation(s)
- Carlos Henríquez-Olguín
- Section of Molecular Physiology, Department of Nutrition, Exercise and Sports (NEXS), Faculty of Science, University of Copenhagen, Copenhagen, Denmark.,Muscle Cell Physiology Laboratory, Center for Exercise, Metabolism, and Cancer, Instituto de Ciencias Biomédicas, Universidad de Chile, Santiago, Chile
| | - Susanna Boronat
- Oxidative Stress and Cell Cycle Group, Universitat Pompeu Fabra, Barcelona, Spain
| | - Claudio Cabello-Verrugio
- Laboratory of Muscle Pathology, Fragility and Aging, Department of Biological Sciences, Faculty of Life Sciences, Universidad Andres Bello, Santiago, Chile.,Millennium Institute on Immunology and Immunotherapy, Santiago, Chile.,Center for the Development of Nanoscience and Nanotechnology (CEDENNA), Universidad de Santiago de Chile, Santiago, Chile
| | - Enrique Jaimovich
- Muscle Cell Physiology Laboratory, Center for Exercise, Metabolism, and Cancer, Instituto de Ciencias Biomédicas, Universidad de Chile, Santiago, Chile
| | - Elena Hidalgo
- Oxidative Stress and Cell Cycle Group, Universitat Pompeu Fabra, Barcelona, Spain
| | - Thomas E Jensen
- Section of Molecular Physiology, Department of Nutrition, Exercise and Sports (NEXS), Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
42
|
Ahn KC, Learman CR, Baker GB, Weaver CL, Chung PS, Kim HG, Song MS. Regulation of Diabetes: a Therapeutic Strategy for Alzheimer's Disease? J Korean Med Sci 2019; 34:e297. [PMID: 31779058 PMCID: PMC6882941 DOI: 10.3346/jkms.2019.34.e297] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Accepted: 10/01/2019] [Indexed: 12/20/2022] Open
Abstract
Accumulated evidence suggests that sporadic cases of Alzheimer's disease (AD) make up more than 95% of total AD patients, and diabetes has been implicated as a strong risk factor for the development of AD. Diabetes shares pathological features of AD, such as impaired insulin signaling, increased oxidative stress, increased amyloid-beta (Aβ) production, tauopathy and cerebrovascular complication. Due to shared pathologies between the two diseases, anti-diabetic drugs may be a suitable therapeutic option for AD treatment. In this article, we will discuss the well-known pathologies of AD, including Aβ plaques and tau tangles, as well as other mechanisms shared in AD and diabetes including reactive glia and the breakdown of blood brain barrier in order to evaluate the presence of any potential, indirect or direct links of pre-diabetic conditions to AD pathology. In addition, clinical evidence of high incidence of diabetic patients to the development of AD are described together with application of anti-diabetic medications to AD patients.
Collapse
Affiliation(s)
- Kee Chan Ahn
- NeuroVIS, Cheonan, Korea
- EnviroBrain, Edmonton, AB, Canada
| | - Cameron R Learman
- Chapman University Physician Assistant Studies Program, Orange, CA, USA
| | - Glen B Baker
- Department of Psychiatry, Neurochemical Research Unit, University of Alberta, Edmonton, AB, Canada
| | - Charles L Weaver
- Department of Health Sciences, Saginaw Valley State University, Saginaw, MI, USA
| | - Phil Sang Chung
- Beckman Laser Institute Korea, Faculty of Medical School, Dankook University, Cheonan, Korea
- Laser Translational Clinical Trial Center, Dankook University Hospital, Cheonan, Korea
| | - Hyung Gun Kim
- NeuroVIS, Cheonan, Korea
- Department of Pharmacology, College of Medicine, Dankook University, Cheonan, Korea
| | - Mee Sook Song
- Beckman Laser Institute Korea, Faculty of Medical School, Dankook University, Cheonan, Korea
- Laser Translational Clinical Trial Center, Dankook University Hospital, Cheonan, Korea.
| |
Collapse
|
43
|
Morris G, Puri BK, Walker AJ, Maes M, Carvalho AF, Bortolasci CC, Walder K, Berk M. Shared pathways for neuroprogression and somatoprogression in neuropsychiatric disorders. Neurosci Biobehav Rev 2019; 107:862-882. [PMID: 31545987 DOI: 10.1016/j.neubiorev.2019.09.025] [Citation(s) in RCA: 77] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Revised: 08/13/2019] [Accepted: 09/16/2019] [Indexed: 12/13/2022]
Abstract
Activated immune-inflammatory, oxidative and nitrosative stress (IO&NS) pathways and consequent mitochondrial aberrations are involved in the pathophysiology of psychiatric disorders including major depression, bipolar disorder and schizophrenia. They offer independent and shared contributions to pathways underpinning medical comorbidities including insulin resistance, metabolic syndrome, obesity and cardiovascular disease - herein conceptualized as somatoprogression. This narrative review of human studies aims to summarize relationships between IO&NS pathways, neuroprogression and somatoprogression. Activated IO&NS pathways, implicated in the neuroprogression of psychiatric disorders, affect the pathogenesis of comorbidities including insulin resistance, dyslipidaemia, obesity and hypertension, and by inference, metabolic syndrome. These conditions activate IO&NS pathways, exacerbating neuroprogression in psychiatric disorders. The processes whereby proinflammatory cytokines, nitrosative and endoplasmic reticulum stress, NADPH oxidase isoforms, PPARγ inactivation, SIRT1 deficiency and intracellular signalling pathways impact lipid metabolism and storage are considered. Through associations between body mass index, chronic neuroinflammation and FTO expression, activation of IO&NS pathways arising from somatoprogression may contribute to neuroprogression. Early evidence highlights the potential of adjuvants targeting IO&NS pathways for treating somatoprogression and neuroprogression.
Collapse
Affiliation(s)
- Gerwyn Morris
- Deakin University, IMPACT Strategic Research Centre, Barwon Health, School of Medicine, Geelong, Victoria, Australia
| | - Basant K Puri
- Department of Medicine, Hammersmith Hospital, Imperial College London, London, UK
| | - Adam J Walker
- Deakin University, IMPACT Strategic Research Centre, Barwon Health, School of Medicine, Geelong, Victoria, Australia
| | - Michael Maes
- Deakin University, IMPACT Strategic Research Centre, Barwon Health, School of Medicine, Geelong, Victoria, Australia
| | - Andre F Carvalho
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada; Centre for Addiction and Mental Health (CAMH), Toronto, ON, Canada
| | - Chiara C Bortolasci
- Deakin University, CMMR Strategic Research Centre, School of Medicine, Geelong, Victoria, Australia
| | - Ken Walder
- Deakin University, CMMR Strategic Research Centre, School of Medicine, Geelong, Victoria, Australia
| | - Michael Berk
- Deakin University, IMPACT Strategic Research Centre, Barwon Health, School of Medicine, Geelong, Victoria, Australia; Deakin University, CMMR Strategic Research Centre, School of Medicine, Geelong, Victoria, Australia; Orygen, The National Centre of Excellence in Youth Mental Health, the Department of Psychiatry and the Florey Institute for Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria, Australia.
| |
Collapse
|
44
|
Martínez-Meza S, Díaz J, Sandoval-Bórquez A, Valenzuela-Valderrama M, Díaz-Valdivia N, Rojas-Celis V, Contreras P, Huilcaman R, Ocaranza MP, Chiong M, Leyton L, Lavandero S, Quest AFG. AT2 Receptor Mediated Activation of the Tyrosine Phosphatase PTP1B Blocks Caveolin-1 Enhanced Migration, Invasion and Metastasis of Cancer Cells. Cancers (Basel) 2019; 11:cancers11091299. [PMID: 31484460 PMCID: PMC6770525 DOI: 10.3390/cancers11091299] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Revised: 08/09/2019] [Accepted: 08/16/2019] [Indexed: 12/29/2022] Open
Abstract
The renin–angiotensin receptor AT2R controls systemic blood pressure and is also suggested to modulate metastasis of cancer cells. However, in the latter case, the mechanisms involved downstream of AT2R remain to be defined. We recently described a novel Caveolin-1(CAV1)/Ras-related protein 5A (Rab5)/Ras-related C3 botulinum toxin substrate 1 (Rac1) signaling axis that promotes metastasis in melanoma, colon, and breast cancer cells. Here, we evaluated whether the anti-metastatic effect of AT2R is connected to inhibition of this pathway. We found that murine melanoma B16F10 cells expressed AT2R, while MDA-MB-231 human breast cancer cells did not. AT2R activation blocked migration, transendothelial migration, and metastasis of B16F10(cav-1) cells, and this effect was lost when AT2R was silenced. Additionally, AT2R activation reduced transendothelial migration of A375 human melanoma cells expressing CAV1. The relevance of AT2R was further underscored by showing that overexpression of the AT2R in MDA-MB-231 cells decreased migration. Moreover, AT2R activation increased non-receptor protein tyrosine phosphatase 1B (PTP1B) activity, decreased phosphorylation of CAV1 on tyrosine-14 as well as Rab5/Rac1 activity, and reduced lung metastasis of B16F10(cav-1) cells in C57BL/6 mice. Thus, AT2R activation reduces migration, invasion, and metastasis of cancer cells by PTP1B-mediated CAV1 dephosphorylation and inhibition of the CAV1/Rab5/Rac-1 pathway. In doing so, these observations open up interesting, novel therapeutic opportunities to treat metastatic cancer disease.
Collapse
Affiliation(s)
- Samuel Martínez-Meza
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Chemical & Pharmaceutical Sciences and Faculty of Medicine, University of Chile, Santiago 8380000, Chile.
- Center for Studies on Exercise, Metabolism and Cancer (CEMC), Institute of Biomedical Sciences (ICBM), Faculty of Medicine, University of Chile, Santiago 8380000, Chile.
| | - Jorge Díaz
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Chemical & Pharmaceutical Sciences and Faculty of Medicine, University of Chile, Santiago 8380000, Chile.
- Center for Studies on Exercise, Metabolism and Cancer (CEMC), Institute of Biomedical Sciences (ICBM), Faculty of Medicine, University of Chile, Santiago 8380000, Chile.
| | - Alejandra Sandoval-Bórquez
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Chemical & Pharmaceutical Sciences and Faculty of Medicine, University of Chile, Santiago 8380000, Chile.
- Center for Studies on Exercise, Metabolism and Cancer (CEMC), Institute of Biomedical Sciences (ICBM), Faculty of Medicine, University of Chile, Santiago 8380000, Chile.
| | - Manuel Valenzuela-Valderrama
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Chemical & Pharmaceutical Sciences and Faculty of Medicine, University of Chile, Santiago 8380000, Chile.
- Instituto de Innovación e Investigación en Salud, Facultad de Ciencias de la Salud, Universidad Central de Chile, Santiago 8320000, Chile.
| | - Natalia Díaz-Valdivia
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Chemical & Pharmaceutical Sciences and Faculty of Medicine, University of Chile, Santiago 8380000, Chile.
- Center for Studies on Exercise, Metabolism and Cancer (CEMC), Institute of Biomedical Sciences (ICBM), Faculty of Medicine, University of Chile, Santiago 8380000, Chile.
| | - Victoria Rojas-Celis
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Chemical & Pharmaceutical Sciences and Faculty of Medicine, University of Chile, Santiago 8380000, Chile.
- Center for Studies on Exercise, Metabolism and Cancer (CEMC), Institute of Biomedical Sciences (ICBM), Faculty of Medicine, University of Chile, Santiago 8380000, Chile.
| | - Pamela Contreras
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Chemical & Pharmaceutical Sciences and Faculty of Medicine, University of Chile, Santiago 8380000, Chile.
- Center for Studies on Exercise, Metabolism and Cancer (CEMC), Institute of Biomedical Sciences (ICBM), Faculty of Medicine, University of Chile, Santiago 8380000, Chile.
| | - Ricardo Huilcaman
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Chemical & Pharmaceutical Sciences and Faculty of Medicine, University of Chile, Santiago 8380000, Chile.
- Center for Studies on Exercise, Metabolism and Cancer (CEMC), Institute of Biomedical Sciences (ICBM), Faculty of Medicine, University of Chile, Santiago 8380000, Chile.
| | - María Paz Ocaranza
- Division of Cardiovascular Diseases, Advanced Center for Chronic Diseases (ACCDiS), Facultad de medicina, Pontificia Universidad Catolica de Chile, Santiago 8330024, Chile.
| | - Mario Chiong
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Chemical & Pharmaceutical Sciences and Faculty of Medicine, University of Chile, Santiago 8380000, Chile.
- Center for Studies on Exercise, Metabolism and Cancer (CEMC), Institute of Biomedical Sciences (ICBM), Faculty of Medicine, University of Chile, Santiago 8380000, Chile.
| | - Lisette Leyton
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Chemical & Pharmaceutical Sciences and Faculty of Medicine, University of Chile, Santiago 8380000, Chile.
- Center for Studies on Exercise, Metabolism and Cancer (CEMC), Institute of Biomedical Sciences (ICBM), Faculty of Medicine, University of Chile, Santiago 8380000, Chile.
| | - Sergio Lavandero
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Chemical & Pharmaceutical Sciences and Faculty of Medicine, University of Chile, Santiago 8380000, Chile.
- Center for Studies on Exercise, Metabolism and Cancer (CEMC), Institute of Biomedical Sciences (ICBM), Faculty of Medicine, University of Chile, Santiago 8380000, Chile.
- Cardiology Division, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas TX75390, Texas, USA.
- Corporación Centro de Estudios Científicos de las Enfermedades Crónicas (CECEC), Santiago 7860201, Chile.
| | - Andrew F G Quest
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Chemical & Pharmaceutical Sciences and Faculty of Medicine, University of Chile, Santiago 8380000, Chile.
- Center for Studies on Exercise, Metabolism and Cancer (CEMC), Institute of Biomedical Sciences (ICBM), Faculty of Medicine, University of Chile, Santiago 8380000, Chile.
- Corporación Centro de Estudios Científicos de las Enfermedades Crónicas (CECEC), Santiago 7860201, Chile.
| |
Collapse
|
45
|
Ikeda KN, Freeman M. Spatial proteomics reveal that the protein phosphatase PTP1B interacts with and may modify tyrosine phosphorylation of the rhomboid protease RHBDL4. J Biol Chem 2019; 294:11486-11497. [PMID: 31177093 PMCID: PMC6663880 DOI: 10.1074/jbc.ra118.007074] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2018] [Revised: 06/03/2019] [Indexed: 12/23/2022] Open
Abstract
Rhomboid-like proteins are evolutionarily conserved, ubiquitous polytopic membrane proteins, including the canonical rhomboid intramembrane serine proteases and also others that have lost protease activity during evolution. We still have much to learn about their cellular roles, and evidence suggests that some may have more than one function. For example, RHBDL4 (rhomboid-like protein 4) is an endoplasmic reticulum (ER)-resident protease that forms a ternary complex with ubiquitinated substrates and p97/VCP (valosin-containing protein), a major driver of ER-associated degradation (ERAD). RHBDL4 is required for ERAD of some substrates, such as the pre-T-cell receptor α chain (pTα) and has also been shown to cleave amyloid precursor protein to trigger its secretion. In another case, RHBDL4 enables the release of full-length transforming growth factor α in exosomes. Using the proximity proteomic method BioID, here we screened for proteins that interact with or are in close proximity to RHBDL4. Bioinformatics analyses revealed that BioID hits of RHBDL4 overlap with factors related to protein stress at the ER, including proteins that interact with p97/VCP. PTP1B (protein-tyrosine phosphatase nonreceptor type 1, also called PTPN1) was also identified as a potential proximity factor and interactor of RHBDL4. Analysis of RHBDL4 peptides highlighted the presence of tyrosine phosphorylation at the cytoplasmic RHBDL4 C terminus. Site-directed mutagenesis targeting these tyrosine residues revealed that their phosphorylation modifies binding of RHBDL4 to p97/VCP and Lys63-linked ubiquitinated proteins. Our work lays a critical foundation for future mechanistic studies of the roles of RHBDL4 in ERAD and other important cellular pathways.
Collapse
Affiliation(s)
- Kyojiro N Ikeda
- Dunn School of Pathology, University of Oxford, Oxford OX1 3RE, United Kingdom
| | - Matthew Freeman
- Dunn School of Pathology, University of Oxford, Oxford OX1 3RE, United Kingdom
| |
Collapse
|
46
|
Hussein UK, Park HS, Bae JS, Kim KM, Chong YJ, Kim CY, Kwon KS, Chung MJ, Lee H, Kang MJ, Moon WS, Jang KY. Expression of oxidized protein tyrosine phosphatase and γH2AX predicts poor survival of gastric carcinoma patients. BMC Cancer 2018; 18:836. [PMID: 30126387 PMCID: PMC6102926 DOI: 10.1186/s12885-018-4752-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Accepted: 08/14/2018] [Indexed: 11/25/2022] Open
Abstract
Background Oxidative stress induces various intracellular damage, which might be correlated with tumorigenesis. Accumulated oxidative stresses might inactivate protein tyrosine phosphatase (PTP) by oxidizing it, and inducing the phosphorylation of H2AX (γH2AX) in response to DNA damage. Methods We evaluated the clinical significance of the expression of oxidized-PTP and γH2AX in 169 gastric carcinomas. Results Immunohistochemical expression of nuclear oxidized-PTP, cytoplasmic oxidized-PTP, and γH2AX expression were significantly associated with each other, and their expressions predicted shorter survival of gastric carcinoma patients. In multivariate analysis, nuclear oxidized-PTP (overall survival; p < 0.001, relapse-free survival; P < 0.001) was an independent indicator of poor prognosis of gastric carcinoma patients. In addition, co-expression patterns of nuclear oxidized-PTP and γH2AX were independent indicators of poor prognosis of gastric carcinoma patients (overall survival; P < 0.001, relapse-free survival; P < 0.001). Conclusions This study suggests that oxidative stress-mediated oxidation of PTP might be involved in the progression of gastric carcinomas. In addition, this study suggests that individual and co-expression pattern of nuclear oxidized-PTP and γH2AX might be used as a prognostic marker of gastric carcinomas.
Collapse
Affiliation(s)
- Usama Khamis Hussein
- Department of Pathology, Chonbuk National University Medical School, Research Institute of Clinical Medicine of Chonbuk, National University-Biomedical Research Institute of Chonbuk National University Hospital, Jeonju, Republic of Korea.,Research Institute for Endocrine Sciences, Chonbuk National University Medical School, Jeonju, Republic of Korea.,Faculty of Science, Beni-Suef University, Beni-Suef, Egypt
| | - Ho Sung Park
- Department of Pathology, Chonbuk National University Medical School, Research Institute of Clinical Medicine of Chonbuk, National University-Biomedical Research Institute of Chonbuk National University Hospital, Jeonju, Republic of Korea.,Research Institute for Endocrine Sciences, Chonbuk National University Medical School, Jeonju, Republic of Korea
| | - Jun Sang Bae
- Department of Pathology, Chonbuk National University Medical School, Research Institute of Clinical Medicine of Chonbuk, National University-Biomedical Research Institute of Chonbuk National University Hospital, Jeonju, Republic of Korea.,Research Institute for Endocrine Sciences, Chonbuk National University Medical School, Jeonju, Republic of Korea
| | - Kyoung Min Kim
- Department of Pathology, Chonbuk National University Medical School, Research Institute of Clinical Medicine of Chonbuk, National University-Biomedical Research Institute of Chonbuk National University Hospital, Jeonju, Republic of Korea.,Research Institute for Endocrine Sciences, Chonbuk National University Medical School, Jeonju, Republic of Korea
| | - Yun Jo Chong
- Center for University-wide Research Facilities, Chonbuk National University, Jeonju, Republic of Korea
| | - Chan Young Kim
- Department of Surgery, Chonbuk National University Medical School, Jeonju, Republic of Korea
| | - Keun Sang Kwon
- Department of Preventive Medicine, Chonbuk National University Medical School, Jeonju, Republic of Korea
| | - Myoung Ja Chung
- Department of Pathology, Chonbuk National University Medical School, Research Institute of Clinical Medicine of Chonbuk, National University-Biomedical Research Institute of Chonbuk National University Hospital, Jeonju, Republic of Korea.,Research Institute for Endocrine Sciences, Chonbuk National University Medical School, Jeonju, Republic of Korea
| | - Ho Lee
- Department of Forensic Medicine, Chonbuk National University Medical School, Jeonju, Republic of Korea
| | - Myoung Jae Kang
- Department of Pathology, Chonbuk National University Medical School, Research Institute of Clinical Medicine of Chonbuk, National University-Biomedical Research Institute of Chonbuk National University Hospital, Jeonju, Republic of Korea.,Research Institute for Endocrine Sciences, Chonbuk National University Medical School, Jeonju, Republic of Korea
| | - Woo Sung Moon
- Department of Pathology, Chonbuk National University Medical School, Research Institute of Clinical Medicine of Chonbuk, National University-Biomedical Research Institute of Chonbuk National University Hospital, Jeonju, Republic of Korea.,Research Institute for Endocrine Sciences, Chonbuk National University Medical School, Jeonju, Republic of Korea
| | - Kyu Yun Jang
- Department of Pathology, Chonbuk National University Medical School, Research Institute of Clinical Medicine of Chonbuk, National University-Biomedical Research Institute of Chonbuk National University Hospital, Jeonju, Republic of Korea. .,Research Institute for Endocrine Sciences, Chonbuk National University Medical School, Jeonju, Republic of Korea.
| |
Collapse
|
47
|
Abstract
SIGNIFICANCE Hydrogen peroxide (H2O2) is a powerful effector of redox signaling. It is able to oxidize cysteine residues, metal ion centers, and lipids. Understanding H2O2-mediated signaling requires, to some extent, measurement of H2O2 level. Recent Advances: Chemically and genetically encoded fluorescent probes for the detection of H2O2 are currently the most sensitive and popular. Novel probes are constantly being developed, with the latest progress particular with boronates and genetically encoded probes. CRITICAL ISSUES All currently available probes display limitations in terms of sensitivity, local and temporal resolution, and specificity in the detection of low H2O2 concentrations. In this review, we discuss the power of fluorescent probes and the systems in which they have been successfully employed. Moreover, we recommend approaches for overcoming probe limitations and for the avoidance of artifacts. FUTURE DIRECTIONS Constant improvements will lead to the generation of probes that are not only more sensitive but also specifically tailored to individual cellular compartments. Antioxid. Redox Signal. 29, 585-602.
Collapse
Affiliation(s)
- Flávia Rezende
- Institute for Cardiovascular Physiology, Goethe-University , Frankfurt am Main, Germany
| | - Ralf P Brandes
- Institute for Cardiovascular Physiology, Goethe-University , Frankfurt am Main, Germany
| | - Katrin Schröder
- Institute for Cardiovascular Physiology, Goethe-University , Frankfurt am Main, Germany
| |
Collapse
|
48
|
Casey GR, Stains CI. Interrogating Protein Phosphatases with Chemical Activity Probes. Chemistry 2018; 24:7810-7824. [PMID: 29338103 PMCID: PMC5986605 DOI: 10.1002/chem.201705194] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Indexed: 12/30/2022]
Abstract
Protein phosphatases, while long overlooked, have recently become appreciated as drivers of both normal- and disease-associated signaling events. As a result, the spotlight is now turning torwards this enzyme family and efforts geared towards the development of modern chemical tools for studying these enzymes are well underway. This Minireview focuses on the evolution of chemical activity probes, both optical and covalent, for the study of protein phosphatases. Small-molecule probes, global monitoring of phosphatase activity through the use of covalent modifiers, and targeted fluorescence-based activity probes are discussed. We conclude with an overview of open questions in the field and highlight the potential impact of chemical tools for studying protein phosphatases.
Collapse
Affiliation(s)
- Garrett R Casey
- Department of Chemistry, University of Nebraska-Lincoln, Lincoln, NE, 68588, USA
| | - Cliff I Stains
- Department of Chemistry, University of Nebraska-Lincoln, Lincoln, NE, 68588, USA
| |
Collapse
|
49
|
Kanuri BN, Rebello SC, Pathak P, Agarwal H, Kanshana JS, Awasthi D, Gupta AP, Gayen JR, Jagavelu K, Dikshit M. Glucose and lipid metabolism alterations in liver and adipose tissue pre-dispose p47 phox knockout mice to systemic insulin resistance. Free Radic Res 2018; 52:568-582. [PMID: 29544378 DOI: 10.1080/10715762.2018.1453136] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Oxidative stress due to enhanced production or reduced scavenging of reactive oxygen species (ROS) has been associated with diet (dyslipidemia) induced obesity and insulin resistance (IR). The present study was undertaken to assess the role of p47phox in IR using wild type (WT) and p47phox-/- mice, fed with different diets (HFD, LFD or Chow). Augmented body weight, glucose intolerance and reduced insulin sensitivity were observed in p47phox-/- mice fed with 45% HFD and 10% LFD. Further, body fat and circulating lipids were increased significantly with 5 weeks LFD feeding in p47phox-/- mice, while parameters of energy homeostasis were reduced as compared with WT mice. LFD fed knockout (KO) mice showed an enhanced hepatic glycogenolysis, and reduced insulin signalling in liver and adipose tissue, while skeletal muscle tissue remained unaffected. A significant increase in hepatic lipids, adiposity, as well as expression of genes regulating lipid synthesis, breakdown and efflux were observed in LFD fed p47phox-/- mice after 5 weeks. On the other hand, mice lacking p47phox demonstrated altered glucose tolerance and tissue insulin sensitivity after 5 weeks chow feeding, while changes in body weight, respiratory exchange ratio (RER) and heat production are non-significant. Our data demonstrate that lack of p47phox is sufficient to induce IR through altered glucose and lipid utilization by the liver and adipose tissue.
Collapse
Affiliation(s)
- Babu Nageswararao Kanuri
- a Division of Pharmacology , Central Drug Research Institute, Council of Scientific and Industrial Research , Lucknow , India.,b Academy of Scientific and Innovative Research , New Delhi , India
| | - Sanjay C Rebello
- a Division of Pharmacology , Central Drug Research Institute, Council of Scientific and Industrial Research , Lucknow , India
| | - Priya Pathak
- a Division of Pharmacology , Central Drug Research Institute, Council of Scientific and Industrial Research , Lucknow , India
| | - Hobby Agarwal
- a Division of Pharmacology , Central Drug Research Institute, Council of Scientific and Industrial Research , Lucknow , India
| | - Jitendra S Kanshana
- a Division of Pharmacology , Central Drug Research Institute, Council of Scientific and Industrial Research , Lucknow , India
| | - Deepika Awasthi
- a Division of Pharmacology , Central Drug Research Institute, Council of Scientific and Industrial Research , Lucknow , India
| | - Anand P Gupta
- c Division of Pharmacokinetics and Metabolism , Central Drug Research Institute, Council of Scientific and Industrial Research , Lucknow , India
| | - Jiaur R Gayen
- c Division of Pharmacokinetics and Metabolism , Central Drug Research Institute, Council of Scientific and Industrial Research , Lucknow , India
| | - Kumaravelu Jagavelu
- a Division of Pharmacology , Central Drug Research Institute, Council of Scientific and Industrial Research , Lucknow , India
| | - Madhu Dikshit
- d Bioscience and Bioengineering , Indian Institute of Technology Jodhpur , Jodhpur , India
| |
Collapse
|
50
|
Alhawiti NM, Al Mahri S, Aziz MA, Malik SS, Mohammad S. TXNIP in Metabolic Regulation: Physiological Role and Therapeutic Outlook. Curr Drug Targets 2018; 18:1095-1103. [PMID: 28137209 PMCID: PMC5543564 DOI: 10.2174/1389450118666170130145514] [Citation(s) in RCA: 149] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Revised: 01/04/2017] [Accepted: 01/25/2017] [Indexed: 12/20/2022]
Abstract
Background & Objective: Thioredoxin-interacting protein (TXNIP) also known as thioredoxin binding protein-2 is a ubiquitously expressed protein that interacts and negatively regulates expression and function of Thioredoxin (TXN). Over the last few years, TXNIP has attracted considerable attention due to its wide-ranging functions impacting several aspects of energy metabolism. TXNIP acts as an important regulator of glucose and lipid metabolism through pleiotropic actions including regulation of β-cell function, hepatic glucose production, peripheral glucose uptake, adipogenesis, and substrate utilization. Overexpression of TXNIP in animal models has been shown to induce apoptosis of pancreatic β-cells, reduce insulin sensitivity in peripheral tissues like skeletal muscle and adipose, and decrease energy expenditure. On the contrary, TXNIP deficient animals are protected from diet induced insulin resistance and type 2 diabetes. Summary: Consequently, targeting TXNIP is thought to offer novel therapeutic opportunity and TXNIP inhibitors have the potential to become a powerful therapeutic tool for the treatment of diabetes mellitus. Here we summarize the current state of our understanding of TXNIP biology, highlight its role in metabolic regulation and raise critical questions that could help future research to exploit TXNIP as a therapeutic target.
Collapse
Affiliation(s)
- Naif Mohammad Alhawiti
- Experimental Medicine, King Abdullah International Medical Research Center (KAIMRC), King Saud Bin Abdulaziz University for Health Sciences (KSAU-HS), Ministry of National Guard Health Affairs (NGHA), Riyadh, Saudi Arabia
| | - Saeed Al Mahri
- Experimental Medicine, King Abdullah International Medical Research Center (KAIMRC), King Saud Bin Abdulaziz University for Health Sciences (KSAU-HS), Ministry of National Guard Health Affairs (NGHA), Riyadh, Saudi Arabia
| | - Mohammad Azhar Aziz
- Colorectal Cancer Research Program, King Abdullah International Medical Research Center (KAIMRC), King Saud Bin Abdulaziz University for Health Sciences (KSAU-HS), Ministry of National Guard Health Affairs (NGHA), Riyadh, Saudi Arabia
| | - Shuja Shafi Malik
- Experimental Medicine, King Abdullah International Medical Research Center (KAIMRC), King Saud Bin Abdulaziz University for Health Sciences (KSAU-HS), Ministry of National Guard Health Affairs (NGHA), Riyadh, Saudi Arabia
| | - Sameer Mohammad
- Experimental Medicine, King Abdullah International Medical Research Center (KAIMRC), King Saud Bin Abdulaziz University for Health Sciences (KSAU-HS), Ministry of National Guard Health Affairs (NGHA), Riyadh, Saudi Arabia
| |
Collapse
|