1
|
Aimeur S, Fas BA, Serfaty X, Santuz H, Sacquin-Mora S, Bizouarn T, Taly A, Baciou L. Structural profiles of the full phagocyte NADPH oxidase unveiled by combining computational biology and experimental knowledge. J Biol Chem 2024:107943. [PMID: 39481598 DOI: 10.1016/j.jbc.2024.107943] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 10/05/2024] [Accepted: 10/23/2024] [Indexed: 11/02/2024] Open
Abstract
The phagocyte NADPH oxidase (NOX2) is an enzyme, crucial for innate immune defense, producing reactive oxygen species necessary for pathogen destruction. Its activation requires the assembly of soluble proteins (p47phox, p40phox, p67phox, and Rac) with the membrane-bound flavocytochrome b558 (cytb558). We combined circular-dichroism analyses, with decades of experimental data, to filter structural models of the NADPH oxidase complex generated by the artificial intelligence program AlphaFold2 (AF2). The predicted patterns tend to closely resemble the active states of the proteins, as shown by the compact structure of the cytb558, whose dehydrogenase domain is stabilized closer to the membrane. The modeling of the interaction of p47phox with cytb558, which is the initial assembly and activation steps of the NADPH oxidase, enables us to describe how the C-terminus of p47phox interacts with the cytb558. Combining the AF2 cytb558 -p47phox model and its classical molecular dynamics simulations, we highlighted new hydrophobic lipid insertions of p47phox, particularly at residues Trp80-Phe81 of its PX domain. The AF2 models also revealed the implications of intrinsically disordered regions, such as the fragment between the PX domain and the SH3 regions of p47phox, in ensuring distant protein-protein and membrane-protein interactions. Finally, the AF2 prediction of the cytb558-Trimera model highlighted the importance of leaving Rac1 as a separate protein to reach an active state of the NADPH oxidase complex. Altogether, our step-by-step approach provides a structural model of the active complex showing how disordered regions and specific lipid and protein interactions can enable and stabilize the multi-subunit assembly.
Collapse
Affiliation(s)
- Sana Aimeur
- Institut de Chimie Physique, UMR 8000, CNRS, Université Paris Saclay, 91405 Orsay Cedex, France
| | - Burcu Aykac Fas
- Laboratoire de Biochimie Théorique, CNRS, UPR 9080, Université Paris-Cité, 75005, Paris, France
| | - Xavier Serfaty
- Institut de Chimie Physique, UMR 8000, CNRS, Université Paris Saclay, 91405 Orsay Cedex, France
| | - Hubert Santuz
- Laboratoire de Biochimie Théorique, CNRS, UPR 9080, Université Paris-Cité, 75005, Paris, France
| | - Sophie Sacquin-Mora
- Laboratoire de Biochimie Théorique, CNRS, UPR 9080, Université Paris-Cité, 75005, Paris, France
| | - Tania Bizouarn
- Institut de Chimie Physique, UMR 8000, CNRS, Université Paris Saclay, 91405 Orsay Cedex, France
| | - Antoine Taly
- Laboratoire de Biochimie Théorique, CNRS, UPR 9080, Université Paris-Cité, 75005, Paris, France
| | - Laura Baciou
- Institut de Chimie Physique, UMR 8000, CNRS, Université Paris Saclay, 91405 Orsay Cedex, France.
| |
Collapse
|
2
|
Suzuki S, Hayashi T, Egawa T. Advanced glycation end products promote ROS production via PKC/p47 phox axis in skeletal muscle cells. J Physiol Sci 2024; 74:51. [PMID: 39369187 PMCID: PMC11452979 DOI: 10.1186/s12576-024-00944-1] [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: 06/19/2024] [Accepted: 09/25/2024] [Indexed: 10/07/2024]
Abstract
Advanced glycation end products (AGEs) are risk factors for various diseases, including sarcopenia. One of the deleterious effects of AGEs is the induction of abnormal reactive oxygen species (ROS) production in skeletal muscle. However, the underlying mechanism remains poorly understood. Therefore, the aim of this study was to elucidate how AGEs induce ROS production in skeletal muscle cells. This study demonstrated that AGEs treatment promoted ROS production in myoblasts and myotubes while PKC inhibitor abolished ROS production by AGEs stimulation. Phosphorylation of p47 phox by kinases such as PKCα is required to form the Nox2 complex, which induces ROS production. In this study, AGEs treatment promoted the phosphorylation of PKCα and p47 phox in myoblasts and myotubes. Our findings suggest that AGEs promote ROS production through the phosphorylation of PKCα and p47 phox in skeletal muscle cells.
Collapse
Affiliation(s)
- Shinichiro Suzuki
- Laboratory of Molecular Adaptations to Exercise, Graduate School of Human and Environmental Studies, Kyoto University, Yoshida-Nihonmatsu-Cho, Sakyo-Ku, Kyoto, 606-8501, Japan
| | - Tatsuya Hayashi
- Laboratory of Sports and Exercise Medicine, Graduate School of Human and Environmental Studies, Kyoto University, Yoshida-Nihonmatsu-Cho, Sakyo-Ku, Kyoto, 606-8501, Japan
| | - Tatsuro Egawa
- Laboratory of Molecular Adaptations to Exercise, Graduate School of Human and Environmental Studies, Kyoto University, Yoshida-Nihonmatsu-Cho, Sakyo-Ku, Kyoto, 606-8501, Japan.
| |
Collapse
|
3
|
Silveira THR, Silva FH, Hill WG, Antunes E, de Oliveira MG. Targeting NADPH Oxidase as an Approach for Diabetic Bladder Dysfunction. Antioxidants (Basel) 2024; 13:1155. [PMID: 39456409 PMCID: PMC11504422 DOI: 10.3390/antiox13101155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 06/26/2024] [Accepted: 06/27/2024] [Indexed: 10/28/2024] Open
Abstract
Diabetic bladder dysfunction (DBD) is the most prevalent complication of diabetes mellitus (DM), affecting >50% of all patients. Currently, no specific treatment is available for this condition. In the early stages of DBD, patients typically complain of frequent urination and often have difficulty sensing when their bladders are full. Over time, bladder function deteriorates to a decompensated state in which incontinence develops. Based on studies of diabetic changes in the eye, kidney, heart, and nerves, it is now recognized that DM causes tissue damage by altering redox signaling in target organs. NADPH oxidase (NOX), whose sole function is the production of reactive oxygen species (ROS), plays a pivotal role in other well-known and bothersome diabetic complications. However, there is a substantial gap in understanding how NOX controls bladder function in health and the impact of NOX on DBD. The current review provides a thorough overview of the various NOX isoforms and their roles in bladder function and discusses the importance of further investigating the role of NOXs as a key contributor to DBD pathogenesis, either as a trigger and/or an effector and potentially as a target.
Collapse
Affiliation(s)
| | - Fábio Henrique Silva
- Laboratory of Pharmacology, São Francisco University (USF), Bragança Paulista, Sao Paulo 12916-900, Brazil; (T.H.R.S.); (F.H.S.)
| | - Warren G. Hill
- Laboratory of Voiding Dysfunction, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA;
| | - Edson Antunes
- Department of Translational Medicine, Pharmacology Area, Faculty of Medical Sciences, University of Campinas (UNICAMP), Campinas, Sao Paulo 13083-970, Brazil;
| | - Mariana G. de Oliveira
- Laboratory of Pharmacology, São Francisco University (USF), Bragança Paulista, Sao Paulo 12916-900, Brazil; (T.H.R.S.); (F.H.S.)
| |
Collapse
|
4
|
Liu H, Zhang J, Wang L, Liu H, Yu C, Li H. Regulation of the RCK1 gene on the oxidative tolerance of Saccharomyces cerevisiae. Free Radic Biol Med 2024; 225:15-23. [PMID: 39326682 DOI: 10.1016/j.freeradbiomed.2024.09.040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2024] [Revised: 09/17/2024] [Accepted: 09/23/2024] [Indexed: 09/28/2024]
Abstract
Our previous work indicated that the quorum sensing (QS) effect could regulate the oxidative tolerance of Saccharomyces cerevisiae, and QS may impact oxidative and antioxidative metabolisms of S. cerevisiae by regulating the RCK1 gene. Therefore, this work proposed a reasonable logic that RCK1 could play roles in regulating the oxidative and antioxidative metabolisms of yeast cells. The results presented here suggested that the overexpression of RCK1 has a regulatory effect on the reduction of ROS level and the promotion of oxidative tolerance of S. cerevisiae. The overexpression of RCK1 promoted the ROS generation through activating the MAPK pathway; on the other hand, RCK1-regulated antioxidative metabolism played a more significant role to realize lower ROS level and higher oxidative tolerance of S288c-RCK1 and ΔARO80-RCK1 strains. To improve the fermentation performance of yeast while circumventing metabolic burden, a recombinant strain with over time-controlled overexpression of the RCK1 gene (i.e., S288c'-RCK1 strain) derived from S288c strain was successfully constructed to achieve artificial regulation of yeast oxidative tolerance. Transcriptomics analysis was further performed on both S. cerevisiae wild-type and S288c'-RCK1 strains to identify differentially expressed genes and analyze their functional pathway classification. This work is instructive for artificially modulating the oxidative tolerance of strains to enhance the fermentation performance of yeast.
Collapse
Affiliation(s)
- Hui Liu
- School of Public Health, Jining Medical University, Jining, 272067, PR China
| | - Jiaxuan Zhang
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, 100029, PR China
| | - Lei Wang
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, 100029, PR China
| | - Huan Liu
- School of Public Health, Jining Medical University, Jining, 272067, PR China
| | - Changyuan Yu
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, 100029, PR China
| | - Hao Li
- School of Public Health, Jining Medical University, Jining, 272067, PR China.
| |
Collapse
|
5
|
Afroz R, Goodwin JE. Wnt Signaling in Atherosclerosis: Mechanisms to Therapeutic Implications. Biomedicines 2024; 12:276. [PMID: 38397878 PMCID: PMC10886882 DOI: 10.3390/biomedicines12020276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 01/08/2024] [Accepted: 01/11/2024] [Indexed: 02/25/2024] Open
Abstract
Atherosclerosis is a vascular disease in which inflammation plays a pivotal role. Receptor-mediated signaling pathways regulate vascular inflammation and the pathophysiology of atherosclerosis. Emerging evidence has revealed the role of the Wnt pathway in atherosclerosis progression. The Wnt pathway influences almost all stages of atherosclerosis progression, including endothelial dysfunction, monocyte infiltration, smooth muscle cell proliferation and migration, and plaque formation. Targeting the Wnt pathway to treat atherosclerosis represents a promising therapeutic approach that remains understudied. Blocking Wnt signaling utilizing small molecule inhibitors, recombinant proteins, and/or neutralizing antibodies ameliorates atherosclerosis in preclinical models. The Wnt pathway can be potentially manipulated through targeting Wnt ligands, receptors, co-receptors, and downstream signaling molecules. However, there are challenges associated with developing a real world therapeutic compound that targets the Wnt pathway. This review focuses on the role of Wnt signaling in atherosclerosis development, and the rationale for targeting this pathway for the treatment of atherosclerosis.
Collapse
Affiliation(s)
- Rizwana Afroz
- Department of Pediatrics, Yale University School of Medicine, New Haven, CT 06520, USA;
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Julie E. Goodwin
- Department of Pediatrics, Yale University School of Medicine, New Haven, CT 06520, USA;
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT 06520, USA
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT 06520, USA
| |
Collapse
|
6
|
Singh RK, Kumar S, Kumar S, Shukla A, Kumar N, Patel AK, Yadav LK, Kaushalendra, Antiwal M, Acharya A. Potential implications of protein kinase Cα in pathophysiological conditions and therapeutic interventions. Life Sci 2023; 330:121999. [PMID: 37536614 DOI: 10.1016/j.lfs.2023.121999] [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: 03/23/2023] [Revised: 07/31/2023] [Accepted: 07/31/2023] [Indexed: 08/05/2023]
Abstract
PKCα is a molecule with many functions that play an important role in cell survival and death to maintain cellular homeostasis. Alteration in the normal functioning of PKCα is responsible for the complicated etiology of many pathologies, including cancer, cardiovascular diseases, kidney complications, neurodegenerative diseases, diabetics, and many others. Several studies have been carried out over the years on this kinase's function, and regulation in normal physiology and pathological conditions. A lot of data with antithetical results have therefore accumulated over time to create a complex framework of physiological implications connected to the PKCα function that needs comprehensive elucidation. In light of this information, we critically analyze the multiple roles played by PKCα in basic cellular processes and their molecular mechanism during various pathological conditions. This review further discusses the current approaches to manipulating PKCα signaling amplitude in the patient's favour and proposed PKCα as a therapeutic target to reverse pathological states.
Collapse
Affiliation(s)
- Rishi Kant Singh
- Lab of Hematopoiesis and Leukemia, KSBS, Indian Institute of Technology, Delhi, New Delhi 110016, India; Cancer Immunology Lab, Department of Zoology, Banaras Hindu University, Varanasi 221005, India
| | - Sanjay Kumar
- Cancer Immunology Lab, Department of Zoology, Banaras Hindu University, Varanasi 221005, India
| | - Sandeep Kumar
- Cancer Immunology Lab, Department of Zoology, Banaras Hindu University, Varanasi 221005, India
| | - Alok Shukla
- Cancer Immunology Lab, Department of Zoology, Banaras Hindu University, Varanasi 221005, India
| | - Naveen Kumar
- Cancer Immunology Lab, Department of Zoology, Banaras Hindu University, Varanasi 221005, India
| | - Anand Kumar Patel
- Cancer Immunology Lab, Department of Zoology, Banaras Hindu University, Varanasi 221005, India
| | - Lokesh Kumar Yadav
- Cancer Immunology Lab, Department of Zoology, Banaras Hindu University, Varanasi 221005, India
| | - Kaushalendra
- Department of Zoology, Pachhunga University College Campus, Mizoram University, Aizawl 796001, India
| | - Meera Antiwal
- Institute of Medical Sciences, Banaras Hindu University, Varanasi 221005, India
| | - Arbind Acharya
- Cancer Immunology Lab, Department of Zoology, Banaras Hindu University, Varanasi 221005, India.
| |
Collapse
|
7
|
Bhosle VK, Sun C, Patel S, Ho TWW, Westman J, Ammendolia DA, Langari FM, Fine N, Toepfner N, Li Z, Sharma M, Glogauer J, Capurro MI, Jones NL, Maynes JT, Lee WL, Glogauer M, Grinstein S, Robinson LA. The chemorepellent, SLIT2, bolsters innate immunity against Staphylococcus aureus. eLife 2023; 12:e87392. [PMID: 37773612 PMCID: PMC10541174 DOI: 10.7554/elife.87392] [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: 03/03/2023] [Accepted: 09/10/2023] [Indexed: 10/01/2023] Open
Abstract
Neutrophils are essential for host defense against Staphylococcus aureus (S. aureus). The neuro-repellent, SLIT2, potently inhibits neutrophil chemotaxis, and might, therefore, be expected to impair antibacterial responses. We report here that, unexpectedly, neutrophils exposed to the N-terminal SLIT2 (N-SLIT2) fragment kill extracellular S. aureus more efficiently. N-SLIT2 amplifies reactive oxygen species production in response to the bacteria by activating p38 mitogen-activated protein kinase that in turn phosphorylates NCF1, an essential subunit of the NADPH oxidase complex. N-SLIT2 also enhances the exocytosis of neutrophil secondary granules. In a murine model of S. aureus skin and soft tissue infection (SSTI), local SLIT2 levels fall initially but increase subsequently, peaking at 3 days after infection. Of note, the neutralization of endogenous SLIT2 worsens SSTI. Temporal fluctuations in local SLIT2 levels may promote neutrophil recruitment and retention at the infection site and hasten bacterial clearance by augmenting neutrophil oxidative burst and degranulation. Collectively, these actions of SLIT2 coordinate innate immune responses to limit susceptibility to S. aureus.
Collapse
Affiliation(s)
- Vikrant K Bhosle
- Cell Biology Program, The Hospital for Sick Children Research InstituteTorontoCanada
| | - Chunxiang Sun
- Faculty of Dentistry, University of TorontoTorontoCanada
| | - Sajedabanu Patel
- Cell Biology Program, The Hospital for Sick Children Research InstituteTorontoCanada
| | - Tse Wing Winnie Ho
- The Keenan Research Centre for Biomedical Science, Unity Health TorontoTorontoCanada
- Department of Laboratory Medicine & Pathobiology, Medical Sciences Building, University of TorontoTorontoCanada
| | - Johannes Westman
- Cell Biology Program, The Hospital for Sick Children Research InstituteTorontoCanada
| | - Dustin A Ammendolia
- Cell Biology Program, The Hospital for Sick Children Research InstituteTorontoCanada
- Department of Molecular Genetics, Medical Sciences Building, University of TorontoTorontoCanada
| | - Fatemeh Mirshafiei Langari
- Program in Molecular Medicine, The Hospital for Sick Children Research InstituteTorontoCanada
- Department of Biochemistry, Medical Sciences Building, University of TorontoTorontoCanada
| | - Noah Fine
- Faculty of Dentistry, University of TorontoTorontoCanada
| | - Nicole Toepfner
- Department of Pediatrics, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität DresdenDresdenGermany
| | - Zhubing Li
- Cell Biology Program, The Hospital for Sick Children Research InstituteTorontoCanada
| | - Manraj Sharma
- Cell Biology Program, The Hospital for Sick Children Research InstituteTorontoCanada
| | - Judah Glogauer
- Cell Biology Program, The Hospital for Sick Children Research InstituteTorontoCanada
- Faculty of Dentistry, University of TorontoTorontoCanada
| | - Mariana I Capurro
- Cell Biology Program, The Hospital for Sick Children Research InstituteTorontoCanada
| | - Nicola L Jones
- Cell Biology Program, The Hospital for Sick Children Research InstituteTorontoCanada
- Division of Gastroenterology, Hepatology and Nutrition, The Hospital for Sick ChildrenTorontoCanada
- Department of Physiology, Medical Sciences Building, University of TorontoTorontoCanada
- Department of Paediatrics, Temerty Faculty of Medicine, University of TorontoTorontoCanada
| | - Jason T Maynes
- Program in Molecular Medicine, The Hospital for Sick Children Research InstituteTorontoCanada
- Department of Anesthesia and Pain Medicine, The Hospital for Sick ChildrenTorontoCanada
- Department of Anesthesiology & Pain Medicine, Temerty Faculty of Medicine, University of TorontoTorontoCanada
| | - Warren L Lee
- The Keenan Research Centre for Biomedical Science, Unity Health TorontoTorontoCanada
- Department of Laboratory Medicine & Pathobiology, Medical Sciences Building, University of TorontoTorontoCanada
- Department of Biochemistry, Medical Sciences Building, University of TorontoTorontoCanada
- Department of Medicine and Interdepartmental Division of Critical Care Medicine, Temerty Faculty of Medicine, University of TorontoTorontoCanada
| | - Michael Glogauer
- Faculty of Dentistry, University of TorontoTorontoCanada
- Department of Dental Oncology and Maxillofacial Prosthetics, University Health Network, Princess Margaret Cancer CentreTorontoCanada
- Centre for Advanced Dental Research and Care, Mount Sinai HospitalTorontoCanada
| | - Sergio Grinstein
- Cell Biology Program, The Hospital for Sick Children Research InstituteTorontoCanada
- The Keenan Research Centre for Biomedical Science, Unity Health TorontoTorontoCanada
- Department of Biochemistry, Medical Sciences Building, University of TorontoTorontoCanada
| | - Lisa A Robinson
- Cell Biology Program, The Hospital for Sick Children Research InstituteTorontoCanada
- Department of Paediatrics, Temerty Faculty of Medicine, University of TorontoTorontoCanada
- Institute of Medical Science, University of Toronto, Medical Sciences Building, University of TorontoTorontoCanada
- Division of Nephrology, The Hospital for Sick ChildrenTorontoCanada
| |
Collapse
|
8
|
Wu Y, Lu K, Lu Y, Liao J, Zhang S, Yang S, Zhao N, Dong Q, Chen L, Wu Q, Du Y. Transient receptor potential vanilloid 4 (TRPV4) in neutrophils enhances myocardial ischemia/reperfusion injury. J Leukoc Biol 2023; 114:266-279. [PMID: 37232941 DOI: 10.1093/jleuko/qiad063] [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: 10/24/2022] [Revised: 05/07/2023] [Accepted: 05/12/2023] [Indexed: 05/27/2023] Open
Abstract
The Ca2+-permeable TRPV4 cation channel is expressed in neutrophils and contributes to myocardial ischemia/reperfusion injury. Here we tested the hypotheses that TRPV4 promotes neutrophil activation and subsequently aggregates myocardial ischemia/reperfusion injury. TRPV4 protein was confirmed in neutrophils, and its function was assessed by the current and intracellular Ca2+ concentration elevations evoked by TRPV4 agonists. Furthermore, TRPV4 agonists dose-dependently promoted migration toward fMLP, reactive oxygen species production, and myeloperoxidase release, which were prevented by pretreatment with a selective TRPV4 antagonist, in neutrophils from TRPV4 knockout mice, Ca2+-free medium, or BAPTA-AM + Ca2+-free medium. Blockade of TRPV4 also inhibited the effects of commonly used neutrophil activators fMLP and PMA. Mechanically, TRPV4 regulated neutrophil activation, particularly reactive oxygen species production, by affecting PKCα, P38, and AKT via Ca2+ signaling. In addition, isolated hearts infused with neutrophils from wild-type mice showed additional myocardial ischemia/reperfusion injuries but not those infused with TRPV4 knockout. Our study reveals that TRPV4-mediated neutrophil activation enhances myocardial ischemia/reperfusion injury, and it might be a novel therapeutic target for myocardial ischemia/reperfusion injury and other neutrophil-mediated inflammatory diseases.
Collapse
Affiliation(s)
- Yuwei Wu
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, China
- Research Center of Ion Channelopathy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, China
- Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, China
| | - Kai Lu
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, China
- Research Center of Ion Channelopathy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, China
- Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, China
- Department of Cardiology, The First College of Clinical Medical Science, China Three Gorges University, 183 Yiling Avenue, Yichang 443003, China
| | - Yang Lu
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, China
- Research Center of Ion Channelopathy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, China
- Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, China
| | - Jie Liao
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, China
- Research Center of Ion Channelopathy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, China
- Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, China
| | - Shaoshao Zhang
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, China
- Research Center of Ion Channelopathy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, China
- Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, China
| | - Shuaitao Yang
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, China
- Research Center of Ion Channelopathy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, China
- Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, China
| | - Ning Zhao
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, China
- Research Center of Ion Channelopathy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, China
- Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, China
| | - Qian Dong
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, China
- Research Center of Ion Channelopathy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, China
- Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, China
| | - Lei Chen
- Department of Physiology, Nanjing Medical University, 101 Longmian Avenue, Jiangning District, Nanjing 211166, China
| | - Qiongfeng Wu
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, China
- Research Center of Ion Channelopathy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, China
- Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, China
| | - Yimei Du
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, China
- Research Center of Ion Channelopathy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, China
- Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, China
| |
Collapse
|
9
|
Bode K, Hauri-Hohl M, Jaquet V, Weyd H. Unlocking the power of NOX2: A comprehensive review on its role in immune regulation. Redox Biol 2023; 64:102795. [PMID: 37379662 DOI: 10.1016/j.redox.2023.102795] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 06/15/2023] [Accepted: 06/21/2023] [Indexed: 06/30/2023] Open
Abstract
Reactive oxygen species (ROS) are a family of highly reactive molecules with numerous, often pleiotropic functions within the cell and the organism. Due to their potential to destroy biological structures such as membranes, enzymes and organelles, ROS have long been recognized as harmful yet unavoidable by-products of cellular metabolism leading to "oxidative stress" unless counterbalanced by cellular anti-oxidative defense mechanisms. Phagocytes utilize this destructive potential of ROS released in high amounts to defend against invading pathogens. In contrast, a regulated and fine-tuned release of "signaling ROS" (sROS) provides essential intracellular second messengers to modulate central aspects of immunity, including antigen presentation, activation of antigen presenting cells (APC) as well as the APC:T cell interaction during T cell activation. This regulated release of sROS is foremost attributed to the specialized enzyme NADPH-oxidase (NOX) 2 expressed mainly in myeloid cells such as neutrophils, macrophages and dendritic cells (DC). NOX-2-derived sROS are primarily involved in immune regulation and mediate protection against autoimmunity as well as maintenance of self-tolerance. Consequently, deficiencies in NOX2 not only result in primary immune-deficiencies such as Chronic Granulomatous Disease (CGD) but also lead to auto-inflammatory diseases and autoimmunity. A comprehensive understanding of NOX2 activation and regulation will be key for successful pharmaceutical interventions of such ROS-related diseases in the future. In this review, we summarize recent progress regarding immune regulation by NOX2-derived ROS and the consequences of its deregulation on the development of immune disorders.
Collapse
Affiliation(s)
- Kevin Bode
- Section for Islet Cell & Regenerative Biology, Joslin Diabetes Center, Harvard Medical School, Boston, MA, 02215, USA
| | - Mathias Hauri-Hohl
- Division of Stem Cell Transplantation, University Children's Hospital Zurich - Eleonore Foundation & Children`s Research Center (CRC), Zurich, Switzerland
| | - Vincent Jaquet
- Department of Pathology & Immunology, Centre Médical Universitaire, Rue Michel Servet 1, 1211, Genève 4, Switzerland
| | - Heiko Weyd
- Clinical Cooperation Unit Applied Tumor Immunity D120, German Cancer Research Center, 69120, Heidelberg, Germany.
| |
Collapse
|
10
|
Role of c-Src and reactive oxygen species in cardiovascular diseases. Mol Genet Genomics 2023; 298:315-328. [PMID: 36700976 DOI: 10.1007/s00438-023-01992-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Accepted: 01/04/2023] [Indexed: 01/27/2023]
Abstract
Oxidative stress, caused by the over production of oxidants or inactivity of antioxidants, can modulate the redox state of several target proteins such as tyrosine kinases, mitogen-activated protein kinases and tyrosine phosphatases. c-Src is one such non-receptor tyrosine kinase which activates NADPH oxidases (Noxs) in response to various growth factors and shear stress. Interaction between c-Src and Noxs is influenced by cell type and primary messengers such as angiotensin II, which binds to G-protein coupled receptor and activates the intracellular signaling cascade. c-Src stimulated activation of Noxs results in elevated release of intracellular and extracellular reactive oxygen species (ROS). These ROS species disturb vascular homeostasis and cause cardiac hypertrophy, coronary artery disease, atherosclerosis and hypertension. Interaction between c-Src and ROS in the pathobiology of cardiac fibrosis is hypothesized to be influenced by cell type and stimuli. c-Src and ROS have a bidirectional relationship, thus increased ROS levels due to c-Src mediated activation of Noxs can further activate c-Src by promoting the oxidation and sulfenylation of critical cysteine residues. This review highlights the role of c-Src and ROS in mediating downstream signaling pathways underlying cardiovascular diseases. Furthermore, due to the central role of c-Src in activation of various signaling proteins involved in differentiation, migration, proliferation, and cytoskeletal reorganization of vascular cells, it is presented as therapeutic target for treating cardiovascular diseases except cardiac fibrosis.
Collapse
|
11
|
Akoumianakis I, Polkinghorne M, Antoniades C. Non-canonical WNT signalling in cardiovascular disease: mechanisms and therapeutic implications. Nat Rev Cardiol 2022; 19:783-797. [PMID: 35697779 PMCID: PMC9191761 DOI: 10.1038/s41569-022-00718-5] [Citation(s) in RCA: 50] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/25/2022] [Indexed: 12/15/2022]
Abstract
WNT signalling comprises a diverse spectrum of receptor-mediated pathways activated by a large family of WNT ligands and influencing fundamental biological processes. WNT signalling includes the β-catenin canonical pathway and the non-canonical pathways, namely the planar cell polarity and the calcium-dependent pathways. Advances over the past decade have linked non-canonical WNT signalling with key mechanisms of atherosclerosis, including oxidative stress, endothelial dysfunction, macrophage activation and vascular smooth muscle cell phenotype regulation. In addition, non-canonical WNT signalling is involved in crucial aspects of myocardial biology, from fibrosis to hypertrophy and oxidative stress. Importantly, non-canonical WNT signalling activation has complex effects in adipose tissue in the context of obesity, thereby potentially linking metabolic and vascular diseases. Tissue-specific targeting of non-canonical WNT signalling might be associated with substantial risks of off-target tumorigenesis, challenging its therapeutic potential. However, novel technologies, such as monoclonal antibodies, recombinant decoy receptors, tissue-specific gene silencing with small interfering RNAs and gene editing with CRISPR-Cas9, might enable more efficient therapeutic targeting of WNT signalling in the cardiovascular system. In this Review, we summarize the components of non-canonical WNT signalling, their links with the main mechanisms of atherosclerosis, heart failure and arrhythmias, and the rationale for targeting individual components of non-canonical WNT signalling for the treatment of cardiovascular disease.
Collapse
Affiliation(s)
- Ioannis Akoumianakis
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Murray Polkinghorne
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Charalambos Antoniades
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK.
- Acute Vascular Imaging Centre, Radcliffe Department of Medicine, University of Oxford, Oxford, UK.
| |
Collapse
|
12
|
Tikhonova IV, Grinevich AA, Kosyakova NI, Safronova VG. The effect of high temperature on kinetics of reactive species generation in patients with type 2 diabetes. Free Radic Biol Med 2022; 192:235-245. [PMID: 36198342 DOI: 10.1016/j.freeradbiomed.2022.09.031] [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: 08/12/2022] [Revised: 09/26/2022] [Accepted: 09/29/2022] [Indexed: 11/19/2022]
Abstract
The excessive amount of reactive species under chronic inflammation, which are accompanied by an increase body temperature, lead to diabetic complications. Phagocyte NADPH oxidase is the key enzyme in these processes. The role of high temperature in its regulation in diabetes is not clear. The aim was to investigate the effect of high temperature on NADPH-oxidase-dependent generation of reactive species in diabetic patients. Chemiluminescent method was applied to assess respiratory burst kinetics initiated by opsonized zymosan in blood or phorbol ester in isolated granulocytes. Analyzing ROC curves, the main predictors and changes in stages of activation of NADPH oxidase were determined. Phosphoisoforms of p47phox and p67phox were quantified by immunoblotting. Response to opsonized zymosan was lower in all subjects at 40 °C vs 37 °C, its kinetic parameters (except Tmax) were higher in blood of patients vs controls. Response rate was the main significant predictor to distinguish groups of subjects at 40 °C indicating NADPH oxidase upregulation in diabetes. Ca2+-dependent generation of reactive species by cells increased in both groups at 40 °C vs 37 °C, kinetic parameters were higher in patients. Initial phospho-p47phox level was higher in patient cells vs ones in controls. It was increased by ionomycin, phorbol ester, or 40 °C in control cells and unchanged in patient ones. Phospho-p67phox level was unchangeable in intact cells of healthy donors and patients at both temperatures. Excessive amounts of reactive species in patient cells were the consequence of granulocyte priming due to p47phox phosphorylation. Thus, high temperature decreased phagocytosis- and enhanced Ca2+-dependent generation of reactive species making the differences between controls and patients less pronounced. The effect of temperature on the generation of reactive species in blood granulocytes is associated with activity of NADPH oxidase that can be a prospective therapeutic target for pathologies accompanied by inflammation including type 2 diabetes.
Collapse
Affiliation(s)
- Irina V Tikhonova
- Institute of Cell Biophysics of Russian Academy of Sciences, Institutskaya st., 3 Pushchino, 142290, Russia.
| | - Andrei A Grinevich
- Institute of Cell Biophysics of Russian Academy of Sciences, Institutskaya st., 3 Pushchino, 142290, Russia
| | - Ninel I Kosyakova
- Hospital of Pushchino Scientific Centre of Russian Academy of Sciences, Institutskaya st., 1 Pushchino, 142290, Russia
| | - Valentina G Safronova
- Institute of Cell Biophysics of Russian Academy of Sciences, Institutskaya st., 3 Pushchino, 142290, Russia
| |
Collapse
|
13
|
Guo CL. Self-Sustained Regulation or Self-Perpetuating Dysregulation: ROS-dependent HIF-YAP-Notch Signaling as a Double-Edged Sword on Stem Cell Physiology and Tumorigenesis. Front Cell Dev Biol 2022; 10:862791. [PMID: 35774228 PMCID: PMC9237464 DOI: 10.3389/fcell.2022.862791] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Accepted: 04/29/2022] [Indexed: 12/19/2022] Open
Abstract
Organ development, homeostasis, and repair often rely on bidirectional, self-organized cell-niche interactions, through which cells select cell fate, such as stem cell self-renewal and differentiation. The niche contains multiplexed chemical and mechanical factors. How cells interpret niche structural information such as the 3D topology of organs and integrate with multiplexed mechano-chemical signals is an open and active research field. Among all the niche factors, reactive oxygen species (ROS) have recently gained growing interest. Once considered harmful, ROS are now recognized as an important niche factor in the regulation of tissue mechanics and topology through, for example, the HIF-YAP-Notch signaling pathways. These pathways are not only involved in the regulation of stem cell physiology but also associated with inflammation, neurological disorder, aging, tumorigenesis, and the regulation of the immune checkpoint molecule PD-L1. Positive feedback circuits have been identified in the interplay of ROS and HIF-YAP-Notch signaling, leading to the possibility that under aberrant conditions, self-organized, ROS-dependent physiological regulations can be switched to self-perpetuating dysregulation, making ROS a double-edged sword at the interface of stem cell physiology and tumorigenesis. In this review, we discuss the recent findings on how ROS and tissue mechanics affect YAP-HIF-Notch-PD-L1 signaling, hoping that the knowledge can be used to design strategies for stem cell-based and ROS-targeting therapy and tissue engineering.
Collapse
Affiliation(s)
- Chin-Lin Guo
- Institute of Physics, Academia Sinica, Taipei, Taiwan
| |
Collapse
|
14
|
Parny M, Coste A, Aubouy A, Rahabi M, Prat M, Pipy B, Treilhou M. Differential immunomodulatory effects of six pesticides of different chemical classes on human monocyte-derived macrophage functions. Food Chem Toxicol 2022; 163:112992. [PMID: 35395341 DOI: 10.1016/j.fct.2022.112992] [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: 12/10/2021] [Revised: 03/04/2022] [Accepted: 04/03/2022] [Indexed: 01/19/2023]
Abstract
Exposure to pesticides through eyes, skin, ingestion and inhalation may affects human health by interfering with immune cells, such as macrophages. We evaluated, in vitro, the effect of six pesticides widely used in apple arboriculture on the functions of human monocyte-derived macrophages (hMDMs). hMDMs were cultured for 4 or 24 h with or without pesticides (0.01, 0.1, 1, 10 μmol.L-1). We showed that chlorpyrifos, thiacloprid, thiophanate, boscalid, and captan had little toxic effect at the tested concentrations, while dithianon had low-cytotoxicity at 10 μmol.L-1. While boscalid showed no effect on hMDMs function, thiophanate (0.01 μmol.L-1) stimulated with TPA and thiacloprid (1, 10 μmol.L-1) stimulated with zymosan activated ROS production. Chlorpyrifos, dithianon, and captan inhibited ROS production and TNF-α, IL-1β pro-inflammatory cytokines. We established that dithianon (0.01-1 μmol.L-1) and captan (0.1, 1 μmol.L-1) induced mRNA expression of NQO1 and HMOX1 antioxidant enzymes. Dithianon also induced the mRNA expression of catalase, superoxide dismutase-2 at 10 μmol.L-1. Together, these results show that exposure to chlorpyrifos, dithianon, and captan induce immunomodulatory effects that may influence the disease fighting properties of monocytes/macrophages while pesticides such as thiacloprid, thiophanate and boscalid have little influence.
Collapse
Affiliation(s)
- Melissa Parny
- EA7417, BTSB, Université Fédérale Toulouse Midi-Pyrénées, INU Champollion, Albi, France; PHARMADEV UMR 152, Institut de Recherche pour le Développement (IRD), Université Paul Sabatier Toulouse 3, Toulouse, France.
| | - Agnès Coste
- PHARMADEV UMR 152, Institut de Recherche pour le Développement (IRD), Université Paul Sabatier Toulouse 3, Toulouse, France.
| | - Agnès Aubouy
- PHARMADEV UMR 152, Institut de Recherche pour le Développement (IRD), Université Paul Sabatier Toulouse 3, Toulouse, France.
| | - Mouna Rahabi
- PHARMADEV UMR 152, Institut de Recherche pour le Développement (IRD), Université Paul Sabatier Toulouse 3, Toulouse, France.
| | - Melissa Prat
- PHARMADEV UMR 152, Institut de Recherche pour le Développement (IRD), Université Paul Sabatier Toulouse 3, Toulouse, France.
| | - Bernard Pipy
- PHARMADEV UMR 152, Institut de Recherche pour le Développement (IRD), Université Paul Sabatier Toulouse 3, Toulouse, France.
| | - Michel Treilhou
- EA7417, BTSB, Université Fédérale Toulouse Midi-Pyrénées, INU Champollion, Albi, France.
| |
Collapse
|
15
|
Gupta V, Garg A, Tomar R, Arora MK. Oxidative Stress: Meeting Multiple Targets in Pathogenesis of Vascular Endothelial Dysfunction. Curr Drug Targets 2022; 23:902-912. [PMID: 35240954 DOI: 10.2174/1389450123666220303090413] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 10/20/2021] [Accepted: 12/31/2021] [Indexed: 11/22/2022]
Abstract
Vascular endothelium is the innermost lining of blood vessels, which maintains vasoconstriction and vasodilation. Loss of vascular tone is a hallmark for cardiovascular disorders. Though there are numerous factors, such as over activation of renin angiotensin aldosterone system, kinases, growth factors, etc. play crucial role in induction and progression of vascular abrasion. Interestingly, dysregulation of these pathways either enhances the intensity of oxidative stress, or these pathways are affected by oxidative stress. Thus, oxidative stress has been considered a key culprit in the progression of vascular endothelial dysfunction. Oxidative stress induced by reactive oxygen and nitrogen species causes abnormal gene expression, alteration in signal transduction, and the activation of pathways leading to induction and progression of vascular injury. In addition, numerous antioxidants have been noted to possess promising therapeutic potential in preventing the development of vascular endothelial dysfunction. Therefore, we have focused on current perspectives in oxidative stress signalling to evaluate common biological processes whereby oxidative stress plays a crucial role in the progression of vascular endothelial dysfunction.
Collapse
Affiliation(s)
- Vardan Gupta
- Department of Pharmacology, KIET School of Pharmacy, Ghaziabad-250005, Uttar Pradesh, India
| | - Anchal Garg
- Department of Pharmacology, KIET School of Pharmacy, Ghaziabad-250005, Uttar Pradesh, India
| | - Ritu Tomar
- School of Pharmaceutical and Population Health Informatics, DIT University, Dehradun-248009, Uttarakhand, India
| | - Mandeep Kumar Arora
- School of Pharmaceutical and Population Health Informatics, DIT University, Dehradun-248009, Uttarakhand, India
| |
Collapse
|
16
|
Prevention of Lipotoxicity in Pancreatic Islets with Gammahydroxybutyrate. Cells 2022; 11:cells11030545. [PMID: 35159354 PMCID: PMC8833960 DOI: 10.3390/cells11030545] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 01/28/2022] [Accepted: 01/30/2022] [Indexed: 02/06/2023] Open
Abstract
Oxidative stress caused by the exposure of pancreatic ß-cells to high levels of fatty acids impairs insulin secretion. This lipotoxicity is thought to play an important role in ß-cell failure in type 2 diabetes and can be prevented by antioxidants. Gamma-hydroxybutyrate (GHB), an endogenous antioxidant and energy source, has previously been shown to protect mice from streptozotocin and alloxan-induced diabetes; both compounds are generators of oxidative stress and yield models of type-1 diabetes. We sought to determine whether GHB could protect mouse islets from lipotoxicity caused by palmitate, a model relevant to type 2 diabetes. We found that GHB prevented the generation of palmitate-induced reactive oxygen species and the associated lipotoxic inhibition of glucose-stimulated insulin secretion while increasing the NADPH/NADP+ ratio. GHB may owe its antioxidant and insulin secretory effects to the formation of NADPH.
Collapse
|
17
|
Sun X, Xiao H, Li S, Chen R, Lin Z, Yang Y, Chen Z, Deng L, Huang H. Connexin32 ameliorates epithelial-to-mesenchymal-transition in diabetic renal tubular via inhibiting NOX4. Pharmacol Res 2022; 176:106084. [PMID: 35051590 DOI: 10.1016/j.phrs.2022.106084] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 12/31/2021] [Accepted: 01/11/2022] [Indexed: 12/22/2022]
Abstract
Renal tubulointerstitial fibrosis (RIF), characterized by epithelial-to-mesenchymal transition (EMT) of renal tubular epithelial cells (TECs), is the main cause of diabetic renal fibrosis. Oxidative stress plays a pivotal role in the development of diabetic RIF. Connexin32 (Cx32), prominently expressed in renal TECs, has emerged as an important player in the regulation of oxidative stress. However, the role of Cx32 in diabetic RIF has not been explored yet. Here, we showed that adenovirus-mediated Cx32 overexpression suppressed EMT to ameliorate RIF and renal function in STZ-induced diabetic mice, while knockout (KO) of Cx32 exacerbated RIF in diabetic mice. Moreover, overexpression of Cx32 inhibited EMT and the production of extra cellular matrix (ECM) in high glucose (HG) induced NRK-52E cells, whereas knockdown of Cx32 showed the opposite effects. Furthermore, we showed that NOX4, the main source of ROS in renal tubular, was down-regulated by Cx32. Mechanistically, Cx32 down-regulated the expression of PKC alpha in a carboxyl-terminal-dependent manner, thereby inhibiting the phosphorylation at Thr147 of p22phox triggered by PKC alpha, which ultimately repressed the formation of the p22phox-NOX4 complex to reduce the protein level of NOX4. Thus, we establish Cx32 as a novel target and confirm the protection mechanism in RIF.
Collapse
Affiliation(s)
- Xiaohong Sun
- Laboratory of Pharmacology & Toxicology, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Haiming Xiao
- Laboratory of Pharmacology & Toxicology, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Shanshan Li
- Laboratory of Pharmacology & Toxicology, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Rui Chen
- Laboratory of Pharmacology & Toxicology, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Zeyuan Lin
- Laboratory of Pharmacology & Toxicology, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Yan Yang
- Laboratory of Pharmacology & Toxicology, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Zhiquan Chen
- Department of Pharmacology, School of Pharmacy, Guangxi Medical University, Nanning 530021, China.
| | - Li Deng
- College of Traditional Chinese Medicine, Jinan University, Guangzhou 510632, China.
| | - Heqing Huang
- Laboratory of Pharmacology & Toxicology, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China.
| |
Collapse
|
18
|
Huang K, Luo X, Zhong Y, Deng L, Feng J. New insights into the role of melatonin in diabetic cardiomyopathy. Pharmacol Res Perspect 2022; 10:e00904. [PMID: 35005848 PMCID: PMC8929360 DOI: 10.1002/prp2.904] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Accepted: 11/25/2021] [Indexed: 12/13/2022] Open
Abstract
Diabetic cardiovascular complications and impaired cardiac function are considered to be the main causes of death in diabetic patients worldwide, especially patients with type 2 diabetes mellitus (T2DM). An increasing number of studies have shown that melatonin, as the main product secreted by the pineal gland, plays a vital role in the occurrence and development of diabetes. Melatonin improves myocardial cell metabolism, reduces vascular endothelial cell death, reverses microcirculation disorders, reduces myocardial fibrosis, reduces oxidative and endoplasmic reticulum stress, regulates cell autophagy and apoptosis, and improves mitochondrial function, all of which are the characteristics of diabetic cardiomyopathy (DCM). This review focuses on the role of melatonin in DCM. We also discuss new molecular findings that might facilitate a better understanding of the underlying mechanism. Finally, we propose potential new therapeutic strategies for patients with T2DM.
Collapse
Affiliation(s)
- Keming Huang
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou, China.,Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China
| | - Xianling Luo
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou, China.,Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China
| | - Yi Zhong
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou, China.,Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China
| | - Li Deng
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou, China.,Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China
| | - Jian Feng
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou, China.,Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China
| |
Collapse
|
19
|
Romero-Pinedo S, Barros DIR, Ruiz-Magaña MJ, Maganto-García E, Moreno de Lara L, Abadía-Molina F, Terhorst C, Abadía-Molina AC. SLAMF8 Downregulates Mouse Macrophage Microbicidal Mechanisms via PI3K Pathways. Front Immunol 2022; 13:910112. [PMID: 35837407 PMCID: PMC9273976 DOI: 10.3389/fimmu.2022.910112] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 05/11/2022] [Indexed: 11/13/2022] Open
Abstract
Signaling lymphocytic activation molecule family 8 (SLAMF8) is involved in the negative modulation of NADPH oxidase activation. However, the impact of SLAMF8 downregulation on macrophage functionality and the microbicide mechanism remains elusive. To study this in depth, we first analyzed NADPH oxidase activation pathways in wild-type and SLAMF8-deficient macrophages upon different stimulus. Herein, we describe increased phosphorylation of the Erk1/2 and p38 MAP kinases, as well as increased phosphorylation of NADPH oxidase subunits in SLAMF8-deficient macrophages. Furthermore, using specific inhibitors, we observed that specific PI3K inhibition decreased the differences observed between wild-type and SLAMF8-deficient macrophages, stimulated with either PMA, LPS, or Salmonella typhimurium infection. Consequently, SLAMF8-deficient macrophages also showed increased recruitment of small GTPases such as Rab5 and Rab7, and the p47phox subunit to cytoplasmic Salmonella, suggesting an impairment of Salmonella-containing vacuole (SCV) progression in SLAMF8-deficient macrophages. Enhanced iNOS activation, NO production, and IL-6 expression were also observed in the absence of SLAMF8 upon Salmonella infection, either in vivo or in vitro, while overexpression of SLAMF8 in RAW264.7 macrophages showed the opposite phenotype. In addition, SLAMF8-deficient macrophages showed increased activation of Src kinases and reduced SHP-1 phosphate levels upon IFNγ and Salmonella stimuli in comparison to wild-type macrophages. In agreement with in vitro results, Salmonella clearance was augmented in SLAMF8-deficient mice compared to that in wild-type mice. Therefore, in conclusion, SLAMF8 intervention upon bacterial infection downregulates mouse macrophage activation, and confirmed that SLAMF8 receptor could be a potential therapeutic target for the treatment of severe or unresolved inflammatory conditions.
Collapse
Affiliation(s)
- Salvador Romero-Pinedo
- Unidad de Inmunología, Instituto de Biopatología y Medicina Regenerativa (IBIMER), Centro de Investigación Biomédica (CIBM), Universidad de Granada, Granada, Spain
| | - Domingo I Rojas Barros
- Unidad de Inmunología, Instituto de Biopatología y Medicina Regenerativa (IBIMER), Centro de Investigación Biomédica (CIBM), Universidad de Granada, Granada, Spain
| | - María José Ruiz-Magaña
- Unidad de Inmunología, Instituto de Biopatología y Medicina Regenerativa (IBIMER), Centro de Investigación Biomédica (CIBM), Universidad de Granada, Granada, Spain
| | - Elena Maganto-García
- Centro de Biología Molecular "Severo Ochoa" Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid (CSIC-UAM), Universidad Autónoma de Madrid, Cantoblanco, Madrid, Spain
| | - Laura Moreno de Lara
- Unidad de Inmunología, Instituto de Biopatología y Medicina Regenerativa (IBIMER), Centro de Investigación Biomédica (CIBM), Universidad de Granada, Granada, Spain
| | - Francisco Abadía-Molina
- Departamento de Biología Celular, Facultad de Ciencias, Universidad de Granada, Granada, Spain.,Instituto de Nutrición Y Tecnología de los Alimentos "José Mataix", (INYTIA), Centro de Investigación Biomédica (CIBM), Universidad de Granada, Granada, Spain
| | - Cox Terhorst
- Division of Immunology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| | - Ana C Abadía-Molina
- Unidad de Inmunología, Instituto de Biopatología y Medicina Regenerativa (IBIMER), Centro de Investigación Biomédica (CIBM), Universidad de Granada, Granada, Spain.,Departamento de Bioqu´ımica y Biolog´ıa Molecular III e Inmunolog´ıa, Facultad de Medicina, Universidad de Granada, Granada, Spain
| |
Collapse
|
20
|
Uthman L, Li X, Baartscheer A, Schumacher CA, Baumgart P, Hermanides J, Preckel B, Hollmann MW, Coronel R, Zuurbier CJ, Weber NC. Empagliflozin reduces oxidative stress through inhibition of the novel inflammation/NHE/[Na +] c/ROS-pathway in human endothelial cells. Biomed Pharmacother 2021; 146:112515. [PMID: 34896968 DOI: 10.1016/j.biopha.2021.112515] [Citation(s) in RCA: 55] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 11/30/2021] [Accepted: 12/06/2021] [Indexed: 02/07/2023] Open
Abstract
Inflammation causing oxidative stress in endothelial cells contributes to heart failure development. Sodium/glucose cotransporter 2 inhibitors (SGLT2i's) were shown to reduce heart failure hospitalization and oxidative stress. However, how inflammation causes oxidative stress in endothelial cells, and how SGLT2i's can reduce this is unknown. Here we hypothesized that 1) TNF-α activates the Na+/H+ exchanger (NHE) and raises cytoplasmatic Na+ ([Na+]c), 2) increased [Na+]c causes reactive oxygen species (ROS) production, and 3) empagliflozin (EMPA) reduces inflammation-induced ROS through NHE inhibition and lowering of [Na+]c in human endothelial cells. Human umbilical vein endothelial cells (HUVECs) and human coronary artery endothelial cells (HCAECs) were incubated with vehicle (V), 10 ng/ml TNF-α, 1 µM EMPA or the NHE inhibitor Cariporide (CARI, 10 µM) and NHE activity, intracellular [Na+]c and ROS were analyzed. TNF-α enhanced NHE activity in HCAECs and HUVECs by 92% (p < 0.01) and 51% (p < 0.05), respectively, and increased [Na+]c from 8.2 ± 1.6 to 11.2 ± 0.1 mM (p < 0.05) in HCAECs. Increasing [Na+]c by ouabain elevated ROS generation in both HCAECs and HUVECs. EMPA inhibited NHE activity in HCAECs and in HUVECs. EMPA concomitantly lowered [Na+]c in both cell types. In both cell types, TNF α-induced ROS was lowered by EMPA or CARI, with no further ROS lowering by EMPA in the presence of CARI, indicating EMPA attenuated ROS through NHE inhibition. In conclusion, inflammation induces oxidative stress in human endothelial cells through NHE activation causing elevations in [Na+]c, a process that is inhibited by EMPA through NHE inhibition.
Collapse
Affiliation(s)
- Laween Uthman
- Department of Anesthesiology, Laboratory of Experimental Intensive Care and Anesthesiology (L.E.I.C.A.), Amsterdam UMC, location Academic Medical Centre (AMC), University of Amsterdam, Cardiovascular Sciences, Amsterdam, The Netherlands; Department of Physiology and Cardiology, Radboud Institute for Molecular Life Sciences, Radboud UMC, Nijmegen, The Netherlands
| | - Xiaoling Li
- Department of Anesthesiology, Laboratory of Experimental Intensive Care and Anesthesiology (L.E.I.C.A.), Amsterdam UMC, location Academic Medical Centre (AMC), University of Amsterdam, Cardiovascular Sciences, Amsterdam, The Netherlands
| | - Antonius Baartscheer
- Department of Experimental Cardiology, Amsterdam UMC, location Academic Medical Centre (AMC), University of Amsterdam, Cardiovascular Sciences, Amsterdam, The Netherlands
| | - Cees A Schumacher
- Department of Experimental Cardiology, Amsterdam UMC, location Academic Medical Centre (AMC), University of Amsterdam, Cardiovascular Sciences, Amsterdam, The Netherlands
| | - Patricia Baumgart
- Department of Anesthesiology, Laboratory of Experimental Intensive Care and Anesthesiology (L.E.I.C.A.), Amsterdam UMC, location Academic Medical Centre (AMC), University of Amsterdam, Cardiovascular Sciences, Amsterdam, The Netherlands
| | - Jeroen Hermanides
- Department of Anesthesiology, Laboratory of Experimental Intensive Care and Anesthesiology (L.E.I.C.A.), Amsterdam UMC, location Academic Medical Centre (AMC), University of Amsterdam, Cardiovascular Sciences, Amsterdam, The Netherlands
| | - Benedikt Preckel
- Department of Anesthesiology, Laboratory of Experimental Intensive Care and Anesthesiology (L.E.I.C.A.), Amsterdam UMC, location Academic Medical Centre (AMC), University of Amsterdam, Cardiovascular Sciences, Amsterdam, The Netherlands
| | - Markus W Hollmann
- Department of Anesthesiology, Laboratory of Experimental Intensive Care and Anesthesiology (L.E.I.C.A.), Amsterdam UMC, location Academic Medical Centre (AMC), University of Amsterdam, Cardiovascular Sciences, Amsterdam, The Netherlands
| | - Ruben Coronel
- Department of Experimental Cardiology, Amsterdam UMC, location Academic Medical Centre (AMC), University of Amsterdam, Cardiovascular Sciences, Amsterdam, The Netherlands
| | - Coert J Zuurbier
- Department of Anesthesiology, Laboratory of Experimental Intensive Care and Anesthesiology (L.E.I.C.A.), Amsterdam UMC, location Academic Medical Centre (AMC), University of Amsterdam, Cardiovascular Sciences, Amsterdam, The Netherlands.
| | - Nina C Weber
- Department of Anesthesiology, Laboratory of Experimental Intensive Care and Anesthesiology (L.E.I.C.A.), Amsterdam UMC, location Academic Medical Centre (AMC), University of Amsterdam, Cardiovascular Sciences, Amsterdam, The Netherlands
| |
Collapse
|
21
|
Filina Y, Gabdoulkhakova A, Rizvanov A, Safronova V. MAP kinases in regulation of NOX activity stimulated through two types of formyl peptide receptors in murine bone marrow granulocytes. Cell Signal 2021; 90:110205. [PMID: 34826588 DOI: 10.1016/j.cellsig.2021.110205] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 10/29/2021] [Accepted: 11/19/2021] [Indexed: 11/16/2022]
Abstract
The functional activity of the phagocytes, as well as the development and resolution of the inflammation, is determined by formylpeptide receptors (FPRs) signaling. There is a growing data on the signaling pathways from two major types of formylpeptide receptors, FPR1 and FPR2, which could be activated by different sets of ligands to provide certain defense functions. Generation of reactive oxygen species (ROS) by the membrane enzyme NADPH oxidase is the most important among them. One of the most studied and significant mechanism for the regulation of activity of NADPH oxidase is phosphorylation by a variety of kinases, including MAP kinases. The question arose whether the role of MAPKs differ in the activation of NADPH oxidase through FPR1 and FPR2. We have studied Fpr1- and Fpr2-induced phosphorylation of p38, ERK, and JNK kinases and their role in the activation of the respiratory burst in isolated mice bone marrow granulocytes. Data has shown distinct patterns of MAP kinase activity for Fpr1 and Fpr2: JNK was involved in both Fpr1 and Fpr2 mediated activation of ROS production, while p38 MAPK and ERK were involved in Fpr1 induced ROS generation only.
Collapse
Affiliation(s)
- Yuliya Filina
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russian Federation.
| | - Aida Gabdoulkhakova
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russian Federation; Kazan State Medical Academy, Federal State Budgetary Educational Institution of Further Professional Education "Russian Medical Academy of Continuous Professional Education" of the Ministry of Healthcare of the Russian Federation, Kazan, Russian Federation
| | - Albert Rizvanov
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russian Federation
| | - Valentina Safronova
- Institute of Cell Biophysics of Russian Academy of Sciences, Pushchino, Russian Federation
| |
Collapse
|
22
|
Villegas L, Nørremølle A, Freude K, Vilhardt F. Nicotinamide Adenine Dinucleotide Phosphate Oxidases Are Everywhere in Brain Disease, but Not in Huntington's Disease? Front Aging Neurosci 2021; 13:736734. [PMID: 34803655 PMCID: PMC8602359 DOI: 10.3389/fnagi.2021.736734] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Accepted: 10/08/2021] [Indexed: 11/13/2022] Open
Abstract
Huntington's disease (HD) is an inherited neurodegenerative disorder characterized by neuronal loss and tissue atrophy mainly in the striatum and cortex. In the early stages of the disease, impairment of neuronal function, synaptic dysfunction and white matter loss precedes neuronal death itself. Relative to other neurodegenerative diseases such as Alzheimer's and Parkinson's disease and Amyotrophic Lateral Sclerosis, where the effects of either microglia or NADPH oxidases (NOXs) are recognized as important contributors to disease pathogenesis and progression, there is a pronounced lack of information in HD. This information void contrasts with evidence from human HD patients where blood monocytes and microglia are activated well before HD clinical symptoms (PET scans), and the clear signs of oxidative stress and inflammation in post mortem HD brain. Habitually, NOX activity and oxidative stress in the central nervous system (CNS) are equated with microglia, but research of the last two decades has carved out important roles for NOX enzyme function in neurons. Here, we will convey recent information about the function of NOX enzymes in neurons, and contemplate on putative roles of neuronal NOX in HD. We will focus on NOX-produced reactive oxygen species (ROS) as redox signaling molecules in/among neurons, and the specific roles of NOXs in important processes such as neurogenesis and lineage specification, neurite outgrowth and growth cone dynamics, and synaptic plasticity where NMDAR-dependent signaling, and long-term depression/potentiation are redox-regulated phenomena. HD animal models and induced pluripotent stem cell (iPSC) studies have made it clear that the very same physiological processes are also affected in HD, and we will speculate on possible roles for NOX in the pathogenesis and development of disease. Finally, we also take into account the limited information on microglia in HD and relate this to any contribution of NOX enzymes.
Collapse
Affiliation(s)
- Luisana Villegas
- Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Anne Nørremølle
- Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Kristine Freude
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Frederik Vilhardt
- Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
23
|
Chauhan-Puri AK, Lee KH, Magoski NS. Hydrogen peroxide and phosphoinositide metabolites synergistically regulate a cation current to influence neuroendocrine cell bursting. J Physiol 2021; 599:5281-5300. [PMID: 34676545 DOI: 10.1113/jp282302] [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: 08/27/2021] [Accepted: 10/07/2021] [Indexed: 11/08/2022] Open
Abstract
In various neurons, including neuroendocrine cells, non-selective cation channels elicit plateau potentials and persistent firing. Reproduction in the marine snail Aplysia californica is initiated when the neuroendocrine bag cell neurons undergo an afterdischarge, that is, a prolonged period of enhanced excitability and spiking during which egg-laying hormone is released into the blood. The afterdischarge is associated with both the production of hydrogen peroxide (H2 O2 ) and activation of phospholipase C (PLC), which hydrolyses phosphatidylinositol-4,5-bisphosphate into diacylglycerol (DAG) and inositol trisphosphate (IP3 ). We previously demonstrated that H2 O2 gates a voltage-dependent cation current and evokes spiking in bag cell neurons. The present study tests if DAG and IP3 impact the H2 O2 -induced current and excitability. In whole-cell voltage-clamped cultured bag cell neurons, bath-application of 1-oleoyl-2-acetyl-sn-glycerol (OAG), a DAG analogue, enhanced the H2 O2 -induced current, which was amplified by the inclusion of IP3 in the pipette. A similar outcome was produced by the PLC activator, N-(3-trifluoromethylphenyl)-2,4,6-trimethylbenzenesulfonamide. In current-clamp, OAG or OAG plus IP3 , elevated the frequency of H2 O2 -induced bursting. PKC is also triggered during the afterdischarge; when PKC was stimulated with phorbol 12-myristate 13-acetate, it caused a voltage-dependent inward current with a reversal potential similar to the H2 O2 -induced current. Furthermore, PKC activation followed by H2 O2 reduced the onset latency and increased the duration of action potential firing. Finally, inhibiting nicotinamide adenine dinucleotide phosphate oxidase with 3-benzyl-7-(2-benzoxazolyl)thio-1,2,3-triazolo[4,5-d]pyrimidine diminished evoked bursting in isolated bag cell neuron clusters. These results suggest that reactive oxygen species and phosphoinostide metabolites may synergize and contribute to reproductive behaviour by promoting neuroendocrine cell firing. KEY POINTS: Aplysia bag cell neurons secrete reproductive hormone during a lengthy burst of action potentials, known as the afterdischarge. During the afterdischarge, phospholipase C (PLC) hydrolyses phosphatidylinositol-4,5-bisphosphate into diacylglycerol (DAG) and inositol trisphosphate (IP3 ). Subsequent activation of protein kinase C (PKC) leads to H2 O2 production. H2 O2 evokes a voltage-dependent inward current and action potential firing. Both a DAG analogue, 1-oleoyl-2-acetyl-sn-glycerol (OAG), and IP3 enhance the H2 O2 -induced current, which is mimicked by the PLC activator, N-(3-trifluoromethylphenyl)-2,4,6-trimethylbenzenesulfonamide. The frequency of H2 O2 -evoked afterdischarge-like bursting is augmented by OAG or OAG plus IP3 . Stimulating PKC with phorbol 12-myristate 13-acetate shortens the latency and increases the duration of H2 O2 -induced bursts. The nicotinamide adenine dinucleotide phosphate oxidase inhibitor, 3-benzyl-7-(2-benzoxazolyl)thio-1,2,3-triazolo[4,5-d]pyrimidine, attenuates burst firing in bag cell neuron clusters.
Collapse
Affiliation(s)
- Alamjeet K Chauhan-Puri
- Department of Biomedical and Molecular Sciences, Experimental Medicine Graduate Program, Queen's University, Kingston, Ontario, Canada
| | - Kelly H Lee
- Department of Biomedical and Molecular Sciences, Experimental Medicine Graduate Program, Queen's University, Kingston, Ontario, Canada
| | - Neil S Magoski
- Department of Biomedical and Molecular Sciences, Experimental Medicine Graduate Program, Queen's University, Kingston, Ontario, Canada
| |
Collapse
|
24
|
Neehus AL, Moriya K, Nieto-Patlán A, Le Voyer T, Lévy R, Özen A, Karakoc-Aydiner E, Baris S, Yildiran A, Altundag E, Roynard M, Haake K, Migaud M, Dorgham K, Gorochov G, Abel L, Lachmann N, Dogu F, Haskologlu S, İnce E, El-Benna J, Uzel G, Kiykim A, Boztug K, Roderick MR, Shahrooei M, Brogan PA, Abolhassani H, Hancioglu G, Parvaneh N, Belot A, Ikinciogullari A, Casanova JL, Puel A, Bustamante J. Impaired respiratory burst contributes to infections in PKCδ-deficient patients. J Exp Med 2021; 218:e20210501. [PMID: 34264265 PMCID: PMC8288504 DOI: 10.1084/jem.20210501] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 05/14/2021] [Accepted: 06/21/2021] [Indexed: 12/24/2022] Open
Abstract
Patients with autosomal recessive protein kinase C δ (PKCδ) deficiency suffer from childhood-onset autoimmunity, including systemic lupus erythematosus. They also suffer from recurrent infections that overlap with those seen in patients with chronic granulomatous disease (CGD), a disease caused by defects of the phagocyte NADPH oxidase and a lack of reactive oxygen species (ROS) production. We studied an international cohort of 17 PKCδ-deficient patients and found that their EBV-B cells and monocyte-derived phagocytes produced only small amounts of ROS and did not phosphorylate p40phox normally after PMA or opsonized Staphylococcus aureus stimulation. Moreover, the patients' circulating phagocytes displayed abnormally low levels of ROS production and markedly reduced neutrophil extracellular trap formation, altogether suggesting a role for PKCδ in activation of the NADPH oxidase complex. Our findings thus show that patients with PKCδ deficiency have impaired NADPH oxidase activity in various myeloid subsets, which may contribute to their CGD-like infectious phenotype.
Collapse
Affiliation(s)
- Anna-Lena Neehus
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Institut National de la Santé et de la Recherche Médicale UMR 1163, Paris, France
- University of Paris, Imagine Institute, Paris, France
- Institute of Experimental Hematology, REBIRTH Research Center for Translational and Regenerative Medicine, Hannover Medical School, Hannover, Germany
| | - Kunihiko Moriya
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Institut National de la Santé et de la Recherche Médicale UMR 1163, Paris, France
- University of Paris, Imagine Institute, Paris, France
| | - Alejandro Nieto-Patlán
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Institut National de la Santé et de la Recherche Médicale UMR 1163, Paris, France
- University of Paris, Imagine Institute, Paris, France
- Research and Development in Bioprocess Unit, National School of Biological Sciences, National Polytechnic Institute, Mexico City, Mexico
- National Laboratory for Specialized Services of Investigation, Development and Innovation for Pharma Chemicals and Biotechnological Products, LANSEIDI-FarBiotec-CONACyT, Mexico City, Mexico
| | - Tom Le Voyer
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Institut National de la Santé et de la Recherche Médicale UMR 1163, Paris, France
- University of Paris, Imagine Institute, Paris, France
| | - Romain Lévy
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Institut National de la Santé et de la Recherche Médicale UMR 1163, Paris, France
- University of Paris, Imagine Institute, Paris, France
- Pediatric Hematology-Immunology Unit, Necker Hospital for Sick Children, Paris, France
| | - Ahmet Özen
- Department of Pediatrics, Division of Allergy and Immunology, Marmara University, School of Medicine, Istanbul, Turkey
| | - Elif Karakoc-Aydiner
- Department of Pediatrics, Division of Allergy and Immunology, Marmara University, School of Medicine, Istanbul, Turkey
| | - Safa Baris
- Department of Pediatrics, Division of Allergy and Immunology, Marmara University, School of Medicine, Istanbul, Turkey
| | - Alisan Yildiran
- Department of Pediatric Immunology and Allergy, Ondokuz Mayıs University School of Medicine, Samsun, Turkey
| | - Engin Altundag
- Department of Medical Genetics, Ondokuz Mayis University Faculty of Medicine, Samsun, Turkey
| | - Manon Roynard
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Institut National de la Santé et de la Recherche Médicale UMR 1163, Paris, France
- University of Paris, Imagine Institute, Paris, France
| | - Kathrin Haake
- Institute of Experimental Hematology, REBIRTH Research Center for Translational and Regenerative Medicine, Hannover Medical School, Hannover, Germany
| | - Mélanie Migaud
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Institut National de la Santé et de la Recherche Médicale UMR 1163, Paris, France
- University of Paris, Imagine Institute, Paris, France
| | - Karim Dorgham
- Sorbonne University, Institut National de la Santé et de la Recherche Médicale, Center for Immunology and Microbial Infections, CIMI-Paris, Assistance Publique–Hôpitaux de Paris, Pitié-Salpêtrière Hospital, Department of Immunology, Paris, France
| | - Guy Gorochov
- Sorbonne University, Institut National de la Santé et de la Recherche Médicale, Center for Immunology and Microbial Infections, CIMI-Paris, Assistance Publique–Hôpitaux de Paris, Pitié-Salpêtrière Hospital, Department of Immunology, Paris, France
| | - Laurent Abel
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Institut National de la Santé et de la Recherche Médicale UMR 1163, Paris, France
- University of Paris, Imagine Institute, Paris, France
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY
| | - Nico Lachmann
- Institute of Experimental Hematology, REBIRTH Research Center for Translational and Regenerative Medicine, Hannover Medical School, Hannover, Germany
| | - Figen Dogu
- Department of Pediatric Immunology and Allergy, Ankara University School of Medicine, Ankara, Turkey
| | - Sule Haskologlu
- Department of Pediatric Immunology and Allergy, Ankara University School of Medicine, Ankara, Turkey
| | - Erdal İnce
- Department of Pediatric Infectious Disease, Ankara University School of Medicine, Ankara, Turkey
| | - Jamel El-Benna
- University of Paris, Institut National de la Santé et de la Recherche Médical U1149, Centre National de la Recherche Scientifique-ERL8252, Paris, France
- Center for Research on Inflammation, Laboratory of Excellence Inflamex, Faculty of Medicine, Xavier Bichat, Paris, France
| | - Gulbu Uzel
- Laboratory of Clinical Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Ayca Kiykim
- Pediatric Allergy and Immunology, Marmara University Pediatric Training and Research Hospital, Istanbul, Turkey
- Division of Pediatric Allergy and Immunology, Cerrahpasa School of Medicine, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Kaan Boztug
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
- St. Anna Children’s Hospital, Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, Vienna, Austria
- Ludwig Boltzmann Institute for Rare and Undiagnosed Diseases, Vienna, Austria
- St. Anna Children’s Cancer Research Institute, Vienna, Austria
| | - Marion R. Roderick
- Pediatric Immunology and Infectious Disease, Bristol Royal Hospital for Children, Bristol, UK
| | - Mohammad Shahrooei
- Specialized Immunology Laboratory of Dr. Shahrooei, Sina Medical Complex, Ahvaz, Iran
- Department of Microbiology and Immunology, Clinical and Diagnostic Immunology, Katholieke Universiteit Leuven, Leuven, Belgium
| | - Paul A. Brogan
- Infection, Inflammation, and Rheumatology Section, Infection, Immunity, Inflammation and Physiological Medicine Programme, University College London Institute of Child Health, London, UK
| | - Hassan Abolhassani
- Department of Laboratory Medicine, Division of Clinical Immunology, Karolinska Institute at Karolinska University Hospital Huddinge, Stockholm, Sweden
- Research Center for Immunodeficiencies, Tehran University of Medical Sciences, Tehran, Iran
| | - Gonca Hancioglu
- Department of Pediatric Immunology and Allergy, Ondokuz Mayıs University School of Medicine, Samsun, Turkey
| | - Nima Parvaneh
- Department of Pediatrics, Division of Allergy and Clinical Immunology, Tehran University of Medical Sciences, Tehran, Iran
- Research Center for Immunodeficiencies, Tehran University of Medical Sciences, Tehran, Iran
| | - Alexandre Belot
- Reference Center for Rare Rheumatic and Autoimmune Diseases in Children, Pediatric Rheumatology, Hospices Civils de Lyon, Centre International de Recherche en Infectiologie, Institut National de la Santé et de la Recherche Médicale U1111, UMS3444/US8 Lyon University, Lyon, France
| | - Aydan Ikinciogullari
- Department of Pediatric Immunology and Allergy, Ankara University School of Medicine, Ankara, Turkey
| | - Jean-Laurent Casanova
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Institut National de la Santé et de la Recherche Médicale UMR 1163, Paris, France
- University of Paris, Imagine Institute, Paris, France
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY
- Howard Hughes Medical Institute, New York, NY
| | - Anne Puel
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Institut National de la Santé et de la Recherche Médicale UMR 1163, Paris, France
- University of Paris, Imagine Institute, Paris, France
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY
| | - Jacinta Bustamante
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Institut National de la Santé et de la Recherche Médicale UMR 1163, Paris, France
- University of Paris, Imagine Institute, Paris, France
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY
- Center for the Study of Primary Immunodeficiencies, Necker Hospital for Sick Children, Assistance Publique–Hôpitaux de Paris, Paris, France
| |
Collapse
|
25
|
Liu F, Fan LM, Geng L, Li JM. p47 phox-Dependent Oxidant Signalling through ASK1, MKK3/6 and MAPKs in Angiotensin II-Induced Cardiac Hypertrophy and Apoptosis. Antioxidants (Basel) 2021; 10:1363. [PMID: 34572995 PMCID: PMC8468498 DOI: 10.3390/antiox10091363] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 08/22/2021] [Accepted: 08/23/2021] [Indexed: 02/07/2023] Open
Abstract
The p47phox is a key regulatory subunit of Nox2-containing NADPH oxidase (Nox2) that by generating reactive oxygen species (ROS) plays an important role in Angiotensin II (AngII)-induced cardiac hypertrophy and heart failure. However, the signalling pathways of p47phox in the heart remains unclear. In this study, we used wild-type (WT) and p47phox knockout (KO) mice (C57BL/6, male, 7-month-old, n = 9) to investigate p47phox-dependent oxidant-signalling in AngII infusion (0.8 mg/kg/day, 14 days)-induced cardiac hypertrophy and cardiomyocyte apoptosis. AngII infusion resulted in remarkable high blood pressure and cardiac hypertrophy in WT mice. However, these AngII-induced pathological changes were significantly reduced in p47phox KO mice. In WT hearts, AngII infusion increased significantly the levels of superoxide production, the expressions of Nox subunits, the expression of PKCα and C-Src and the activation of ASK1 (apoptosis signal-regulating kinase 1), MKK3/6, ERK1/2, p38 MAPK and JNK signalling pathways together with an elevated expression of apoptotic markers, i.e., γH2AX and p53 in the cardiomyocytes. However, in the absence of p47phox, although PKCα expression was increased in the hearts after AngII infusion, there was no significant activation of ASK1, MKK3/6 and MAPKs signalling pathways and no increase in apoptosis biomarker expression in cardiomyocytes. In conclusion, p47phox-dependent redox-signalling through ASK1, MKK3/6 and MAPKs plays a crucial role in AngII-induced cardiac hypertrophy and cardiomyocyte apoptosis.
Collapse
Affiliation(s)
- Fangfei Liu
- School of Biological Sciences, University of Reading, Reading RG6 6AS, UK; (F.L.); (L.G.)
| | - Lampson M. Fan
- The Royal Wolverhampton NHS Trust, Wolverhampton WV10 0QP, UK;
| | - Li Geng
- School of Biological Sciences, University of Reading, Reading RG6 6AS, UK; (F.L.); (L.G.)
- Faculty of Health and Medical Sciences, University of Surrey, Guildford GU2 7XH, UK
| | - Jian-Mei Li
- School of Biological Sciences, University of Reading, Reading RG6 6AS, UK; (F.L.); (L.G.)
- Faculty of Health and Medical Sciences, University of Surrey, Guildford GU2 7XH, UK
| |
Collapse
|
26
|
Merő B, Koprivanacz K, Cserkaszky A, Radnai L, Vas V, Kudlik G, Gógl G, Sok P, Póti ÁL, Szeder B, Nyitray L, Reményi A, Geiszt M, Buday L. Characterization of the Intramolecular Interactions and Regulatory Mechanisms of the Scaffold Protein Tks4. Int J Mol Sci 2021; 22:ijms22158103. [PMID: 34360869 PMCID: PMC8348221 DOI: 10.3390/ijms22158103] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 07/19/2021] [Accepted: 07/26/2021] [Indexed: 01/01/2023] Open
Abstract
The scaffold protein Tks4 is a member of the p47phox-related organizer superfamily. It plays a key role in cell motility by being essential for the formation of podosomes and invadopodia. In addition, Tks4 is involved in the epidermal growth factor (EGF) signaling pathway, in which EGF induces the translocation of Tks4 from the cytoplasm to the plasma membrane. The evolutionarily-related protein p47phox and Tks4 share many similarities in their N-terminal region: a phosphoinositide-binding PX domain is followed by two SH3 domains (so called “tandem SH3”) and a proline-rich region (PRR). In p47phox, the PRR is followed by a relatively short, disordered C-terminal tail region containing multiple phosphorylation sites. These play a key role in the regulation of the protein. In Tks4, the PRR is followed by a third and a fourth SH3 domain connected by a long (~420 residues) unstructured region. In p47phox, the tandem SH3 domain binds the PRR while the first SH3 domain interacts with the PX domain, thereby preventing its binding to the membrane. Based on the conserved structural features of p47phox and Tks4 and the fact that an intramolecular interaction between the third SH3 and the PX domains of Tks4 has already been reported, we hypothesized that Tks4 is similarly regulated by autoinhibition. In this study, we showed, via fluorescence-based titrations, MST, ITC, and SAXS measurements, that the tandem SH3 domain of Tks4 binds the PRR and that the PX domain interacts with the third SH3 domain. We also investigated a phosphomimicking Thr-to-Glu point mutation in the PRR as a possible regulator of intramolecular interactions. Phosphatidylinositol-3-phosphate (PtdIns(3)P) was identified as the main binding partner of the PX domain via lipid-binding assays. In truncated Tks4 fragments, the presence of the tandem SH3, together with the PRR, reduced PtdIns(3)P binding, while the presence of the third SH3 domain led to complete inhibition.
Collapse
Affiliation(s)
- Balázs Merő
- Research Centre for Natural Sciences, Institute of Enzymology, 1117 Budapest, Hungary; (B.M.); (K.K.); (A.C.); (L.R.); (V.V.); (G.K.); (B.S.)
| | - Kitti Koprivanacz
- Research Centre for Natural Sciences, Institute of Enzymology, 1117 Budapest, Hungary; (B.M.); (K.K.); (A.C.); (L.R.); (V.V.); (G.K.); (B.S.)
| | - Anna Cserkaszky
- Research Centre for Natural Sciences, Institute of Enzymology, 1117 Budapest, Hungary; (B.M.); (K.K.); (A.C.); (L.R.); (V.V.); (G.K.); (B.S.)
| | - László Radnai
- Research Centre for Natural Sciences, Institute of Enzymology, 1117 Budapest, Hungary; (B.M.); (K.K.); (A.C.); (L.R.); (V.V.); (G.K.); (B.S.)
| | - Virag Vas
- Research Centre for Natural Sciences, Institute of Enzymology, 1117 Budapest, Hungary; (B.M.); (K.K.); (A.C.); (L.R.); (V.V.); (G.K.); (B.S.)
| | - Gyöngyi Kudlik
- Research Centre for Natural Sciences, Institute of Enzymology, 1117 Budapest, Hungary; (B.M.); (K.K.); (A.C.); (L.R.); (V.V.); (G.K.); (B.S.)
| | - Gergő Gógl
- Department of Biochemistry, Eötvös Loránd University, 1117 Budapest, Hungary; (G.G.); (L.N.)
| | - Péter Sok
- Research Centre for Natural Sciences, Institute of Organic Chemistry, 1117 Budapest, Hungary; (P.S.); (Á.L.P.); (A.R.)
| | - Ádám L. Póti
- Research Centre for Natural Sciences, Institute of Organic Chemistry, 1117 Budapest, Hungary; (P.S.); (Á.L.P.); (A.R.)
| | - Bálint Szeder
- Research Centre for Natural Sciences, Institute of Enzymology, 1117 Budapest, Hungary; (B.M.); (K.K.); (A.C.); (L.R.); (V.V.); (G.K.); (B.S.)
| | - László Nyitray
- Department of Biochemistry, Eötvös Loránd University, 1117 Budapest, Hungary; (G.G.); (L.N.)
| | - Attila Reményi
- Research Centre for Natural Sciences, Institute of Organic Chemistry, 1117 Budapest, Hungary; (P.S.); (Á.L.P.); (A.R.)
| | - Miklós Geiszt
- Department of Physiology, Semmelweis University, 1094 Budapest, Hungary;
| | - László Buday
- Department of Biochemistry, Eötvös Loránd University, 1117 Budapest, Hungary; (G.G.); (L.N.)
- Department of Molecular Biology, Semmelweis University Medical School, 1094 Budapest, Hungary
- Correspondence:
| |
Collapse
|
27
|
Palmer TM, Salt IP. Nutrient regulation of inflammatory signalling in obesity and vascular disease. Clin Sci (Lond) 2021; 135:1563-1590. [PMID: 34231841 DOI: 10.1042/cs20190768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 06/10/2021] [Accepted: 06/28/2021] [Indexed: 11/17/2022]
Abstract
Despite obesity and diabetes markedly increasing the risk of developing cardiovascular diseases, the molecular and cellular mechanisms that underlie this association remain poorly characterised. In the last 20 years it has become apparent that chronic, low-grade inflammation in obese adipose tissue may contribute to the risk of developing insulin resistance and type 2 diabetes. Furthermore, increased vascular pro-inflammatory signalling is a key event in the development of cardiovascular diseases. Overnutrition exacerbates pro-inflammatory signalling in vascular and adipose tissues, with several mechanisms proposed to mediate this. In this article, we review the molecular and cellular mechanisms by which nutrients are proposed to regulate pro-inflammatory signalling in adipose and vascular tissues. In addition, we examine the potential therapeutic opportunities that these mechanisms provide for suppression of inappropriate inflammation in obesity and vascular disease.
Collapse
Affiliation(s)
- Timothy M Palmer
- Centre for Atherothrombosis and Metabolic Disease, Hull York Medical School, University of Hull, Hull HU6 7RX, United Kingdom
| | - Ian P Salt
- Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, United Kingdom
| |
Collapse
|
28
|
Kang X, Gao Z, Zheng L, Zhang X, Li H. Regulation of Lactobacillus plantarum on the reactive oxygen species related metabolisms of Saccharomyces cerevisiae. Lebensm Wiss Technol 2021. [DOI: 10.1016/j.lwt.2021.111492] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
29
|
Nunes Marsiglio-Librais G, Aparecida Vilas-Boas E, Carlein C, Hoffmann MDA, Roma LP, Carpinelli AR. Evidence for NADPH oxidase activation by GPR40 in pancreatic β-cells. Redox Rep 2021; 25:41-50. [PMID: 32354273 PMCID: PMC7241480 DOI: 10.1080/13510002.2020.1757877] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Objective: Investigate the involvement of the fatty acids receptor GPR40 in the assembly and activation of NADPH oxidase and the implications on pancreatic β-cell function. Methods: BRIN-BD11 β-cells were exposed to GPR40 agonist (GW9508) or linoleic acid in different glucose concentrations. Superoxide and H2O2 were analyzed, respectively, by DHE fluorescence and by fluorescence of the H2O2 sensor, roGFP2-Orp1. Protein contents of p47phox in plasma membrane and cytosol were analyzed by western blot. NADPH oxidase role was evaluated by p22phox siRNA or by pharmacological inhibition with VAS2870. NOX2 KO islets were used to measure total cytosolic calcium and insulin secretion. Results: GW9508 and linoleic acid increased superoxide and H2O2 contents at 5.6 and 8.3 mM of glucose. In addition, in 5.6 mM, but not at 16.7 mM of glucose, activation of GPR40 led to the translocation of p47phox to the plasma membrane. Knockdown of p22phox abolished the increase in superoxide after GW9508 and linoleic acid. No differences in insulin secretion were found between wild type and NOX2 KO islets treated with GW9508 or linoleic acid. Discussion: We report for the first time that acute activation of GPR40 leads to NADPH oxidase activation in pancreatic β-cells, without impact on insulin secretion.
Collapse
Affiliation(s)
| | - Eloisa Aparecida Vilas-Boas
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo (USP), São Paulo, Brazil.,Department of Biophysics, Center for Human and Molecular Biology, CIPMM, Saarland University, Homburg/Saar, Germany
| | - Christopher Carlein
- Department of Biophysics, Center for Human and Molecular Biology, CIPMM, Saarland University, Homburg/Saar, Germany
| | | | - Leticia Prates Roma
- Department of Biophysics, Center for Human and Molecular Biology, CIPMM, Saarland University, Homburg/Saar, Germany
| | - Angelo Rafael Carpinelli
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo (USP), São Paulo, Brazil
| |
Collapse
|
30
|
Vermot A, Petit-Härtlein I, Smith SME, Fieschi F. NADPH Oxidases (NOX): An Overview from Discovery, Molecular Mechanisms to Physiology and Pathology. Antioxidants (Basel) 2021; 10:890. [PMID: 34205998 PMCID: PMC8228183 DOI: 10.3390/antiox10060890] [Citation(s) in RCA: 260] [Impact Index Per Article: 86.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 05/21/2021] [Accepted: 05/26/2021] [Indexed: 01/17/2023] Open
Abstract
The reactive oxygen species (ROS)-producing enzyme NADPH oxidase (NOX) was first identified in the membrane of phagocytic cells. For many years, its only known role was in immune defense, where its ROS production leads to the destruction of pathogens by the immune cells. NOX from phagocytes catalyzes, via one-electron trans-membrane transfer to molecular oxygen, the production of the superoxide anion. Over the years, six human homologs of the catalytic subunit of the phagocyte NADPH oxidase were found: NOX1, NOX3, NOX4, NOX5, DUOX1, and DUOX2. Together with the NOX2/gp91phox component present in the phagocyte NADPH oxidase assembly itself, the homologs are now referred to as the NOX family of NADPH oxidases. NOX are complex multidomain proteins with varying requirements for assembly with combinations of other proteins for activity. The recent structural insights acquired on both prokaryotic and eukaryotic NOX open new perspectives for the understanding of the molecular mechanisms inherent to NOX regulation and ROS production (superoxide or hydrogen peroxide). This new structural information will certainly inform new investigations of human disease. As specialized ROS producers, NOX enzymes participate in numerous crucial physiological processes, including host defense, the post-translational processing of proteins, cellular signaling, regulation of gene expression, and cell differentiation. These diversities of physiological context will be discussed in this review. We also discuss NOX misregulation, which can contribute to a wide range of severe pathologies, such as atherosclerosis, hypertension, diabetic nephropathy, lung fibrosis, cancer, or neurodegenerative diseases, giving this family of membrane proteins a strong therapeutic interest.
Collapse
Affiliation(s)
- Annelise Vermot
- Univ. Grenoble Alpes, CNRS, CEA, Institut de Biologie Structurale, 38000 Grenoble, France; (A.V.); (I.P.-H.)
| | - Isabelle Petit-Härtlein
- Univ. Grenoble Alpes, CNRS, CEA, Institut de Biologie Structurale, 38000 Grenoble, France; (A.V.); (I.P.-H.)
| | - Susan M. E. Smith
- Department of Molecular and Cellular Biology, Kennesaw State University, Kennesaw, GA 30144, USA;
| | - Franck Fieschi
- Univ. Grenoble Alpes, CNRS, CEA, Institut de Biologie Structurale, 38000 Grenoble, France; (A.V.); (I.P.-H.)
| |
Collapse
|
31
|
Rap1a Regulates Cardiac Fibroblast Contraction of 3D Diabetic Collagen Matrices by Increased Activation of the AGE/RAGE Cascade. Cells 2021; 10:cells10061286. [PMID: 34067282 PMCID: PMC8224555 DOI: 10.3390/cells10061286] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 05/12/2021] [Accepted: 05/20/2021] [Indexed: 01/11/2023] Open
Abstract
Cardiovascular disease is a common diabetic complication that can arise when cardiac fibroblasts transition into myofibroblasts. Myofibroblast transition can be induced by advanced glycated end products (AGEs) present in the extracellular matrix (ECM) activating RAGE (receptor for advanced glycated end products) to elicit intracellular signaling. The levels of AGEs are higher under diabetic conditions due to the hyperglycemic conditions present in diabetics. AGE/RAGE signaling has been shown to alter protein expression and ROS production in cardiac fibroblasts, resulting in changes in cellular function, such as migration and contraction. Recently, a small GTPase, Rap1a, has been identified to overlap the AGE/RAGE signaling cascade and mediate changes in protein expression. While Rap1a has been shown to impact AGE/RAGE-induced protein expression, there are currently no data examining the impact Rap1a has on AGE/RAGE-induced cardiac fibroblast function. Therefore, we aimed to determine the impact of Rap1a on AGE/RAGE-mediated cardiac fibroblast contraction, as well as the influence isolated diabetic ECM has on facilitating these effects. In order to address this idea, genetically different cardiac fibroblasts were embedded in 3D collagen matrices consisting of collagen isolated from either non-diabetic of diabetic mice. Fibroblasts were treated with EPAC and/or exogenous AGEs, which was followed by assessment of matrix contraction, protein expression (α-SMA, SOD-1, and SOD-2), and hydrogen peroxide production. The results showed Rap1a overlaps the AGE/RAGE cascade to increase the myofibroblast population and generation of ROS production. The increase in myofibroblasts and oxidative stress appeared to contribute to increased matrix contraction, which was further exacerbated by diabetic conditions. Based off these results, we determined that Rap1a was essential in mediating the response of cardiac fibroblasts to AGEs within diabetic collagen.
Collapse
|
32
|
Maksouri H, Darif D, Estaquier J, Riyad M, Desterke C, Lemrani M, Dang PMC, Akarid K. The Modulation of NADPH Oxidase Activity in Human Neutrophils by Moroccan Strains of Leishmania major and Leishmania tropica Is Not Associated with p47 phox Phosphorylation. Microorganisms 2021; 9:1025. [PMID: 34068760 PMCID: PMC8151549 DOI: 10.3390/microorganisms9051025] [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: 02/28/2021] [Revised: 04/26/2021] [Accepted: 04/28/2021] [Indexed: 11/30/2022] Open
Abstract
Polymorphonuclear neutrophils (PMNs) are the first phagocyte recruited and infected by Leishmania. They synthetize superoxide anions (O2-) under the control of the NADPH oxidase complex. In Morocco, Leishmania major and L. tropica are the main species responsible for cutaneous leishmaniasis (CL). The impact of these parasites on human PMN functions is still unclear. We evaluated the in vitro capacity of primary Moroccan strains of L. major and L. tropica to modulate PMN O2- production and p47phox phosphorylation status of the NADPH oxidase complex. PMNs were isolated from healthy blood donors, and their infection rate was measured by microscopy. O2- production was measured by superoxide dismutase-inhibitable reduction of cytochrome C. P47phox phosphorylation was analyzed by Western blot using specific antibodies against Ser328 and Ser345 sites. Whereas we did not observe any difference in PMN infectivity rate, our results indicated that only L. tropica promastigotes inhibited both fMLF- and PMA-mediated O2- production independently of p47phox phosphorylation. Leishmania soluble antigens (SLAs) from both species significantly inhibited O2- induced by fMLF or PMA. However, they only decreased PMA-induced p47phox phosphorylation. L. major and L. tropica modulated differently O2- production by human PMNs independently of p47phox phosphorylation. The inhibition of ROS production by L. tropica could be a mechanism of its survival within PMNs that might explain the reported chronic pathogenicity of L. tropica CL.
Collapse
Affiliation(s)
- Hasnaa Maksouri
- Research Team on Immunopathology of Infectious and Systemic Diseases, Laboratory of Cellular and Molecular Pathology, Faculty of Medicine and Pharmacy (FMPC), Hassan II University of Casablanca (UH2C), 20000 Casablanca, Morocco; (H.M.); (M.R.)
- Molecular Genetics and Immunophysiopathology Research Team, Health and Environment Laboratory, Aïn Chock Faculty of Sciences, UH2C, 20000 Casablanca, Morocco;
| | - Dounia Darif
- Molecular Genetics and Immunophysiopathology Research Team, Health and Environment Laboratory, Aïn Chock Faculty of Sciences, UH2C, 20000 Casablanca, Morocco;
| | - Jerome Estaquier
- INSERM U1124, Paris University, 75006 Paris, France
- Centre Hospitalier Universitaire (CHU) de Québec Research Center, Faculty of Medicine, Laval University, Quebec City, QC G1V0A6, Canada
| | - Myriam Riyad
- Research Team on Immunopathology of Infectious and Systemic Diseases, Laboratory of Cellular and Molecular Pathology, Faculty of Medicine and Pharmacy (FMPC), Hassan II University of Casablanca (UH2C), 20000 Casablanca, Morocco; (H.M.); (M.R.)
| | - Christophe Desterke
- Faculty of Medicine of the Kremlin-Bicêtre, University Paris-Sud, 94270 Paris, France;
| | - Meryem Lemrani
- Laboratory of Parasitology and Vector-Borne-Diseases, Institut Pasteur du Maroc, 20250 Casablanca, Morocco;
| | - Pham My-Chan Dang
- INSERM-U1149, CNRS-ERL8252, Inflammation Research Center, 75018 Paris, France;
- Inflamex Laboratory of Excellence, Faculty of Medicine, Site Xavier Bichat, University of Paris, 75018 Paris, France
| | - Khadija Akarid
- Molecular Genetics and Immunophysiopathology Research Team, Health and Environment Laboratory, Aïn Chock Faculty of Sciences, UH2C, 20000 Casablanca, Morocco;
| |
Collapse
|
33
|
Xu Z, Liang Y, Delaney MK, Zhang Y, Kim K, Li J, Bai Y, Cho J, Ushio-Fukai M, Cheng N, Du X. Shear and Integrin Outside-In Signaling Activate NADPH-Oxidase 2 to Promote Platelet Activation. Arterioscler Thromb Vasc Biol 2021; 41:1638-1653. [PMID: 33691478 PMCID: PMC8057529 DOI: 10.1161/atvbaha.120.315773] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Accepted: 03/01/2021] [Indexed: 11/18/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Zheng Xu
- Department of Pharmacology, University of Illinois at Chicago (Z.X., Y.L., M.K.D., Y.Z., K.K., J.L., Y.B., J.C., M.U.-F., N.C., X.D.)
| | - Ying Liang
- Department of Pharmacology, University of Illinois at Chicago (Z.X., Y.L., M.K.D., Y.Z., K.K., J.L., Y.B., J.C., M.U.-F., N.C., X.D.)
| | - M. Keegan Delaney
- Department of Pharmacology, University of Illinois at Chicago (Z.X., Y.L., M.K.D., Y.Z., K.K., J.L., Y.B., J.C., M.U.-F., N.C., X.D.)
- Dupage Medical Technology, Inc (M.K.D.)
| | - Yaping Zhang
- Department of Pharmacology, University of Illinois at Chicago (Z.X., Y.L., M.K.D., Y.Z., K.K., J.L., Y.B., J.C., M.U.-F., N.C., X.D.)
| | - Kyungho Kim
- Department of Pharmacology, University of Illinois at Chicago (Z.X., Y.L., M.K.D., Y.Z., K.K., J.L., Y.B., J.C., M.U.-F., N.C., X.D.)
- Korean Medicine-Application Center, Korea Institute of Oriental Medicine, Daegu (K.K.)
| | - Jing Li
- Department of Pharmacology, University of Illinois at Chicago (Z.X., Y.L., M.K.D., Y.Z., K.K., J.L., Y.B., J.C., M.U.-F., N.C., X.D.)
| | - Yanyan Bai
- Department of Pharmacology, University of Illinois at Chicago (Z.X., Y.L., M.K.D., Y.Z., K.K., J.L., Y.B., J.C., M.U.-F., N.C., X.D.)
| | - Jaehyung Cho
- Department of Pharmacology, University of Illinois at Chicago (Z.X., Y.L., M.K.D., Y.Z., K.K., J.L., Y.B., J.C., M.U.-F., N.C., X.D.)
| | - Masuko Ushio-Fukai
- Department of Pharmacology, University of Illinois at Chicago (Z.X., Y.L., M.K.D., Y.Z., K.K., J.L., Y.B., J.C., M.U.-F., N.C., X.D.)
- Department of Medicine (Cardiology), Vascular Biology Center, Medical College of Georgia at Augusta University (M.U.-F.)
| | - Ni Cheng
- Department of Pharmacology, University of Illinois at Chicago (Z.X., Y.L., M.K.D., Y.Z., K.K., J.L., Y.B., J.C., M.U.-F., N.C., X.D.)
| | - Xiaoping Du
- Department of Pharmacology, University of Illinois at Chicago (Z.X., Y.L., M.K.D., Y.Z., K.K., J.L., Y.B., J.C., M.U.-F., N.C., X.D.)
| |
Collapse
|
34
|
Moghadam ZM, Henneke P, Kolter J. From Flies to Men: ROS and the NADPH Oxidase in Phagocytes. Front Cell Dev Biol 2021; 9:628991. [PMID: 33842458 PMCID: PMC8033005 DOI: 10.3389/fcell.2021.628991] [Citation(s) in RCA: 62] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Accepted: 02/26/2021] [Indexed: 12/16/2022] Open
Abstract
The cellular formation of reactive oxygen species (ROS) represents an evolutionary ancient antimicrobial defense system against microorganisms. The NADPH oxidases (NOX), which are predominantly localized to endosomes, and the electron transport chain in mitochondria are the major sources of ROS. Like any powerful immunological process, ROS formation has costs, in particular collateral tissue damage of the host. Moreover, microorganisms have developed defense mechanisms against ROS, an example for an arms race between species. Thus, although NOX orthologs have been identified in organisms as diverse as plants, fruit flies, rodents, and humans, ROS functions have developed and diversified to affect a multitude of cellular properties, i.e., far beyond direct antimicrobial activity. Here, we focus on the development of NOX in phagocytic cells, where the so-called respiratory burst in phagolysosomes contributes to the elimination of ingested microorganisms. Yet, NOX participates in cellular signaling in a cell-intrinsic and -extrinsic manner, e.g., via the release of ROS into the extracellular space. Accordingly, in humans, the inherited deficiency of NOX components is characterized by infections with bacteria and fungi and a seemingly independently dysregulated inflammatory response. Since ROS have both antimicrobial and immunomodulatory properties, their tight regulation in space and time is required for an efficient and well-balanced immune response, which allows for the reestablishment of tissue homeostasis. In addition, distinct NOX homologs expressed by non-phagocytic cells and mitochondrial ROS are interlinked with phagocytic NOX functions and thus affect the overall redox state of the tissue and the cellular activity in a complex fashion. Overall, the systematic and comparative analysis of cellular ROS functions in organisms of lower complexity provides clues for understanding the contribution of ROS and ROS deficiency to human health and disease.
Collapse
Affiliation(s)
- Zohreh Mansoori Moghadam
- Institute for Immunodeficiency, Center for Chronic Immunodeficiency (CCI), Medical Center – University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Philipp Henneke
- Institute for Immunodeficiency, Center for Chronic Immunodeficiency (CCI), Medical Center – University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Center for Pediatrics and Adolescent Medicine, Medical Center – University of Freiburg, Freiburg, Germany
| | - Julia Kolter
- Institute for Immunodeficiency, Center for Chronic Immunodeficiency (CCI), Medical Center – University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| |
Collapse
|
35
|
Karmacharya U, Chaudhary P, Lim D, Dahal S, Awasthi BP, Park HD, Kim JA, Jeong BS. Synthesis and anticancer evaluation of 6-azacyclonol-2,4,6-trimethylpyridin-3-ol derivatives: M3 muscarinic acetylcholine receptor-mediated anticancer activity of a cyclohexyl derivative in androgen-refractory prostate cancer. Bioorg Chem 2021; 110:104805. [PMID: 33725508 DOI: 10.1016/j.bioorg.2021.104805] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2021] [Revised: 02/20/2021] [Accepted: 03/02/2021] [Indexed: 12/24/2022]
Abstract
We recently reported 2,4,5-trimethylpyridin-3-ol with C(6)-azacyclonol, whose code name is BJ-1207, showing a promising anticancer activity by inhibiting NOX-derived ROS in A549 human lung cancer cells. The present study was focused on structural modification of the azacyclonol moiety of BJ-1207 to find a compound with better anticancer activity. Ten new compounds (3A-3J) were prepared and evaluated their inhibitory actions against proliferation of eighteen cancer cell lines as a primary screening. Among the ten derivatives of BJ-1207, the effects of compounds 3A and 3J on DU145 and PC-3, androgen-refractory cancer cell lines (ARPC), were greater than the parent compound, and compound 3A showed better activity than 3J. Antitumor activity of compound 3A was also observed in DU145-xenografted chorioallantoic membrane (CAM) tumor model. In addition, the ligand-based target prediction and molecular docking study using DeepZema® server showed compound 3A was a ligand to M3 muscarinic acetylcholine receptor (M3R) which is overexpressed in ARPC. Carbachol, a muscarinic receptor agonist, concentration dependently increased proliferation of DU145 in the absence of serum, and it also activated NADPH oxidase (NOX). The carbachol-induced proliferation and NOX activity was significantly blocked by compounds 3A in a concentration-dependent manner. This finding might become a new milestone in the development of pyridinol-based anti-cancer agents against ARPC.
Collapse
Affiliation(s)
- Ujjwala Karmacharya
- College of Pharmacy, Yeungnam University, 280 Daehak-ro, Gyeongsan 38541, Republic of Korea
| | - Prakash Chaudhary
- College of Pharmacy, Yeungnam University, 280 Daehak-ro, Gyeongsan 38541, Republic of Korea
| | - Dongchul Lim
- Innovo Therapeutics Inc., Daeduck Biz Center C-313, 17 Techno 4-ro, Yuseong-gu, Daejeon 34013, Republic of Korea
| | - Sadan Dahal
- College of Pharmacy, Yeungnam University, 280 Daehak-ro, Gyeongsan 38541, Republic of Korea
| | - Bhuwan Prasad Awasthi
- College of Pharmacy, Yeungnam University, 280 Daehak-ro, Gyeongsan 38541, Republic of Korea
| | - Hee Dong Park
- Innovo Therapeutics Inc., Daeduck Biz Center C-313, 17 Techno 4-ro, Yuseong-gu, Daejeon 34013, Republic of Korea
| | - Jung-Ae Kim
- College of Pharmacy, Yeungnam University, 280 Daehak-ro, Gyeongsan 38541, Republic of Korea.
| | - Byeong-Seon Jeong
- College of Pharmacy, Yeungnam University, 280 Daehak-ro, Gyeongsan 38541, Republic of Korea.
| |
Collapse
|
36
|
Burr SD, Stewart JA. Rap1a Overlaps the AGE/RAGE Signaling Cascade to Alter Expression of α-SMA, p-NF-κB, and p-PKC-ζ in Cardiac Fibroblasts Isolated from Type 2 Diabetic Mice. Cells 2021; 10:cells10030557. [PMID: 33806572 PMCID: PMC8000763 DOI: 10.3390/cells10030557] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 02/28/2021] [Accepted: 02/28/2021] [Indexed: 12/12/2022] Open
Abstract
Cardiovascular disease, specifically heart failure, is a common complication for individuals with type 2 diabetes mellitus. Heart failure can arise with stiffening of the left ventricle, which can be caused by “active” cardiac fibroblasts (i.e., myofibroblasts) remodeling the extracellular matrix (ECM). Differentiation of fibroblasts to myofibroblasts has been demonstrated to be an outcome of AGE/RAGE signaling. Hyperglycemia causes advanced glycated end products (AGEs) to accumulate within the body, and this process is greatly accelerated under chronic diabetic conditions. AGEs can bind and activate their receptor (RAGE) to trigger multiple downstream outcomes, such as altering ECM remodeling, inflammation, and oxidative stress. Previously, our lab has identified a small GTPase, Rap1a, that possibly overlaps the AGE/RAGE signaling cascade to affect the downstream outcomes. Rap1a acts as a molecular switch connecting extracellular signals to intracellular responses. Therefore, we hypothesized that Rap1a crosses the AGE/RAGE cascade to alter the expression of AGE/RAGE associated signaling proteins in cardiac fibroblasts in type 2 diabetic mice. To delineate this cascade, we used genetically different cardiac fibroblasts from non-diabetic, diabetic, non-diabetic RAGE knockout, diabetic RAGE knockout, and Rap1a knockout mice and treated them with pharmacological modifiers (exogenous AGEs, EPAC, Rap1a siRNA, and pseudosubstrate PKC-ζ). We examined changes in expression of proteins implicated as markers for myofibroblasts (α-SMA) and inflammation/oxidative stress (NF-κB and SOD-1). In addition, oxidative stress was also assessed by measuring hydrogen peroxide concentration. Our results indicated that Rap1a connects to the AGE/RAGE cascade to promote and maintain α-SMA expression in cardiac fibroblasts. Moreover, Rap1a, in conjunction with activation of the AGE/RAGE cascade, increased NF-κB expression as well as hydrogen peroxide concentration, indicating a possible oxidative stress response. Additionally, knocking down Rap1a expression resulted in an increase in SOD-1 expression suggesting that Rap1a can affect oxidative stress markers independently of the AGE/RAGE signaling cascade. These results demonstrated that Rap1a contributes to the myofibroblast population within the heart via AGE/RAGE signaling as well as promotes possible oxidative stress. This study offers a new potential therapeutic target that could possibly reduce the risk for developing diabetic cardiovascular complications attributed to AGE/RAGE signaling.
Collapse
|
37
|
Rabani R, Cossette C, Graham F, Powell WS. Protein kinase C activates NAD kinase in human neutrophils. Free Radic Biol Med 2020; 161:50-59. [PMID: 33011272 DOI: 10.1016/j.freeradbiomed.2020.09.022] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2020] [Revised: 08/14/2020] [Accepted: 09/26/2020] [Indexed: 12/23/2022]
Abstract
NAD kinase (NADK) is required for the de novo synthesis of NADP+ from NAD+. In neutrophils, NADK plays an essential role by providing sufficient levels of NADPH to support a robust oxidative burst. Activation of NADPH oxidase-2 (NOX-2) in neutrophils by stimulators of protein kinase C (PKC), such as phorbol myristate acetate (PMA), results in the rapid generation of superoxide at the expense of oxidation of NADPH to NADP+. In this study, we measured the levels of pyridine nucleotides following the addition of PMA to neutrophils. PMA elicited a rapid increase in NADP+ in neutrophils, which was not due to oxidation of NADPH, the levels of which also rose. This was mirrored by a rapid reduction in NAD+ levels, suggesting that NADK had been activated. PMA-induced depletion of NAD+ in neutrophils was blocked by PKC inhibitors, but was not dependent on NOX-2, as it was not blocked by the NOX inhibitor, diphenyleneiodonium. PMA also increased NADK activity in neutrophil lysates as well as NADK phosphorylation, as revealed by a monoclonal antibody selective for phospho-NADK. Human recombinant NADK was phosphorylated by PKCδ, resulting in increased immunoreactivity, but unchanged enzyme activity, suggesting that PKC-induced phosphorylation alone is insufficient to increase NADK activity in neutrophils. This leads us to speculate that phosphorylation of NADK promotes the dissociation of an inhibitory molecule from a complex, thereby increasing enzyme activity. Activation of NADK by PKC in phagocytic cells could be critical for the rapid provision of sufficient levels of superoxide for host defence against invading microorganisms.
Collapse
Affiliation(s)
- Razieh Rabani
- Meakins-Christie Laboratories, Centre for Translational Biology, Research Institute of the McGill University Health Centre, 1001 Decarie Blvd, Montreal, QC H4A 3J1, Canada
| | - Chantal Cossette
- Meakins-Christie Laboratories, Centre for Translational Biology, Research Institute of the McGill University Health Centre, 1001 Decarie Blvd, Montreal, QC H4A 3J1, Canada
| | - François Graham
- Meakins-Christie Laboratories, Centre for Translational Biology, Research Institute of the McGill University Health Centre, 1001 Decarie Blvd, Montreal, QC H4A 3J1, Canada
| | - William S Powell
- Meakins-Christie Laboratories, Centre for Translational Biology, Research Institute of the McGill University Health Centre, 1001 Decarie Blvd, Montreal, QC H4A 3J1, Canada.
| |
Collapse
|
38
|
Raad H, Mouawia H, Hassan H, El-Seblani M, Arabi-Derkawi R, Boussetta T, Gougerot-Pocidalo MA, Dang PMC, El-Benna J. The protein kinase A negatively regulates reactive oxygen species production by phosphorylating gp91phox/NOX2 in human neutrophils. Free Radic Biol Med 2020; 160:19-27. [PMID: 32758662 DOI: 10.1016/j.freeradbiomed.2020.07.021] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 07/01/2020] [Accepted: 07/11/2020] [Indexed: 02/06/2023]
Abstract
Superoxide anion production by neutrophils is essential for host defense against microbes. Superoxide anion generates other reactive oxygen species (ROS) that are very toxic for microbes and host cells, therefore their excessive production could induce inflammatory reactions and tissue injury. Cyclic adenosine monophosphate (cAMP) elevating agents are considered to be physiological inhibitors of superoxide production by neutrophils but the mechanisms involved in this inhibitory effect are poorly understood. Superoxide is produced by the phagocyte NADPH oxidase, a complex enzyme composed of two membrane subunits, gp91phox or NOX2 and p22phox, and four cytosolic components p47phox, p67phox, p40phox, and Rac2. Except Rac2, these proteins are known to be phosphorylated upon neutrophil stimulation. Here we show that forskolin, an activator of the adenylate cyclase-cAMP-PKA pathway, induced phosphorylation of gp91phox/NOX2 and inhibited fMLF-induced NADPH oxidase activation in human neutrophils. H89, a PKA inhibitor prevented the forskolin-induced phosphorylation of gp91phox and restored NADPH oxidase activation. Furthermore, PKA phosphorylated the recombinant gp91phox/NOX2-cytosolic C-terminal region in vitro only on a few specific peptides containing serine residues, as compared to PKC. Interestingly, phosphorylation of NOX2-Cter by PKA alone did not induce interaction with the cytosolic components p47phox, p67phox and Rac2, however it induced inhibition of PKC-induced interaction. Furthermore, PKA alone did not induce NOX2 electron transfer activity, however it inhibited PKC-induced activation. These results suggest that PKA phosphorylates NOX2 in human neutrophils, a process essential to limit ROS production and inflammation under physiological conditions. Our data identify the cAMP-PKA-NOX2-axis as a critical gatekeeper of neutrophil ROS production.
Collapse
Affiliation(s)
- Houssam Raad
- Université de Paris, Centre de Recherche sur L'Inflammation (CRI), INSERM-U1149, CNRS-ERL8252, Laboratoire D'Excellence Inflamex, Faculté de Médecine Xavier Bichat, 75018, Paris, France; Medical Care Laboratory, Lebanese University, Faculty of the Public Health IV, Zahle, Lebanon
| | - Hussein Mouawia
- Medical Care Laboratory, Lebanese University, Faculty of the Public Health IV, Zahle, Lebanon
| | - Hamad Hassan
- Medical Care Laboratory, Lebanese University, Faculty of the Public Health IV, Zahle, Lebanon
| | - Mohamed El-Seblani
- Medical Care Laboratory, Lebanese University, Faculty of the Public Health IV, Zahle, Lebanon
| | - Riad Arabi-Derkawi
- Université de Paris, Centre de Recherche sur L'Inflammation (CRI), INSERM-U1149, CNRS-ERL8252, Laboratoire D'Excellence Inflamex, Faculté de Médecine Xavier Bichat, 75018, Paris, France
| | - Tarek Boussetta
- Université de Paris, Centre de Recherche sur L'Inflammation (CRI), INSERM-U1149, CNRS-ERL8252, Laboratoire D'Excellence Inflamex, Faculté de Médecine Xavier Bichat, 75018, Paris, France
| | - Marie-Anne Gougerot-Pocidalo
- Université de Paris, Centre de Recherche sur L'Inflammation (CRI), INSERM-U1149, CNRS-ERL8252, Laboratoire D'Excellence Inflamex, Faculté de Médecine Xavier Bichat, 75018, Paris, France
| | - Pham My-Chan Dang
- Université de Paris, Centre de Recherche sur L'Inflammation (CRI), INSERM-U1149, CNRS-ERL8252, Laboratoire D'Excellence Inflamex, Faculté de Médecine Xavier Bichat, 75018, Paris, France
| | - Jamel El-Benna
- Université de Paris, Centre de Recherche sur L'Inflammation (CRI), INSERM-U1149, CNRS-ERL8252, Laboratoire D'Excellence Inflamex, Faculté de Médecine Xavier Bichat, 75018, Paris, France.
| |
Collapse
|
39
|
Zhang X, Zhang Y, Ji Z, Wang F, Zhang L, Song M, Li H. Oxidative damage mechanism in Saccharomyces cerevisiae cells exposed to tetrachlorobisphenol A. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2020; 80:103507. [PMID: 33007436 DOI: 10.1016/j.etap.2020.103507] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Revised: 09/05/2020] [Accepted: 09/25/2020] [Indexed: 06/11/2023]
Abstract
Tetrachlorobisphenol A (TCBPA) can promote intracellular reactive oxygen species (ROS) accumulation. However, limited attention has been given to mechanisms underlying TCBPA exposure-associated ROS accumulation. Here, such mechanisms were explored in the simple eukaryotic model organism Saccharomyces cerevisiae exposed to multiple concentrations of TCBPA. Addition of diphenyleneiodonium, a specific inhibitor of NADPH oxidase, blocked TCBPA treatment-associated intracellular ROS accumulation. NADPH oxidase can be activated by calcineurin, mitogen-activated protein kinase (MAPK), and tyrosine kinase. Therefore, corresponding specific inhibition respectively on these three kinases was performed and results suggested that the Ca2+ signaling pathway, MAPK pathway, and tyrosine kinase pathway all contributed to the TCBPA exposure-associated intracellular ROS accumulation. In addition, TCBPA exposure-associated up-regulation of genes involved in ROS production and down-regulation of catalase promoted ROS accumulation in S. cerevisiae. To sum up, our current results provide insights into the understanding of TCBPA exposure-associated ROS accumulation.
Collapse
Affiliation(s)
- Xiaoru Zhang
- Beijing Key Laboratory of Bioprocess, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, PR China
| | - Yaxian Zhang
- Beijing Key Laboratory of Bioprocess, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, PR China
| | - Zhihua Ji
- Beijing Key Laboratory of Bioprocess, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, PR China
| | - Fengbang Wang
- Key Laboratory of Environmental Nanotechnology and Health Effects, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, PR China
| | - Lei Zhang
- Beijing Key Laboratory of Bioprocess, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, PR China
| | - Maoyong Song
- Key Laboratory of Environmental Nanotechnology and Health Effects, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, PR China.
| | - Hao Li
- Beijing Key Laboratory of Bioprocess, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, PR China.
| |
Collapse
|
40
|
EphA2 Is a Neutrophil Receptor for Candida albicans that Stimulates Antifungal Activity during Oropharyngeal Infection. Cell Rep 2020; 28:423-433.e5. [PMID: 31291578 PMCID: PMC6638578 DOI: 10.1016/j.celrep.2019.06.020] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Revised: 05/03/2019] [Accepted: 06/05/2019] [Indexed: 12/23/2022] Open
Abstract
During oropharyngeal candidiasis (OPC), Candida albicans proliferates and invades the superficial oral epithelium. Ephrin type-A receptor 2 (EphA2) functions as an oral epithelial cell β-glucan receptor that triggers the production of proinflammatory mediators in response to fungal infection. Because EphA2 is also expressed by neutrophils, we investigated its role in neutrophil candidacidal activity during OPC. We found that EphA2 on stromal cells is required for the accumulation of phagocytes in the oral mucosa of mice with OPC. EphA2 on neutrophils is also central to host defense against OPC. The interaction of neutrophil EphA2 with serum- opsonized C. albicans yeast activates the MEK-ERK signaling pathway, leading to NADPH subunit p47phox site-specific phospho-priming. This priming increases intracellular reactive oxygen species production and enhances fungal killing. Thus, in neutrophils, EphA2 serves as a receptor for β-glucans that augments Fcγ receptor-mediated antifungal activity and controls early fungal proliferation during OPC. In oral epithelial cells, EphA2 functions as a β-glucan receptor that triggers the production of proinflammatory mediators in response to oropharyngeal candidiasis. Here, Swidergall et al. show that, in neutrophils, EphA2 recognition of β-glucans augments Fcγ receptor-mediated antifungal activity and prevents fungal proliferation during the initiation of oropharyngeal infection.
Collapse
|
41
|
Tozour J, Hughes F, Carrier A, Vieau D, Delahaye F. Prenatal Hyperglycemia Exposure and Cellular Stress, a Sugar-Coated View of Early Programming of Metabolic Diseases. Biomolecules 2020; 10:E1359. [PMID: 32977673 PMCID: PMC7598660 DOI: 10.3390/biom10101359] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 09/15/2020] [Accepted: 09/16/2020] [Indexed: 12/21/2022] Open
Abstract
Worldwide, the number of people with diabetes has quadrupled since 1980 reaching 422 million in 2014 (World Health Organization). This distressing rise in diabetes also affects pregnant women and thus, in regard to early programming of adult diseases, creates a vicious cycle of metabolic dysfunction passed from one generation to another. Metabolic diseases are complex and caused by the interplay between genetic and environmental factors. High-glucose exposure during in utero development, as observed with gestational diabetes mellitus (GDM), is an established risk factor for metabolic diseases. Despite intense efforts to better understand this phenomenon of early memory little is known about the molecular mechanisms associating early exposure to long-term diseases risk. However, evidence promotes glucose associated oxidative stress as one of the molecular mechanisms able to influence susceptibility to metabolic diseases. Thus, we decided here to further explore the relationship between early glucose exposure and cellular stress in the context of early development, and focus on the concept of glycemic memory, its consequences, and sexual dimorphic and epigenetic aspects.
Collapse
Affiliation(s)
- Jessica Tozour
- Department of Obstetrics and Gynecology, NYU Winthrop Hospital, Mineola, NY 11501, USA;
| | - Francine Hughes
- Obstetrics & Gynecology and Women’s Health, Division of Maternal-Fetal Medicine, Montefiore Medical Center/Albert Einstein College of Medicine, Bronx, NY 10461, USA;
| | - Arnaud Carrier
- Institut Pasteur de Lille, U1283-UMR 8199 EGID, Université de Lille, Inserm, CNRS, CHU Lille, F-59000 Lille, France;
| | - Didier Vieau
- BiologyDepartment, LilNCog Lille Neurosciences and Cognition U 1172, Université de Lille, Inserm, CHU Lille, F-59000 Lille, France;
| | - Fabien Delahaye
- Institut Pasteur de Lille, U1283-UMR 8199 EGID, Université de Lille, Inserm, CNRS, CHU Lille, F-59000 Lille, France;
| |
Collapse
|
42
|
Brier MI, Mundell JW, Yu X, Su L, Holmann A, Squeri J, Zhang B, Stanley SA, Friedman JM, Dordick JS. Uncovering a possible role of reactive oxygen species in magnetogenetics. Sci Rep 2020; 10:13096. [PMID: 32753716 PMCID: PMC7403421 DOI: 10.1038/s41598-020-70067-1] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Accepted: 07/07/2020] [Indexed: 02/06/2023] Open
Abstract
Recent reports have shown that intracellular, (super)paramagnetic ferritin nanoparticles can gate TRPV1, a non-selective cation channel, in a magnetic field. Here, we report the effects of differing field strength and frequency as well as chemical inhibitors on channel gating using a Ca2+-sensitive promoter to express a secreted embryonic alkaline phosphatase (SEAP) reporter. Exposure of TRPV1-ferritin-expressing HEK-293T cells at 30 °C to an alternating magnetic field of 501 kHz and 27.1 mT significantly increased SEAP secretion by ~ 82% relative to control cells, with lesser effects at other field strengths and frequencies. Between 30-32 °C, SEAP production was strongly potentiated 3.3-fold by the addition of the TRPV1 agonist capsaicin. This potentiation was eliminated by the competitive antagonist AMG-21629, the NADPH oxidase assembly inhibitor apocynin, and the reactive oxygen species (ROS) scavenger N-acetylcysteine, suggesting that ROS contributes to magnetogenetic TRPV1 activation. These results provide a rational basis to address the heretofore unknown mechanism of magnetogenetics.
Collapse
Affiliation(s)
- Matthew I Brier
- Department of Chemical and Biological Engineering, and Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY, 12180, USA
| | - Jordan W Mundell
- Department of Chemical and Biological Engineering, and Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY, 12180, USA
| | - Xiaofei Yu
- Laboratory of Molecular Genetics, Rockefeller University, New York, NY, 10065, USA
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, 200438, China
| | - Lichao Su
- State Key Laboratory Breeding Base of Nonferrous Metals and Specific Materials Processing, College of Material Science and Engineering, Guilin University of Technology, Jian Gan Road 12, Guilin, 541004, China
| | - Alexander Holmann
- Department of Chemical and Biological Engineering, and Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY, 12180, USA
| | - Jessica Squeri
- Department of Chemical and Biological Engineering, and Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY, 12180, USA
| | - Baolin Zhang
- State Key Laboratory Breeding Base of Nonferrous Metals and Specific Materials Processing, College of Material Science and Engineering, Guilin University of Technology, Jian Gan Road 12, Guilin, 541004, China
| | - Sarah A Stanley
- Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine At Mount Sinai, New York, NY, 10029, USA
| | - Jeffrey M Friedman
- Laboratory of Molecular Genetics, Rockefeller University, New York, NY, 10065, USA
- Howard Hughes Medical Institute, New York, NY, 10065, USA
| | - Jonathan S Dordick
- Department of Chemical and Biological Engineering, and Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY, 12180, USA.
- Departments of Biomedical Engineering and Biological Sciences, Rensselaer Polytechnic Institute, Troy, NY, 12180, USA.
| |
Collapse
|
43
|
Comparative study of the effects of ziram and disulfiram on human monocyte-derived macrophage functions and polarization: involvement of zinc. Cell Biol Toxicol 2020; 37:379-400. [PMID: 32712770 DOI: 10.1007/s10565-020-09540-6] [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] [Received: 03/02/2020] [Accepted: 06/03/2020] [Indexed: 10/23/2022]
Abstract
Ziram, a zinc dithiocarbamate is widely used worldwide as a fungicide in agriculture. In order to investigate ziram-induced changes in macrophage functions and polarization, human monocytes-derived macrophages in culture were treated with ziram at 0.01-10 μmol.L-1 for 4-24 h. To characterize zinc involvement in these changes, we also determined the effects of disulfiram alone (dithiocarbamate without zinc) or in co-incubation with ZnSO4. We have shown that ziram and disulfiram at 0.01 μmol.L-1 increased zymosan phagocytosis. In contrast, ziram at 10 μmol.L-1 completely inhibited this phagocytic process, the oxidative burst triggered by zymosan and the production of TNF-α, IL-1β, IL-6, and CCL2 triggered by LPS. Disulfiram had the same effects on these macrophages functions only when combined with zinc (10 μmol.L-1). In contrast, at 10 μmol.L-1 ziram and zinc associated-disulfiram induced expression of several antioxidants genes HMOX1, SOD2, and catalase, which could suggest the induction of oxidative stress. This oxidative stress could be involved in the increase in late apoptosis induced by ziram (10 μmol.L-1) and zinc associated-disulfiram. Concerning gene expression profiles of membrane markers of macrophage polarization, ziram at 10 μmol.L-1 had two opposite effects. It inhibited the gene expression of M2 markers (CD36, CD163) in the same way as the disulfiram-zinc co-treatment. Conversely, ziram induced gene expression of other M2 markers CD209, CD11b, and CD16 in the same way as treatment with zinc alone. Disulfiram-zinc association had no significant effects on these markers. These results taken together show that ziram via zinc modulates macrophages to M2-like anti-inflammatory phenotype which is often associated with various diseases.
Collapse
|
44
|
Li Y, Li M, Weigel B, Mall M, Werth VP, Liu ML. Nuclear envelope rupture and NET formation is driven by PKCα-mediated lamin B disassembly. EMBO Rep 2020; 21:e48779. [PMID: 32537912 DOI: 10.15252/embr.201948779] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 05/22/2020] [Accepted: 05/25/2020] [Indexed: 12/23/2022] Open
Abstract
The nuclear lamina is essential for the structural integration of the nuclear envelope. Nuclear envelope rupture and chromatin externalization is a hallmark of the formation of neutrophil extracellular traps (NETs). NET release was described as a cellular lysis process; however, this notion has been questioned recently. Here, we report that during NET formation, nuclear lamin B is not fragmented by destructive proteolysis, but rather disassembled into intact full-length molecules. Furthermore, we demonstrate that nuclear translocation of PKCα, which serves as the kinase to induce lamin B phosphorylation and disassembly, results in nuclear envelope rupture. Decreasing lamin B phosphorylation by PKCα inhibition, genetic deletion, or by mutating the PKCα consensus sites on lamin B attenuates extracellular trap formation. In addition, strengthening the nuclear envelope by lamin B overexpression attenuates NET release in vivo and reduces levels of NET-associated inflammatory cytokines in UVB-irradiated skin of lamin B transgenic mice. Our findings advance the mechanistic understanding of NET formation by showing that PKCα-mediated lamin B phosphorylation drives nuclear envelope rupture for chromatin release in neutrophils.
Collapse
Affiliation(s)
- Yubin Li
- Corporal Michael J. Crescenz VAMC, Philadelphia, PA, USA.,Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Minghui Li
- Corporal Michael J. Crescenz VAMC, Philadelphia, PA, USA.,Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.,Department of Rheumatology and Immunology, Tianjin Medical University General Hospital, Tianjin, China
| | - Bettina Weigel
- Cell Fate Engineering and Disease Modeling Group, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, Heidelberg, Germany.,HITBR Hector Institute for Translational Brain Research GmbH, Heidelberg, Germany.,Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Moritz Mall
- Cell Fate Engineering and Disease Modeling Group, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, Heidelberg, Germany.,HITBR Hector Institute for Translational Brain Research GmbH, Heidelberg, Germany.,Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Victoria P Werth
- Corporal Michael J. Crescenz VAMC, Philadelphia, PA, USA.,Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ming-Lin Liu
- Corporal Michael J. Crescenz VAMC, Philadelphia, PA, USA.,Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
45
|
Griffiths HR, Rooney MCO, Perrie Y. Does Dysregulation of Redox State Underpin the Decline of Innate Immunity with Aging? Antioxid Redox Signal 2020; 32:1014-1030. [PMID: 31989832 DOI: 10.1089/ars.2020.8021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Significance: Antibacterial defense invokes the innate immune system as a first responder, with neutrophils phagocytozing and forming neutrophil extracellular traps around pathogens in a reactive oxygen species (ROS)-dependent manner. Increased NOX2 activity and mitochondrial ROS production in phagocytic, antigen-presenting cells (APCs) affect local cytokine secretion and proteolysis of antigens for presentation to T cells at the immune synapse. Uncontrolled oxidative post-translational modifications to surface and cytoplasmic proteins in APCs during aging can impair innate immunity. Recent Advances: NOX2 plays a role in the maturation of dendritic cells, but paradoxically NOX2 activity has also been shown to promote viral pathogenicity. Accumulating evidence suggests that a reducing environment is essential to inhibit pathogen proliferation, facilitate antigenic processing in the endosomal lumen, and enable an effective immune synapse between APCs and T cells. This suggests that the kinetics and location of ROS production and reducing potential are important for effective innate immunity. Critical Issues: During aging, innate immune cells are less well able to phagocytoze, kill bacteria/viruses, and process proteins into antigenic peptides-three key steps that are necessary for developing a specific targeted response to protect against future exposure. Aberrant control of ROS production and impaired Nrf2-dependent reducing potential may contribute to age-associated immune decline. Future Directions: Local changes in redox potential may be achieved through adjuvant formulations to improve innate immunity. Further work is needed to understand the timing of delivery for redox modulators to facilitate innate immune cell recruitment, survival, antigen processing and presentation activity without disrupting essential ROS-dependent bacterial killing.
Collapse
Affiliation(s)
- Helen R Griffiths
- Faculty of Health and Medical Sciences, University of Surrey, Guildford, United Kingdom
| | - Matthew C O Rooney
- Faculty of Health and Medical Sciences, University of Surrey, Guildford, United Kingdom
| | - Yvonne Perrie
- Department of Pharmacy, University of Strathclyde, Glasgow, Scotland
| |
Collapse
|
46
|
Hann J, Bueb JL, Tolle F, Bréchard S. Calcium signaling and regulation of neutrophil functions: Still a long way to go. J Leukoc Biol 2019; 107:285-297. [PMID: 31841231 DOI: 10.1002/jlb.3ru0719-241r] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Revised: 11/08/2019] [Accepted: 12/01/2019] [Indexed: 12/22/2022] Open
Abstract
Neutrophils are the most abundant leukocytes in blood and disruption in their functions often results in an increased risk of serious infections and inflammatory autoimmune diseases. Following recent discoveries in their influence over disease progression, a resurgence of interest for neutrophil biology has taken place. The multitude of signaling pathways activated by the engagement of numerous types of receptors, with which neutrophils are endowed, reflects the functional complexity of these cells. It is therefore not surprising that there remains a huge lack in the understanding of molecular mechanisms underlining neutrophil functions. Moreover, studies on neutrophils are undoubtedly limited by the difficulty to efficiently edit the cell's genome. Over the past 30 years, compelling evidence has clearly highlighted that Ca2+ -signaling is governing the key processes associated with neutrophil functions. The confirmation of the role of an elevation of intracellular Ca2+ concentration has come from studies on NADPH oxidase activation and phagocytosis. In this review, we give an overview and update of our current knowledge on the role of Ca2+ mobilization in the regulation of pro-inflammatory functions of neutrophils. In particular, we stress the importance of Ca2+ in the formation of NETs and cytokine secretion in the light of newest findings. This will allow us to embrace how much further we have to go to understand the complex dynamics of Ca2+ -dependent mechanisms in order to gain more insights into the role of neutrophils in the pathogenesis of inflammatory diseases. The potential for therapeutics to regulate the neutrophil functions, such as Ca2+ influx inhibitors to prevent autoimmune and chronic inflammatory diseases, has been discussed in the last part of the review.
Collapse
Affiliation(s)
- J Hann
- Life Sciences Research Unit, Immune Cells and Inflammatory Diseases Group, University of Luxembourg, Belvaux, Luxembourg
| | - J-L Bueb
- Life Sciences Research Unit, Immune Cells and Inflammatory Diseases Group, University of Luxembourg, Belvaux, Luxembourg
| | - F Tolle
- Life Sciences Research Unit, Immune Cells and Inflammatory Diseases Group, University of Luxembourg, Belvaux, Luxembourg
| | - S Bréchard
- Life Sciences Research Unit, Immune Cells and Inflammatory Diseases Group, University of Luxembourg, Belvaux, Luxembourg
| |
Collapse
|
47
|
Hydrogen Peroxide Gates a Voltage-Dependent Cation Current in Aplysia Neuroendocrine Cells. J Neurosci 2019; 39:9900-9913. [PMID: 31676600 DOI: 10.1523/jneurosci.1460-19.2019] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Revised: 10/07/2019] [Accepted: 10/27/2019] [Indexed: 11/21/2022] Open
Abstract
Nonselective cation channels promote persistent spiking in many neurons from a diversity of animals. In the hermaphroditic marine-snail, Aplysia californica, synaptic input to the neuroendocrine bag cell neurons triggers various cation channels, causing an ∼30 min afterdischarge of action potentials and the secretion of egg-laying hormone. During the afterdischarge, protein kinase C is also activated, which in turn elevates hydrogen peroxide (H2O2), likely by stimulating nicotinamide adenine dinucleotide phosphate oxidase. The present study investigated whether H2O2 regulates cation channels to drive the afterdischarge. In single, cultured bag cell neurons, H2O2 elicited a prolonged, concentration- and voltage-dependent inward current, associated with an increase in membrane conductance and a reversal potential of ∼+30 mV. Compared with normal saline, the presence of Ca2+-free, Na+-free, or Na+/Ca2+-free extracellular saline, lowered the current amplitude and left-shifted the reversal potential, consistent with a nonselective cationic conductance. Preventing H2O2 reduction with the glutathione peroxidase inhibitor, mercaptosuccinate, enhanced the H2O2-induced current, while boosting glutathione production with its precursor, N-acetylcysteine, or adding the reducing agent, dithiothreitol, lessened the response. Moreover, the current generated by the alkylating agent, N-ethylmaleimide, occluded the effect of H2O2 The H2O2-induced current was inhibited by tetrodotoxin as well as the cation channel blockers, 9-phenanthrol and clotrimazole. In current-clamp, H2O2 stimulated burst firing, but this was attenuated or prevented altogether by the channel blockers. Finally, H2O2 evoked an afterdischarge from whole bag cell neuron clusters recorded ex vivo by sharp-electrode. H2O2 may regulate a cation channel to influence long-term changes in activity and ultimately reproduction.SIGNIFICANCE STATEMENT Hydrogen peroxide (H2O2) is often studied in a pathological context, such as ischemia or inflammation. However, H2O2 also physiologically modulates synaptic transmission and gates certain transient receptor potential channels. That stated, the effect of H2O2 on neuronal excitability remains less well defined. Here, we examine how H2O2 influences Aplysia bag cell neurons, which elicit ovulation by releasing hormones during an afterdischarge. These neuroendocrine cells are uniquely identifiable and amenable to recording as individual cultured neurons or a cluster from the nervous system. In both culture and the cluster, H2O2 evokes prolonged, afterdischarge-like bursting by gating a nonselective voltage-dependent cationic current. Thus, H2O2, which is generated in response to afterdischarge-associated second messengers, may prompt the firing necessary for hormone secretion and procreation.
Collapse
|
48
|
Knock GA. NADPH oxidase in the vasculature: Expression, regulation and signalling pathways; role in normal cardiovascular physiology and its dysregulation in hypertension. Free Radic Biol Med 2019; 145:385-427. [PMID: 31585207 DOI: 10.1016/j.freeradbiomed.2019.09.029] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Revised: 08/29/2019] [Accepted: 09/23/2019] [Indexed: 02/06/2023]
Abstract
The last 20-25 years have seen an explosion of interest in the role of NADPH oxidase (NOX) in cardiovascular function and disease. In vascular smooth muscle and endothelium, NOX generates reactive oxygen species (ROS) that act as second messengers, contributing to the control of normal vascular function. NOX activity is altered in response to a variety of stimuli, including G-protein coupled receptor agonists, growth-factors, perfusion pressure, flow and hypoxia. NOX-derived ROS are involved in smooth muscle constriction, endothelium-dependent relaxation and smooth muscle growth, proliferation and migration, thus contributing to the fine-tuning of blood flow, arterial wall thickness and vascular resistance. Through reversible oxidative modification of target proteins, ROS regulate the activity of protein tyrosine phosphatases, kinases, G proteins, ion channels, cytoskeletal proteins and transcription factors. There is now considerable, but somewhat contradictory evidence that NOX contributes to the pathogenesis of hypertension through oxidative stress. Specific NOX isoforms have been implicated in endothelial dysfunction, hyper-contractility and vascular remodelling in various animal models of hypertension, pulmonary hypertension and pulmonary arterial hypertension, but also have potential protective effects, particularly NOX4. This review explores the multiplicity of NOX function in the healthy vasculature and the evidence for and against targeting NOX for antihypertensive therapy.
Collapse
Affiliation(s)
- Greg A Knock
- Dpt. of Inflammation Biology, School of Immunology & Microbial Sciences, Faculty of Life Sciences & Medicine, King's College London, UK.
| |
Collapse
|
49
|
NADPH oxidases and oxidase crosstalk in cardiovascular diseases: novel therapeutic targets. Nat Rev Cardiol 2019; 17:170-194. [PMID: 31591535 DOI: 10.1038/s41569-019-0260-8] [Citation(s) in RCA: 311] [Impact Index Per Article: 62.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/19/2019] [Indexed: 02/07/2023]
Abstract
Reactive oxygen species (ROS)-dependent production of ROS underlies sustained oxidative stress, which has been implicated in the pathogenesis of cardiovascular diseases such as hypertension, aortic aneurysm, hypercholesterolaemia, atherosclerosis, diabetic vascular complications, cardiac ischaemia-reperfusion injury, myocardial infarction, heart failure and cardiac arrhythmias. Interactions between different oxidases or oxidase systems have been intensively investigated for their roles in inducing sustained oxidative stress. In this Review, we discuss the latest data on the pathobiology of each oxidase component, the complex crosstalk between different oxidase components and the consequences of this crosstalk in mediating cardiovascular disease processes, focusing on the central role of particular NADPH oxidase (NOX) isoforms that are activated in specific cardiovascular diseases. An improved understanding of these mechanisms might facilitate the development of novel therapeutic agents targeting these oxidase systems and their interactions, which could be effective in the prevention and treatment of cardiovascular disorders.
Collapse
|
50
|
Burtenshaw D, Kitching M, Redmond EM, Megson IL, Cahill PA. Reactive Oxygen Species (ROS), Intimal Thickening, and Subclinical Atherosclerotic Disease. Front Cardiovasc Med 2019; 6:89. [PMID: 31428618 PMCID: PMC6688526 DOI: 10.3389/fcvm.2019.00089] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Accepted: 06/14/2019] [Indexed: 12/14/2022] Open
Abstract
Arteriosclerosis causes significant morbidity and mortality worldwide. Central to this process is the development of subclinical non-atherosclerotic intimal lesions before the appearance of pathologic intimal thickening and advanced atherosclerotic plaques. Intimal thickening is associated with several risk factors, including oxidative stress due to reactive oxygen species (ROS), inflammatory cytokines and lipid. The main ROS producing systems in-vivo are reduced nicotinamide dinucleotide phosphate (NADPH) oxidase (NOX). ROS effects are context specific. Exogenous ROS induces apoptosis and senescence, whereas intracellular ROS promotes stem cell differentiation, proliferation, and migration. Lineage tracing studies using murine models of subclinical atherosclerosis have revealed the contributory role of medial smooth muscle cells (SMCs), resident vascular stem cells, circulating bone-marrow progenitors and endothelial cells that undergo endothelial-mesenchymal-transition (EndMT). This review will address the putative physiological and patho-physiological roles of ROS in controlling vascular cell fate and ROS contribution to vascular regeneration and disease progression.
Collapse
Affiliation(s)
- Denise Burtenshaw
- Vascular Biology & Therapeutics, School of Biotechnology, Dublin City University, Dublin, Ireland
| | | | - Eileen M Redmond
- Department of Surgery, University of Rochester, Rochester, NY, United States
| | - Ian L Megson
- Centre for Health Science, UHI Institute of Health Research and Innovation, Inverness, United Kingdom
| | - Paul A Cahill
- Vascular Biology & Therapeutics, School of Biotechnology, Dublin City University, Dublin, Ireland
| |
Collapse
|