1
|
Kervella M, Bertile F, Bouillaud F, Criscuolo F. The cell origin of reactive oxygen species and its implication for evolutionary trade-offs. Open Biol 2025; 15:240312. [PMID: 40237040 PMCID: PMC12001088 DOI: 10.1098/rsob.240312] [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: 10/21/2024] [Revised: 01/08/2025] [Accepted: 02/09/2025] [Indexed: 04/17/2025] Open
Abstract
The allocation of resources in animals is shaped by adaptive trade-offs aimed at maximizing fitness. At the heart of these trade-offs, lies metabolism and the conversion of food resources into energy, a process mostly occurring in mitochondria. Yet, the conversion of nutrients to utilizable energy molecules (adenosine triphosphate) inevitably leads to the by-production of reactive oxygen species (ROS) that may cause damage to important biomolecules such as proteins or lipids. The 'ROS theory of ageing' has thus proposed that the relationship between lifespan and metabolic rate may be mediated by ROS production. However, the relationship is not as straightforward as it may seem: not only are mitochondrial ROS crucial for various cellular functions, but mitochondria are also actually equipped with antioxidant systems, and many extra-mitochondrial sources also produce ROS. In this review, we discuss how viewing the mitochondrion as a regulator of cellular oxidative homeostasis, not merely a ROS producer, may provide new insights into the role of oxidative stress in the reproduction-survival trade-off. We suggest several avenues to test how mitochondrial oxidative buffering capacity might complement current bioenergetic and evolutionary studies.
Collapse
|
2
|
Wang K, Zhang L, Deng B, Zhao K, Chen C, Wang W. Mitochondrial uncoupling protein 2: a central player in pancreatic disease pathophysiology. Mol Med 2024; 30:259. [PMID: 39707176 DOI: 10.1186/s10020-024-01027-y] [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: 10/07/2024] [Accepted: 12/03/2024] [Indexed: 12/23/2024] Open
Abstract
Pancreatic diseases pose considerable health challenges due to their complex etiology and limited therapeutic options. Mitochondrial uncoupling protein 2 (UCP2), highly expressed in pancreatic tissue, participates in numerous physiological processes and signaling pathways, indicating its potential relevance in these diseases. Despite this, UCP2's role in acute pancreatitis (AP) remains underexplored, and its functions in chronic pancreatitis (CP) and pancreatic steatosis are largely unknown. Additionally, the mechanisms connecting various pancreatic diseases are intricate and not yet fully elucidated. Given UCP2's diverse functionality, broad expression in pancreatic tissue, and the distinct pathophysiological features of pancreatic diseases, this review offers a comprehensive analysis of current findings on UCP2's involvement in these conditions. We discuss recent insights into UCP2's complex regulatory mechanisms, propose that UCP2 may serve as a central regulatory factor in pancreatic disease progression, and hypothesize that UCP2 dysfunction could significantly contribute to disease pathogenesis. Understanding UCP2's role and mechanisms in pancreatic diseases may pave the way for innovative therapeutic and diagnostic approaches.
Collapse
Affiliation(s)
- Kunpeng Wang
- Department of General Surgery, Renmin Hospital of Wuhan University, Wuhan, China
- General Surgery Laboratory, Renmin Hospital of Wuhan University, Wuhan, China
| | - Lilong Zhang
- Department of General Surgery, Renmin Hospital of Wuhan University, Wuhan, China
- General Surgery Laboratory, Renmin Hospital of Wuhan University, Wuhan, China
| | - Beiying Deng
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Kailiang Zhao
- Department of General Surgery, Renmin Hospital of Wuhan University, Wuhan, China
- General Surgery Laboratory, Renmin Hospital of Wuhan University, Wuhan, China
| | - Chen Chen
- Department of General Surgery, Renmin Hospital of Wuhan University, Wuhan, China.
- General Surgery Laboratory, Renmin Hospital of Wuhan University, Wuhan, China.
| | - Weixing Wang
- Department of General Surgery, Renmin Hospital of Wuhan University, Wuhan, China.
- General Surgery Laboratory, Renmin Hospital of Wuhan University, Wuhan, China.
| |
Collapse
|
3
|
Nesci S, Rubattu S. UCP2, a Member of the Mitochondrial Uncoupling Proteins: An Overview from Physiological to Pathological Roles. Biomedicines 2024; 12:1307. [PMID: 38927514 PMCID: PMC11201685 DOI: 10.3390/biomedicines12061307] [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: 05/12/2024] [Revised: 06/07/2024] [Accepted: 06/10/2024] [Indexed: 06/28/2024] Open
Abstract
UCP2 is an uncoupling protein homolog to UCP1. Unlike UCP1, which participates in non-shivering thermogenesis by uncoupling oxidative phosphorylation (OXPHOS), UCP2 does not perform a canonical H+ leak, consuming the protonmotive force (Δp) through the inner mitochondrial membrane. The UCP2 biological role is elusive. It can counteract oxidative stress, acting with a "mild uncoupling" process to reduce ROS production, and, in fact, UCP2 activities are related to inflammatory processes, triggering pathological conditions. However, the Δp dissipation by UCP2 activity reduces the mitochondrial ATP production and rewires the bioenergetic metabolism of the cells. In all likelihood, UCP2 works as a carrier of metabolites with four carbon atoms (C4), reversing the anaerobic glycolysis-dependent catabolism to OXPHOS. Indeed, UCP2 can perform catalysis in dual mode: mild uncoupling of OXPHOS and metabolite C4 exchange of mitochondria. In vivo, the UCP2 features in the biology of mitochondria promote healthy ageing, increased lifespan, and can assure cerebro- and cardiovascular protection. However, the pathological conditions responsible for insulin secretion suppression are dependent on UCP2 activity. On balance, the uncertain biochemical mechanisms dependent on UCP2 do not allow us to depict the protective role in mitochondrial bioenergetics.
Collapse
Affiliation(s)
- Salvatore Nesci
- Department of Veterinary Medical Sciences, Alma Mater Studiorum University of Bologna, 40064 Ozzano Emilia, Italy;
| | - Speranza Rubattu
- Department of Clinical and Molecular Medicine, School of Medicine and Psychology, “Sapienza” University of Rome, 00189 Rome, Italy
- IRCCS Neuromed, 86077 Pozzilli, Italy
| |
Collapse
|
4
|
Lang L, Zheng D, Jiang Q, Meng T, Ma X, Yang Y. Uncoupling protein 2 modulates polarization and metabolism of human primary macrophages via glycolysis and the NF‑κB pathway. Exp Ther Med 2023; 26:583. [PMID: 38023353 PMCID: PMC10665990 DOI: 10.3892/etm.2023.12282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2023] [Accepted: 08/17/2023] [Indexed: 12/01/2023] Open
Abstract
Metabolic abnormalities, particularly the M1/M2 macrophage imbalance, play a critical role in the development of various diseases, leading to severe inflammatory responses. The present study aimed to investigate the role of uncoupling protein 2 (UCP2) in regulating macrophage polarization, glycolysis, metabolic reprogramming, reactive oxygen species (ROS) and inflammation. Primary human macrophages were first polarized into M1 and M2 subtypes, and these two subtypes were infected by lentivirus-mediated UCP2 overexpression or knockdown, followed by enzyme-linked immunosorbent assay, reverse transcription-quantitative PCR, western blotting and flow cytometry to analyze the effects of UCP2 on glycolysis, oxidative phosphorylation (OXPHOS), ROS production and cytokine secretion, respectively. The results demonstrated that UCP2 expression was suppressed in M1 macrophages and increased in M2 macrophages, suggesting its regulatory role in macrophage polarization. UCP2 overexpression decreased macrophage glycolysis, increased OXPHOS, decreased ROS production, and led to the conversion of M1 polarization to M2 polarization. This process involved NF-κB signaling to regulate the secretion profile of cytokines and chemokines and affected the expression of key enzymes of glycolysis and a key factor for maintaining mitochondrial homeostasis (nuclear respiratory factor 1). UCP2 knockdown in M2 macrophages exacerbated inflammation and oxidative stress by promoting glycolysis, which was attenuated by the glycolysis inhibitor 2-deoxyglucose. These findings highlight the critical role of UCP2 in regulating macrophage polarization, metabolism, inflammation and oxidative stress through its effects on glycolysis, providing valuable insights into potential therapeutic strategies for macrophage-driven inflammatory and metabolic diseases.
Collapse
Affiliation(s)
- Liguo Lang
- Department of Cardiology, People's Hospital of Ningxia Hui Autonomous Region, Yinchuan, Ningxia Hui Autonomous Region 750001, P.R. China
| | - Dongju Zheng
- Department of Cardiology, People's Hospital of Ningxia Hui Autonomous Region, Yinchuan, Ningxia Hui Autonomous Region 750001, P.R. China
| | - Qingjun Jiang
- Department of Cardiology, People's Hospital of Ningxia Hui Autonomous Region, Yinchuan, Ningxia Hui Autonomous Region 750001, P.R. China
| | - Ting Meng
- Department of Cardiology, People's Hospital of Ningxia Hui Autonomous Region, Yinchuan, Ningxia Hui Autonomous Region 750001, P.R. China
| | - Xiaohu Ma
- Department of Cardiology, People's Hospital of Ningxia Hui Autonomous Region, Yinchuan, Ningxia Hui Autonomous Region 750001, P.R. China
| | - Yang Yang
- Department of Cardiology, People's Hospital of Ningxia Hui Autonomous Region, Yinchuan, Ningxia Hui Autonomous Region 750001, P.R. China
| |
Collapse
|
5
|
A lncRNA-encoded mitochondrial micropeptide exacerbates microglia-mediated neuroinflammation in retinal ischemia/reperfusion injury. Cell Death Dis 2023; 14:126. [PMID: 36792584 PMCID: PMC9932084 DOI: 10.1038/s41419-023-05617-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 01/20/2023] [Accepted: 01/25/2023] [Indexed: 02/17/2023]
Abstract
As a common pathology of many ocular disorders such as diabetic retinopathy and glaucoma, retinal ischemia/reperfusion (IR) triggers inflammation and microglia activation that lead to irreversible retinal damage. The detailed molecular mechanism underlying retinal IR injury, however, remains poorly understood at present. Here we report the bioinformatic identification of a lncRNA 1810058I24Rik (181-Rik) that was shown to encode a mitochondrion-located micropeptide Stmp1. Its deficiency in mice protected retinal ganglion cells from retinal IR injury by attenuating the activation of microglia and the Nlrp3 inflammasome pathway. Moreover, its genetic knockout in mice or knockdown in primary microglia promoted mitochondrial fusion, impaired mitochondrial membrane potential, and reactive oxygen species (ROS) production, diminished aerobic glycolysis, and ameliorated inflammation. It appears that 181-Rik may trigger the Nlrp3 inflammasome activation by controlling mitochondrial functions through inhibiting expression of the metabolic sensor uncoupling protein 2 (Ucp2) and activating expression of the Ca2+ sensors S100a8/a9. Together, our findings shed new light on the molecular pathogenesis of retinal IR injury and may provide a fresh therapeutic target for IR-associated neurodegenerative diseases.
Collapse
|
6
|
Luby A, Alves-Guerra MC. UCP2 as a Cancer Target through Energy Metabolism and Oxidative Stress Control. Int J Mol Sci 2022; 23:ijms232315077. [PMID: 36499405 PMCID: PMC9735768 DOI: 10.3390/ijms232315077] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 11/25/2022] [Accepted: 11/27/2022] [Indexed: 12/02/2022] Open
Abstract
Despite numerous therapies, cancer remains one of the leading causes of death worldwide due to the lack of markers for early detection and response to treatment in many patients. Technological advances in tumor screening and renewed interest in energy metabolism have allowed us to identify new cellular players in order to develop personalized treatments. Among the metabolic actors, the mitochondrial transporter uncoupling protein 2 (UCP2), whose expression is increased in many cancers, has been identified as an interesting target in tumor metabolic reprogramming. Over the past decade, a better understanding of its biochemical and physiological functions has established a role for UCP2 in (1) protecting cells from oxidative stress, (2) regulating tumor progression through changes in glycolytic, oxidative and calcium metabolism, and (3) increasing antitumor immunity in the tumor microenvironment to limit cancer development. With these pleiotropic roles, UCP2 can be considered as a potential tumor biomarker that may be interesting to target positively or negatively, depending on the type, metabolic status and stage of tumors, in combination with conventional chemotherapy or immunotherapy to control tumor development and increase response to treatment. This review provides an overview of the latest published science linking mitochondrial UCP2 activity to the tumor context.
Collapse
|
7
|
Sancerni T, Renoult O, Luby A, Caradeuc C, Lenoir V, Croyal M, Ransy C, Aguilar E, Postic C, Bertho G, Dentin R, Prip-Buus C, Pecqueur C, Alves-Guerra MC. UCP2 silencing restrains leukemia cell proliferation through glutamine metabolic remodeling. Front Immunol 2022; 13:960226. [PMID: 36275699 PMCID: PMC9582289 DOI: 10.3389/fimmu.2022.960226] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Accepted: 09/20/2022] [Indexed: 11/30/2022] Open
Abstract
T-cell acute lymphoblastic leukemia (T-ALL) is an aggressive hematologic malignancy derived from early T cell progenitors. Since relapsed T-ALL is associated with a poor prognosis improving initial treatment of patients is essential to avoid resistant selection of T-ALL. During initiation, development, metastasis and even in response to chemotherapy, tumor cells face strong metabolic challenges. In this study, we identify mitochondrial UnCoupling Protein 2 (UCP2) as a tricarboxylic acid (TCA) cycle metabolite transporter controlling glutamine metabolism associated with T-ALL cell proliferation. In T-ALL cell lines, we show that UCP2 expression is controlled by glutamine metabolism and is essential for their proliferation. Our data show that T-ALL cell lines differ in their substrate dependency and their energetic metabolism (glycolysis and oxidative). Thus, while UCP2 silencing decreases cell proliferation in all leukemia cells, it also alters mitochondrial respiration of T-ALL cells relying on glutamine-dependent oxidative metabolism by rewiring their cellular metabolism to glycolysis. In this context, the function of UCP2 in the metabolite export of malate enables appropriate TCA cycle to provide building blocks such as lipids for cell growth and mitochondrial respiration. Therefore, interfering with UCP2 function can be considered as an interesting strategy to decrease metabolic efficiency and proliferation rate of leukemia cells.
Collapse
Affiliation(s)
| | | | - Angèle Luby
- Université Paris Cité, CNRS, INSERM, Institut Cochin, Paris, France
| | | | - Véronique Lenoir
- Université Paris Cité, CNRS, INSERM, Institut Cochin, Paris, France
| | - Mikael Croyal
- Nantes Université, INSERM, CNRS, CRCI2NA, Nantes, France
| | - Céline Ransy
- Université Paris Cité, CNRS, INSERM, Institut Cochin, Paris, France
| | - Esther Aguilar
- Asociación Española Contra el Cáncer (AECC), Fundación Científica AECC, Madrid, Spain
| | - Catherine Postic
- Université Paris Cité, CNRS, INSERM, Institut Cochin, Paris, France
| | | | - Renaud Dentin
- Université Paris Cité, CNRS, INSERM, Institut Cochin, Paris, France
| | - Carina Prip-Buus
- Université Paris Cité, CNRS, INSERM, Institut Cochin, Paris, France
| | | | - Marie-Clotilde Alves-Guerra
- Université Paris Cité, CNRS, INSERM, Institut Cochin, Paris, France
- *Correspondence: Marie-Clotilde Alves-Guerra,
| |
Collapse
|
8
|
Shi L, Bian Z, Kidder K, Liang H, Liu Y. Non-Lyn Src Family Kinases Activate SIRPα-SHP-1 to Inhibit PI3K-Akt2 and Dampen Proinflammatory Macrophage Polarization. THE JOURNAL OF IMMUNOLOGY 2021; 207:1419-1427. [PMID: 34348974 DOI: 10.4049/jimmunol.2100266] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Accepted: 06/24/2021] [Indexed: 12/24/2022]
Abstract
Macrophage functional plasticity plays a central role in responding to proinflammatory stimuli. The molecular basis underlying the dynamic phenotypic activation of macrophages, however, remains incompletely understood. In this article, we report that SIRPα is a chief negative regulator of proinflammatory macrophage polarization. In response to TLR agonists, proinflammatory cytokines, or canonical M1 stimulation, Src family kinases (SFK) excluding Lyn phosphorylate SIRPα ITIMs, leading to the preferential recruitment and activation of SHP-1, but not SHP-2. Solely extracellular ligation of SIRPα by CD47 does not greatly induce phosphorylation of SIRPα ITIMs, but it enhances proinflammatory stimuli-induced SIRPα phosphorylation. Examination of downstream signaling elicited by IFN-γ and TLR3/4/9 agonists found that SIRPα-activated SHP-1 moderately represses STAT1, NF-κB, and MAPK signaling but markedly inhibits Akt2, resulting in dampened proinflammatory cytokine production and expression of Ag presentation machinery. Pharmacological inhibition of SHP-1 or deficiency of SIRPα conversely attenuates SIRPα-mediated inhibition and, as such, augments macrophage proinflammatory polarization that in turn exacerbates proinflammation in mouse models of type I diabetes and peritonitis. Our results reveal an SFK-SIRPα-SHP-1 mechanism that fine-tunes macrophage proinflammatory phenotypic activation via inhibition of PI3K-Akt2, which controls the transcription and translation of proinflammatory cytokines, Ag presentation machinery, and other cellular programs.
Collapse
Affiliation(s)
- Lei Shi
- Program of Immunology and Molecular Cellular Biology, Department of Biology, Center for Diagnostics and Therapeutics, Center of Inflammation, Immunity and Infection, Georgia State University, Atlanta, GA
| | - Zhen Bian
- Program of Immunology and Molecular Cellular Biology, Department of Biology, Center for Diagnostics and Therapeutics, Center of Inflammation, Immunity and Infection, Georgia State University, Atlanta, GA
| | - Koby Kidder
- Program of Immunology and Molecular Cellular Biology, Department of Biology, Center for Diagnostics and Therapeutics, Center of Inflammation, Immunity and Infection, Georgia State University, Atlanta, GA
| | - Hongwei Liang
- Program of Immunology and Molecular Cellular Biology, Department of Biology, Center for Diagnostics and Therapeutics, Center of Inflammation, Immunity and Infection, Georgia State University, Atlanta, GA
| | - Yuan Liu
- Program of Immunology and Molecular Cellular Biology, Department of Biology, Center for Diagnostics and Therapeutics, Center of Inflammation, Immunity and Infection, Georgia State University, Atlanta, GA
| |
Collapse
|
9
|
Dang CP, Issara-Amphorn J, Charoensappakit A, Udompornpitak K, Bhunyakarnjanarat T, Saisorn W, Sae-Khow K, Leelahavanichkul A. BAM15, a Mitochondrial Uncoupling Agent, Attenuates Inflammation in the LPS Injection Mouse Model: An Adjunctive Anti-Inflammation on Macrophages and Hepatocytes. J Innate Immun 2021; 13:359-375. [PMID: 34062536 PMCID: PMC8613553 DOI: 10.1159/000516348] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Accepted: 03/30/2021] [Indexed: 11/19/2022] Open
Abstract
Controlof immune responses through the immunometabolism interference is interesting for sepsis treatment. Then, expression of immunometabolism-associated genes and BAM15, a mitochondrial uncoupling agent, was explored in a proinflammatory model using lipopolysaccharide (LPS) injection. Accordingly, the decreased expression of mitochondrial uncoupling proteins was demonstrated by transcriptomic analysis on metabolism-associated genes in macrophages (RAW246.7) and by polymerase chain reaction in LPS-stimulated RAW246.7 and hepatocytes (Hepa 1-6). Pretreatment with BAM15 at 24 h prior to LPS in macrophages attenuated supernatant inflammatory cytokines (IL-6, TNF-α, and IL-10), downregulated genes of proinflammatory M1 polarization (iNOS and IL-1β), upregulated anti-inflammatory M2 polarization (Arg1 and FIZZ), and decreased cell energy status (extracellular flux analysis and ATP production). Likewise, BAM15 decreased expression of proinflammatory genes (IL-6, TNF-α, IL-10, and iNOS) and reduced cell energy in hepatocytes. In LPS-administered mice, BAM15 attenuated serum cytokines, organ injury (liver enzymes and serum creatinine), and tissue cytokines (livers and kidneys), in part, through the enhanced phosphorylated αAMPK, a sensor of ATP depletion with anti-inflammatory property, in the liver, and reduced inflammatory monocytes/macrophages (Ly6C +ve, CD11b +ve) in the liver as detected by Western blot and flow cytometry, respectively. In conclusion, a proof of concept for inflammation attenuation of BAM15 through metabolic interference-induced anti-inflammation on macrophages and hepatocytes was demonstrated as a new strategy of anti-inflammation in sepsis.
Collapse
Affiliation(s)
- Cong Phi Dang
- Medical Microbiology, Interdisciplinary and International Program, Graduate School, Chulalongkorn University, Bangkok, Thailand,
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand,
| | | | - Awirut Charoensappakit
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Kanyarat Udompornpitak
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | | | - Wilasinee Saisorn
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Kritsanawan Sae-Khow
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Asada Leelahavanichkul
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Department of Microbiology, Translational Research in Inflammation and Immunology Research Unit (TRIRU), Chulalongkorn University, Bangkok, Thailand
| |
Collapse
|
10
|
Sha W, Hu F, Bu S. Mitochondrial dysfunction and pancreatic islet β-cell failure (Review). Exp Ther Med 2020; 20:266. [PMID: 33199991 PMCID: PMC7664595 DOI: 10.3892/etm.2020.9396] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Accepted: 07/17/2020] [Indexed: 02/07/2023] Open
Abstract
Pancreatic β-cells are the only source of insulin in humans. Mitochondria uses pyruvate to produce ATP as an intermediate link between glucose intake and insulin secretion in β-cells, in a process known as glucose-stimulated insulin secretion (GSIS). Previous studies have demonstrated that GSIS is negatively regulated by various factors in the mitochondria, including tRNALeu mutations, high p58 expression, reduced nicotinamide nucleotide transhydrogenase activity, abnormal levels of uncoupling proteins and reduced expression levels of transcription factors A, B1 and B2. Additionally, oxidative stress damages mitochondria and impairs antioxidant defense mechanisms, leading to the increased production of reactive oxygen species, which induces β-cell dysfunction. Inflammation in islets can also damage β-cell physiology. Inflammatory cytokines trigger the release of cytochrome c from the mitochondria via the NF-κB pathway. The present review examined the potential factors underlying mitochondrial dysfunction and their association with islet β-cell failure, which may offer novel insights regarding future strategies for the preservation of mitochondrial function and enhancement of antioxidant activity for individuals with diabetes mellitus.
Collapse
Affiliation(s)
- Wenxin Sha
- Diabetes Research Center, School of Medicine, Ningbo University, Ningbo, Zhejiang 315211, P.R. China
| | - Fei Hu
- Diabetes Research Center, School of Medicine, Ningbo University, Ningbo, Zhejiang 315211, P.R. China
| | - Shizhong Bu
- Diabetes Research Center, School of Medicine, Ningbo University, Ningbo, Zhejiang 315211, P.R. China
| |
Collapse
|
11
|
Ponnalagu D, Singh H. Insights Into the Role of Mitochondrial Ion Channels in Inflammatory Response. Front Physiol 2020; 11:258. [PMID: 32327997 PMCID: PMC7160495 DOI: 10.3389/fphys.2020.00258] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Accepted: 03/05/2020] [Indexed: 12/14/2022] Open
Abstract
Mitochondria are the source of many pro-inflammatory signals that cause the activation of the immune system and generate inflammatory responses. They are also potential targets of pro-inflammatory mediators, thus triggering a severe inflammatory response cycle. As mitochondria are a central hub for immune system activation, their dysfunction leads to many inflammatory disorders. Thus, strategies aiming at regulating mitochondrial dysfunction can be utilized as a therapeutic tool to cure inflammatory disorders. Two key factors that determine the structural and functional integrity of mitochondria are mitochondrial ion channels and transporters. They are not only important for maintaining the ionic homeostasis of the cell, but also play a role in regulating reactive oxygen species generation, ATP production, calcium homeostasis and apoptosis, which are common pro-inflammatory signals. The significance of the mitochondrial ion channels in inflammatory response is still not clearly understood and will need further investigation. In this article, we review the different mechanisms by which mitochondria can generate the inflammatory response as well as highlight how mitochondrial ion channels modulate these mechanisms and impact the inflammatory processes.
Collapse
Affiliation(s)
- Devasena Ponnalagu
- Department of Physiology and Cell Biology, Davis Heart and Lung Research Institute, The Ohio State University, Wexner Medical Center, Columbus, OH, United States
| | - Harpreet Singh
- Department of Physiology and Cell Biology, Davis Heart and Lung Research Institute, The Ohio State University, Wexner Medical Center, Columbus, OH, United States
| |
Collapse
|
12
|
Kaabi YA, Mansor AS, Alfagih AS, Hakami AM, Summ MA, Mjery YA, Alzughbi MN, Habibullah MM. Frequency of UCP2 45-bp Ins/Del polymorphism in Saudi population from Jazan area and its association with autoimmune hypothyroidism UCP2 45-bp Ins/Del frequency in hypothyroidism. Int J Health Sci (Qassim) 2020; 14:11-16. [PMID: 32694967 PMCID: PMC7346973] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
OBJECTIVES Autoimmune hypothyroidism (AHT) is a common endocrine disorder. Although the exact cause of AHT is not yet understood, genetic factors may play a major role. Uncoupling protein 2 (UCP2) is a member of mitochondrial protein family involved in the regulation of cellular metabolism. An important functional polymorphism in the UCP2 gene, 45-bp insertion/deletion (ins/del) polymorphism, has been linked to certain clinical conditions. However, an association between the 45-bp ins/del polymorphism and AHT has not yet been established. METHODS In this study, about 259 blood samples were collected from, patients with AHT and age-matched healthy control subjects. DNA was extracted for UCP2 45-bp ins/del polymorphisms genotyping, using a standard polymerase chain reaction technique. The distribution of different genotypes was determined in both groups and possible association with AHT was also assessed by logistic regression analysis using the Del/Del variant as a reference genotype. RESULTS The frequency of the UCP2 45-bp ins/del polymorphism in the total study population was 49.04%, 40.15%, and 10.81% for Del/Del, Ins/Del, and Ins/Ins genotypes, respectively. The logistic regression analysis showed crude odds ratios (ORs), respectively, with their 95% confidence intervals (CIs) and P-values in codominant (Del/Ins) (OR = 1.53, CI = 0.89-2.60, P = 0.17), codominant (Ins/Ins) (OR = 0.75, CI = 0.34-1.74, P = 0.53), dominant (OR = 1.30, CI = 0.79-2.16, P = 0.37), and recessive (OR = 0.62, CI = 0.29-1.36, P = 0.30) inheritance models tested, where none of which were statistically significant. CONCLUSION Our data revealed the distribution of the UCP2 45-bp ins/del polymorphisms in Jazan area and confirmed the lack of association between these genetic variants and the development of AHT.
Collapse
Affiliation(s)
- Yahia A. Kaabi
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, Jazan University, Jazan, Kingdom of Saudi Arabia
| | - Abdullah S. Mansor
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, Jazan University, Jazan, Kingdom of Saudi Arabia
| | - Ashwag S. Alfagih
- Endocrinology and Diabetes Center, King Fahad Central Hospital, Jazan, Kingdom of Saudi Arabia
| | - Alhussain M. Hakami
- Endocrinology and Diabetes Center, King Fahad Central Hospital, Jazan, Kingdom of Saudi Arabia
| | - Mohammed A. Summ
- Endocrinology and Diabetes Center, King Fahad Central Hospital, Jazan, Kingdom of Saudi Arabia
| | - Yahia A. Mjery
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, Jazan University, Jazan, Kingdom of Saudi Arabia,Address for correspondence: Yahia A. Kaabi, Department of Clinical Chemistry, Faculty of Applied Medical Sciences, Jazan University, Kingdom of Saudi Arabia. Mobile: +966 549918001. E-mail:
| | - Mona N. Alzughbi
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, Jazan University, Jazan, Kingdom of Saudi Arabia
| | - Mahmoud M. Habibullah
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, Jazan University, Jazan, Kingdom of Saudi Arabia
| |
Collapse
|
13
|
Uncoupling protein-2 regulates M1 macrophage infiltration of gingiva with periodontitis. Cent Eur J Immunol 2020; 45:9-21. [PMID: 32425675 PMCID: PMC7226558 DOI: 10.5114/ceji.2020.94664] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Accepted: 02/15/2019] [Indexed: 12/17/2022] Open
Abstract
Periodontitis is an inflammatory disease accompanied by alveolar bone loss. Moreover, M1 macrophages play a critical role in the development of periodontal disease. Uncoupling protein-2 (UCP2) is a mitochondrial transporter protein that controls M1 macrophage activation by modulating reactive oxygen species (ROS) production. We investigated the role of UCP2 in M1 macrophage infiltration in gingival tissues with periodontitis. We found that the expression of UCP2 was upregulated in M1 macrophages infiltrating human periodontal tissues with periodontitis. Macrophage-specific knockout of UCP2 could increase the infiltration of macrophage and exacerbate inflammatory response in a mouse gingiva affected with periodontitis, induced by Porphyromonas gingivalis-LPS (Pg-LPS) injection. The loss of UCP2 may contribute to the enhanced abilities of proliferation, migration, pro-inflammatory cytokine secretion, and ROS production in Pg-LPS-treated macrophages. Our results indicate that UCP2 has an important role in M1 macrophage polarization in the periodontal tissue with periodontitis. It might be helpful to provide theoretical basis for design of new therapeutic strategies for periodontitis.
Collapse
|
14
|
Mice overexpressing chromogranin A display hypergranulogenic adrenal glands with attenuated ATP levels contributing to the hypertensive phenotype. J Hypertens 2019; 36:1115-1128. [PMID: 29389743 DOI: 10.1097/hjh.0000000000001678] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVE Elevated circulating chromogranin A (CHGA) is observed in human hypertension. CHGA is critical for granulogenesis and exocytosis of catecholamine stores from secretory large dense core vesicles (LDCV). This study aims to understand the morphological, molecular and phenotypic changes because of excess CHGA and the mechanistic link eventuating in hyper-adrenergic hypertension. METHODS Blood pressure and heart rate was monitored in mouse models expressing normal and elevated level of CHGA by telemetry. Catecholamine and oxidative stress radicals were measured. Adrenal ultrastructure, LDCV content and mitochondrial abundance were compared and respiration analyzed by Seahorse assay. Effect of CHGA dosage on adrenal ATP content, electron transport chain components and uncoupling protein 2 (UCP-2) were compared in vivo and in vitro. RESULTS Mice with excess-CHGA displayed hypertensive phenotype, higher heart rate and increased sympathetic tone. They had elevated plasma catecholamine and adrenal ROS levels. Excess-CHGA caused an increase in size and abundance of LDCV and adrenal mitochondria. Nonetheless, they had attenuated levels of ATP. Isolated adrenal mitochondria from mice with elevated CHGA showed higher maximal respiration rates in the presence of protonophore, which uncouples oxidative phosphorylation. Elevated CHGA resulted in overexpression of UCP2 and diminished ATP. In vitro in chromaffin cells overexpressing CHGA, concomitant increase in UCP2 protein and decreased ATP was detected. CONCLUSION Elevated CHGA expression resulted in underlying bioenergetic dysfunction in ATP production despite higher mitochondrial mass. The outcome was unregulated negative feedback of LDCV exocytosis and secretion, resulting in elevated levels of circulating catecholamine and consequently the hypertensive phenotype.
Collapse
|
15
|
Pei Y, Cui F, Du X, Shang G, Xiao W, Yang X, Cui Q. Antioxidative nanofullerol inhibits macrophage activation and development of osteoarthritis in rats. Int J Nanomedicine 2019; 14:4145-4155. [PMID: 31239673 PMCID: PMC6559768 DOI: 10.2147/ijn.s202466] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Accepted: 04/16/2019] [Indexed: 12/28/2022] Open
Abstract
Background: There is emerging evidence which suggests that cellular ROS including nitric oxide (NO) are important mediators for inflammation and osteoarthritis (OA). Water-soluble polyhydroxylated fullerene C60 (fullerol) nanoparticle has been demonstrated to have an outstanding ability to scavenge ROS. Purpose: The objective of this study is to assess the effects of fullerol on inflammation and OA by in vitro and in vivo studies. Methods: For in vitro experiments, primary mouse peritoneal macrophages and a macrophage cell line RAW264.7 were stimulated to inflammatory phenotypes by lipopolysaccharide (LPS) in the presence of fullerol. For the animal study, OA model was created by intra-articular injection of monoiodoacetate into the knee joints of rats and fullerol was intravenously injected immediately after OA induction. Results: NO production and pro-inflammatory gene expression induced by LPS was significantly diminished by fullerol in both macrophage cell types. Meanwhile, fullerol could remarkably reduce phosphorylation of p38 mitogen-activated protein kinase, and protein level of transcription factors nuclear factor-kappaB and forkhead box transcription factor 1 within the nucleus. The animal study delineated that systematic administration of fullerol prevented OA, inhibiting inflammation of synovial membranes and the damage toward the cartilage chondrocytes in the OA joints. Conclusion: Antioxidative fullerol may have a potential therapeutic application for OA.
Collapse
Affiliation(s)
- Yilun Pei
- Orthopaedic Research Lab, University of Virginia, Charlottesville, VA, USA
| | - Fuai Cui
- Orthopaedic Research Lab, University of Virginia, Charlottesville, VA, USA.,Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong, China
| | - Xuejun Du
- Orthopaedic Research Lab, University of Virginia, Charlottesville, VA, USA
| | - Guowei Shang
- Orthopaedic Research Lab, University of Virginia, Charlottesville, VA, USA
| | - Wanan Xiao
- Orthopaedic Research Lab, University of Virginia, Charlottesville, VA, USA
| | - Xinlin Yang
- Orthopaedic Research Lab, University of Virginia, Charlottesville, VA, USA
| | - Quanjun Cui
- Orthopaedic Research Lab, University of Virginia, Charlottesville, VA, USA
| |
Collapse
|
16
|
Nigro M, De Sanctis C, Formisano P, Stanzione R, Forte M, Capasso G, Gigliotti G, Rubattu S, Viggiano D. Cellular and subcellular localization of uncoupling protein 2 in the human kidney. J Mol Histol 2018; 49:437-445. [DOI: 10.1007/s10735-018-9782-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Accepted: 06/18/2018] [Indexed: 01/20/2023]
|
17
|
Kapnick SM, Pacheco SE, McGuire PJ. The emerging role of immune dysfunction in mitochondrial diseases as a paradigm for understanding immunometabolism. Metabolism 2018; 81:97-112. [PMID: 29162500 PMCID: PMC5866745 DOI: 10.1016/j.metabol.2017.11.010] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Revised: 11/07/2017] [Accepted: 11/11/2017] [Indexed: 01/08/2023]
Abstract
Immunometabolism aims to define the role of intermediary metabolism in immune cell function, with bioenergetics and the mitochondria recently taking center stage. To date, the medical literature on mitochondria and immune function extols the virtues of mouse models in exploring this biologic intersection. While the laboratory mouse has become a standard for studying mammalian biology, this model comprises part of a comprehensive approach. Humans, with their broad array of inherited phenotypes, serve as a starting point for studying immunometabolism; specifically, patients with mitochondrial disease. Using this top-down approach, the mouse as a model organism facilitates further exploration of the consequences of mutations involved in mitochondrial maintenance and function. In this review, we will discuss the emerging phenotype of immune dysfunction in mitochondrial disease as a model for understanding the role of the mitochondria in immune function in available mouse models.
Collapse
Affiliation(s)
- Senta M Kapnick
- Metabolism, Infection and Immunity Section, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Susan E Pacheco
- Department of Pediatrics, The University of Texas Health Science Center, Houston, TX, USA
| | - Peter J McGuire
- Metabolism, Infection and Immunity Section, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
18
|
Broche B, Ben Fradj S, Aguilar E, Sancerni T, Bénard M, Makaci F, Berthault C, Scharfmann R, Alves-Guerra MC, Duvillié B. Mitochondrial Protein UCP2 Controls Pancreas Development. Diabetes 2018; 67:78-84. [PMID: 29079704 DOI: 10.2337/db17-0118] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Accepted: 10/23/2017] [Indexed: 11/13/2022]
Abstract
The mitochondrial carrier uncoupling protein (UCP) 2 belongs to the family of the UCPs. Despite its name, it is now accepted that UCP2 is rather a metabolite transporter than a UCP. UCP2 can regulate oxidative stress and/or energetic metabolism. In rodents, UCP2 is involved in the control of α- and β-cell mass as well as insulin and glucagon secretion. Our aim was to determine whether the effects of UCP2 observed on β-cell mass have an embryonic origin. Thus, we used Ucp2 knockout mice. We found an increased size of the pancreas in Ucp2-/- fetuses at embryonic day 16.5, associated with a higher number of α- and β-cells. This phenotype was caused by an increase of PDX1+ progenitor cells. Perinatally, an increase in the proliferation of endocrine cells also participates in their expansion. Next, we analyzed the oxidative stress in the pancreata. We quantified an increased nuclear translocation of nuclear factor erythroid 2-related factor 2 (NRF2) in the mutant, suggesting an increased production of reactive oxygen species (ROS). Phosphorylation of AKT, an ROS target, was also activated in the Ucp2-/- pancreata. Finally, administration of the antioxidant N-acetyl-l-cysteine to Ucp2-/- pregnant mice alleviated the effect of knocking out UCP2 on pancreas development. Together, these data demonstrate that UCP2 controls pancreas development through the ROS-AKT signaling pathway.
Collapse
Affiliation(s)
- Benjamin Broche
- INSERM, U1016, Institut Cochin, Paris, France
- CNRS, UMR8104, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Selma Ben Fradj
- INSERM, U1016, Institut Cochin, Paris, France
- CNRS, UMR8104, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Esther Aguilar
- INSERM, U1016, Institut Cochin, Paris, France
- CNRS, UMR8104, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Tiphaine Sancerni
- INSERM, U1016, Institut Cochin, Paris, France
- CNRS, UMR8104, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France
- Université Paris Diderot, Sorbonne Paris Cité, Paris, France
| | - Matthieu Bénard
- INSERM, U1016, Institut Cochin, Paris, France
- CNRS, UMR8104, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Fatna Makaci
- INSERM, U1016, Institut Cochin, Paris, France
- CNRS, UMR8104, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Claire Berthault
- INSERM, U1016, Institut Cochin, Paris, France
- CNRS, UMR8104, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Raphaël Scharfmann
- INSERM, U1016, Institut Cochin, Paris, France
- CNRS, UMR8104, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Marie-Clotilde Alves-Guerra
- INSERM, U1016, Institut Cochin, Paris, France
- CNRS, UMR8104, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Bertrand Duvillié
- INSERM, U1016, Institut Cochin, Paris, France
- CNRS, UMR8104, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| |
Collapse
|
19
|
Sreedhar A, Zhao Y. Uncoupling protein 2 and metabolic diseases. Mitochondrion 2017; 34:135-140. [PMID: 28351676 PMCID: PMC5477468 DOI: 10.1016/j.mito.2017.03.005] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Revised: 03/22/2017] [Accepted: 03/24/2017] [Indexed: 02/06/2023]
Abstract
Mitochondria are fascinating organelles involved in various cellular-metabolic activities that are integral for mammalian development. Although they perform diverse, yet interconnected functions, mitochondria are remarkably regulated by complex signaling networks. Therefore, it is not surprising that mitochondrial dysfunction is involved in plethora of diseases, including neurodegenerative and metabolic disorders. One of the many factors that lead to mitochondrial-associated metabolic diseases is the uncoupling protein-2, a family of mitochondrial anion proteins present in the inner mitochondrial membrane. Since their discovery, uncoupling proteins have attracted considerable attention due to their involvement in mitochondrial-mediated oxidative stress and energy metabolism. This review attempts to provide a summary of recent developments in the field of uncoupling protein 2 relating to mitochondrial associated metabolic diseases.
Collapse
Affiliation(s)
- Annapoorna Sreedhar
- Department of Pharmacology, Toxicology & Neuroscience, LSU Health Sciences Center in Shreveport, Shreveport, LA 71130, USA
| | - Yunfeng Zhao
- Department of Pharmacology, Toxicology & Neuroscience, LSU Health Sciences Center in Shreveport, Shreveport, LA 71130, USA.
| |
Collapse
|
20
|
Iacobazzi V, Infantino V, Castegna A, Menga A, Palmieri EM, Convertini P, Palmieri F. Mitochondrial carriers in inflammation induced by bacterial endotoxin and cytokines. Biol Chem 2017; 398:303-317. [PMID: 27727142 DOI: 10.1515/hsz-2016-0260] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Accepted: 10/02/2016] [Indexed: 12/18/2022]
Abstract
Significant metabolic changes occur in the shift from resting to activated cellular status in inflammation. Thus, changes in expression of a large number of genes and extensive metabolic reprogramming gives rise to acquisition of new functions (e.g. production of cytokines, intermediates for biosynthesis, lipid mediators, PGE, ROS and NO). In this context, mitochondrial carriers, which catalyse the transport of solute across mitochondrial membrane, change their expression to transport mitochondrially produced molecules, among which citrate and succinate, to be used as intracellular signalling molecules in inflammation. This review summarises the mitochondrial carriers studied so far that are, directly or indirectly, involved in inflammation.
Collapse
|
21
|
Wu R, Nakatsu G, Zhang X, Yu J. Pathophysiological mechanisms and therapeutic potentials of macrophages in non-alcoholic steatohepatitis. Expert Opin Ther Targets 2016; 20:615-26. [PMID: 26609894 DOI: 10.1517/14728222.2016.1125883] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
INTRODUCTION Non-alcoholic steatohepatitis (NASH), a hepatic manifestation of metabolic syndrome, is a major cause of morbidity and healthcare burden worldwide. While the molecular pathogenesis of NASH remains unclear and therapeutic options are limited, inflammation is recognized as an essential factor for NASH development. Factors that link NASH to inflammation are macrophages and their secreted cytokines. AREAS COVERED This review summarizes the current knowledge of macrophage-mediated molecular pathways in NASH to shed insights on potential pharmacotherapeutic applications. EXPERT OPINION Macrophages are not only known for their role of phagocytosis in innate immunity, but also for both extrinsic and intrinsic regulation of inflammatory functions of many cytokines. Recent advances have revealed the effects of macrophage recruitment and polarization on the development of NASH. We and others have shown that the proliferation of hepatic macrophages and the subsequent production of pro-inflammatory cytokines initiates inflammatory cascades, orchestrates activities of transcription factors involved in lipid metabolism/translocation, and modulates programmed cell death. Together, these findings support the pathophysiological role of macrophages in the pathogenesis of NASH. Thus, evaluating potential therapeutic targets against the infiltration and/or polarization of specific macrophage subtypes is of clinical interest for alleviation of early-stage NASH, with the goal of halting disease progression.
Collapse
Affiliation(s)
- Ruonan Wu
- a Institute of Digestive Disease and the Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences , CUHK-Shenzhen Research Institute, The Chinese University of Hong Kong , Shatin , Hong Kong
| | - Geicho Nakatsu
- a Institute of Digestive Disease and the Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences , CUHK-Shenzhen Research Institute, The Chinese University of Hong Kong , Shatin , Hong Kong
| | - Xiang Zhang
- a Institute of Digestive Disease and the Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences , CUHK-Shenzhen Research Institute, The Chinese University of Hong Kong , Shatin , Hong Kong
| | - Jun Yu
- a Institute of Digestive Disease and the Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences , CUHK-Shenzhen Research Institute, The Chinese University of Hong Kong , Shatin , Hong Kong
| |
Collapse
|
22
|
Jahansouz C. Adipocyte Dysfunction, Inflammation, and Insulin Resistance in Obesity. METABOLIC SYNDROME AND DIABETES 2016:61-80. [DOI: 10.1007/978-1-4939-3220-7_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
23
|
Aslan Y, Kader Ş, Mutlu M, Sarıaydın M, Aran T, Alver A, Kahraman C. The effect of delivery type on uncoupling protein-2 levels. J Matern Fetal Neonatal Med 2015; 29:2940-3. [PMID: 26479203 DOI: 10.3109/14767058.2015.1109619] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
OBJECTIVE Uncoupling proteins (UCPs) are carrier proteins located in the mitochondrial inner membrane that disturb the proton gradient by re-transporting protons and that thus inhibit ATP synthesis. UCP-2 is found in in several tissues, particularly the brain. This study was performed to examine the effects of mode of delivery on UCP-2 in humans. METHODS The study was performed prospectively. Cord blood specimens were collected for measurement of blood gasses, full-blood count, total and direct bilirubin levels and UCP-2. UCP-2 levels were measured using an enzyme-linked immunosorbent assay (ELISA) kit. Results were expressed as nanogram per milliliter. RESULTS The study was performed with 120 healthy term babies, 60 born by normal spontaneous vaginal delivery (NSVD) and 60 by cesarean/section (C/S). There was significant difference in UCP-2 levels between the two groups. UCP-2 levels were significantly higher in the cases born by NSVD then in the cases born by C/S. CONCLUSION This study showed that a correlation exists between mode of delivery and UCP-2 in humans. As UCP-2 is described as playing a significant role in the formation of nerve cells and deficiency of this protein during development of the brain may lead to behavioral problems extending to adulthood, we think that increasing UCP-2 levels through normal delivery will protect all organs, and particularly the brain, against oxidative damage and play a role in preventing organ dysfunctions.
Collapse
Affiliation(s)
- Yakup Aslan
- a Department of Neonatology , Karadeniz Technical University, Faculty of Medicine , Trabzon , Turkey
| | - Şebnem Kader
- a Department of Neonatology , Karadeniz Technical University, Faculty of Medicine , Trabzon , Turkey
| | - Mehmet Mutlu
- a Department of Neonatology , Karadeniz Technical University, Faculty of Medicine , Trabzon , Turkey
| | - Mehmet Sarıaydın
- a Department of Neonatology , Karadeniz Technical University, Faculty of Medicine , Trabzon , Turkey
| | - Turhan Aran
- b Department of Gynecology and Obstetrics , Karadeniz Technical University, Faculty of Medicine , Trabzon , Turkey , and
| | - Ahmet Alver
- c Department of Medical Biochemistry , Karadeniz Technical University, Faculty of Medicine , Trabzon , Turkey
| | - Cemil Kahraman
- c Department of Medical Biochemistry , Karadeniz Technical University, Faculty of Medicine , Trabzon , Turkey
| |
Collapse
|
24
|
Jain D, Weber G, Eberhard D, Mehana AE, Eglinger J, Welters A, Bartosinska B, Jeruschke K, Weiss J, Päth G, Ariga H, Seufert J, Lammert E. DJ-1 Protects Pancreatic Beta Cells from Cytokine- and Streptozotocin-Mediated Cell Death. PLoS One 2015; 10:e0138535. [PMID: 26422139 PMCID: PMC4589499 DOI: 10.1371/journal.pone.0138535] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2015] [Accepted: 09/01/2015] [Indexed: 01/07/2023] Open
Abstract
A hallmark feature of type 1 and type 2 diabetes mellitus is the progressive dysfunction and loss of insulin-producing pancreatic beta cells, and inflammatory cytokines are known to trigger beta cell death. Here we asked whether the anti-oxidant protein DJ-1 encoded by the Parkinson’s disease gene PARK7 protects islet cells from cytokine- and streptozotocin-mediated cell death. Wild type and DJ-1 knockout mice (KO) were treated with multiple low doses of streptozotocin (MLDS) to induce inflammatory beta cell stress and cell death. Subsequently, glucose tolerance tests were performed, and plasma insulin as well as fasting and random blood glucose concentrations were monitored. Mitochondrial morphology and number of insulin granules were quantified in beta cells. Moreover, islet cell damage was determined in vitro after streptozotocin and cytokine treatment of isolated wild type and DJ-1 KO islets using calcein AM/ethidium homodimer-1 staining and TUNEL staining. Compared to wild type mice, DJ-1 KO mice became diabetic following MLDS treatment. Insulin concentrations were substantially reduced, and fasting blood glucose concentrations were significantly higher in MLDS-treated DJ-1 KO mice compared to equally treated wild type mice. Rates of beta cell apoptosis upon MLDS treatment were twofold higher in DJ-1 KO mice compared to wild type mice, and in vitro inflammatory cytokines led to twice as much beta cell death in pancreatic islets from DJ-1 KO mice versus those of wild type mice. In conclusion, this study identified the anti-oxidant protein DJ-1 as being capable of protecting pancreatic islet cells from cell death induced by an inflammatory and cytotoxic setting.
Collapse
Affiliation(s)
- Deepak Jain
- Institute of Metabolic Physiology, Heinrich Heine University, Düsseldorf, Germany
- Institute for Beta Cell Biology, German Diabetes Center at Heinrich Heine University, Leibniz Center for Diabetes Research, Düsseldorf, Germany
- German Center for Diabetes Research (DZD e.V.), Düsseldorf Partner Institute, Düsseldorf, Germany
| | - Gesine Weber
- Institute of Metabolic Physiology, Heinrich Heine University, Düsseldorf, Germany
| | - Daniel Eberhard
- Institute of Metabolic Physiology, Heinrich Heine University, Düsseldorf, Germany
- Institute for Beta Cell Biology, German Diabetes Center at Heinrich Heine University, Leibniz Center for Diabetes Research, Düsseldorf, Germany
| | - Amir E. Mehana
- Division of Endocrinology and Diabetology, Department of Internal Medicine II, University Hospital of Freiburg, Freiburg, Germany
- Department of Zoology, Faculty of Science, Suez Canal University, Ismailia, Egypt
| | - Jan Eglinger
- Institute of Metabolic Physiology, Heinrich Heine University, Düsseldorf, Germany
- Institute for Beta Cell Biology, German Diabetes Center at Heinrich Heine University, Leibniz Center for Diabetes Research, Düsseldorf, Germany
- German Center for Diabetes Research (DZD e.V.), Düsseldorf Partner Institute, Düsseldorf, Germany
| | - Alena Welters
- Institute of Metabolic Physiology, Heinrich Heine University, Düsseldorf, Germany
- Institute for Beta Cell Biology, German Diabetes Center at Heinrich Heine University, Leibniz Center for Diabetes Research, Düsseldorf, Germany
- German Center for Diabetes Research (DZD e.V.), Düsseldorf Partner Institute, Düsseldorf, Germany
- Department of General Pediatrics, Neonatology and Pediatric Cardiology, University Children’s Hospital Düsseldorf, Düsseldorf, Germany
| | - Barbara Bartosinska
- Institute of Metabolic Physiology, Heinrich Heine University, Düsseldorf, Germany
| | - Kay Jeruschke
- Institute of Clinical Biochemistry and Pathobiochemistry, German Diabetes Center at Heinrich Heine University, Leibniz Center for Diabetes Research, Düsseldorf, Germany
| | - Jürgen Weiss
- Institute of Clinical Biochemistry and Pathobiochemistry, German Diabetes Center at Heinrich Heine University, Leibniz Center for Diabetes Research, Düsseldorf, Germany
| | - Günter Päth
- Division of Endocrinology and Diabetology, Department of Internal Medicine II, University Hospital of Freiburg, Freiburg, Germany
| | - Hiroyoshi Ariga
- Graduate School of Pharmaceutical Sciences, Hokkaido University, Kita-Ku, N12 W6, Sapporo, Japan
| | - Jochen Seufert
- Division of Endocrinology and Diabetology, Department of Internal Medicine II, University Hospital of Freiburg, Freiburg, Germany
| | - Eckhard Lammert
- Institute of Metabolic Physiology, Heinrich Heine University, Düsseldorf, Germany
- Institute for Beta Cell Biology, German Diabetes Center at Heinrich Heine University, Leibniz Center for Diabetes Research, Düsseldorf, Germany
- German Center for Diabetes Research (DZD e.V.), Düsseldorf Partner Institute, Düsseldorf, Germany
- * E-mail:
| |
Collapse
|
25
|
Rajanbabu V, Galam L, Fukumoto J, Enciso J, Tadikonda P, Lane TN, Bandyopadhyay S, Parthasarathy PT, Cho Y, Cho SH, Lee YC, Lockey RF, Kolliputi N. Genipin suppresses NLRP3 inflammasome activation through uncoupling protein-2. Cell Immunol 2015; 297:40-5. [PMID: 26123077 DOI: 10.1016/j.cellimm.2015.06.002] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2014] [Revised: 06/06/2015] [Accepted: 06/09/2015] [Indexed: 11/26/2022]
Abstract
Incomplete clearance of apoptotic cells and reactive oxygen species (ROS) release are known to trigger inflammasome activation causing severe inflammation in acute lung injury and various metabolic and autoimmune diseases. Moreover, it has been reported that apoptotic cell clearance and ROS-mediated apoptosis critically depend on mitochondrial uncoupling protein-2 (UCP2). However, the relationship between UCP2 and inflammasome activation has not been studied. This report investigates the role of UCP2 in the expression and activation of NACHT, LRR and PYD domains-containing protein 3 (NLRP3) inflammasome in human macrophages. We found that UCP2 overexpression significantly enhanced the expression levels of NLRP3. The NLRP3 expression levels were significantly suppressed in THP1 cells treated with genipin, a UCP2 inhibitor, compared to controls. In addition, genipin altered adenosine triphosphate (ATP)- and hydrogen peroxide (H2O2)-mediated interleukin-1 beta (IL-1β) secretion and significantly suppressed caspase-1 activity in inflammasome-activated human macrophages. Taken together, our results suggest that genipin modulates NLRP3 inflammasome activation and ATP- or H2O2-mediated IL-1β release.
Collapse
Affiliation(s)
- Venugopal Rajanbabu
- Division of Allergy and Immunology, Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
| | - Lakshmi Galam
- Division of Allergy and Immunology, Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
| | - Jutaro Fukumoto
- Division of Allergy and Immunology, Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
| | - Juan Enciso
- Division of Allergy and Immunology, Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
| | - Pratima Tadikonda
- Division of Allergy and Immunology, Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
| | - Troy N Lane
- Division of Allergy and Immunology, Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
| | - Sayantani Bandyopadhyay
- Division of Allergy and Immunology, Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
| | - Prasanna Tamarapu Parthasarathy
- Division of Allergy and Immunology, Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
| | - Young Cho
- Division of Allergy and Immunology, Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
| | - Seong Ho Cho
- Division of Allergy and Immunology, Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
| | - Yong Chul Lee
- Department of Internal Medicine, Research Center for Pulmonary Disorders, Chonbuk National University Medical School, Jeonju, South Korea
| | - Richard F Lockey
- Division of Allergy and Immunology, Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
| | - Narasaiah Kolliputi
- Division of Allergy and Immunology, Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA.
| |
Collapse
|
26
|
Alteration of UCP2 and ZO-1 expression in trabecular meshwork of neovascular glaucoma patients. J Glaucoma 2015; 24:291-6. [PMID: 23835672 DOI: 10.1097/ijg.0b013e31829d9b91] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
PURPOSE The aim of this study was to investigate the expression of uncoupling protein 2 (UCP2) and zonula occludens-1 (ZO-1) in the trabecular meshwork (TM) of neovascular glaucoma (NVG) patients treated with trabeculectomy. MATERIALS AND METHODS Six eyes with NVG underwent trabeculectomy for therapeutic purposes. The data consisted of patient demographics, presurgical and postsurgical visual acuity, intraocular pressure, gonioscopy, and neovascularization of iris and/or the anterior chamber angle. TM samples were obtained from the NVG eyes that had undergone surgery. Immunofluorescence and confocal laser scanning microscopy were carried out to determine the expression of UCP2 and ZO-1 in the TM cells. RESULTS The baseline median visual acuity was light perception, and the mean intraocular pressure (standard error) was 52.5 (8.3) mm Hg. All eyes displayed neovascularization of the iris and the anterior chamber angle. The expression of UCP2 was significantly decreased in TM cells of NVG compared with the control (P=0.000), whereas increase in ZO-1 expression was detected in staining cells with NVG in comparison with the control (P=0.000). The necrotic cells in the TM were increased (P=0.000), whereas the viable cells were reduced (P=0.000) in comparison with the control. CONCLUSIONS The decreased UCP2 expression and increased ZO-1 expression suggest that the oxidative stress-induced mitochondrial dysfunction and tight junction formation may play pivotal roles in the progress of NVG.
Collapse
|
27
|
Abstract
Congenital hyperinsulinism (CHI) is a complex heterogeneous condition in which insulin secretion from pancreatic β-cells is unregulated and inappropriate for the level of blood glucose. The inappropriate insulin secretion drives glucose into the insulin-sensitive tissues, such as the muscle, liver and adipose tissue, leading to severe hyperinsulinaemic hypoglycaemia (HH). At a molecular level, genetic abnormalities in nine different genes (ABCC8, KCNJ11, GLUD1, GCK, HNF4A, HNF1A, SLC16A1, UCP2 and HADH) have been identified which cause CHI. Autosomal recessive and dominant mutations in ABCC8/KCNJ11 are the commonest cause of medically unresponsive CHI. Mutations in GLUD1 and HADH lead to leucine-induced HH, and these two genes encode the key enzymes glutamate dehydrogenase and short chain 3-hydroxyacyl-CoA dehydrogenase which play a key role in amino acid and fatty acid regulation of insulin secretion respectively. Genetic abnormalities in HNF4A and HNF1A lead to a dual phenotype of HH in the newborn period and maturity onset-diabetes later in life. This state of the art review provides an update on the molecular basis of CHI.
Collapse
Affiliation(s)
- Sofia A Rahman
- Genetics and Genomic MedicineUCL Institute of Child Health, 30 Guilford Street, London WC1N 1EH, UKDepartment of Paediatric EndocrinologyGreat Ormond Street Hospital for Children NHS, 30 Guilford Street, London WC1N 1EH, UK
| | - Azizun Nessa
- Genetics and Genomic MedicineUCL Institute of Child Health, 30 Guilford Street, London WC1N 1EH, UKDepartment of Paediatric EndocrinologyGreat Ormond Street Hospital for Children NHS, 30 Guilford Street, London WC1N 1EH, UK
| | - Khalid Hussain
- Genetics and Genomic MedicineUCL Institute of Child Health, 30 Guilford Street, London WC1N 1EH, UKDepartment of Paediatric EndocrinologyGreat Ormond Street Hospital for Children NHS, 30 Guilford Street, London WC1N 1EH, UK Genetics and Genomic MedicineUCL Institute of Child Health, 30 Guilford Street, London WC1N 1EH, UKDepartment of Paediatric EndocrinologyGreat Ormond Street Hospital for Children NHS, 30 Guilford Street, London WC1N 1EH, UK
| |
Collapse
|
28
|
Flouris AD, Piantoni C. Links between thermoregulation and aging in endotherms and ectotherms. Temperature (Austin) 2014; 2:73-85. [PMID: 27226994 PMCID: PMC4843886 DOI: 10.4161/23328940.2014.989793] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2014] [Revised: 10/21/2014] [Accepted: 11/13/2014] [Indexed: 01/22/2023] Open
Abstract
While the link between thermoregulation and aging is generally accepted, much further research, reflection, and debate is required to elucidate the physiological and molecular pathways that generate the observed thermal-induced changes in lifespan. Our aim in this review is to present, discuss, and scrutinize the thermoregulatory mechanisms that are implicated in the aging process in endotherms and ectotherms. Our analysis demonstrates that low body temperature benefits lifespan in both endothermic and ectothermic organisms. Research in endotherms has delved deeper into the physiological and molecular mechanisms linking body temperature and longevity. While research in ectotherms has been steadily increasing during the past decades, further mechanistic work is required in order to fully elucidate the underlying phenomena. What is abundantly clear is that both endotherms and ectotherms have a specific temperature zone at which they function optimally. This zone is defended through both physiological and behavioral means and plays a major role on organismal senescence. That low body temperature may be beneficial for lifespan is contrary to conventional medical theory where reduced body temperature is usually considered as a sign of underlying pathology. Regardless, this phenomenon has been targeted by scientists with the expectation that advancements may compress morbidity, as well as lower disease and mortality risk. The available evidence suggests that lowered body temperature may prolong life span, yet finding the key to temperature regulation remains the problem. While we are still far from a complete understanding of the mechanisms linking body temperature and longevity, we are getting closer.
Collapse
Affiliation(s)
- Andreas D Flouris
- FAME Laboratory; Department of Exercise Science; University of Thessaly ; Trikala, Greece
| | - Carla Piantoni
- University of Sao Paulo; Department of Physiology ; Sao Paulo, Brazil
| |
Collapse
|
29
|
Jantsch J, Schödel J. Hypoxia and hypoxia-inducible factors in myeloid cell-driven host defense and tissue homeostasis. Immunobiology 2014; 220:305-14. [PMID: 25439732 DOI: 10.1016/j.imbio.2014.09.009] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2014] [Revised: 08/01/2014] [Accepted: 09/05/2014] [Indexed: 02/08/2023]
Abstract
The impact of tissue oxygenation and hypoxia on immune cells has been recognized as a major determinant of host defense and tissue homeostasis. In this review, we will summarize the available data on tissue oxygenation in inflamed and infected tissue and the effect of low tissue oxygenation on myeloid cell function. Furthermore, we will highlight effects of the master regulators of the cellular hypoxic response, hypoxia-inducible transcription factors (HIF), in myeloid cells in antimicrobial defense and tissue homeostasis.
Collapse
Affiliation(s)
- Jonathan Jantsch
- Institut für Klinische Mikrobiologie und Hygiene, Universitätsklinikum Regensburg, Germany; Mikrobiologisches Institut - Klinische Mikrobiologie, Immunologie und Hygiene, Universitätsklinikum Erlangen und Friedrich-Alexander Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany.
| | - Johannes Schödel
- Medizinische Klinik 4, Universitätsklinikum Erlangen und Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany; Translational Research Center, Universitätsklinikum Erlangen und Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany.
| |
Collapse
|
30
|
Donadelli M, Dando I, Fiorini C, Palmieri M. UCP2, a mitochondrial protein regulated at multiple levels. Cell Mol Life Sci 2014; 71:1171-90. [PMID: 23807210 PMCID: PMC11114077 DOI: 10.1007/s00018-013-1407-0] [Citation(s) in RCA: 125] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2013] [Revised: 05/16/2013] [Accepted: 06/10/2013] [Indexed: 12/11/2022]
Abstract
An ever-increasing number of studies highlight the role of uncoupling protein 2 (UCP2) in a broad range of physiological and pathological processes. The knowledge of the molecular mechanisms of UCP2 regulation is becoming fundamental in both the comprehension of UCP2-related physiological events and the identification of novel therapeutic strategies based on UCP2 modulation. The study of UCP2 regulation is a fast-moving field. Recently, several research groups have made a great effort to thoroughly understand the various molecular mechanisms at the basis of UCP2 regulation. In this review, we describe novel findings concerning events that can occur in a concerted manner at various levels: Ucp2 gene mutation (single nucleotide polymorphisms), UCP2 mRNA and protein expression (transcriptional, translational, and protein turn-over regulation), UCP2 proton conductance (ligands and post-transcriptional modifications), and nutritional and pharmacological regulation of UCP2.
Collapse
Affiliation(s)
- Massimo Donadelli
- Section of Biochemistry, Deparment of Life and Reproduction Sciences, University of Verona, Strada Le Grazie 8, 37134, Verona, Italy,
| | | | | | | |
Collapse
|
31
|
Uncoupling protein 2 deficiency aggravates astrocytic endoplasmic reticulum stress and nod-like receptor protein 3 inflammasome activation. Neurobiol Aging 2014; 35:421-30. [DOI: 10.1016/j.neurobiolaging.2013.08.015] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2013] [Revised: 08/02/2013] [Accepted: 08/16/2013] [Indexed: 01/07/2023]
|
32
|
Cao T, Dong Y, Tang R, Chen J, Zhang CY, Zen K. Mitochondrial uncoupling protein 2 protects splenocytes from oxidative stress-induced apoptosis during pathogen activation. Cell Immunol 2013; 286:39-44. [PMID: 24291389 DOI: 10.1016/j.cellimm.2013.10.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2013] [Revised: 09/30/2013] [Accepted: 10/10/2013] [Indexed: 11/27/2022]
Abstract
Accumulating evidences suggested that mitochondrial uncoupling protein 2 (UCP2) is involved in host defense in parasite infection, inflammation, and autoimmune responses. However, it remains unknown whether UCP2 is participated in the modulation of humoral immune response. Here we used quantitative PCR, ELISA, TUNEL assay, flow cytometry, etc. to study the role of UCP2 in spleen B lymphocytes during pathogen activation and obtained following results. First, UCP2 is highly expressed in splenocytes and its expression level in splenocytes is rapidly increased when the cells are activated by lipopolysaccharide (LPS) in vivo or by LPS plus cytokines in vitro. Second, in contrast to the wild type (WT) littermates, the UCP2 knockout (UCP2-KO) mice show an impaired humoral immune response when they are challenged by pathogen. Although UCP2-KO mice produce a normal level of IgM, the levels of IgGs are significantly less than those of WT littermates. Third, splenocytes from UCP2-KO mice are more susceptible to pathogen activation-induced apoptosis, and the high level of reactive oxygen species (ROS) in UCP2-KO mice may be the cause for the apoptosis. In conclusion, our study demonstrates that mitochondrial UCP2 plays a critical role in protecting splenocytes from oxidative stress-induced apoptosis during pathogen activation.
Collapse
Affiliation(s)
- Ting Cao
- Jiangsu Engineering Research Center for MicroRNA Biology and Biotechnology, State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Jiangsu 210093, China; Marine Biology Lab, School of Life Sciences, Nanjing University, Jiangsu 210093, China
| | - Yeyan Dong
- Jiangsu Engineering Research Center for MicroRNA Biology and Biotechnology, State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Jiangsu 210093, China
| | - Rui Tang
- Jiangsu Engineering Research Center for MicroRNA Biology and Biotechnology, State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Jiangsu 210093, China; Marine Biology Lab, School of Life Sciences, Nanjing University, Jiangsu 210093, China
| | - Junyuan Chen
- Marine Biology Lab, School of Life Sciences, Nanjing University, Jiangsu 210093, China
| | - Chen-Yu Zhang
- Jiangsu Engineering Research Center for MicroRNA Biology and Biotechnology, State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Jiangsu 210093, China
| | - Ke Zen
- Jiangsu Engineering Research Center for MicroRNA Biology and Biotechnology, State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Jiangsu 210093, China.
| |
Collapse
|
33
|
Patel N, Barrientos A, Landgraf R. The growth factor receptor ERBB2 regulates mitochondrial activity on a signaling time scale. J Biol Chem 2013; 288:35253-65. [PMID: 24142693 DOI: 10.1074/jbc.m113.478271] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Overexpression of the ERBB2 receptor tyrosine kinase and the mitochondrial inner membrane protein UCP2 occurs frequently in aggressive cancers with dysfunctional mitochondria. Overexpressed ERBB2 signals constitutively and elevated UCP2 can uncouple mitochondria and alleviate oxidative stress. However, the physiological contributions of UCP2 and ERBB2 at the low expression levels that are typical of most tissues, as well as the path to oncogenic deregulation, are poorly understood. We now show that ERBB2 directly controls UCP2 levels, both at low physiological levels and oncogenic overexpression. At low levels of receptor and UCP2, ligand stimulation creates a distinct temporal response pattern driven by the opposing forces of translational suppression of the exceptionally short lived UCP2 protein and a time delayed transcriptional up-regulation. The latter becomes dominant through constitutive signaling by overexpressed ERBB2, resulting in high levels of UCP2 that contribute mitochondrial uncoupling. By contrast, ligand stimulation of non-overexpressed ERBB2 transiently removes UCP2 and paradoxically reduces the mitochondrial membrane potential, oxygen consumption, and OXPHOS on a signaling time scale. However, neither the transporter activity nor down-regulation of already low UCP2 levels drive this reduction in mitochondrial activity. Instead, UCP2 is required to establish mitochondria that are capable of responding to ligand. UCP2 knockdown impairs proliferation at high glucose but its absence specifically impairs ligand-induced growth when glucose levels fluctuate. These findings demonstrate the ability of growth factor signaling to control oxidative phosphorylation on a signaling time scale and point toward a non-transporter role for low levels of UCP2 in establishing dynamic response capability.
Collapse
Affiliation(s)
- Nirav Patel
- From the Department of Biochemistry and Molecular Biology
| | | | | |
Collapse
|
34
|
Zoonens M, Comer J, Masscheleyn S, Pebay-Peyroula E, Chipot C, Miroux B, Dehez F. Dangerous liaisons between detergents and membrane proteins. The case of mitochondrial uncoupling protein 2. J Am Chem Soc 2013; 135:15174-82. [PMID: 24021091 DOI: 10.1021/ja407424v] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
The extraction of membrane proteins from their native environment by detergents is central to their biophysical characterization. Recent studies have emphasized that detergents may perturb the structure locally and modify the dynamics of membrane proteins. However, it remains challenging to determine whether these perturbations are negligible or could be responsible for misfolded conformations, altering the protein's function. In this work, we propose an original strategy combining functional studies and molecular simulations to address the physiological relevance of membrane protein structures obtained in the presence of detergents. We apply our strategy to a structure of isoform 2 of an uncoupling protein (UCP2) binding an inhibitor recently obtained in dodecylphosphocholine detergent micelles. Although this structure shares common traits with the ADP/ATP carrier, a member of the same protein family, its functional and biological significance remains to be addressed. In the present investigation, we demonstrate how dodecylphosphocholine severely alters the structure as well as the function of UCPs. The proposed original strategy opens new vistas for probing the physiological relevance of three-dimensional structures of membrane proteins obtained in non-native environments.
Collapse
Affiliation(s)
- Manuela Zoonens
- CNRS UMR 7099, Institut de Biologie Physico Chimique (IBPC), 75005 Paris, France
| | | | | | | | | | | | | |
Collapse
|
35
|
Coenzyme Q10 depletion in medical and neuropsychiatric disorders: potential repercussions and therapeutic implications. Mol Neurobiol 2013; 48:883-903. [PMID: 23761046 DOI: 10.1007/s12035-013-8477-8] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2013] [Accepted: 05/29/2013] [Indexed: 12/18/2022]
Abstract
Coenzyme Q10 (CoQ10) is an antioxidant, a membrane stabilizer, and a vital cofactor in the mitochondrial electron transport chain, enabling the generation of adenosine triphosphate. It additionally regulates gene expression and apoptosis; is an essential cofactor of uncoupling proteins; and has anti-inflammatory, redox modulatory, and neuroprotective effects. This paper reviews the known physiological role of CoQ10 in cellular metabolism, cell death, differentiation and gene regulation, and examines the potential repercussions of CoQ10 depletion including its role in illnesses such as Parkinson's disease, depression, myalgic encephalomyelitis/chronic fatigue syndrome, and fibromyalgia. CoQ10 depletion may play a role in the pathophysiology of these disorders by modulating cellular processes including hydrogen peroxide formation, gene regulation, cytoprotection, bioenegetic performance, and regulation of cellular metabolism. CoQ10 treatment improves quality of life in patients with Parkinson's disease and may play a role in delaying the progression of that disorder. Administration of CoQ10 has antidepressive effects. CoQ10 treatment significantly reduces fatigue and improves ergonomic performance during exercise and thus may have potential in alleviating the exercise intolerance and exhaustion displayed by people with myalgic encepholamyletis/chronic fatigue syndrome. Administration of CoQ10 improves hyperalgesia and quality of life in patients with fibromyalgia. The evidence base for the effectiveness of treatment with CoQ10 may be explained via its ability to ameliorate oxidative stress and protect mitochondria.
Collapse
|
36
|
Yang CS, Yuk JM, Kim JJ, Hwang JH, Lee CH, Kim JM, Oh GT, Choi HS, Jo EK. Small heterodimer partner-targeting therapy inhibits systemic inflammatory responses through mitochondrial uncoupling protein 2. PLoS One 2013; 8:e63435. [PMID: 23704907 PMCID: PMC3660347 DOI: 10.1371/journal.pone.0063435] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2013] [Accepted: 03/28/2013] [Indexed: 01/15/2023] Open
Abstract
The orphan nuclear receptor, small heterodimer partner (SHP), appears to play a negative regulatory role in innate immune signaling. Emerging evidence warrants further study on the therapeutic targeting of SHP to suppress excessive and deleterious inflammation. Here we show that fenofibrate, which targets SHP, is required for inhibiting systemic inflammation via mitochondrial uncoupling protein 2 (UCP2). In vivo administration of fenofibrate ameliorated systemic inflammatory responses and increased survival upon experimental sepsis through SHP. An abundance of SHP was observed in mice fed fenofibrate and in cultured macrophages through LKB1-dependent activation of the AMP-activated protein kinase pathway. Fenofibrate significantly blocked endotoxin-triggered inflammatory signaling responses via SHP, but not via peroxisome proliferator-activated receptor (PPAR)-α. In addition to the known mechanism by which SHP modulates innate signaling, we identify a new role of fenofibrate-induced SHP on UCP2 induction, which is required for the suppression of inflammatory responses through modulation of mitochondrial ROS production. These data strongly suggest that the SHP-inducing drug fenofibrate paves the way for novel therapies for systemic inflammation by targeting SHP.
Collapse
Affiliation(s)
- Chul-Su Yang
- Department of Microbiology, Chungnam National University School of Medicine, Daejeon, S. Korea
- Infection Signaling Network Research Center, Chungnam National University School of Medicine, Daejeon, S. Korea
| | - Jae-Min Yuk
- Department of Microbiology, Chungnam National University School of Medicine, Daejeon, S. Korea
- Infection Signaling Network Research Center, Chungnam National University School of Medicine, Daejeon, S. Korea
| | - Jwa-Jin Kim
- Department of Microbiology, Chungnam National University School of Medicine, Daejeon, S. Korea
- Infection Signaling Network Research Center, Chungnam National University School of Medicine, Daejeon, S. Korea
| | - Jung Hwan Hwang
- Laboratory Animal Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, S. Korea
| | - Chul-Ho Lee
- Laboratory Animal Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, S. Korea
| | - Jin-Man Kim
- Department of Pathology, Chungnam National University School of Medicine, Daejeon, S. Korea
- Infection Signaling Network Research Center, Chungnam National University School of Medicine, Daejeon, S. Korea
| | - Goo Taeg Oh
- Division of Life and Pharmaceutical Science, Ewha Womans University, Seoul, S. Korea
| | - Hueng-Sik Choi
- National Creative Research Initiatives Center for Nuclear Receptor Signals, Hormone Research Center, School of Biological Sciences and Technology, Chonnam National University, Gwangju, S. Korea
| | - Eun-Kyeong Jo
- Department of Microbiology, Chungnam National University School of Medicine, Daejeon, S. Korea
- Infection Signaling Network Research Center, Chungnam National University School of Medicine, Daejeon, S. Korea
- * E-mail:
| |
Collapse
|
37
|
Wu J, Zhang F, Yan M, Wu D, Yu Q, Zhang Y, Zhou B, McBurney MW, Zhai Q. WldS enhances insulin transcription and secretion via a SIRT1-dependent pathway and improves glucose homeostasis. Diabetes 2011; 60:3197-207. [PMID: 21998399 PMCID: PMC3219932 DOI: 10.2337/db11-0232] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
UNLABELLED OBJECTIVE Wld(S) (Wallerian degeneration slow), a fusion protein from a spontaneous mutation containing full-length nicotinamide mononucleotide adenylyltransferase 1, has NAD biosynthesis activity and protects axon from degeneration robustly. NAD biosynthesis is also implicated in insulin secretion in β-cells. The aim of this study was to investigate the effect of Wld(S) on β-cells and glucose homeostasis. RESEARCH DESIGN AND METHODS Using the Wld(S) mice, we measured the expression of Wld(S) in pancreas and analyzed the effect of Wld(S) on glucose homeostasis. The direct effect of Wld(S) on insulin transcription and secretion and the related mechanisms was measured in isolated islets or β-cell lines. Silent information regulator 1 (SIRT1), an NAD-dependent protein deacetylase, is involved in insulin secretion. Thus, Wld(S) mice with SIRT1 deficiency were generated to study whether the SIRT1-dependent pathway is involved. RESULTS Wld(S) is highly expressed in the pancreas and improves glucose homeostasis. Wld(S) mice are resistant to high-fat diet-induced glucose intolerance and streptozotocin (STZ)-induced hyperglycemia. Wld(S) increases insulin transcription dependent on its NAD biosynthesis activity and enhances insulin secretion. SIRT1 is required for the improved insulin transcription, secretion, and resistance to STZ-induced hyperglycemia caused by Wld(S). Moreover, Wld(S) associates with SIRT1 and increases NAD levels in the pancreas, causing the enhanced SIRT1 activity to downregulate uncoupling protein 2 (UCP2) expression and upregulate ATP levels. CONCLUSIONS Our results demonstrate that Wld(S) combines an insulinotropic effect with protection against β-cell failure and suggest that enhancing NAD biosynthesis in β-cells to increase SIRT1 activity could be a potential therapeutic approach for diabetes.
Collapse
Affiliation(s)
- Jingxia Wu
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Graduate School of the Chinese Academy of Sciences, Shanghai, China
| | - Fang Zhang
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Graduate School of the Chinese Academy of Sciences, Shanghai, China
| | - Menghong Yan
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Graduate School of the Chinese Academy of Sciences, Shanghai, China
| | - Dongmei Wu
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Graduate School of the Chinese Academy of Sciences, Shanghai, China
| | - Qiujing Yu
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Graduate School of the Chinese Academy of Sciences, Shanghai, China
| | - Yi Zhang
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Graduate School of the Chinese Academy of Sciences, Shanghai, China
| | - Ben Zhou
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Graduate School of the Chinese Academy of Sciences, Shanghai, China
| | - Michael W. McBurney
- Center for Cancer Therapeutics, Ottawa Hospital Research Institute, and Department of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Qiwei Zhai
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Graduate School of the Chinese Academy of Sciences, Shanghai, China
- Corresponding author: Qiwei Zhai,
| |
Collapse
|
38
|
Divakaruni AS, Brand MD. The regulation and physiology of mitochondrial proton leak. Physiology (Bethesda) 2011; 26:192-205. [PMID: 21670165 DOI: 10.1152/physiol.00046.2010] [Citation(s) in RCA: 304] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Mitochondria couple respiration to ATP synthesis through an electrochemical proton gradient. Proton leak across the inner membrane allows adjustment of the coupling efficiency. The aim of this review is threefold: 1) introduce the unfamiliar reader to proton leak and its physiological significance, 2) review the role and regulation of uncoupling proteins, and 3) outline the prospects of proton leak as an avenue to treat obesity, diabetes, and age-related disease.
Collapse
Affiliation(s)
- Ajit S Divakaruni
- Medical Research Council Mitochondrial Biology Unit, Wellcome Trust/MRC Building, Cambridge, United Kingdom
| | | |
Collapse
|
39
|
Ramadan JW, Steiner SR, O'Neill CM, Nunemaker CS. The central role of calcium in the effects of cytokines on beta-cell function: implications for type 1 and type 2 diabetes. Cell Calcium 2011; 50:481-90. [PMID: 21944825 DOI: 10.1016/j.ceca.2011.08.005] [Citation(s) in RCA: 72] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2011] [Revised: 07/20/2011] [Accepted: 08/16/2011] [Indexed: 12/29/2022]
Abstract
The appropriate regulation of intracellular calcium is a requirement for proper cell function and survival. This review focuses on the effects of proinflammatory cytokines on calcium regulation in the insulin-producing pancreatic beta-cell and how normal stimulus-secretion coupling, organelle function, and overall beta-cell viability are impacted. Proinflammatory cytokines are increasingly thought to contribute to beta-cell dysfunction not only in type 1 diabetes (T1D), but also in the progression of type 2 diabetes (T2D). Cytokine-induced disruptions in calcium handling result in reduced insulin release in response to glucose stimulation. Cytokines can alter intracellular calcium levels by depleting calcium from the endoplasmic reticulum (ER) and by increasing calcium influx from the extracellular space. Depleting ER calcium leads to protein misfolding and activation of the ER stress response. Disrupting intracellular calcium may also affect organelles, including the mitochondria and the nucleus. As a chronic condition, cytokine-induced calcium disruptions may lead to beta-cell death in T1D and T2D, although possible protective effects are also discussed. Calcium is thus central to both normal and pathological cell processes. Because the tight regulation of intracellular calcium is crucial to homeostasis, measuring the dynamics of calcium may serve as a good indicator of overall beta-cell function.
Collapse
Affiliation(s)
- James W Ramadan
- Department of Medicine, University of Virginia, Charlottesville, United States
| | | | | | | |
Collapse
|
40
|
Deletion of UCP2 in iNOS deficient mice reduces the severity of the disease during experimental autoimmune encephalomyelitis. PLoS One 2011; 6:e22841. [PMID: 21857957 PMCID: PMC3152556 DOI: 10.1371/journal.pone.0022841] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2011] [Accepted: 07/05/2011] [Indexed: 12/11/2022] Open
Abstract
Uncoupling protein 2 is a member of the mitochondrial anion carrier family that is widely expressed in neurons and the immune cells of humans. Deletion of Ucp2 gene in mice pre-activates the immune system leading to higher resistance toward infection and to an increased susceptibility to develop chronic inflammatory diseases as previously exemplified with the Experimental Autoimmune Encephalomyelitis (EAE), a mouse model for multiple sclerosis. Given that oxidative stress is enhanced in Ucp2−/− mice and that nitric oxide (NO) also plays a critical function in redox balance and in chronic inflammation, we generated mice deficient for both Ucp2 and iNos genes and submitted them to EAE. Mice lacking iNos gene exhibited the highest clinical score (3.4+/−0.5 p<0.05). Surprisingly, mice deficient for both genes developed milder disease with reduced immune cell infiltration, cytokines and ROS production as compared to iNos−/− mice.
Collapse
|
41
|
Gao P, Jiao Y, Xiong Q, Wang CY, Gerling I, Gu W. Genetic and Molecular Basis of QTL of Diabetes in Mouse: Genes and Polymorphisms. Curr Genomics 2011; 9:324-37. [PMID: 19471607 PMCID: PMC2685644 DOI: 10.2174/138920208785133253] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2008] [Revised: 04/14/2008] [Accepted: 04/17/2008] [Indexed: 12/14/2022] Open
Abstract
A systematic study has been conducted of all available reports in PubMed and OMIM (Online Mendelian Inheritance in Man) to examine the genetic and molecular basis of quantitative genetic loci (QTL) of diabetes with the main focus on genes and polymorphisms. The major question is, What can the QTL tell us? Specifically, we want to know whether those genome regions differ from other regions in terms of genes relevant to diabetes. Which genes are within those QTL regions, and, among them, which genes have already been linked to diabetes? whether more polymorphisms have been associated with diabetes in the QTL regions than in the non-QTL regions. Our search revealed a total of 9038 genes from 26 type 1 diabetes QTL, which cover 667,096,006 bp of the mouse genomic sequence. On one hand, a large number of candidate genes are in each of these QTL; on the other hand, we found that some obvious candidate genes of QTL have not yet been investigated. Thus, the comprehensive search of candidate genes for known QTL may provide unexpected benefit for identifying QTL genes for diabetes.
Collapse
Affiliation(s)
- Peng Gao
- Departments of Orthopaedic Surgery, Campbell Clinic and Pathology, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | | | | | | | | | | |
Collapse
|
42
|
Sebert SP, Dellschaft NS, Chan LLY, Street H, Henry M, Francois C, Sharma V, Fainberg HP, Patel N, Roda J, Keisler D, Budge H, Symonds ME. Maternal nutrient restriction during late gestation and early postnatal growth in sheep differentially reset the control of energy metabolism in the gastric mucosa. Endocrinology 2011; 152:2816-26. [PMID: 21558318 PMCID: PMC3192420 DOI: 10.1210/en.2011-0169] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Fetal growth restriction followed by accelerated postnatal growth contributes to impaired metabolic function in adulthood. The extent to which these outcomes may be mediated centrally within the hypothalamus, as opposed to in the periphery within the digestive tract, remains unknown. In a sheep model, we achieved intrauterine growth restriction experimentally by maternal nutrient restriction (R) that involved a 40% reduction in food intake through late gestation. R offspring were then either reared singly to accelerate postnatal growth (RA) or as twins and compared with controls also reared singly. From weaning, all offspring were maintained indoors until adulthood. A reduced litter size accelerated postnatal growth for only the first month of lactation. Independently from postnatal weight gain and later fat mass, R animals developed insulin resistance as adults. However, restricted accelerated offspring compared with both the control accelerated and restricted restricted offspring ate less and had higher fasting plasma leptin as adults, an adaptation which was accompanied by changes in energy sensing and cell proliferation within the abomasum. Additionally, although fetal restriction down-regulated gene expression of mammalian target of rapamycin and carnitine palmitoyltransferase 1-dependent pathways in the abomasum, RA offspring compensated for this by exhibiting greater activity of AMP-activated kinase-dependent pathways. This study demonstrates a role for perinatal nutrition in the peripheral control of food intake and in energy sensing in the gastric mucosal and emphasizes the importance of diet in early life in regulating energy metabolism during adulthood.
Collapse
Affiliation(s)
- S P Sebert
- Academic Child Health, School of Clinical Sciences, University Hospital, University of Nottingham, Nottingham, NG7 2UH United Kingdom
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Sun XL, Liu Y, Dai T, Ding JH, Hu G. Uncoupling protein 2 knockout exacerbates depression-like behaviors in mice via enhancing inflammatory response. Neuroscience 2011; 192:507-14. [PMID: 21729739 DOI: 10.1016/j.neuroscience.2011.05.047] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2011] [Revised: 05/19/2011] [Accepted: 05/19/2011] [Indexed: 11/29/2022]
Abstract
Mitochondrial uncoupling protein 2 (UCP2) has been recognized as an important protein to regulate reactive oxygen species (ROS) production. The absence of UCP2 has the potential to promote ROS accumulation and thereby induces oxidative damages and inflammatory response. Increasing evidence strongly reveals that depression is accompanied by oxidative stress, so the present study was to investigate the impacts of UCP2 on the etiology of depression. Wild-type and UCP2 knockout mice were used to establish chronic mild stress (CMS)-induced anhedonia model of depression. The results showed that CMS led to more severe depressive responses in UCP2 knockout mice, characterized by exacerbated depression-like behaviors, increased corticosterone level and significant loss of weight. Moreover, CMS resulted in a higher mortality in UCP2 knockout mice. Our further study showed that UCP2 knockout enhanced CMS-induced activation of nuclear factor κB (NF-κB) p65 and increased mRNA expression of tumor necrosis factor alpha (TNF-α) in hypothalamus. And the levels of TNF-α of serum and spleen in UCP2 knockout mice are remarkably enhanced by CMS, even under basal conditions. Therefore, our findings suggest that UCP2 knockout-induced inflammation may contribute to the development of depressive symptoms.
Collapse
Affiliation(s)
- X-L Sun
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, 140 Hanzhong Road, Nanjing, Jiangsu 210029, PR China
| | | | | | | | | |
Collapse
|
44
|
Lo Iacono L, Boczkowski J, Zini R, Salouage I, Berdeaux A, Motterlini R, Morin D. A carbon monoxide-releasing molecule (CORM-3) uncouples mitochondrial respiration and modulates the production of reactive oxygen species. Free Radic Biol Med 2011; 50:1556-64. [PMID: 21382478 DOI: 10.1016/j.freeradbiomed.2011.02.033] [Citation(s) in RCA: 122] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2011] [Revised: 02/25/2011] [Accepted: 02/28/2011] [Indexed: 01/17/2023]
Abstract
Carbon monoxide (CO), produced during the degradation of heme by the enzyme heme oxygenase, is an important signaling mediator in mammalian cells. Here we show that precise delivery of CO to isolated heart mitochondria using a water-soluble CO-releasing molecule (CORM-3) uncouples respiration. Addition of low-micromolar concentrations of CORM-3 (1-20 μM), but not an inactive compound that does not release CO, significantly increased mitochondrial oxygen consumption rate (State 2 respiration) in a concentration-dependent manner. In contrast, higher concentrations of CORM-3 (100 μM) suppressed ADP-dependent respiration through inhibition of cytochrome c oxidase. The uncoupling effect mediated by CORM-3 was inhibited in the presence of the CO scavenger myoglobin. Moreover, this effect was associated with a gradual decrease in membrane potential (ψ) over time and was partially reversed by malonate, an inhibitor of complex II activity. Similarly, inhibition of uncoupling proteins or blockade of adenine nucleotide transporter attenuated the effect of CORM-3 on both State 2 respiration and Δψ. Hydrogen peroxide (H₂O₂) produced by mitochondria respiring from complex I-linked substrates (pyruvate/malate) was increased by CORM-3. However, respiration initiated via complex II using succinate resulted in a fivefold increase in H₂O₂ production and this effect was significantly inhibited by CORM-3. These findings disclose a counterintuitive action of CORM-3 suggesting that CO at low levels acts as an important regulator of mitochondrial respiration.
Collapse
Affiliation(s)
- Luisa Lo Iacono
- Department of Drug Discovery and Development, Italian Institute of Technology, 16163 Genoa, Italy
| | | | | | | | | | | | | |
Collapse
|
45
|
Bellenger J, Bellenger S, Bataille A, Massey KA, Nicolaou A, Rialland M, Tessier C, Kang JX, Narce M. High pancreatic n-3 fatty acids prevent STZ-induced diabetes in fat-1 mice: inflammatory pathway inhibition. Diabetes 2011; 60:1090-9. [PMID: 21330635 PMCID: PMC3064083 DOI: 10.2337/db10-0901] [Citation(s) in RCA: 122] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
OBJECTIVE Because of confounding factors, the effects of dietary n-3 polyunsaturated fatty acids (PUFA) on type 1 diabetes remain to be clarified. We therefore evaluated whether fat-1 transgenic mice, a well-controlled experimental model endogenously synthesizing n-3 PUFA, were protected against streptozotocin (STZ)-induced diabetes. We then aimed to elucidate the in vivo response at the pancreatic level. RESEARCH DESIGN AND METHODS β-Cell destruction was produced by multiple low-doses STZ (MLD-STZ). Blood glucose level, plasma insulin level, and plasma lipid analysis were then performed. Pancreatic mRNA expression of cytokines, the monocyte chemoattractant protein, and GLUT2 were evaluated as well as pancreas nuclear factor (NF)-κB p65 and inhibitor of κB (IκB) protein expression. Insulin and cleaved caspase-3 immunostaining and lipidomic analysis were performed in the pancreas. RESULTS STZ-induced fat-1 mice did not develop hyperglycemia compared with wild-type mice, and β-cell destruction was prevented as evidenced by lack of histological pancreatic damage or reduced insulin level. The prevention of β-cell destruction was associated with no proinflammatory cytokine induction (tumor necrosis factor-α, interleukin-1β, inducible nitric oxide synthase) in the pancreas, a decreased NF-κB, and increased IκB pancreatic protein expression. In the fat-1-treated mice, proinflammatory arachidonic-derived mediators as prostaglandin E₂ and 12-hydroxyeicosatetraenoic acid were decreased and the anti-inflammatory lipoxin A₄ was detected. Moreover, the 18-hydroxyeicosapentaenoic acid, precursor of the anti-inflammatory resolvin E1, was highly increased. CONCLUSIONS Collectively, these findings indicate that fat-1 mice were protected against MLD-STZ-induced diabetes and pointed out for the first time in vivo the beneficial effects of n-3 PUFA at the pancreatic level, on each step of the development of the pathology-inflammation, β-cell damage-through cytokine response and lipid mediator production.
Collapse
Affiliation(s)
- Jérôme Bellenger
- Université de Bourgogne, UFR Sciences de la Vie, de la Terre et de l’Environnement, Dijon, France
- UMR 866 Physiopathologie des Dyslipidémies, Faculté des Sciences Gabriel, Université de Bourgogne, Dijon, France
| | - Sandrine Bellenger
- Université de Bourgogne, UFR Sciences de la Vie, de la Terre et de l’Environnement, Dijon, France
- UMR 866 Physiopathologie des Dyslipidémies, Faculté des Sciences Gabriel, Université de Bourgogne, Dijon, France
| | - Amandine Bataille
- UMR 866, IFR100 Imagerie Cellulaire–Histologie, Faculté de Médecine et de Pharmacie, Dijon, France
| | | | - Anna Nicolaou
- School of Pharmacy, University of Bradford, Bradford, U.K
| | - Mickaël Rialland
- Université de Bourgogne, UFR Sciences de la Vie, de la Terre et de l’Environnement, Dijon, France
- UMR 866 Physiopathologie des Dyslipidémies, Faculté des Sciences Gabriel, Université de Bourgogne, Dijon, France
| | - Christian Tessier
- Université de Bourgogne, UFR Sciences de la Vie, de la Terre et de l’Environnement, Dijon, France
- UMR 866 Physiopathologie des Dyslipidémies, Faculté des Sciences Gabriel, Université de Bourgogne, Dijon, France
| | - Jing X. Kang
- Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Michel Narce
- Université de Bourgogne, UFR Sciences de la Vie, de la Terre et de l’Environnement, Dijon, France
- UMR 866 Physiopathologie des Dyslipidémies, Faculté des Sciences Gabriel, Université de Bourgogne, Dijon, France
- Corresponding author: Michel Narce,
| |
Collapse
|
46
|
Affourtit C, Jastroch M, Brand MD. Uncoupling protein-2 attenuates glucose-stimulated insulin secretion in INS-1E insulinoma cells by lowering mitochondrial reactive oxygen species. Free Radic Biol Med 2011; 50:609-16. [PMID: 21172424 PMCID: PMC3036803 DOI: 10.1016/j.freeradbiomed.2010.12.020] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2010] [Revised: 11/30/2010] [Accepted: 12/11/2010] [Indexed: 01/26/2023]
Abstract
Glucose-stimulated insulin secretion (GSIS) by pancreatic β cells is regulated by mitochondrial uncoupling protein-2 (UCP2), but opposing phenotypes, GSIS improvement and impairment, have been reported for different Ucp2-ablated mouse models. By measuring mitochondrial bioenergetics in attached INS-1E insulinoma cells with and without UCP2, we show that UCP2 contributes to proton leak and attenuates glucose-induced rises in both respiratory activity and the coupling efficiency of oxidative phosphorylation. Strikingly, the GSIS improvement seen upon UCP2 knockdown in INS-1E cells is annulled completely by the cell-permeative antioxidant MnTMPyP. Consistent with this observation, UCP2 lowers mitochondrial reactive oxygen species at high glucose levels. We conclude that UCP2 plays both regulatory and protective roles in β cells by acutely lowering GSIS and chronically preventing oxidative stress. Our findings thus provide a mechanistic explanation for the apparently discrepant findings in the field.
Collapse
Key Words
- ∆ψ, mitochondrial membrane potential
- dapi, 4′,6-diamidino-2-phenylindole
- dhe, hydroethidine
- fcs, fetal calf serum
- fccp, carbonyl cyanide p-trifluoromethoxyphenylhydrazone
- gsis, glucose-stimulated insulin secretion
- krh, hepes-buffered krebs–ringer medium
- mitosox, mitochondria-targeted hydroethidine
- mntbap, manganese tetrakis-(4-benzoic acid) porphyrin
- mntmpyp, manganese tetrakis-(n-methyl-4-pyridyl) porphyrin
- ros, reactive oxygen species
- ttnpb, 4-[(e)-2-(5,6,7,8-tetrahydro-5,5,8,8-tetramethyl-2-napthalenyl)-1-propenyl]benzoic acid
- ucp2, uncoupling protein-2
- pancreatic β cells
- glucose-stimulated insulin secretion
- uncoupling protein 2
- mitochondrial respiration
- reactive oxygen species
- coupling efficiency of oxidative phosphorylation
- type 2 diabetes
- metabolic syndrome
- free radicals
Collapse
Affiliation(s)
- Charles Affourtit
- Mitochondrial Biology Unit, Medical Research Council, Cambridge CB2 0XY,
| | | | | |
Collapse
|
47
|
Zheng D, Wang Y, Cao Q, Lee VWS, Zheng G, Sun Y, Tan TK, Wang Y, Alexander SI, Harris DCH. Transfused macrophages ameliorate pancreatic and renal injury in murine diabetes mellitus. NEPHRON. EXPERIMENTAL NEPHROLOGY 2011; 118:e87-e99. [PMID: 21311199 DOI: 10.1159/000321034] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
BACKGROUND Alternatively activated macrophages (M2 macrophages) are able to reduce renal injury in murine adriamycin nephropathy. However, the effect of M2 macrophages in other renal diseases such as diabetic nephropathy remains unknown. METHODS Macrophages were separated from splenocytes and polarized with IL-4 and IL-13 into a protective phenotype. Mice underwent adoptive transfer with M2 macrophages, and then diabetes was induced by tail vein injection with streptozotocin (STZ). Blood glucose levels were monitored daily. Mice were sacrificed at week 10 after STZ. Renal function and histopathological injury were assessed quantitatively. RESULTS Transfused M2 macrophages accumulated progressively in kidneys for up to 10 weeks after STZ. Kidneys from diabetic mice transfused with M2 macrophages had less tubular atrophy, glomerular hypertrophy and interstitial expansion than did control diabetic mice. M2 macrophages suppressed the development of interstitial fibrosis. In addition, the degree of pancreatic islet injury, as assessed by insulin staining, haemoglobin A1c and blood glucose was reduced after transfusion of M2 macrophages. In vivo, activation of kidney endogenous macrophage cytokine expression was inhibited by M2 macrophages. CONCLUSION Our findings show that M2 macrophages can protect against islet and renal injury in streptozotocin-induced diabetes, providing a potential therapeutic strategy for diabetes and diabetic nephropathy.
Collapse
Affiliation(s)
- Dong Zheng
- Centre for Transplant and Renal Research, University of Sydney, Westmead Millennium Institute, Sydney, NSW 2145, Australia.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Yu Q, Yasuda M, Takahashi T, Nomura M, Hagino N, Kobayashi S. Effects of Bofutsushosan and Gardeniae Fructus on Diabetic Serum Parameters in Streptozotocin-Induced Diabetic Mice. Chin Med 2011. [DOI: 10.4236/cm.2011.24022] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|
49
|
Carrillo AE, Flouris AD. Caloric restriction and longevity: effects of reduced body temperature. Ageing Res Rev 2011; 10:153-62. [PMID: 20969980 DOI: 10.1016/j.arr.2010.10.001] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2010] [Revised: 09/27/2010] [Accepted: 10/01/2010] [Indexed: 01/22/2023]
Abstract
Caloric restriction (CR) causes a reduction in body temperature (T(b)) which is suggested to contribute to changes that increase lifespan. Moreover, low T(b) has been shown to improve health and longevity independent of CR. In this review we examine the connections between CR, T(b) and mechanisms that influence longevity and ageing. Recent findings regarding the overlapping mechanisms of CR and T(b) that benefit longevity are discussed, including changes in body composition, hormone regulation, and gene expression, as well as reductions in low-level inflammation and reactive oxygen species-induced molecular damage. This information is summarized in a model describing how CR and low T(b), both synergistically and independently, increase lifespan. Moreover, the nascent notion that the rate of ageing may be pre-programmed in response to environmental influences at critical periods of early development is also considered. Based on current evidence, it is concluded that low T(b) plays an integral role in mediating the effects of CR on health and longevity, and that low T(b) may exert independent biological changes that increase lifespan. Our understanding of the overlap between CR- and T(b)-mediated longevity remains incomplete and should be explored in future research.
Collapse
|
50
|
Brand MD, Parker N, Affourtit C, Mookerjee SA, Azzu V. Mitochondrial uncoupling protein 2 in pancreatic β-cells. Diabetes Obes Metab 2010; 12 Suppl 2:134-40. [PMID: 21029310 DOI: 10.1111/j.1463-1326.2010.01264.x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Pancreatic β-cells have remarkable bioenergetics in which increased glucose supply upregulates the cytosolic ATP/ADP ratio and increases insulin secretion. This arrangement allows glucose-stimulated insulin secretion (GSIS) to be regulated by the coupling efficiency of oxidative phosphorylation. Uncoupling protein 2 (UCP2) modulates coupling efficiency and may regulate GSIS. Initial measurements of GSIS and glucose tolerance in Ucp2(-/-) mice supported this model, but recent studies show confounding effects of genetic background. Importantly, however, the enhancement of GSIS is robustly recapitulated with acute UCP2 knockdown in INS-1E insulinoma cells. UCP2 protein level in these cells is dynamically regulated, over at least a fourfold concentration range, by rapid proteolysis (half-life less than 1 h) opposing regulated gene transcription and mRNA translation. Degradation is catalysed by the cytosolic proteasome in an unprecedented pathway that is currently known to act only on UCP2 and UCP3. Evidence for proteasomal turnover of UCP2 includes sensitivity of degradation to classic proteasome inhibitors in cells, and reconstitution of degradation in vitro in mitochondria incubated with ubiquitin and the cytosolic 26S proteasome. These dynamic changes in UCP2 content may provide a fine level of control over GSIS in β-cells.
Collapse
Affiliation(s)
- M D Brand
- Buck Institute for Age Research, Novato, CA 94945, USA.
| | | | | | | | | |
Collapse
|