1
|
Jiang Y, Xu L, Zhu X, Zhu X, Xu X, Li J. Hyperglycemic stress induces oxidative damage of enteric glial cells by triggering redoxosomes/p66SHC activation. Redox Rep 2024; 29:2324234. [PMID: 38444386 PMCID: PMC10919305 DOI: 10.1080/13510002.2024.2324234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/07/2024] Open
Abstract
OBJECTIVES Diabetic gastrointestinal dysfunction (DGD) is a serious complication of diabetic mellitus (DM), affecting the enteric nervous system (ENS), particular enteric glial cells (EGCs). This study aimed to elucidate the effects and underlying molecular mechanisms of hyperglycemic stress on EGCs in in vitro and in vivo models of DM. METHODS In in vitro studies, enteric glial cell line CRL-2690 was exposed to hyperglycemia stress, and cell viability, cell apoptosis and oxidative damage were assessed. In in vivo studies, STZ-induced diabetic mice were constructed, and cell apoptosis and oxidative damage of EGCs in the duodenum of DM mice were assessed. RESULTS The results showed that hyperglycemic stress markedly induced oxidative damage of EGCs in in vitro and in vivo models of DM. This damage was found to be dependent on the activation of redoxosomes, which involved the phosphorylation of SRC and Vav2, the up-regulation of active RAC1-GTP, and the activation of NADPH oxidase (NOX). Moreover, inhibitors of redoxosomes, such as the RAC1 inhibitor NSC23766 and the NOX inhibitor VAS2870, effectively mitigated the hyperglycemic stress-induced oxidative damage of EGCs. Additionally, inhibition of p66SHC, a downstream target of redoxosomes, attenuated oxidative damage of EGCs under hyperglycemic stress. DISCUSSION Our findings suggest that the redoxosomes/p66SHC signaling is involved in the oxidative damage of EGCs during the pathological process of DGD. This signaling cascade may represent a potential therapeutic target for the treatment of DGD.
Collapse
Affiliation(s)
- Yanmin Jiang
- Department of Endocrinology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, People’s Republic of China
- Department of Endocrinology, Wuxi Medical Center, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Nanjing Medical University, Wuxi, People’s Republic of China
| | - Lan Xu
- Department of Endocrinology, Wuxi Medical Center, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Nanjing Medical University, Wuxi, People’s Republic of China
| | - Xue Zhu
- National Health Commission (NHC) Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, People’s Republic of China
| | - Xiaowei Zhu
- Department of Endocrinology, Wuxi Medical Center, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Nanjing Medical University, Wuxi, People’s Republic of China
| | - Xiang Xu
- Department of Endocrinology, Wuxi Medical Center, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Nanjing Medical University, Wuxi, People’s Republic of China
| | - Jianbo Li
- Department of Endocrinology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, People’s Republic of China
| |
Collapse
|
2
|
Luo Y, Zhu J, Hu Z, Luo W, Du X, Hu H, Peng S. Progress in the Pathogenesis of Diabetic Encephalopathy: The Key Role of Neuroinflammation. Diabetes Metab Res Rev 2024; 40:e3841. [PMID: 39295168 DOI: 10.1002/dmrr.3841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 04/29/2024] [Accepted: 06/27/2024] [Indexed: 09/21/2024]
Abstract
Diabetic encephalopathy (DE) is a severe complication that occurs in the central nervous system (CNS) and leads to cognitive impairment. DE involves various pathophysiological processes, and its pathogenesis is still unclear. This review summarised current research on the pathogenesis of diabetic encephalopathy, which involves neuroinflammation, oxidative stress, iron homoeostasis, blood-brain barrier disruption, altered gut microbiota, insulin resistance, etc. Among these pathological mechanisms, neuroinflammation has been focused on. This paper summarises some of the molecular mechanisms involved in neuroinflammation, including the Mammalian Target of Rapamycin (mTOR), Lipocalin-2 (LCN-2), Pyroptosis, Advanced Glycosylation End Products (AGEs), and some common pro-inflammatory factors. In addition, we discuss recent advances in the study of potential therapeutic targets for the treatment of DE against neuroinflammation. The current research on the pathogenesis of DE is progressing slowly, and more research is needed in the future. Further study of neuroinflammation as a mechanism is conducive to the discovery of more effective treatments for DE in the future.
Collapse
Affiliation(s)
- Yifan Luo
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
- Department of Clinical Medicine, The Second Clinical Medical College of Nanchang University, Nanchang, China
| | - Jinxi Zhu
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
- Department of Clinical Medicine, The Second Clinical Medical College of Nanchang University, Nanchang, China
| | - Ziyan Hu
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
- Department of Clinical Medicine, The Second Clinical Medical College of Nanchang University, Nanchang, China
| | - Wei Luo
- Department of Sports Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Xiaohong Du
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Haijun Hu
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Shengliang Peng
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| |
Collapse
|
3
|
Chamgordani MK, Bardestani A, Ebrahimpour S, Esmaeili A. In diabetic male Wistar rats, quercetin-conjugated superparamagnetic iron oxide nanoparticles have an effect on the SIRT1/p66Shc-mediated pathway related to cognitive impairment. BMC Pharmacol Toxicol 2023; 24:81. [PMID: 38129872 PMCID: PMC10734159 DOI: 10.1186/s40360-023-00725-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2023] [Accepted: 12/12/2023] [Indexed: 12/23/2023] Open
Abstract
BACKGROUND Quercetin (QC) possesses a variety of health-promoting effects in pure and in conjugation with nanoparticles. Since the mRNA-SIRT1/p66Shc pathway and microRNAs (miRNAs) are implicated in the oxidative process, we aimed to compare the effects of QC and QC-conjugated superparamagnetic iron oxide nanoparticles (QCSPIONs) on this pathway. METHODS Through the use of the chemical coprecipitation technique (CPT), SPIONs were synthesized, coated with dextran, and conjugated with quercetin. Adult male Wistar rats were given intraperitoneal injections of streptozotocin to look for signs of type 1 diabetes (T1D). The animals were randomized into five groups: the control group got deionized water (DI), free QC solution (25 mg/kg), SPIONs (25 mg/kg), and QCSPIONs (25 mg/kg), and all groups received repeat doses administered orally over 35 days. Real-time quantitative PCR was used to assess the levels of miR-34a, let-7a-p5, SIRT1, p66Shc, CASP3, and PARP1 expression in the hippocampus of diabetic rats. RESULTS In silico investigations identified p66Shc, CASP3, and PARP1 as targets of let-7a-5p and miR-34a as possible regulators of SIRT1 genes. The outcomes demonstrated that diabetes elevated miR-34a, p66Shc, CASP3, and PARP1 and downregulated let-7a-5p and SIRT1 expression. In contrast to the diabetic group, QCSPIONs boosted let-7a-5p expression levels and consequently lowered p66Shc, CASP3, and PARP1 expression levels. QCSPIONs also reduced miR-34a expression, which led to an upsurge in SIRT1 expression. CONCLUSION Our results suggest that QCSPIONs can regulate the SIRT1/p66Shc-mediated signaling pathway and can be considered a promising candidate for ameliorating the complications of diabetes.
Collapse
Affiliation(s)
- Mahnaz Karami Chamgordani
- Department of Cell and Molecular Biology & Microbiology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, P.O. Box: 8174673441, Iran
| | - Akram Bardestani
- Department of Cell and Molecular Biology & Microbiology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, P.O. Box: 8174673441, Iran
| | - Shiva Ebrahimpour
- Department of Cell and Molecular Biology & Microbiology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, P.O. Box: 8174673441, Iran
| | - Abolghasem Esmaeili
- Department of Cell and Molecular Biology & Microbiology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, P.O. Box: 8174673441, Iran.
| |
Collapse
|
4
|
Biondi G, Marrano N, Borrelli A, Rella M, D’Oria R, Genchi VA, Caccioppoli C, Cignarelli A, Perrini S, Laviola L, Giorgino F, Natalicchio A. The p66 Shc Redox Protein and the Emerging Complications of Diabetes. Int J Mol Sci 2023; 25:108. [PMID: 38203279 PMCID: PMC10778847 DOI: 10.3390/ijms25010108] [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: 10/31/2023] [Revised: 12/11/2023] [Accepted: 12/15/2023] [Indexed: 01/12/2024] Open
Abstract
Diabetes mellitus is a chronic metabolic disease, the prevalence of which is constantly increasing worldwide. It is often burdened by disabling comorbidities that reduce the quality and expectancy of life of the affected individuals. The traditional complications of diabetes are generally described as macrovascular complications (e.g., coronary heart disease, peripheral arterial disease, and stroke), and microvascular complications (e.g., diabetic kidney disease, retinopathy, and neuropathy). Recently, due to advances in diabetes management and the increased life expectancy of diabetic patients, a strong correlation between diabetes and other pathological conditions (such as liver diseases, cancer, neurodegenerative diseases, cognitive impairments, and sleep disorders) has emerged. Therefore, these comorbidities have been proposed as emerging complications of diabetes. P66Shc is a redox protein that plays a role in oxidative stress, apoptosis, glucose metabolism, and cellular aging. It can be regulated by various stressful stimuli typical of the diabetic milieu and is involved in various types of organ and tissue damage under diabetic conditions. Although its role in the pathogenesis of diabetes remains controversial, there is strong evidence regarding the involvement of p66Shc in the traditional complications of diabetes. In this review, we will summarize the evidence supporting the role of p66Shc in the pathogenesis of diabetes and its complications, focusing for the first time on the emerging complications of diabetes.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Francesco Giorgino
- Department of Precision and Regenerative Medicine and Ionian Area, Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, University of Bari Aldo Moro, 70124 Bari, Italy (M.R.); (R.D.); (V.A.G.)
| | | |
Collapse
|
5
|
Veselov IM, Vinogradova DV, Maltsev AV, Shevtsov PN, Spirkova EA, Bachurin SO, Shevtsova EF. Mitochondria and Oxidative Stress as a Link between Alzheimer's Disease and Diabetes Mellitus. Int J Mol Sci 2023; 24:14450. [PMID: 37833898 PMCID: PMC10572926 DOI: 10.3390/ijms241914450] [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: 08/14/2023] [Revised: 09/15/2023] [Accepted: 09/20/2023] [Indexed: 10/15/2023] Open
Abstract
This review is devoted to the problems of the common features linking metabolic disorders and type 2 diabetes with the development of Alzheimer's disease. The pathogenesis of Alzheimer's disease closely intersects with the mechanisms of type 2 diabetes development, and an important risk factor for both pathologies is aging. Common pathological mechanisms include both factors in the development of oxidative stress, neuroinflammation, insulin resistance, and amyloidosis, as well as impaired mitochondrial dysfunctions and increasing cell death. The currently available drugs for the treatment of type 2 diabetes and Alzheimer's disease have limited therapeutic efficacy. It is important to note that drugs used to treat Alzheimer's disease, in particular acetylcholinesterase inhibitors, show a positive therapeutic potential in the treatment of type 2 diabetes, while drugs used in the treatment of type 2 diabetes can also prevent a number of pathologies characteristic for Alzheimer's disease. A promising direction in the search for a strategy for the treatment of type 2 diabetes and Alzheimer's disease may be the creation of complex multi-target drugs that have neuroprotective potential and affect specific common targets for type 2 diabetes and Alzheimer's disease.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Elena F. Shevtsova
- Institute of Physiologically Active Compounds at Federal Research Center of Problems of Chemical Physics and Medicinal Chemistry, Russian Academy of Sciences (IPAC RAS), Chernogolovka 142432, Russia; (I.M.V.); (A.V.M.); (P.N.S.); (E.A.S.); (S.O.B.)
| |
Collapse
|
6
|
Chadwick W, Maudsley S, Hull W, Havolli E, Boshoff E, Hill MDW, Goetghebeur PJD, Harrison DC, Nizami S, Bedford DC, Coope G, Real K, Thiemermann C, Maycox P, Carlton M, Cole SL. The oDGal Mouse: A Novel, Physiologically Relevant Rodent Model of Sporadic Alzheimer's Disease. Int J Mol Sci 2023; 24:ijms24086953. [PMID: 37108119 PMCID: PMC10138655 DOI: 10.3390/ijms24086953] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 03/17/2023] [Accepted: 03/30/2023] [Indexed: 04/29/2023] Open
Abstract
Sporadic Alzheimer's disease (sAD) represents a serious and growing worldwide economic and healthcare burden. Almost 95% of current AD patients are associated with sAD as opposed to patients presenting with well-characterized genetic mutations that lead to AD predisposition, i.e., familial AD (fAD). Presently, the use of transgenic (Tg) animals overexpressing human versions of these causative fAD genes represents the dominant research model for AD therapeutic development. As significant differences in etiology exist between sAD and fAD, it is perhaps more appropriate to develop novel, more sAD-reminiscent experimental models that would expedite the discovery of effective therapies for the majority of AD patients. Here we present the oDGal mouse model, a novel model of sAD that displays a range of AD-like pathologies as well as multiple cognitive deficits reminiscent of AD symptomology. Hippocampal cognitive impairment and pathology were delayed with N-acetyl-cysteine (NaC) treatment, which strongly suggests that reactive oxygen species (ROS) are the drivers of downstream pathologies such as elevated amyloid beta and hyperphosphorylated tau. These features demonstrate a desired pathophenotype that distinguishes our model from current transgenic rodent AD models. A preclinical model that presents a phenotype of non-genetic AD-like pathologies and cognitive deficits would benefit the sAD field, particularly when translating therapeutics from the preclinical to the clinical phase.
Collapse
Affiliation(s)
- Wayne Chadwick
- Takeda Cambridge, 418 Cambridge Science Park, Cambridge CB4 0PZ, UK
| | - Stuart Maudsley
- Receptor Biology Lab, University of Antwerp, 2000 Antwerp, Belgium
| | - William Hull
- William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Centre for Translational Medicine and Therapeutics, Queen Mary University of London, London E1 4NS, UK
| | - Enes Havolli
- Takeda Cambridge, 418 Cambridge Science Park, Cambridge CB4 0PZ, UK
| | - Eugene Boshoff
- Takeda Cambridge, 418 Cambridge Science Park, Cambridge CB4 0PZ, UK
| | - Mark D W Hill
- Takeda Cambridge, 418 Cambridge Science Park, Cambridge CB4 0PZ, UK
| | | | - David C Harrison
- Takeda Cambridge, 418 Cambridge Science Park, Cambridge CB4 0PZ, UK
| | - Sohaib Nizami
- Takeda Cambridge, 418 Cambridge Science Park, Cambridge CB4 0PZ, UK
| | - David C Bedford
- Takeda Cambridge, 418 Cambridge Science Park, Cambridge CB4 0PZ, UK
| | - Gareth Coope
- Takeda Cambridge, 418 Cambridge Science Park, Cambridge CB4 0PZ, UK
| | - Katia Real
- Takeda Cambridge, 418 Cambridge Science Park, Cambridge CB4 0PZ, UK
| | - Christoph Thiemermann
- William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Centre for Translational Medicine and Therapeutics, Queen Mary University of London, London E1 4NS, UK
| | - Peter Maycox
- Takeda Cambridge, 418 Cambridge Science Park, Cambridge CB4 0PZ, UK
| | - Mark Carlton
- Takeda Cambridge, 418 Cambridge Science Park, Cambridge CB4 0PZ, UK
| | - Sarah L Cole
- Takeda Cambridge, 418 Cambridge Science Park, Cambridge CB4 0PZ, UK
| |
Collapse
|
7
|
Chen Q, Dai J, Nan F, Xu J, Chen S. P66shc in the spinal cord is an important contributor in complete Freund's adjuvant induced inflammatory pain in mice. Biochem Biophys Res Commun 2023; 656:63-69. [PMID: 36958256 DOI: 10.1016/j.bbrc.2023.03.028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Accepted: 03/10/2023] [Indexed: 03/13/2023]
Abstract
PURPOSE The aim of this study is to investigate whether p66shc is involved in inflammatory pain and the potential molecular mechanisms of p66shc in inflammatory pain. METHODS Inflammatory pain model was established by complete Freund's adjuvant (CFA) injection. Paw withdrawal latency (PWL) and paw withdrawal frequency (PWF) was recorded. The expression of spinal p66shc were determined by immunohistochemical staining, immunofluorescence staining. P66shc knockdown was performed by an adeno-associated virus (AAV) vector infusion. NLRP3 inflammasome complexes were determined by Western blot. DHE staining was used to evaluate reactive oxygen species (ROS) generation. RESULTS P66Shc expression was progressively elevated in spinal cord of inflammatory pain mice, and p66Shc knockdown in vivo significantly attenuated CFA injection triggers hyperalgesia. Furthermore, knockdown of p66Shc significantly inhibited ROS production and NOD-like receptor protein 3 (NLRP3) inflammasome activation, which were reversed by a ROS donor (t-BOOH). However, post-treatment with nigericin, a agonist of NLRP3, reversed AAV-shP66shc analgesic effect. CONCLUSION Spinal p66shc may facilitate the development of inflammatory pain by promoting the activation of NLRP3 inflammasome through ROS.
Collapse
Affiliation(s)
- Qianqian Chen
- Reproductive Medicine Center, Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Juji Dai
- Department of Colorectal and Anal Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Fubei Nan
- Department of Anesthesiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jun Xu
- Department of Gastroenterology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Shuangdong Chen
- Department of Anesthesiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China.
| |
Collapse
|
8
|
Vargas-Soria M, García-Alloza M, Corraliza-Gómez M. Effects of diabetes on microglial physiology: a systematic review of in vitro, preclinical and clinical studies. J Neuroinflammation 2023; 20:57. [PMID: 36869375 PMCID: PMC9983227 DOI: 10.1186/s12974-023-02740-x] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Accepted: 02/16/2023] [Indexed: 03/05/2023] Open
Abstract
Diabetes mellitus is a heterogeneous chronic metabolic disorder characterized by the presence of hyperglycemia, commonly preceded by a prediabetic state. The excess of blood glucose can damage multiple organs, including the brain. In fact, cognitive decline and dementia are increasingly being recognized as important comorbidities of diabetes. Despite the largely consistent link between diabetes and dementia, the underlying causes of neurodegeneration in diabetic patients remain to be elucidated. A common factor for almost all neurological disorders is neuroinflammation, a complex inflammatory process in the central nervous system for the most part orchestrated by microglial cells, the main representatives of the immune system in the brain. In this context, our research question aimed to understand how diabetes affects brain and/or retinal microglia physiology. We conducted a systematic search in PubMed and Web of Science to identify research items addressing the effects of diabetes on microglial phenotypic modulation, including critical neuroinflammatory mediators and their pathways. The literature search yielded 1327 records, including 18 patents. Based on the title and abstracts, 830 papers were screened from which 250 primary research papers met the eligibility criteria (original research articles with patients or with a strict diabetes model without comorbidities, that included direct data about microglia in the brain or retina), and 17 additional research papers were included through forward and backward citations, resulting in a total of 267 primary research articles included in the scoping systematic review. We reviewed all primary publications investigating the effects of diabetes and/or its main pathophysiological traits on microglia, including in vitro studies, preclinical models of diabetes and clinical studies on diabetic patients. Although a strict classification of microglia remains elusive given their capacity to adapt to the environment and their morphological, ultrastructural and molecular dynamism, diabetes modulates microglial phenotypic states, triggering specific responses that include upregulation of activity markers (such as Iba1, CD11b, CD68, MHC-II and F4/80), morphological shift to amoeboid shape, secretion of a wide variety of cytokines and chemokines, metabolic reprogramming and generalized increase of oxidative stress. Pathways commonly activated by diabetes-related conditions include NF-κB, NLRP3 inflammasome, fractalkine/CX3CR1, MAPKs, AGEs/RAGE and Akt/mTOR. Altogether, the detailed portrait of complex interactions between diabetes and microglia physiology presented here can be regarded as an important starting point for future research focused on the microglia-metabolism interface.
Collapse
Affiliation(s)
- María Vargas-Soria
- Division of Physiology, School of Medicine, Universidad de Cadiz, Cadiz, Spain.,Instituto de Investigacion e Innovacion en Ciencias Biomedicas de la Provincia de Cadiz (INIBICA), Cadiz, Spain
| | - Mónica García-Alloza
- Division of Physiology, School of Medicine, Universidad de Cadiz, Cadiz, Spain.,Instituto de Investigacion e Innovacion en Ciencias Biomedicas de la Provincia de Cadiz (INIBICA), Cadiz, Spain
| | - Miriam Corraliza-Gómez
- Division of Physiology, School of Medicine, Universidad de Cadiz, Cadiz, Spain. .,Instituto de Investigacion e Innovacion en Ciencias Biomedicas de la Provincia de Cadiz (INIBICA), Cadiz, Spain.
| |
Collapse
|
9
|
Roohbakhsh A, Etemad L, Karimi G. Resolvin D1: A key endogenous inhibitor of neuroinflammation. Biofactors 2022; 48:1005-1026. [PMID: 36176016 DOI: 10.1002/biof.1891] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2022] [Accepted: 08/08/2022] [Indexed: 12/14/2022]
Abstract
After the initiation of inflammation, a series of processes start to resolve the inflammation. A group of endogenous lipid mediators, namely specialized pro-resolving lipid mediators is at the top list of inflammation resolution. Resolvin D1 (RvD1), is one of the lipid mediators with significant anti-inflammatory properties. It is produced from docosahexaenoic acid (omega-3 polyunsaturated fatty acid) in the body. In this article, we aimed to review the most recent findings concerning the pharmacological effects of RvD1 in the central nervous system with a focus on major neurological diseases and dysfunctions. A literature review of the past studies demonstrated that RvD1 plasma level changes during mania, depression, and Parkinson's disease. Furthermore, RVD1 and its epimer, aspirin-triggered RvD1 (AT-RvD1), have significant therapeutic effects on experimental models of ischemic and traumatic brain injuries, memory dysfunction, pain, depression, amyotrophic lateral sclerosis, and Alzheimer's and Parkinson's diseases. Interestingly, the beneficial effects of RvD1 and AT-RvD1 were mostly induced at nanomolar and micromolar concentrations implying the significant potency of these lipid mediators in treating diseases with inflammation.
Collapse
Affiliation(s)
- Ali Roohbakhsh
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Leila Etemad
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Gholamreza Karimi
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
10
|
Miller B, Regner K, Sorokin A. p66Shc signaling does not contribute to tubular damage induced by renal ischemia-reperfusion injury in rat. Biochem Biophys Res Commun 2022; 603:69-74. [PMID: 35278882 PMCID: PMC8969123 DOI: 10.1016/j.bbrc.2022.03.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 03/03/2022] [Indexed: 11/02/2022]
Abstract
Renal ischemia-reperfusion (IR) injury is one of the major causes of acute kidney injury and represents a significant risk factor for renal transplantation. The level of renal damage is influenced by the ischemic duration and is caused by excessive amounts of produced reactive oxygen species (ROS). Adaptor protein p66Shc is known to regulate cellular and organ's sensitivity to oxidative stress and to contribute significantly to mitochondrial production of hydrogen peroxide in stress conditions. Studies carried out in cultured renal cells suggest that p66Shc-mediated mitochondrial dysfunction and ROS production are responsible for renal ischemic injury. We used our genetically modified rats, which either lack p66Shc expression, or express p66Shc variant, which constitutively generates increased quantities of hydrogen peroxide, to evaluate potential contribution of p66Shc signaling to renal damage in ischemia reperfusion rat model. Analysis of outer medulla tubule damage revealed the lack of contribution of either p66Shc expression or its constitutive signaling to IR injury in rat model.
Collapse
|
11
|
Mousavi S, Khazeei Tabari MA, Bagheri A, Samieefar N, Shaterian N, Kelishadi R. The Role of p66Shc in Diabetes: A Comprehensive Review from Bench to Bedside. J Diabetes Res 2022; 2022:7703520. [PMID: 36465704 PMCID: PMC9715346 DOI: 10.1155/2022/7703520] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 11/02/2022] [Accepted: 11/09/2022] [Indexed: 11/27/2022] Open
Abstract
It is well-documented that diabetes is an inflammatory and oxidative disease, with an escalating global burden. Still, there is no definite treatment for diabetes or even prevention of its harmful complications. Therefore, understanding the molecular pathways associated with diabetes might help in finding a solution. p66Shc is a member of Shc family proteins, and it is considered as an oxidative stress sensor and regulator in cells. There are inconsistent data about the role of p66Shc in inducing diabetes, but accumulating evidence supports its role in the pathogenesis of diabetes-related complications, including macro and microangiopathies. There is growing hope that by understanding and targeting molecular pathways involved in this network, prevention of diabetes or its complications would be achievable. This review provides an overview about the role of p66Shc in the development of diabetes and its complications.
Collapse
Affiliation(s)
- SeyedehFatemeh Mousavi
- Student Research Committee, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- USERN Office, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Amin Khazeei Tabari
- Student Research Committee, Mazandaran University of Medical Sciences, Mazandaran, Iran
- USERN Office, Mazandaran University of Medical Sciences, Mazandaran, Iran
| | - Alireza Bagheri
- USERN Office, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Department of Genetics, Faculty of Basic Sciences, Shahrekord University, Shahrekord, Iran
| | - Noosha Samieefar
- Student Research Committee, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- USERN Office, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Negar Shaterian
- Student Research Committee, School of Medicine, Jahrom University of Medical Sciences, Jahrom, Iran
- USERN Office, Jahrom University of Medical Sciences, Jahrom, Iran
| | - Roya Kelishadi
- Child Growth and Development Research Center, Research Institute for Primordial Prevention of Non-Communicable Disease, Isfahan University of Medical Sciences, Isfahan, Iran
- USERN Office, Research Institute for Primordial Prevention of Non-Communicable Disease, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
12
|
Jelinek HF, Helf C, Khalaf K. Human SHC-transforming protein 1 and its isoforms p66shc: A novel marker for prediabetes. J Diabetes Investig 2021; 12:1881-1889. [PMID: 33759377 PMCID: PMC8504898 DOI: 10.1111/jdi.13551] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 03/02/2021] [Accepted: 03/18/2021] [Indexed: 12/15/2022] Open
Abstract
AIMS Prediabetes is a multifactorial condition. Current guidelines for diabetes screening recommend either the use of glycated hemoglobin (HbA1c), or blood glucose level (BGL). This research aimed to identify if p66shc a component of the Human SHC-Transforming Protein 1 (Shc1), a mitochondrial associated oxidative stress biomarker, is significantly altered in patients with elevated BGL. Furthermore, we evaluated if inflammatory and oxidative stress markers, such as p66shc, are a useful addition to the regularly used biomarkers to increase sensitivity for identification of prediabetes. METHODS All participants attended the Diabetic Health Screening at Charles Sturt University (CSU), Australia. The cross-sectional clinical study collected demographic and clinical variables from 346 participants and classified into control or prediabetes based on fasting BGL. Blood and urine samples were analyzed for oxidative stress and inflammation markers. Logistic regression was used to compare multidimensional diagnostic models for prediabetes, including p66shc/Shc1, to the current HbA1c-only model in terms of sensitivity, specificity and predictive accuracy. Significance was set at P ≤ 0.05. RESULTS A significant decrease of p66shc/Shc1 was determined in prediabetes compared to controls (P ≤ 0.05). HbA1c testing resulted in an accuracy of 62%, while adding p66shc and triglycerides increased predictive accuracy to 88.05%. When HbA1c was omitted and Shc1 was combined with 8-hydroxy-2'-deoxyguanosine (8-OHdG) and monocyte chemo-attractant protein-1 (MCP-1), a predictive accuracy of 89.5% was achieved. CONCLUSION Our findings showed a major improvement of sensitivity to identify prediabetes by including oxidative stress and inflammatory biomarkers underlining beneficial diagnostic information, which most likely improves prevention and early treatment options in prediabetes.
Collapse
Affiliation(s)
- Herbert F Jelinek
- Department of Biomedical EngineeringKhalifa UniversityAbu DhabiUnited Arab Emirates
- Health Engineering Innovation CenterKhalifa UniversityAbu DhabiUnited Arab Emirates
- Biotechnology CenterKhalifa UniversityAbu DhabiUnited Arab Emirates
| | | | - Kinda Khalaf
- Department of Biomedical EngineeringKhalifa UniversityAbu DhabiUnited Arab Emirates
- Health Engineering Innovation CenterKhalifa UniversityAbu DhabiUnited Arab Emirates
| |
Collapse
|
13
|
TREM2 Regulates High Glucose-Induced Microglial Inflammation via the NLRP3 Signaling Pathway. Brain Sci 2021; 11:brainsci11070896. [PMID: 34356130 PMCID: PMC8306970 DOI: 10.3390/brainsci11070896] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 06/26/2021] [Accepted: 07/03/2021] [Indexed: 01/04/2023] Open
Abstract
Background: TREM2 expressed on microglia plays an important role in modulating inflammation in neurodegenerative diseases. It remains unknown whether TREM2 modulates hyperglycemia-induced microglial inflammation. Methods: We investigated the molecular function of TREM2 in high glucose-induced microglial inflammation using western blotting, qPCR, ELISA, pulldown, and co-IP methods. Results: Our data showed that in high glucose-induced BV2 cells, TREM2 was increased, and the proinflammatory cytokine IL-1β was increased. TREM2 knockout (KO) attenuated the proinflammatory cytokine IL-1β; conversely, TREM2 overexpression (OE) exacerbated IL-1β expression. Furthermore, we found that high glucose promoted the interaction of TREM2 with NLRP3. TREM2 KO abolished the interaction of TREM2 with NLRP3, while TREM2 OE enhanced the interaction. Moreover, TREM2 KO reduced high glucose-induced NLRP3 inflammasome activation, and TREM2 OE augmented high glucose-induced NLRP3 inflammasome activation, indicating that high glucose enhances the expression of TREM2, which activates the NLRP3 inflammasome. To further clarify whether the NLRP3 signaling pathway mediates the TREM2-regulated inflammatory response, we blocked the NLRP3 inflammasome by knocking out NLRP3 and treating cells with a caspase1 inhibitor, which decreased the levels of the IL-1β proinflammatory cytokine but did not affect the high glucose-induced expression of TREM2. Conclusions: TREM2 modulates high glucose-induced microglial inflammation via the NLRP3 signaling pathway.
Collapse
|
14
|
Mir HA, Ali R, Mushtaq U, Khanday FA. Structure-functional implications of longevity protein p66Shc in health and disease. Ageing Res Rev 2020; 63:101139. [PMID: 32795504 DOI: 10.1016/j.arr.2020.101139] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 07/17/2020] [Accepted: 08/06/2020] [Indexed: 12/15/2022]
Abstract
ShcA (Src homologous- collagen homologue), family of adapter proteins, consists of three isoforms which integrate and transduce external stimuli to different signaling networks. ShcA family consists of p46Shc, p52Shc and p66Shc isoforms, characterized by having multiple protein-lipid and protein-protein interaction domains implying their functional diversity. Among the three isoforms p66Shc is structurally different containing an additional CH2 domain which attributes to its dual functionality in cell growth, mediating both cell proliferation and apoptosis. Besides, p66Shc is also involved in different biological processes including reactive oxygen species (ROS) production, cell migration, ageing, cytoskeletal reorganization and cell adhesion. Moreover, the interplay between p66Shc and ROS is implicated in the pathology of various dreadful diseases. Accordingly, here we discuss the recent structural aspects of all ShcA adaptor proteins but are highlighting the case of p66Shc as model isoform. Furthermore, this review insights the role of p66Shc in progression of chronic age-related diseases like neuro diseases, metabolic disorders (non-alcoholic fatty liver, obesity, diabetes, cardiovascular diseases, vascular endothelial dysfunction) and cancer in relation to ROS. We finally conclude that p66Shc might act as a valuable biomarker for the prognosis of these diseases and could be used as a potential therapeutic target.
Collapse
|
15
|
El-Kott AF, Alshehri AS, Khalifa HS, Abd-Lateif AEKM, Alshehri MA, El-Maksoud MMA, Eid RA, Bin-Meferij MM. Cadmium Chloride Induces Memory Deficits and Hippocampal Damage by Activating the JNK/p 66Shc/NADPH Oxidase Axis. Int J Toxicol 2020; 39:477-490. [PMID: 32856499 DOI: 10.1177/1091581820930651] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
This study investigated whether the mechanism underlying the neurotoxic effects of cadmium chloride (CdCl2) in rats involves p66Shc. This study comprised an initial in vivo experiment followed by an in vitro experiment. For the in vivo experiment, male rats were orally administered saline (vehicle) or CdCl2 (0.05 mg/kg) for 30 days. Thereafter, spatial and retention memory of rats were tested and their hippocampi were used for biochemical and molecular analyses. For the in vitro experiment, control or p66Shc-deficient hippocampal cells were treated with CdCl2 (25 µM) in the presence or absence of SP600125, a c-Jun N-terminal kinase (JNK) inhibitor. Cadmium chloride impaired the spatial learning and retention memory of rats; depleted levels of glutathione and manganese superoxide dismutase; increased reactive oxygen species (ROS), tumor necrosis factor α, and interleukin 6; and induced nuclear factor kappa B activation. Cadmium chloride also decreased the number of pyramidal cells in the CA1 region and induced severe damage to the mitochondria and endoplasmic reticulum of cells in the hippocampi of rats. Moreover, CdCl2 increased the total unphosphorylated p66Shc, phosphorylated (Ser36) p66Shc, phosphorylated JNK, nicotinamide adenine dinucleotide phosphate (NADPH) oxidase, cytochrome c, and cleaved caspase-3. A dose-response increase in cell death, ROS, DNA damage, p66Shc, and NADPH oxidase was also observed in cultured hippocampal cells treated with CdCl2. Of note, all of these biochemical changes were attenuated by silencing p66Shc or inhibiting JNK with SP600125. In conclusion, CdCl2 induces hippocampal ROS generation and apoptosis by promoting the JNK-mediated activation of p66Shc.
Collapse
Affiliation(s)
- Attalla Farag El-Kott
- Biology Department, College of Science, 204574King Khalid University, Abha, Saudi Arabia.,Zoology Department, College of Science, 110144Damanhour University, Damanhour, Egypt
| | - Ali S Alshehri
- Biology Department, College of Science, 204574King Khalid University, Abha, Saudi Arabia
| | - Heba S Khalifa
- Zoology Department, College of Science, 110144Damanhour University, Damanhour, Egypt
| | | | - Mohammad Ali Alshehri
- Biology Department, College of Science, 204574King Khalid University, Abha, Saudi Arabia
| | - Mona M Abd El-Maksoud
- Community of Nursing Care, Nursing College, 204574King Khalid University, Abha, Saudi Arabia.,Community Health Nursing, Faculty of Nursing, Helwan University, Helwan, Egypt
| | - Refaat A Eid
- Department of Pathology, College of Medicine, 204574King Khalid University, Abha, Saudi Arabia
| | | |
Collapse
|
16
|
Mostafa DG, Satti HH. Resolvin D1 Prevents the Impairment in the Retention Memory and Hippocampal Damage in Rats Fed a Corn Oil-Based High Fat Diet by Upregulation of Nrf2 and Downregulation and Inactivation of p 66Shc. Neurochem Res 2020; 45:1576-1591. [PMID: 32253649 DOI: 10.1007/s11064-020-03022-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Revised: 03/27/2020] [Accepted: 04/02/2020] [Indexed: 12/31/2022]
Abstract
This study investigated the effect of a high-fat diet rich in corn oil (CO-HFD) on the memory retention and hippocampal oxidative stress, inflammation, and apoptosis in rats, and examined if the underlying mechanisms involve modulating Resolvin D1 (RvD1) levels and activation of p66Shc. Also, we tested if co-administration of RvD1 could prevent these neural adverse effects induced by CO-HFD. Adult male Wistar rats were divided into 4 groups (n = 18/each) as control fed standard diet (STD) (3.82 kcal/g), STD + RvD1 (0.2 µg/Kg, i.p/twice/week), CO-HFD (5.4 kcal/g), and CO-HFD + RvD1. All treatments were conducted for 8 weeks. With normal fasting glucose levels, CO-HFD induced hyperlipidemia, hyperinsulinemia, increased HOMA-IRI and reduced the rats' memory retention. In parallel, CO-HFD increased levels of reactive oxygen species (ROS), malondialdehyde (MDA), cytoplasmic cytochrome-c, and cleaved caspase-3 and significantly decreased levels of glutathione (GSH), Bcl-2, and manganese superoxide dismutase (MnSOD) in rats' hippocampi. Besides, CO-HFD significantly reduced hippocampal levels of docosahexaenoic acid (DHA) and RvD1, as well as total protein levels of Nrf2 and significantly increased nuclear protein levels of p-NF-κB. Concomitantly, CO-HFD increased hippocampal protein levels of p-JNK, p53, p66Shc, p-p66Shc, and NADPH oxidase. However, without altering plasma and serum levels of glucose, insulin, and lipids, co-administration of RvD1 to CO-HFD completely reversed all these events. It also resulted in similar effects in the STD fed-rats. In conclusion, CO-HFD impairs memory function and induces hippocampal damage by reducing levels of RvD1 and activation of JNK/p53/p66Shc/NADPH oxidase, effects that are prevented by co-administration of RvD1.
Collapse
Affiliation(s)
- Dalia G Mostafa
- Department of Medical Physiology, College of Medicine, Kingdom of Saudi Arabia, King Khalid University, P.O. Box 3340, Abha, 61421, Kingdom of Saudi Arabia. .,Department of Medical Physiology, Faculty of Medicine, Assiut University, Assiut, Egypt.
| | - Huda H Satti
- Department of Pathology, College of Medicine, Kingdom of Saudi Arabia, King Khalid University, P.O.Box 3340, Abha, 61421, Kingdom of Saudi Arabia.,Department of Pathology, Faculty of Medicine, University of Khartoum, Khartoum, Sudan
| |
Collapse
|
17
|
Ide M, Sonoda N, Inoue T, Kimura S, Minami Y, Makimura H, Hayashida E, Hyodo F, Yamato M, Takayanagi R, Inoguchi T. The dipeptidyl peptidase-4 inhibitor, linagliptin, improves cognitive impairment in streptozotocin-induced diabetic mice by inhibiting oxidative stress and microglial activation. PLoS One 2020; 15:e0228750. [PMID: 32032367 PMCID: PMC7006898 DOI: 10.1371/journal.pone.0228750] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Accepted: 01/21/2020] [Indexed: 12/15/2022] Open
Abstract
OBJECTIVE Accumulating epidemiological studies have demonstrated that diabetes is an important risk factor for dementia. However, the underlying pathological and molecular mechanisms, and effective treatment, have not been fully elucidated. Herein, we investigated the effect of the dipeptidyl peptidase-4 (DPP-4) inhibitor, linagliptin, on diabetes-related cognitive impairment. METHOD Streptozotocin (STZ)-induced diabetic mice were treated with linagliptin (3 mg/kg/24 h) for 17 weeks. The radial arm water maze test was performed, followed by evaluation of oxidative stress using DNP-MRI and the expression of NAD(P)H oxidase components and proinflammatory cytokines and of microglial activity. RESULTS Administration of linagliptin did not affect the plasma glucose and body weight of diabetic mice; however, it improved cognitive impairment. Additionally, linagliptin reduced oxidative stress and the mRNA expression of NAD(P)H oxidase component and TNF-α, and the number and body area of microglia, all of which were significantly increased in diabetic mice. CONCLUSIONS Linagliptin may have a beneficial effect on diabetes-related dementia by inhibiting oxidative stress and microglial activation, independently of glucose-lowering.
Collapse
Affiliation(s)
- Makoto Ide
- Department of Internal Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Noriyuki Sonoda
- Department of Internal Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
- Innovation Center for Medical Redox Navigation, Kyushu University, Fukuoka, Japan
- * E-mail:
| | - Tomoaki Inoue
- Department of Internal Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Shinichiro Kimura
- Department of Internal Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yohei Minami
- Department of Internal Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Hiroaki Makimura
- Department of Internal Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Eiichi Hayashida
- Department of Internal Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Fuminori Hyodo
- Innovation Center for Medical Redox Navigation, Kyushu University, Fukuoka, Japan
| | - Mayumi Yamato
- Innovation Center for Medical Redox Navigation, Kyushu University, Fukuoka, Japan
| | - Ryoichi Takayanagi
- Department of Internal Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Toyoshi Inoguchi
- Innovation Center for Medical Redox Navigation, Kyushu University, Fukuoka, Japan
- Fukuoka City Health Promotion Support Center, Fukuoka, Japan
| |
Collapse
|
18
|
Takenoshita N, Shimizu S, Kanetaka H, Sakurai H, Suzuki R, Miwa T, Odawara M, Ishii K, Shimada H, Higuchi M, Suhara T, Hanyu H. Classification of Clinically Diagnosed Alzheimer’s Disease Associated with Diabetes Based on Amyloid and Tau PET Results. J Alzheimers Dis 2019; 71:261-271. [DOI: 10.3233/jad-190620] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Affiliation(s)
- Naoto Takenoshita
- Department of Geriatric Medicine, Tokyo Medical University, Tokyo, Japan
| | - Soichiro Shimizu
- Department of Geriatric Medicine, Tokyo Medical University, Tokyo, Japan
| | - Hidekazu Kanetaka
- Department of Geriatric Medicine, Tokyo Medical University, Tokyo, Japan
| | - Hirofumi Sakurai
- Department of Geriatric Medicine, Tokyo Medical University, Tokyo, Japan
| | - Ryo Suzuki
- Department of Diabetes, Endocrinology and Metabolism, Tokyo Medical University, Tokyo, Japan
| | - Takashi Miwa
- Department of Diabetes, Endocrinology and Metabolism, Tokyo Medical University, Tokyo, Japan
| | - Masato Odawara
- Department of Diabetes, Endocrinology and Metabolism, Tokyo Medical University, Tokyo, Japan
| | - Kenji Ishii
- Research Team for Neuroimaging, Tokyo Metropolitan Institute of Gerontology, Tokyo, Japan
| | - Hitoshi Shimada
- Department of Functional Brain Imaging Research, Clinical Research Cluster, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba-shi, Chiba, Japan
| | - Makoto Higuchi
- Department of Functional Brain Imaging Research, Clinical Research Cluster, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba-shi, Chiba, Japan
| | - Tetsuya Suhara
- Department of Functional Brain Imaging Research, Clinical Research Cluster, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba-shi, Chiba, Japan
| | - Haruo Hanyu
- Department of Geriatric Medicine, Tokyo Medical University, Tokyo, Japan
| |
Collapse
|
19
|
Serine Phosphorylation of IRS1 Correlates with Aβ-Unrelated Memory Deficits and Elevation in Aβ Level Prior to the Onset of Memory Decline in AD. Nutrients 2019; 11:nu11081942. [PMID: 31426549 PMCID: PMC6723493 DOI: 10.3390/nu11081942] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Revised: 08/03/2019] [Accepted: 08/14/2019] [Indexed: 12/16/2022] Open
Abstract
The biological effects of insulin signaling are regulated by the phosphorylation of insulin receptor substrate 1 (IRS1) at serine (Ser) residues. In the brain, phosphorylation of IRS1 at specific Ser sites increases in patients with Alzheimer’s disease (AD) and its animal models. However, whether the activation of Ser sites on neural IRS1 is related to any type of memory decline remains unclear. Here, we show the modifications of IRS1 through its phosphorylation at etiology-specific Ser sites in various animal models of memory decline, such as diabetic, aged, and amyloid precursor protein (APP) knock-in NL-G-F (APPKINL-G-F) mice. Substantial phosphorylation of IRS1 at specific Ser sites occurs in type 2 diabetes- or age-related memory deficits independently of amyloid-β (Aβ). Furthermore, we present the first evidence that, in APPKINL-G-F mice showing Aβ42 elevation, the increased phosphorylation of IRS1 at multiple Ser sites occurs without memory impairment. Our findings suggest that the phosphorylation of IRS1 at specific Ser sites is a potential marker of Aβ-unrelated memory deficits caused by type 2 diabetes and aging; however, in Aβ-related memory decline, the modifications of IRS1 may be a marker of early detection of Aβ42 elevation prior to the onset of memory decline in AD.
Collapse
|
20
|
Dong Y, Wang Q, Yao H, Xiao Y, Wei J, Xie P, Hu J, Chen W, Tang Y, Zhou H, Liu J. A promising structural magnetic resonance imaging assessment in patients with preclinical cognitive decline and diabetes mellitus. J Cell Physiol 2019; 234:16838-16846. [PMID: 30786010 DOI: 10.1002/jcp.28359] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Revised: 01/30/2019] [Accepted: 02/01/2019] [Indexed: 01/18/2023]
Abstract
Subjective cognitive decline (SCD) is frequently reported in diabetic patients. Diabetes mellitus (DM) is associated with changes in the microstructure of the brain arise in diabetic patients, including changes in gray matter volume (GMV). However, the underlying mechanisms of changes in GMV in DM patients with cognitive impairment remain uncertain. Here, we present an overview of amyloid-β-dependent cognitive impairment in DM patients with SCD. Moreover, we review the evolving insights from studies on the GMV changes in GMV and cognitive dysfunction to which provide the mechanisms of cognitive impairment in T2DM. Ultimately, the novel structural magnetic resonance imaging (MRI) protocol was used for detecting neuroimaging biomarkers that can predict the clinical outcomes in diabetic patients with SCD. A reliable MRI protocol would be helpful to detect neurobiomarkers, and to understand the pathological mechanisms of preclinical cognitive impairment in diabetic patients.
Collapse
Affiliation(s)
- Yulan Dong
- Department of Radiology, the First Affiliated Hospital of University of South China, Hengyang, China
| | - Qi Wang
- Department of Radiology, the Hunan Province Hospital, Changsha, China
| | - Hailun Yao
- Institute of Pharmacy and Medical Technology, Hunan Polytechnic of Environment and Biology, Hengyang, Hunan, China
| | - Yawen Xiao
- Department of Radiology, the First Affiliated Hospital of University of South China, Hengyang, China
| | - Jiaohong Wei
- Department of Radiology, the First Affiliated Hospital of University of South China, Hengyang, China
| | - Peihan Xie
- Department of Radiology, the First Affiliated Hospital of University of South China, Hengyang, China
| | - Jun Hu
- Department of Radiology, the First Affiliated Hospital of University of South China, Hengyang, China
| | - Wen Chen
- Department of Radiology, the First Affiliated Hospital of University of South China, Hengyang, China
| | - Yan Tang
- Department of Ultrasound, the First Affiliated Hospital of University of South China, Hengyang, China
| | - Hong Zhou
- Department of Radiology, the First Affiliated Hospital of University of South China, Hengyang, China.,Hengyang Medical College, University of South China, Hengyang, China
| | - Jincai Liu
- Department of Radiology, the First Affiliated Hospital of University of South China, Hengyang, China
| |
Collapse
|
21
|
p66Shc activation promotes increased oxidative phosphorylation and renders CNS cells more vulnerable to amyloid beta toxicity. Sci Rep 2018; 8:17081. [PMID: 30459314 PMCID: PMC6244282 DOI: 10.1038/s41598-018-35114-y] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Accepted: 10/28/2018] [Indexed: 12/26/2022] Open
Abstract
A key pathological feature of Alzheimer's disease (AD) is the accumulation of the neurotoxic amyloid beta (Aβ) peptide within the brains of affected individuals. Previous studies have shown that neuronal cells selected for resistance to Aβ toxicity display a metabolic shift from mitochondrial-dependent oxidative phosphorylation (OXPHOS) to aerobic glycolysis to meet their energy needs. The Src homology/collagen (Shc) adaptor protein p66Shc is a key regulator of mitochondrial function, ROS production and aging. Moreover, increased expression and activation of p66Shc promotes a shift in the cellular metabolic state from aerobic glycolysis to OXPHOS in cancer cells. Here we evaluated the hypothesis that activation of p66Shc in CNS cells promotes both increased OXPHOS and enhanced sensitivity to Aβ toxicity. The effect of altered p66Shc expression on metabolic activity was assessed in rodent HT22 and B12 cell lines of neuronal and glial origin respectively. Overexpression of p66Shc repressed glycolytic enzyme expression and increased both mitochondrial electron transport chain activity and ROS levels in HT22 cells. The opposite effect was observed when endogenous p66Shc expression was knocked down in B12 cells. Moreover, p66Shc activation in both cell lines increased their sensitivity to Aβ toxicity. Our findings indicate that expression and activation of p66Shc renders CNS cells more sensitive to Aβ toxicity by promoting mitochondrial OXPHOS and ROS production while repressing aerobic glycolysis. Thus, p66Shc may represent a potential therapeutically relevant target for the treatment of AD.
Collapse
|