1
|
Karati D, Meur S, Roy S, Mukherjee S, Debnath B, Jha SK, Sarkar BK, Naskar S, Ghosh P. Glycogen synthase kinase 3 (GSK3) inhibition: a potential therapeutic strategy for Alzheimer's disease. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:2319-2342. [PMID: 39432068 DOI: 10.1007/s00210-024-03500-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Accepted: 09/30/2024] [Indexed: 10/22/2024]
Abstract
Alzheimer's disease (AD), the most common type of dementia among older adults, is a chronic neurodegenerative pathology that causes a progressive loss of cognitive functioning with a decline of rational skills. It is well known that AD is multifactorial, so there are many different pharmacological targets that can be pursued. According to estimates from the World Health Organization (WHO), 18 million individuals worldwide suffer from AD. Major initiatives to identify risk factors, enhance care giving, and conduct basic research to delay the beginning of AD were started by the USA, France, Germany, France, and various other nations. Widely recognized as a key player in the development and subsequent progression of AD pathogenesis, glycogen synthase kinase-3 (GSK-3) controls a number of crucial targets associated with neuronal degeneration. GSK-3 inhibition has been linked to reduced tau hyperphosphorylation, β-amyloid formation, and neuroprotective benefits in Alzheimer's disease. Lithium, the very first inhibitor of GSK-3β that was used therapeutically, has been successfully used for many years with remarkable results. A great variety of structurally varied strong GSK-3β blockers have been identified in recent years. The purpose of this thorough review is to cover the biological and structural elements of glycogen synthase kinase, as well as the medicinal chemistry aspects of GSK inhibitors that have been produced in recent years.
Collapse
Affiliation(s)
- Dipanjan Karati
- Department of Pharmaceutical Technology, School of Pharmacy, Techno India University, Kolkata, West Bengal, 700091, India
| | - Shreyasi Meur
- Department of Pharmaceutical Technology, School of Pharmacy, Techno India University, Kolkata, West Bengal, 700091, India
| | - Souvik Roy
- Department of Pharmaceutical Technology, NSHM Knowledge Campus, Kolkata - Group of Institutions, 124, B.L Saha Road, Kolkata, West Bengal, 700053, India
| | - Swarupananda Mukherjee
- Department of Pharmaceutical Technology, NSHM Knowledge Campus, Kolkata - Group of Institutions, 124, B.L Saha Road, Kolkata, West Bengal, 700053, India.
| | - Biplab Debnath
- Department of Pharmaceutical Technology, Bharat Technology, Uluberia, Howrah, West Bengal, 711316, India
| | - Sajal Kumar Jha
- Department of Pharmaceutical Technology, Bengal College of Pharmaceutical Technology, Dubrajpur, West Bengal, 731123, India
| | | | - Saheli Naskar
- Department of Pharmaceutical Technology, NSHM Knowledge Campus, Kolkata - Group of Institutions, 124, B.L Saha Road, Kolkata, West Bengal, 700053, India
| | - Priya Ghosh
- Department of Pharmaceutical Technology, NSHM Knowledge Campus, Kolkata - Group of Institutions, 124, B.L Saha Road, Kolkata, West Bengal, 700053, India
| |
Collapse
|
2
|
Rahman SO, Khan T, Iqubal A, Agarwal S, Akhtar M, Parvez S, Shah ZA, Najmi AK. Association between insulin and Nrf2 signalling pathway in Alzheimer's disease: A molecular landscape. Life Sci 2023:121899. [PMID: 37394097 DOI: 10.1016/j.lfs.2023.121899] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 06/17/2023] [Accepted: 06/27/2023] [Indexed: 07/04/2023]
Abstract
Insulin, a well-known hormone, has been implicated as a regulator of blood glucose levels for almost a century now. Over the past few decades, the non-glycemic actions of insulin i.e. neuronal growth and proliferation have been extensively studied. In 2005, Dr. Suzanne de La Monte and her team reported that insulin might be involved in the pathogenesis of Alzheimer's Disease (AD) and thus coined a term "Type-3 diabetes" This hypothesis was supported by several subsequent studies. The nuclear factor erythroid 2- related factor 2 (Nrf2) triggers a cascade of events under the regulation of distinct mechanisms including protein stability, phosphorylation and nuclear cytoplasmic shuttling, finally leading to the protection against oxidative damage. The Nrf2 pathway has been investigated extensively in relevance to neurodegenerative disorders, particularly AD. Many studies have indicated a strong correlation between insulin and Nrf2 signalling pathways both in the periphery and the brainbut merely few of them have focused on elucidating their inter-connective role in AD. The present review emphasizes key molecular pathways that correlate the role of insulin with Nrf2 during AD. The review has also identified key unexplored areas that could be investigated in future to further establish the insulin and Nrf2 influence in AD.
Collapse
Affiliation(s)
- Syed Obaidur Rahman
- Pharmaceutical Medicine, Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - Tahira Khan
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - Ashif Iqubal
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - Shivani Agarwal
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - Mohd Akhtar
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - Suhel Parvez
- Neurobehavioral Pharmacology Laboratory, Department of Medical Elementology and Toxicology, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi 110062, India
| | - Zahoor Ahmad Shah
- Department of Medicinal and Biological Chemistry, University of Toledo, 3000 Arlington Avenue, Toledo, OH 43614, USA
| | - Abul Kalam Najmi
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India.
| |
Collapse
|
3
|
Mani S, Dubey R, Lai IC, Babu MA, Tyagi S, Swargiary G, Mody D, Singh M, Agarwal S, Iqbal D, Kumar S, Hamed M, Sachdeva P, Almutary AG, Albadrani HM, Ojha S, Singh SK, Jha NK. Oxidative Stress and Natural Antioxidants: Back and Forth in the Neurological Mechanisms of Alzheimer's Disease. J Alzheimers Dis 2023; 96:877-912. [PMID: 37927255 DOI: 10.3233/jad-220700] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2023]
Abstract
Alzheimer's disease (AD) is characterized by the progressive degeneration of neuronal cells. With the increase in aged population, there is a prevalence of irreversible neurodegenerative changes, causing a significant mental, social, and economic burden globally. The factors contributing to AD are multidimensional, highly complex, and not completely understood. However, it is widely known that aging, neuroinflammation, and excessive production of reactive oxygen species (ROS), along with other free radicals, substantially contribute to oxidative stress and cell death, which are inextricably linked. While oxidative stress is undeniably important in AD, limiting free radicals and ROS levels is an intriguing and potential strategy for deferring the process of neurodegeneration and alleviating associated symptoms. Therapeutic compounds from natural sources have recently become increasingly accepted and have been effectively studied for AD treatment. These phytocompounds are widely available and a multitude of holistic therapeutic efficiencies for treating AD owing to their antioxidant, anti-inflammatory, and biological activities. Some of these compounds also function by stimulating cholinergic neurotransmission, facilitating the suppression of beta-site amyloid precursor protein-cleaving enzyme 1, α-synuclein, and monoamine oxidase proteins, and deterring the occurrence of AD. Additionally, various phenolic, flavonoid, and terpenoid phytocompounds have been extensively described as potential palliative agents for AD progression. Preclinical studies have shown their involvement in modulating the cellular redox balance and minimizing ROS formation, displaying them as antioxidant agents with neuroprotective abilities. This review emphasizes the mechanistic role of natural products in the treatment of AD and discusses the various pathological hypotheses proposed for AD.
Collapse
Affiliation(s)
- Shalini Mani
- Centre for Emerging Diseases, Department of Biotechnology, Jaypee Institute of Information Technology, Noida, UP, India
| | - Rajni Dubey
- Division of Cardiology, Department of Internal Medicine, Taipei Medical University Hospital, Taipei, Taiwan
| | - I-Chun Lai
- School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Division of Radiation Oncology, Department of Oncology, Taipei Veterans General Hospital, Taipei, Taiwan
- The Ph.D. Program for Translational Medicine, College of Medical Science and Technology, Taipei Medical University and Academia Sinica, Taipei, Taiwan
| | - M Arockia Babu
- Institute of Pharmaceutical Research, GLA University, Mathura, India
| | - Sakshi Tyagi
- Centre for Emerging Diseases, Department of Biotechnology, Jaypee Institute of Information Technology, Noida, UP, India
| | - Geeta Swargiary
- Centre for Emerging Diseases, Department of Biotechnology, Jaypee Institute of Information Technology, Noida, UP, India
| | - Deepansh Mody
- Centre for Emerging Diseases, Department of Biotechnology, Jaypee Institute of Information Technology, Noida, UP, India
| | - Manisha Singh
- Centre for Emerging Diseases, Department of Biotechnology, Jaypee Institute of Information Technology, Noida, UP, India
| | - Shriya Agarwal
- Department of Molecular Sciences, Macquarie University, Sydney, Australia
| | - Danish Iqbal
- Department of Health Information Management, College of Applied Medical Sciences, Buraydah Private Colleges, Buraydah, Saudi Arabia
| | - Sanjay Kumar
- Department of Life Sciences, School of Basic Sciences and Research (SBSR), Sharda University, Greater Noida, Uttar Pradesh, India
| | - Munerah Hamed
- Department of Pathology, Faculty of Medicine, Umm Al-Qura University, Makkah, Saudi Arabia
| | | | - Abdulmajeed G Almutary
- Department of Biomedical Sciences, College of Health Sciences, Abu Dhabi University, Abu Dhabi, United Arab Emirates
| | - Hind Muteb Albadrani
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Imam Abdulrahman Bin Faisal University, Dammam, Eastern Province, Kingdom of Saudi Arabia
| | - Shreesh Ojha
- Department of Pharmacology and Therapeutics, College of Medicine and Health Sciences, United Arab Emirates University, Abu Dhabi, United Arab Emirates
| | | | - Niraj Kumar Jha
- Department of Biotechnology, School of Engineering & Technology (SET), Sharda University, Greater Noida, Uttar Pradesh, India
- School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, Punjab, India
- Department of Biotechnology, School of Applied & Life Sciences (SALS), Uttaranchal University, Dehradun, Uttarakhand, India
- Department of Biotechnology Engineering and Food Technology, Chandigarh University, Mohali, India
| |
Collapse
|
4
|
El-Sayed NS, Elatrebi S, Said R, Ibrahim HF, Omar EM. Potential mechanisms underlying the association between type II diabetes mellitus and cognitive dysfunction in rats: a link between miRNA-21 and Resveratrol's neuroprotective action. Metab Brain Dis 2022; 37:2375-2388. [PMID: 35781592 PMCID: PMC9581846 DOI: 10.1007/s11011-022-01035-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 06/13/2022] [Indexed: 11/10/2022]
Abstract
Cognitive impairment is considered as a typical feature of neurodegenerative diseases in diabetes mellitus (DM). However, the exact link between cognitive dysfunction and diabetes mellitus is still vague. This study aims to investigate some of the mechanisms underlying cognitive impairment that associates diabetes mellitus and insulin resistance. We investigated the role of resveratrol as well on cognitive function in experimentally induced type 2 diabetes highlighting on its influence on the expression of brain miRNA 21. Resveratrol is a naturally occurring, biologically active compound that has numerous significant impacts on the body. Type 2 diabetes mellitus was induced by high fat diet followed a single dose of streptozotocin. Diabetic rats were treated with resveratrol for four weeks. Rats were sacrificed after neurobehavioral testing. Hippocampal tissues were used to assess expression of miRNA 21, GSK and oxidative stress markers. Serum samples were obtained to determine glucose levels, lipid profile and insulin levels. Hippocampal and serum AGEs were measured as well and HOMA IR was calculated. We detected memory impairment and disturbed insulin signaling in diabetic rats. These derangements were reversed by resveratrol treatment partially due to increased expression of miRNA-21. Our study pins the role of miRNA-21 in modulating brain insulin signaling and hence alleviating cognitive dysfunction accompanying diabetes mellitus.
Collapse
Affiliation(s)
- Norhan S. El-Sayed
- Department of Medical Physiology, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Soha Elatrebi
- Department of Clinical Pharmacology, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Rasha Said
- Department of Medical Biochemistry, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Heba F. Ibrahim
- Department of Histology and Cell Biology, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Eman M. Omar
- Department of Medical Physiology, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| |
Collapse
|
5
|
The Role of Insulin Resistance in Fueling NAFLD Pathogenesis: From Molecular Mechanisms to Clinical Implications. J Clin Med 2022; 11:jcm11133649. [PMID: 35806934 PMCID: PMC9267803 DOI: 10.3390/jcm11133649] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 06/09/2022] [Accepted: 06/21/2022] [Indexed: 02/06/2023] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) represents a predominant hepatopathy that is rapidly becoming the most common cause of hepatocellular carcinoma worldwide. The close association with metabolic syndrome’s extrahepatic components has suggested the nature of the systemic metabolic-related disorder based on the interplay between genetic, nutritional, and environmental factors, creating a complex network of yet-unclarified pathogenetic mechanisms in which the role of insulin resistance (IR) could be crucial. This review detailed the clinical and pathogenetic evidence involved in the NAFLD–IR relationship, presenting both the classic and more innovative models. In particular, we focused on the reciprocal effects of IR, oxidative stress, and systemic inflammation on insulin-sensitivity disruption in critical regions such as the hepatic and the adipose tissue, while considering the impact of genetics/epigenetics on the regulation of IR mechanisms as well as nutrients on specific insulin-related gene expression (nutrigenetics and nutrigenomics). In addition, we discussed the emerging capability of the gut microbiota to interfere with physiological signaling of the hormonal pathways responsible for maintaining metabolic homeostasis and by inducing an abnormal activation of the immune system. The translation of these novel findings into clinical practice could promote the expansion of accurate diagnostic/prognostic stratification tools and tailored pharmacological approaches.
Collapse
|
6
|
Cadamuro M, Lasagni A, Sarcognato S, Guido M, Fabris R, Strazzabosco M, Strain AJ, Simioni P, Villa E, Fabris L. The Neglected Role of Bile Duct Epithelial Cells in NASH. Semin Liver Dis 2022; 42:34-47. [PMID: 34794182 DOI: 10.1055/s-0041-1739455] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is the most prevalent liver disease worldwide, and affects 25% of the population in Western countries. NAFLD is the hepatic manifestation of the metabolic syndrome, linked to insulin resistance, which is the common pathogenetic mechanism. In approximately 40% of NAFLD patients, steatosis is associated with necro-inflammation and fibrosis, resulting in nonalcoholic steatohepatitis (NASH), a severe condition that may progress to cirrhosis and liver cancer. Although the hepatocyte represents the main target of the disease, involvement of the bile ducts occurs in a subset of patients with NASH, and is characterized by ductular reaction and activation of the progenitor cell compartment, which incites portal fibrosis and disease progression. We aim to dissect the multiple biological effects that adipokines and metabolic alterations exert on cholangiocytes to derive novel information on the mechanisms driven by insulin resistance, which promote fibro-inflammation and carcinogenesis in NASH.
Collapse
Affiliation(s)
| | - Alberto Lasagni
- Division of General Medicine, Padua University-Hospital, Padua, Italy
| | | | - Maria Guido
- Department of Pathology, Azienda ULSS2 Marca Trevigiana, Treviso, Italy.,Department of Medicine (DIMED), University of Padua, Padua, Italy
| | - Roberto Fabris
- Division of Clinica Medica 3, Center for the Study and the Integrated Management of Obesity, Padua University-Hospital, Padua, Italy
| | - Mario Strazzabosco
- Department of Internal Medicine, Digestive Disease Section, Liver Center, Yale University, New Haven, Connecticut
| | - Alastair J Strain
- School of Biosciences, University of Birmingham, Birmingham, United Kingdom
| | - Paolo Simioni
- Division of General Medicine, Padua University-Hospital, Padua, Italy.,Department of Medicine (DIMED), University of Padua, Padua, Italy
| | - Erica Villa
- Gastroenterology Unit, Department of Medical Specialties, University of Modena & Reggio Emilia and Modena University-Hospital, Modena, Italy
| | - Luca Fabris
- Department of Molecular Medicine (DMM), University of Padua, Padua, Italy.,Division of General Medicine, Padua University-Hospital, Padua, Italy.,Department of Internal Medicine, Digestive Disease Section, Liver Center, Yale University, New Haven, Connecticut
| |
Collapse
|
7
|
Behl T, Arora A, Sehgal A, Singh S, Sharma N, Bhatia S, Al-Harrasi A, Bungau S, Mostafavi E. Molecular and Biochemical Pathways Encompassing Diabetes Mellitus and Dementia. CNS & NEUROLOGICAL DISORDERS-DRUG TARGETS 2021; 21:542-556. [PMID: 34758720 DOI: 10.2174/1871527320666211110115257] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 08/25/2021] [Accepted: 09/15/2021] [Indexed: 11/22/2022]
Abstract
Diabetes mellitus is a major metabolic disorder that has now emerged as an epidemic, and it affects the brain through an array of pathways. Diabetes mellitus patients can develop pathological changes in the brain, which eventually take the shape of mild cognitive impairment progressing to Alzheimer's Disease. A number of preclinical and clinical studies demonstrate this fact, and it comes out to be those molecular pathways such as amyloidogenesis, oxidative stress, inflammation, and impaired insulin signaling are identical in diabetes mellitus and dementia. However, the critical player involved in the vicious cycle of diabetes mellitus and dementia is insulin, whose signaling, when impaired in diabetes mellitus (both type 1 and 2), leads to a decline in cognition, although other pathways are also essential contributors. Moreover, it is not only that diabetes mellitus patients indicate cognitive decline at a later stage; many Alzheimer's Disease patients also reflect symptoms of diabetes mellitus, thus creating a vicious cycle inculcating a web of complex molecular mechanisms and hence categorizing Alzheimer's Disease as 'brain diabetes'. Thus, it is practical to suggest that anti-diabetic drugs are beneficial in Alzheimer's Disease; but only smaller trials, not the larger ones, have showcased positive outcomes mainly because of the late onset of therapy. Therefore, it is extremely important to develop more of such molecules that target insulin in dementia patients along with such methods that diagnose impaired insulin signaling and the associated cognitive decline so that early therapy may be initiated and the progression of the disease be prevented.
Collapse
Affiliation(s)
- Tapan Behl
- Chitkara College of Pharmacy, Chitkara University, Punjab. India
| | - Arpita Arora
- Chitkara College of Pharmacy, Chitkara University, Punjab. India
| | - Aayush Sehgal
- Chitkara College of Pharmacy, Chitkara University, Punjab. India
| | - Sukhbir Singh
- Chitkara College of Pharmacy, Chitkara University, Punjab. India
| | - Neelam Sharma
- Chitkara College of Pharmacy, Chitkara University, Punjab. India
| | - Saurabh Bhatia
- Amity Institute of Pharmacy, Amity University, Haryana. India
| | - Ahmed Al-Harrasi
- Natural & Medical Sciences Research Centre, University of Nizwa, Nizwa. Oman
| | - Simona Bungau
- Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, Oradea. Romania
| | - Ebrahim Mostafavi
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA. United States
| |
Collapse
|
8
|
Bagaméry F, Varga K, Kecsmár K, Vincze I, Szökő É, Tábi T. The Impact of Differentiation on Cytotoxicity and Insulin Sensitivity in Streptozotocin Treated SH-SY5Y Cells. Neurochem Res 2021; 46:1350-1358. [PMID: 33616807 PMCID: PMC8084777 DOI: 10.1007/s11064-021-03269-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2020] [Revised: 02/01/2021] [Accepted: 02/05/2021] [Indexed: 11/29/2022]
Abstract
Recently neuronal insulin resistance was suggested playing a role in Alzheimer’s disease. Streptozotocin (STZ) is commonly used to induce impairment in insulin metabolism. In our previous work on undifferentiated SH-SY5Y cells the compound exerted cytotoxicity without altering insulin sensitivity. Nevertheless, differentiation of the cells to a more mature neuron-like phenotype may considerably affect the significance of insulin signaling and its sensitivity to STZ. We aimed at studying the influence of STZ treatment on insulin signaling in SH-SY5Y cells differentiated by retinoic acid (RA). Cytotoxicity of STZ or low serum (LS) condition and protective effect of insulin were compared in RA differentiated SH-SY5Y cells. The effect of insulin and an incretin analogue, exendin-4 on insulin signaling was also examined by assessing glycogen synthase kinase-3 (GSK-3) phosphorylation. STZ was found less cytotoxic in the differentiated cells compared to our previous results in undifferentiated SH-SY5Y cells. The cytoprotective concentration of insulin was similar in the STZ and LS groups. However, the right-shifted concentration–response curve of insulin induced GSK-3 phosphorylation in STZ-treated differentiated cells is suggestive of the development of insulin resistance that was further confirmed by the insulin potentiating effect of exendin-4. Differentiation reduced the sensitivity of SH-SY5Y cells for the non-specific cytotoxicity of STZ and enhanced the relative significance of development of insulin resistance. The differentiated cells thus serve as a better model for studying the role of insulin signaling in neuronal survival. However, direct cytotoxicity of STZ also contributes to the cell death.
Collapse
Affiliation(s)
- Fruzsina Bagaméry
- Department of Pharmacodynamics, Semmelweis University, Nagyvárad tér 4, 1089, Budapest, Hungary
| | - Kamilla Varga
- Department of Pharmacodynamics, Semmelweis University, Nagyvárad tér 4, 1089, Budapest, Hungary
| | - Kitti Kecsmár
- Department of Pharmacodynamics, Semmelweis University, Nagyvárad tér 4, 1089, Budapest, Hungary
| | - István Vincze
- Department of Pharmacodynamics, Semmelweis University, Nagyvárad tér 4, 1089, Budapest, Hungary
| | - Éva Szökő
- Department of Pharmacodynamics, Semmelweis University, Nagyvárad tér 4, 1089, Budapest, Hungary
| | - Tamás Tábi
- Department of Pharmacodynamics, Semmelweis University, Nagyvárad tér 4, 1089, Budapest, Hungary.
| |
Collapse
|
9
|
Huang G, Zhang G, Yu Z. Computational prediction and analysis of histone H3k27me1-associated miRNAs. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2020; 1869:140539. [PMID: 32947024 DOI: 10.1016/j.bbapap.2020.140539] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 08/29/2020] [Accepted: 09/10/2020] [Indexed: 12/31/2022]
Abstract
The mono-methylation of histone H3 on lysine 27 (H3K27me1) plays key roles in the cellular processes. The H3K27me1 interacts with the DNA sequence of the miRNAs and regulates the transcription of miRNAs. Therefore, biological roles of the H3K27me1 are closely related to the downstream miRNAs. We proposed a machine learning-based computational method to predict H3K27me1-associated miRNAs and obtained AUCs of 0.6866 and 0.6849 on the leave-one-out and five-fold cross validation, respectively. We also performed enrichment analysis of the transcript factors, GO terms and pathways of H3K27me1-associated miRNAs. Among the top 10 significantly enriched transcription factors, five were unfavorable prognostic marker in renal cancer. The enrichment analysis of molecular function showed that the H3K27me1-associated miRNAs were linked to RNA binding and protein binding which were involved in the transcription and translation regulation. The enrichment of pathway showed that H3K27me1-associated miRNAs were mainly involved in pathways related to cancers, signaling and virus.
Collapse
Affiliation(s)
- Guohua Huang
- Provincial Key Laboratory of Informational Service for Rural Area of Southwestern Hunan, Shaoyang University, Shaoyang 422000, China.
| | - Guiyang Zhang
- Provincial Key Laboratory of Informational Service for Rural Area of Southwestern Hunan, Shaoyang University, Shaoyang 422000, China
| | - Zuguo Yu
- Key Laboratory of Intelligent Computing and Information Processing of Ministry of Education and Hunan Key Laboratory for Computation and Simulation in Science and Engineering, Xiangtan University, Xiangtan, Hunan 411105, China.
| |
Collapse
|
10
|
Oxidative Stress-Mediated Blood-Brain Barrier (BBB) Disruption in Neurological Diseases. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020. [DOI: 10.1155/2020/4356386] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The blood-brain barrier (BBB), as a crucial gate of brain-blood molecular exchange, is involved in the pathogenesis of multiple neurological diseases. Oxidative stress is caused by an imbalance between the production of reactive oxygen species (ROS) and the scavenger system. Since oxidative stress plays a significant role in the production and maintenance of the BBB, the cerebrovascular system is especially vulnerable to it. The pathways that initiate BBB dysfunction include, but are not limited to, mitochondrial dysfunction, excitotoxicity, iron metabolism, cytokines, pyroptosis, and necroptosis, all converging on the generation of ROS. Interestingly, ROS also provide common triggers that directly regulate BBB damage, parameters including tight junction (TJ) modifications, transporters, matrix metalloproteinase (MMP) activation, inflammatory responses, and autophagy. We will discuss the role of oxidative stress-mediated BBB disruption in neurological diseases, such as hemorrhagic stroke, ischemic stroke (IS), Alzheimer’s disease (AD), Parkinson’s disease (PD), traumatic brain injury (TBI), amyotrophic lateral sclerosis (ALS), and cerebral small vessel disease (CSVD). This review will also discuss the latest clinical evidence of potential biomarkers and antioxidant drugs towards oxidative stress in neurological diseases. A deeper understanding of how oxidative stress damages BBB may open up more therapeutic options for the treatment of neurological diseases.
Collapse
|
11
|
Vilariño-García T, Pérez-Pérez A, Santamaría-López E, Prados N, Fernández-Sánchez M, Sánchez-Margalet V. Sam68 mediates leptin signaling and action in human granulosa cells: possible role in leptin resistance in PCOS. Endocr Connect 2020; 9:479-488. [PMID: 32375121 PMCID: PMC7354740 DOI: 10.1530/ec-20-0062] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2020] [Accepted: 05/06/2020] [Indexed: 12/17/2022]
Abstract
INTRODUCTION Polycystic ovary syndrome (PCOS) is a complex metabolic disorder associated with ovulatory dysfunction, hyperandrogenism, obesity, and insulin resistance, that leads to subfertility. Sam68 is an RNA-binding protein with signaling functions that is ubiquitously expressed, including gonads. Sam68 is recruited to leptin signaling, mediating different leptin actions. OBJECTIVE We aimed to investigate the role of Sam68 in leptin signaling, mediating the effect on aromatase expression in granulosa cells and the posible implication of Sam68 in the leptin resistance in PCOS. MATERIALS AND METHODS Granulosa cells were from healthy donors (n = 25) and women with PCOS (n = 25), within the age range of 20 to 40 years, from Valencian Infertility Institute (IVI), Seville, Spain. Sam68 expression was inhibited by siRNA method and overexpressed by expression vector. Expression level was analysed by qPCR and immunoblot. Statistical significance was assessed by ANOVA followed by different post-hoc tests. A P value of <0.05 was considered statistically significant. RESULTS We have found that leptin stimulation increases phosphorylation and expression level of Sam68 and aromatase in granulosa cells from normal donors. Downregulation of Sam68 expression resulted in a lower activation of MAPK and PI3K pathways in response to leptin, whereas overexpression of Sam68 increased leptin stimulation of signaling, enhancing aromatase expression. Granulosa cells from women with PCOS presented lower expression of Sam68 and were resistant to the leptin effect on aromatase expression. CONCLUSIONS These results suggest the participation of Sam68 in leptin receptor signaling, mediating the leptin effect on aromatase expression in granulosa cells, and point to a new target in leptin resistance in PCOS.
Collapse
Affiliation(s)
- Teresa Vilariño-García
- Department of Medical Biochemistry, Molecular Biology and Immunology. Medical School, Virgen Macarena University Hospital, University of Seville, Seville, Spain
| | - Antonio Pérez-Pérez
- Department of Medical Biochemistry, Molecular Biology and Immunology. Medical School, Virgen Macarena University Hospital, University of Seville, Seville, Spain
| | | | | | | | - Víctor Sánchez-Margalet
- Department of Medical Biochemistry, Molecular Biology and Immunology. Medical School, Virgen Macarena University Hospital, University of Seville, Seville, Spain
- Correspondence should be addressed to V Sánchez-Margalet:
| |
Collapse
|
12
|
Berbudi A, Rahmadika N, Tjahjadi AI, Ruslami R. Type 2 Diabetes and its Impact on the Immune System. Curr Diabetes Rev 2020; 16:442-449. [PMID: 31657690 PMCID: PMC7475801 DOI: 10.2174/1573399815666191024085838] [Citation(s) in RCA: 500] [Impact Index Per Article: 100.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Revised: 07/24/2019] [Accepted: 09/16/2019] [Indexed: 02/06/2023]
Abstract
INTRODUCTION Type 2 Diabetes (T2D) is a major health problem worldwide. This metabolic disease is indicated by high blood glucose levels due to insufficient insulin production by the pancreas. An inflammatory response occurs as a result of the immune response to high blood glucose levels as well as the presence of inflammatory mediators produced by adipocytes and macrophages in fat tissue. This low and chronic inflammation damages the pancreatic beta cells and leads to insufficient insulin production, which results in hyperglycemia. Hyperglycemia in diabetes is thought to cause dysfunction of the immune response, which fails to control the spread of invading pathogens in diabetic subjects. Therefore, diabetic subjects are known to more susceptible to infections. The increased prevalence of T2D will increase the incidence of infectious diseases and related comorbidities. OBJECTIVE This review provides an overview of the immunological aspect of T2D and the possible mechanisms that result in increased infections in diabetics. CONCLUSION A better understanding of how immune dysfunctions occur during hyperglycemia can lead to novel treatments and preventions for infectious diseases and T2D comorbidities, thus improving the outcome of infectious disease treatment in T2D patients.
Collapse
Affiliation(s)
- Afiat Berbudi
- Department of Biomedical Sciences, Parasitology Division, Faculty of Medicine, Universitas Padjadjaran, Bandung, Indonesia
- Infectious Disease Research Center, Faculty of Medicine, Universitas Padjadjaran, Bandung, Indonesia
- Address correspondence to this author at the Department of Biomedical Sciences, Faculty of Medicine, Universitas Padjadjaran, Jl. Raya Bandung Sumedang Km. 21, 45363, West Java, Indonesia; E-mail:
| | - Nofri Rahmadika
- Infectious Disease Research Center, Faculty of Medicine, Universitas Padjadjaran, Bandung, Indonesia
| | - Adi Imam Tjahjadi
- Infectious Disease Research Center, Faculty of Medicine, Universitas Padjadjaran, Bandung, Indonesia
- Department of Biomedical Sciences, Microbiology Division, Faculty of Medicine, Universitas Padjadjaran, Bandung, Indonesia
| | - Rovina Ruslami
- Infectious Disease Research Center, Faculty of Medicine, Universitas Padjadjaran, Bandung, Indonesia
- Department of Biomedical Sciences, Pharmacology and Therapy Division, Faculty of Medicine, Universitas Padjadjaran, Bandung, Indonesia
| |
Collapse
|
13
|
Shalaby MA, Nounou HA, Deif MM. The potential value of capsaicin in modulating cognitive functions in a rat model of streptozotocin-induced Alzheimer’s disease. THE EGYPTIAN JOURNAL OF NEUROLOGY, PSYCHIATRY AND NEUROSURGERY 2019. [DOI: 10.1186/s41983-019-0094-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
|
14
|
Bessone F, Razori MV, Roma MG. Molecular pathways of nonalcoholic fatty liver disease development and progression. Cell Mol Life Sci 2019; 76:99-128. [PMID: 30343320 PMCID: PMC11105781 DOI: 10.1007/s00018-018-2947-0] [Citation(s) in RCA: 405] [Impact Index Per Article: 67.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Revised: 10/10/2018] [Accepted: 10/15/2018] [Indexed: 02/06/2023]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is a main hepatic manifestation of metabolic syndrome. It represents a wide spectrum of histopathological abnormalities ranging from simple steatosis to nonalcoholic steatohepatitis (NASH) with or without fibrosis and, eventually, cirrhosis and hepatocellular carcinoma. While hepatic simple steatosis seems to be a rather benign manifestation of hepatic triglyceride accumulation, the buildup of highly toxic free fatty acids associated with insulin resistance-induced massive free fatty acid mobilization from adipose tissue and the increased de novo hepatic fatty acid synthesis from glucose acts as the "first hit" for NAFLD development. NAFLD progression seems to involve the occurrence of "parallel, multiple-hit" injuries, such as oxidative stress-induced mitochondrial dysfunction, endoplasmic reticulum stress, endotoxin-induced, TLR4-dependent release of inflammatory cytokines, and iron overload, among many others. These deleterious factors are responsible for the triggering of a number of signaling cascades leading to inflammation, cell death, and fibrosis, the hallmarks of NASH. This review is aimed at integrating the overwhelming progress made in the characterization of the physiopathological mechanisms of NAFLD at a molecular level, to better understand the factor influencing the initiation and progression of the disease.
Collapse
Affiliation(s)
- Fernando Bessone
- Hospital Provincial del Centenario, Facultad de Ciencias Médicas, Servicio de Gastroenterología y Hepatología, Universidad Nacional de Rosario, Rosario, Argentina
| | - María Valeria Razori
- Instituto de Fisiología Experimental (IFISE-CONICET), Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario, Suipacha 570, 2000, Rosario, Argentina
| | - Marcelo G Roma
- Instituto de Fisiología Experimental (IFISE-CONICET), Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario, Suipacha 570, 2000, Rosario, Argentina.
| |
Collapse
|
15
|
Maternal Cognitive Impairment Associated with Gestational Diabetes Mellitus-A Review of Potential Contributing Mechanisms. Int J Mol Sci 2018; 19:ijms19123894. [PMID: 30563117 PMCID: PMC6321050 DOI: 10.3390/ijms19123894] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2018] [Revised: 11/14/2018] [Accepted: 11/29/2018] [Indexed: 12/16/2022] Open
Abstract
Gestational diabetes mellitus (GDM) carries many risks, where high blood pressure, preeclampsia and future type II diabetes are widely acknowledged, but less focus has been placed on its effect on cognitive function. Although the multifactorial pathogenesis of maternal cognitive impairment is not completely understood, it shares several features with type 2 diabetes mellitus (T2DM). In this review, we discuss some key pathophysiologies of GDM that may lead to cognitive impairment, specifically hyperglycemia, insulin resistance, oxidative stress, and neuroinflammation. We explain how these incidents: (i) impair the insulin-signaling pathway and/or (ii) lead to cognitive impairment through hyperphosphorylation of τ protein, overexpression of amyloid-β and/or activation of microglia. The aforementioned pathologies impair the insulin-signaling pathway primarily through serine phosphorylation of insulin receptor substances (IRS). This then leads to the inactivation of the phosphatidylinositol 3-kinase/Protein kinase B (PI3K/AKT) signaling cascade, which is responsible for maintaining brain homeostasis and normal cognitive functioning. PI3K/AKT is crucial in maintaining normal cognitive function through the inactivation of glycogen synthase kinase 3β (GSκ3β), which hyperphosphorylates τ protein and releases pro-inflammatory cytokines that are neurotoxic. Several biomarkers were also highlighted as potential biomarkers of GDM-related cognitive impairment such as AGEs, serine-phosphorylated IRS-1 and inflammatory markers such as tumor necrosis factor α (TNF-α), high-sensitivity C-reactive protein (hs-CRP), leptin, interleukin 1β (IL-1β), and IL-6. Although GDM is a transient disease, its complications may be long-term, and hence increased mechanistic knowledge of the molecular changes contributing to cognitive impairment may provide important clues for interventional strategies.
Collapse
|
16
|
Shi M, O'Keefe L, Simcocks AC, Su XQ, McAinch AJ. The effect of cyanidin-3-O-β-glucoside and peptides extracted from yoghurt on glucose uptake and gene expression in human primary skeletal muscle myotubes from obese and obese diabetic participants. J Funct Foods 2018. [DOI: 10.1016/j.jff.2018.10.012] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
|
17
|
Liu J, Xu C, Zhang S, Li H, Chen K, Huang P, Guo Z, Xu L. Microcystin-LR disrupts insulin signaling by hyperphosphorylating insulin receptor substrate 1 and glycogen synthase. ENVIRONMENTAL TOXICOLOGY 2018; 33:16-22. [PMID: 28984034 DOI: 10.1002/tox.22456] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/14/2017] [Revised: 07/21/2017] [Accepted: 07/27/2017] [Indexed: 06/07/2023]
Abstract
Microcystin-LR (MC-LR) is a cyanobacteria-derived heptapeptide that has been commonly characterized as a hepatotoxin. Although the liver is a primary organ in glucose homeostasis, the effect of MC-LR on glucose metabolism remains unclear. In this study, the human liver cell line HL7702 and ICR mice were exposed to various concentrations of MC-LR for 24 h, and the proteins involved in insulin signaling were investigated. The results showed that MC-LR treatment induced the hyperphosphorylation of insulin receptor substrate 1 (IRS1) at several serine sites, S307, S323, S636/639, and S1101 in HL7702 cells, and S302, S318, S632/635, and S1097 in mice livers. In addition, the activation of S6K1 was demonstrated to play an important role in MC-LR-induced IRS1 hyperphosphorylation at several serine sites. Decreased levels of total IRS1 were observed in the mice livers, but there was no significant change in HL7702 cells. MC-LR also induced glycogen synthase (GS) hyperphosphorylation at S641 (inactivating GS) both in vitro and in vivo, even glycogen synthase kinase 3, a well-known GS kinase, was inactivated after MC-LR treatment. Moreover, MC-LR could block insulin-induced GS activation. In addition, glucose transport in liver cells was not impacted by MC-LR either with or without insulin stimulation. Our study implies that MC-LR can interfere with the actions of IRS1 and GS in insulin signaling and may have a toxic effect on glucose metabolism in the liver.
Collapse
Affiliation(s)
- Jinghui Liu
- Department of Biochemistry, School of Medicine, Zhejiang University, Hangzhou, 310058, China
| | - Chun Xu
- Department of Endocrinology, General Hospital of the Chinese People's Armed Police Forces, Beijing, 100039, China
| | - Shaofeng Zhang
- Department of Endocrinology, General Hospital of the Chinese People's Armed Police Forces, Beijing, 100039, China
| | - Haoyan Li
- Department of Endocrinology, General Hospital of the Chinese People's Armed Police Forces, Beijing, 100039, China
| | - Kele Chen
- Department of Biochemistry, School of Medicine, Zhejiang University, Hangzhou, 310058, China
| | - Pu Huang
- Department of Biochemistry, School of Medicine, Zhejiang University, Hangzhou, 310058, China
| | - Zonglou Guo
- Department of Biosystem Engineering, College of Biosystem Engineering and Food Science, Zhejiang University, Hangzhou, 310058, China
| | - Lihong Xu
- Department of Biochemistry, School of Medicine, Zhejiang University, Hangzhou, 310058, China
| |
Collapse
|
18
|
Villalobo A, Ishida H, Vogel HJ, Berchtold MW. Calmodulin as a protein linker and a regulator of adaptor/scaffold proteins. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2017; 1865:507-521. [PMID: 29247668 DOI: 10.1016/j.bbamcr.2017.12.004] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Revised: 12/07/2017] [Accepted: 12/08/2017] [Indexed: 01/29/2023]
Abstract
Calmodulin (CaM) is a universal regulator for a huge number of proteins in all eukaryotic cells. Best known is its function as a calcium-dependent modulator of the activity of enzymes, such as protein kinases and phosphatases, as well as other signaling proteins including membrane receptors, channels and structural proteins. However, less well known is the fact that CaM can also function as a Ca2+-dependent adaptor protein, either by bridging between different domains of the same protein or by linking two identical or different target proteins together. These activities are possible due to the fact that CaM contains two independently-folded Ca2+ binding lobes that are able to interact differentially and to some degree separately with targets proteins. In addition, CaM can interact with and regulates several proteins that function exclusively as adaptors. This review provides an overview over our present knowledge concerning the structural and functional aspects of the role of CaM as an adaptor protein and as a regulator of known adaptor/scaffold proteins.
Collapse
Affiliation(s)
- Antonio Villalobo
- Department of Cancer Biology, Instituto de Investigaciones Biomédicas, Consejo Superior de Investigaciones Científicas and Universidad Autónoma de Madrid, Arturo Duperier 4, E-28029 Madrid, Spain.
| | - Hiroaki Ishida
- Department of Biological Sciences, University of Calgary, 2500 University Dr. N.W., Calgary, Alberta T2N 1N4, Canada
| | - Hans J Vogel
- Department of Biological Sciences, University of Calgary, 2500 University Dr. N.W., Calgary, Alberta T2N 1N4, Canada.
| | - Martin W Berchtold
- Department of Biology, University of Copenhagen, 13 Universitetsparken, DK-2100 Copenhagen Ø, Denmark.
| |
Collapse
|
19
|
Meng B, Li H, Sun X, Qu W, Yang B, Cheng F, Shi L, Yuan H. σ-1 receptor stimulation protects against pressure-induced damage through InsR-MAPK signaling in human trabecular meshwork cells. Mol Med Rep 2017; 16:617-624. [PMID: 28560459 PMCID: PMC5482118 DOI: 10.3892/mmr.2017.6647] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2016] [Accepted: 03/21/2017] [Indexed: 01/27/2023] Open
Abstract
The purpose of the present study was to investigate the protective effect of the σ-1 receptor (Sig-1R) agonist (+)‑pentazocin (PTZ) on pressure-induced apoptosis and death of human trabecular meshwork cells (hTMCs). The expression levels of Sig‑1R and insulin receptor (InsR) were examined in hTMCs. Cells were cultured under a pressure of 0, 20, 40, 60 and 80 mmHg for 48 h, and under 80 mmHg for 44 h, after which the cells were treated with (+)‑PTZ (20 µM), N-(2-(3,4-dichlorophenyl)ethyl)-N‑methyl-2‑(dimethylamino) ethylamine (BD‑1063; 20 µM) administered 30 min prior to (+)‑PTZ, or BD‑1063 (20 µM) and then exposed to 80 mmHg again until the 48 h time‑point. The changes of the cells were observed by optical and electron microscopy, the apoptosis and death of hTMCs were detected by ethidium bromide/acridine orange dual staining assay and the expression of Sig‑1R and InsR by reverse transcription‑quantitative polymerase chain reaction and western blot analysis. The phosphorylation of extracellular signal‑regulated kinase (ERK), an important downstream protein of the InsR‑mitogen‑activated protein kinases (MAPK) signaling pathway, was also detected by western blot analysis when (+)‑PTZ and BD‑1063 were added to the 80 mmHg‑treated cells. Sig‑1Rs and InsRs were expressed in hTMCs. The apoptosis and death of hTMCs increased from 40 mmHg with 50% cell death when the pressure was at 80 mmHg and the structure of the cells noticeably changed. The expression of Sig‑1R and InsR increased along with the elevation of pressure. (+)‑PTZ decreased the apoptosis and death of hTMCs and increased the expression of Sig‑1R and InsR, and the phosphorylation of ERK. Such effects were blocked by BD‑1063. The present study suggested that Sig‑1R agonist (+)‑PTZ can protect hTMCs from pressure‑induced apoptosis and death by activating InsR and the MAPK signal pathway.
Collapse
Affiliation(s)
- Bo Meng
- Department of Ophthalmology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150086, P.R. China
| | - Hongyi Li
- Department of Ophthalmology and Otorhinolaryngology, Hospital of Heilongjiang University, Harbin, Heilongjiang 150080, P.R. China
| | - Xian Sun
- Department of Oncology, The Third Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150081, P.R. China
| | - Wei Qu
- Department of Ophthalmology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150086, P.R. China
| | - Binbin Yang
- Department of Ophthalmology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150086, P.R. China
| | - Fang Cheng
- Department of Ophthalmology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150086, P.R. China
| | - Liping Shi
- Department of Ophthalmology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150086, P.R. China
| | - Huiping Yuan
- Department of Ophthalmology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150086, P.R. China
| |
Collapse
|
20
|
Aliper AM, Korzinkin MB, Kuzmina NB, Zenin AA, Venkova LS, Smirnov PY, Zhavoronkov AA, Buzdin AA, Borisov NM. Mathematical Justification of Expression-Based Pathway Activation Scoring (PAS). Methods Mol Biol 2017; 1613:31-51. [PMID: 28849557 DOI: 10.1007/978-1-4939-7027-8_3] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Although modeling of activation kinetics for various cell signaling pathways has reached a high grade of sophistication and thoroughness, most such kinetic models still remain of rather limited practical value for biomedicine. Nevertheless, recent advancements have been made in application of signaling pathway science for real needs of prescription of the most effective drugs for individual patients. The methods for such prescription evaluate the degree of pathological changes in the signaling machinery based on two types of data: first, on the results of high-throughput gene expression profiling, and second, on the molecular pathway graphs that reflect interactions between the pathway members. For example, our algorithm OncoFinder evaluates the activation of molecular pathways on the basis of gene/protein expression data in the objects of the interest.Yet, the question of assessment of the relative importance for each gene product in a molecular pathway remains unclear unless one call for the methods of parameter sensitivity /stiffness analysis in the interactomic kinetic models of signaling pathway activation in terms of total concentrations of each gene product.Here we show two principal points: 1. First, the importance coefficients for each gene in pathways that were obtained using the extremely time- and labor-consuming stiffness analysis of full-scaled kinetic models generally differ from much easier-to-calculate expression-based pathway activation score (PAS) not more than by 30%, so the concept of PAS is kinetically justified. 2. Second, the use of pathway-based approach instead of distinct gene analysis, due to the law of large numbers, allows restoring the correlation between the similar samples that were examined using different transcriptome investigation techniques.
Collapse
Affiliation(s)
- Alexander M Aliper
- Drug Research and Design Department, Pathway Pharmaceuticals, Wan Chai, Hong Kong, Hong Kong SAR
- Department of Personalized Medicine, First Oncology Research and Advisory Center, Moscow, Russia
- Laboratory of Bioinformatics, D. Rogachev Federal Research Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russia
| | - Michael B Korzinkin
- Department of Personalized Medicine, First Oncology Research and Advisory Center, Moscow, Russia
- Laboratory of Bioinformatics, D. Rogachev Federal Research Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russia
| | - Natalia B Kuzmina
- Laboratory of Systems Biology, A.I. Burnazyan Federal Medical Biophysical Center, Moscow, 123182, Russia
| | - Alexander A Zenin
- Laboratory of Systems Biology, A.I. Burnazyan Federal Medical Biophysical Center, Moscow, 123182, Russia
| | - Larisa S Venkova
- Department of Personalized Medicine, First Oncology Research and Advisory Center, Moscow, Russia
- Laboratory of Bioinformatics, D. Rogachev Federal Research Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russia
| | - Philip Yu Smirnov
- Laboratory of Systems Biology, A.I. Burnazyan Federal Medical Biophysical Center, Moscow, 123182, Russia
| | - Alex A Zhavoronkov
- Drug Research and Design Department, Pathway Pharmaceuticals, Wan Chai, Hong Kong, Hong Kong SAR
- Department of Personalized Medicine, First Oncology Research and Advisory Center, Moscow, Russia
- Laboratory of Bioinformatics, D. Rogachev Federal Research Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russia
| | - Anton A Buzdin
- Drug Research and Design Department, Pathway Pharmaceuticals, Wan Chai, Hong Kong, Hong Kong SAR
- Department of Personalized Medicine, First Oncology Research and Advisory Center, Moscow, Russia
- Laboratory of Bioinformatics, D. Rogachev Federal Research Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russia
- Group for Genomic Regulation of Cell Signaling Systems, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow, Russia
- National Research Centre "Kurchatov Institute", Centre for Convergence of Nano-, Bio-, Information and Cognitive Sciences and Technologies, Moscow, Russia
| | - Nikolay M Borisov
- Department of Personalized Medicine, First Oncology Research and Advisory Center, Moscow, Russia.
- Laboratory of Bioinformatics, D. Rogachev Federal Research Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russia.
- National Research Centre "Kurchatov Institute", Centre for Convergence of Nano-, Bio-, Information and Cognitive Sciences and Technologies, Moscow, Russia.
| |
Collapse
|
21
|
Ponce-Lopez T, Hong E, Abascal-Díaz M, Meneses A. Role of GSK3<i>β</i> and PP2A on Regulation of Tau Phosphorylation in Hippocampus and Memory Impairment in ICV-STZ Animal Model of Alzheimer’s Disease. ACTA ACUST UNITED AC 2017. [DOI: 10.4236/aad.2017.61002] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
22
|
Rajasekar N, Nath C, Hanif K, Shukla R. Intranasal Insulin Administration Ameliorates Streptozotocin (ICV)-Induced Insulin Receptor Dysfunction, Neuroinflammation, Amyloidogenesis, and Memory Impairment in Rats. Mol Neurobiol 2016; 54:6507-6522. [PMID: 27730514 DOI: 10.1007/s12035-016-0169-8] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Accepted: 09/27/2016] [Indexed: 12/14/2022]
Abstract
Alzheimer's disease (AD) is associated with reduced insulin level and impairment of insulin receptor (IR) signaling in the brain, which correlates to amyloid pathology, neuroinflammation, and synaptic neurotoxicity. Clinical studies show that intranasal insulin improves memory in AD patients without peripheral hypoglycemia. However, neuroprotective molecular mechanism of the beneficial effect of intranasal insulin in AD pathology is unexplored. Therefore, we investigated the role of intranasal insulin on intracerebroventricular (ICV) streptozotocin (STZ)-induced memory impairment in rats as evaluated in the Morris water maze test. STZ (ICV) treated rats had shown memory impairment along with a significant decrease in IR signaling molecules (IR, pIRS-1, pAkt, and pGSK-3α/β expression) and IDE expression in both hippocampus and cerebral cortex. Intranasal insulin delivery prevented these changes. Moreover, intranasal insulin was found to inhibit significantly glial cell activation (GFAP and Iba-1 expression), neuroinflammation (COX-2 expression, NFκB translocation, TNF-α, and IL-10 level) and amyloidogenic protein expression (BACE-1 and Aβ1-42 expression) in STZ (ICV)-injected rats. STZ (ICV)-induced caspase activation and postsynaptic neurotoxicity were also prevented by treatment with intranasal insulin. Our findings reveal that insulin has the neuroprotective effect and clearly signifies the potential use of intranasal insulin delivery for the treatment of AD. Graphical Abstract Neuroprotective effects of intranasal insulin administration on streptozotocin (ICV)-induced memory impairment in rats.
Collapse
Affiliation(s)
- N Rajasekar
- Divisions of Pharmacology and Toxicology, CSIR-Central Drug Research Institute, Lucknow, 226031, India
- Academy of Scientific and Innovative Research (AcSIR), Chennai, India
| | - Chandishwar Nath
- Divisions of Pharmacology and Toxicology, CSIR-Central Drug Research Institute, Lucknow, 226031, India
- Academy of Scientific and Innovative Research (AcSIR), Chennai, India
| | - Kashif Hanif
- Divisions of Pharmacology and Toxicology, CSIR-Central Drug Research Institute, Lucknow, 226031, India
- Academy of Scientific and Innovative Research (AcSIR), Chennai, India
| | - Rakesh Shukla
- Divisions of Pharmacology and Toxicology, CSIR-Central Drug Research Institute, Lucknow, 226031, India.
- Academy of Scientific and Innovative Research (AcSIR), Chennai, India.
| |
Collapse
|
23
|
Lipina C, Vaanholt LM, Davidova A, Mitchell SE, Storey-Gordon E, Hambly C, Irving AJ, Speakman JR, Hundal HS. CB1 receptor blockade counters age-induced insulin resistance and metabolic dysfunction. Aging Cell 2016; 15:325-35. [PMID: 26757949 PMCID: PMC4783351 DOI: 10.1111/acel.12438] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/24/2015] [Indexed: 12/11/2022] Open
Abstract
The endocannabinoid system can modulate energy homeostasis by regulating feeding behaviour as well as peripheral energy storage and utilization. Importantly, many of its metabolic actions are mediated through the cannabinoid type 1 receptor (CB1R), whose hyperactivation is associated with obesity and impaired metabolic function. Herein, we explored the effects of administering rimonabant, a selective CB1R inverse agonist, upon key metabolic parameters in young (4 month old) and aged (17 month old) adult male C57BL/6 mice. Daily treatment with rimonabant for 14 days transiently reduced food intake in young and aged mice; however, the anorectic response was more profound in aged animals, coinciding with a substantive loss in body fat mass. Notably, reduced insulin sensitivity in aged skeletal muscle and liver concurred with increased CB1R mRNA abundance. Strikingly, rimonabant was shown to improve glucose tolerance and enhance skeletal muscle and liver insulin sensitivity in aged, but not young, adult mice. Moreover, rimonabant‐mediated insulin sensitization in aged adipose tissue coincided with amelioration of low‐grade inflammation and repressed lipogenic gene expression. Collectively, our findings indicate a key role for CB1R in aging‐related insulin resistance and metabolic dysfunction and highlight CB1R blockade as a potential strategy for combating metabolic disorders associated with aging.
Collapse
Affiliation(s)
- Christopher Lipina
- Division of Cell Signalling and Immunology; Sir James Black Centre; School of Life Sciences; University of Dundee; Dundee UK
| | - Lobke M. Vaanholt
- Institute of Biological and Environmental Sciences; University of Aberdeen; Aberdeen UK
| | - Anastasija Davidova
- Division of Cell Signalling and Immunology; Sir James Black Centre; School of Life Sciences; University of Dundee; Dundee UK
| | - Sharon E. Mitchell
- Institute of Biological and Environmental Sciences; University of Aberdeen; Aberdeen UK
| | - Emma Storey-Gordon
- Division of Cell Signalling and Immunology; Sir James Black Centre; School of Life Sciences; University of Dundee; Dundee UK
| | - Catherine Hambly
- Institute of Biological and Environmental Sciences; University of Aberdeen; Aberdeen UK
| | - Andrew J. Irving
- Division of Neuroscience; Medical Research Institute; Ninewells Hospital; University of Dundee; Dundee UK
| | - John R. Speakman
- Institute of Biological and Environmental Sciences; University of Aberdeen; Aberdeen UK
- Institute of Genetics and Developmental Biology; Chinese Academy of Sciences, Chaoyang; Beijing China
| | - Harinder S. Hundal
- Division of Cell Signalling and Immunology; Sir James Black Centre; School of Life Sciences; University of Dundee; Dundee UK
| |
Collapse
|
24
|
Coles CA. Adipokines in Healthy Skeletal Muscle and Metabolic Disease. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 900:133-60. [DOI: 10.1007/978-3-319-27511-6_6] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
25
|
Zeng L, Kuti M, Mujtaba S, Zhou MM. Structural insights into FRS2α PTB domain recognition by neurotrophin receptor TrkB. Proteins 2015; 82:1534-41. [PMID: 24470253 DOI: 10.1002/prot.24523] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2013] [Revised: 12/27/2013] [Accepted: 01/16/2014] [Indexed: 11/07/2022]
Abstract
The fibroblast growth factor receptor (FGFR) substrate 2 (FRS2) family proteins function as scaffolding adapters for receptor tyrosine kinases (RTKs). The FRS2α proteins interact with RTKs through the phosphotyrosine-binding (PTB) domain and transfer signals from the activated receptors to downstream effector proteins. Here, we report the nuclear magnetic resonance structure of the FRS2α PTB domain bound to phosphorylated TrkB. The structure reveals that the FRS2α-PTB domain is comprised of two distinct but adjacent pockets for its mutually exclusive interaction with either nonphosphorylated juxtamembrane region of the FGFR, or tyrosine phosphorylated peptides TrkA and TrkB. The new structural insights suggest rational design of selective small molecules through targeting of the two conjunct pockets in the FRS2α PTB domain.
Collapse
Affiliation(s)
- Lei Zeng
- Department of Structural and Chemical Biology, Icahn School of Medicine at Mount Sinai, New York, New York, 10029
| | | | | | | |
Collapse
|
26
|
Alpha-synuclein overexpression negatively regulates insulin receptor substrate 1 by activating mTORC1/S6K1 signaling. Int J Biochem Cell Biol 2015; 64:25-33. [PMID: 25813876 DOI: 10.1016/j.biocel.2015.03.006] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2014] [Revised: 02/26/2015] [Accepted: 03/09/2015] [Indexed: 12/15/2022]
Abstract
Alpha-synuclein (α-Syn) is a major component of Lewy bodies, a pathological feature of Parkinson's and other neurodegenerative diseases collectively known as synucleinopathies. Among the possible mechanisms of α-Syn-mediated neurotoxicity is interference with cytoprotective pathways such as insulin signaling. Insulin receptor substrate (IRS)-1 is a docking protein linking IRs to downstream signaling pathways such as phosphatidylinositol 3-kinase/Akt and mammalian target of rapamycin (mTOR)/ribosomal protein S6 kinase (S6K)1; the latter exerts negative feedback control on insulin signaling, which is impaired in Alzheimer's disease. Our previous study found that α-Syn overexpression can inhibit protein phosphatase (PP)2A activity, which is involved in the protective mechanism of insulin signaling. In this study, we found an increase in IRS-1 phosphorylation at Ser636 and decrease in tyrosine phosphorylation, which accelerated IRS-1 turnover and reduced insulin-Akt signaling in α-Syn-overexpressing SK-N-SH cells and transgenic mice. The mTOR complex (C)1/S6K1 blocker rapamycin inhibited the phosphorylation of IRS-1 at Ser636 in cells overexpressing α-Syn, suggesting that mTORC1/S6K1 activation by α-Syn causes feedback inhibition of insulin signaling via suppression of IRS-1 function. α-Syn overexpression also inhibited PP2A activity, while the PP2A agonist C2 ceramide suppressed both S6K1 activation and IRS-1 Ser636 phosphorylation upon α-Syn overexpression. Thus, α-Syn overexpression negatively regulated IRS-1 via mTORC1/S6K1 signaling while activation of PP2A reverses this process. These results provide evidence for a link between α-Syn and IRS-1 that may represent a novel mechanism for α-Syn-associated pathogenesis.
Collapse
|
27
|
Gibbs M. Reflections on glycogen and β-amyloid: why does glycogenolytic β2-adrenoceptor stimulation not rescue memory after β-amyloid? Metab Brain Dis 2015; 30:345-52. [PMID: 24810634 DOI: 10.1007/s11011-014-9563-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2014] [Accepted: 04/30/2014] [Indexed: 01/23/2023]
Abstract
Normally noradrenaline release ~30 min after training in the day-old chick is essential for memory consolidation by simultaneously increasing both glycogenolysis, by its stimulation of β2-adrenergic (AR) receptors, and glycogen synthesis, by its stimulation of α2-AR receptors in astrocytes. At the same time noradrenaline stimulation of β3-AR receptors increases glucose uptake solely in astrocytes. Intracerebral injection of small oligomeric β-amyloid protein (Aβ1-42) (Aβ) 45 min before one-trial bead discrimination learning in day-old chicks abolishes consolidation of memory 30 min post-learning. The ensuing memory loss can be rescued by injection of selective β3- and β(2-AR agonists (CL316243 and zinterol), which also have the ability to consolidate weakly-reinforced learning into long-term memory. However, although CL316243 rescues Aβ-induced memory loss over a similar time period to when it consolidates weak learning (up to 25 min post training), zinterol is effective over a more limited time period and unexpectedly it does not rescue at the time it promotes glycogenolysis. Injection of Aβ into the hippocampus and the locus coeruleus (LoC) also produces similar memory deficits and injection of both AR agonists into a cortical area can rescue memory from LoC Aβ. We have previously shown that β3-AR stimulation increases astrocytic glucose uptake and have suggested there may be sensitization or upregulation of the receptor. Since β2-AR stimulation does not rescue memory at the time it promotes glycogenolysis, but the receptor does not appear to be impaired, it is suggested that Aβ may be causing an impairment in the synthesis of readily available glycogen.
Collapse
Affiliation(s)
- Marie Gibbs
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Melbourne, 3052, VIC, Australia,
| |
Collapse
|
28
|
Stuart CA, Howell MEA, Cartwright BM, McCurry MP, Lee ML, Ramsey MW, Stone MH. Insulin resistance and muscle insulin receptor substrate-1 serine hyperphosphorylation. Physiol Rep 2014; 2:2/12/e12236. [PMID: 25472611 PMCID: PMC4332214 DOI: 10.14814/phy2.12236] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Insulin resistance in metabolic syndrome subjects is profound in spite of muscle insulin receptor and insulin-responsive glucose transporter (GLUT4) expression being nearly normal. Insulin receptor tyrosine kinase phosphorylation of insulin receptor substrate-1 (IRS-1) at Tyr896 is a necessary step in insulin stimulation of translocation of GLUT4 to the cell surface. Serine phosphorylation of IRS-1 by some kinases diminishes insulin action in mice. We evaluated the phosphorylation status of muscle IRS-1 in 33 subjects with the metabolic syndrome and seventeen lean controls. Each underwent euglycemic insulin clamps and a thigh muscle biopsy before and after 8 weeks of either strength or endurance training. Muscle IRS-1 phosphorylation at six sites was quantified by immunoblots. Metabolic syndrome muscle IRS-1 had excess phosphorylation at Ser337 and Ser636 but not at Ser307, Ser789, or Ser1101. Ser337 is a target for phosphorylation by glycogen synthase kinase 3 (GSK3) and Ser636 is phosphorylated by c-Jun N-terminal kinase 1 (JNK1). Exercise training without weight loss did not change the IRS-1 serine phosphorylation. These data suggest that baseline hyperphosphorylation of at least two key serines within muscle IRS-1 diminishes the transmission of the insulin signal and thereby decreases the insulin-stimulated translocation of GLUT4. Excess fasting phosphorylation of muscle IRS-1 at Ser636 may be a major cause of the insulin resistance seen in obesity and might prevent improvement in insulin responsiveness when exercise training is not accompanied by weight loss.
Collapse
Affiliation(s)
- Charles A Stuart
- Department of Internal Medicine, Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee
| | - Mary E A Howell
- Department of Internal Medicine, Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee
| | - Brian M Cartwright
- Department of Internal Medicine, Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee
| | - Melanie P McCurry
- Department of Internal Medicine, Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee
| | - Michelle L Lee
- Department of Allied Health, College of Clinical and Rehabilitative Health, East Tennessee State University, Johnson City, Tennessee
| | - Michael W Ramsey
- Department of Exercise and Sports Science, Clemmer College of Education, East Tennessee State University, Johnson City, Tennessee
| | - Michael H Stone
- Department of Exercise and Sports Science, Clemmer College of Education, East Tennessee State University, Johnson City, Tennessee
| |
Collapse
|
29
|
Rajasekar N, Dwivedi S, Nath C, Hanif K, Shukla R. Protection of streptozotocin induced insulin receptor dysfunction, neuroinflammation and amyloidogenesis in astrocytes by insulin. Neuropharmacology 2014; 86:337-52. [PMID: 25158313 DOI: 10.1016/j.neuropharm.2014.08.013] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2014] [Revised: 08/12/2014] [Accepted: 08/14/2014] [Indexed: 01/04/2023]
Abstract
Impaired insulin signaling, amyloid pathology and neuroinflammation are closely associated with neurodegenerative disorder like Alzheimer's disease (AD). Our earlier studies showed that intracerebroventricular streptozotocin (STZ) induces insulin receptor (IR) signaling defect in the hippocampus, which is associated with memory impairment in rats. Astrocytes are the most abundant cells in the brain and play a major role in neuroinflammation. However, involvement of astrocytes in STZ induced IR dysfunction has not received much attention. Therefore, the present study was planned to explore the effect of STZ on IR signaling, proinflammatory markers and amyloidogenesis in rat astrocytoma cell line, (C6). STZ (100 μM) treatment in astrocytes (n = 3) for 24 h, resulted significant decrease in IR mRNA and protein expression, phosphorylation of IRS-1, Akt, GSK-3α and GSK-3β (p < 0.01). Further STZ induced amyloidogenic protein expression as evidenced by the increase in APP, BACE-1 and Aβ1-42 expression (p < 0.05) in astrocytes. STZ also significantly induced astrocytes activation as evidenced by increased expression of GFAP and p-P38 MAPK (p < 0.05). STZ treatment caused enhanced translocation of p65 NF-kB, triggered over expression of TNF-α, IL-1β, COX-2, oxidative/nitrosative stress and caspase activation (p < 0.05) in astrocytes. Insulin (25-100 nM) pretreatment (n = 3) significantly prevented changes in IR signaling, amyloidogenic protein expression and levels of proinflammatory markers (p < 0.05) in STZ treated astroglial cells. In the present study, the protective effect of insulin suggests that, IR dysfunction along with amyloidogenesis and neuroinflammation may have played a major role in STZ induced toxicity in astrocytes which are relevant to AD pathology.
Collapse
Affiliation(s)
- N Rajasekar
- Divisions of Pharmacology and Toxicology, CSIR-Central Drug Research Institute, Lucknow, 226031, India; Academy of Scientific and Innovative Research (AcSIR), India
| | - Subhash Dwivedi
- Divisions of Pharmacology and Toxicology, CSIR-Central Drug Research Institute, Lucknow, 226031, India
| | - Chandishwar Nath
- Divisions of Pharmacology and Toxicology, CSIR-Central Drug Research Institute, Lucknow, 226031, India; Academy of Scientific and Innovative Research (AcSIR), India
| | - Kashif Hanif
- Divisions of Pharmacology and Toxicology, CSIR-Central Drug Research Institute, Lucknow, 226031, India; Academy of Scientific and Innovative Research (AcSIR), India
| | - Rakesh Shukla
- Divisions of Pharmacology and Toxicology, CSIR-Central Drug Research Institute, Lucknow, 226031, India; Academy of Scientific and Innovative Research (AcSIR), India.
| |
Collapse
|
30
|
Morán J, Garrido P, Cabello E, Alonso A, González C. Effects of estradiol and genistein on the insulin signaling pathway in the cerebral cortex of aged female rats. Exp Gerontol 2014; 58:104-12. [PMID: 25086228 DOI: 10.1016/j.exger.2014.07.018] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2014] [Revised: 07/04/2014] [Accepted: 07/30/2014] [Indexed: 01/29/2023]
Abstract
Menopause leads to a decrease in estrogen production that increases central insulin resistance, contributing to the development of neurodegenerative diseases. We have evaluated the influence of aging and estradiol or genistein treatments on some key stages of the insulin signaling pathway in the cerebral cortex. Young and aged female Wistar rats were ovariectomized and treated acutely with 17β-estradiol (1.4μg/kg body weight), two doses of genistein (10 or 40mg/kg body weight), or vehicle. The cortical expression of several key insulin signaling pathway components was analyzed by western blotting. Our results showed an age-related deterioration in the interactions between the regulatory subunit of phosphatidylinositol 3-kinase (p85α) and the activated form of insulin receptor substrate 1 (p-IRS1tyr612), as well as between p85α and the 46kDa isoform of the estrogen receptor α (ERα46). Moreover, aging also decreased the translocation of glucose transporter-4 (GLUT4) to the plasma membrane. 17β-Estradiol but not genistein reduced the negative impact of aging on central insulin sensitivity by favoring this GLUT4 translocation, and therefore could be neuroprotective against the associated neurodegenerative diseases. However, protein kinase B (Akt) activation by genistein suggests that other possible mechanisms are involved in the neuroprotective effects of this phytoestrogen during the aging process.
Collapse
Affiliation(s)
- Javier Morán
- Department of Functional Biology, Physiology Area, University of Oviedo, Av. Julián Clavería, No. 6, 33006 Oviedo, Spain.
| | - Pablo Garrido
- Department of Functional Biology, Physiology Area, University of Oviedo, Av. Julián Clavería, No. 6, 33006 Oviedo, Spain.
| | - Estefanía Cabello
- Department of Functional Biology, Physiology Area, University of Oviedo, Av. Julián Clavería, No. 6, 33006 Oviedo, Spain.
| | - Ana Alonso
- Department of Functional Biology, Physiology Area, University of Oviedo, Av. Julián Clavería, No. 6, 33006 Oviedo, Spain.
| | - Celestino González
- Department of Functional Biology, Physiology Area, University of Oviedo, Av. Julián Clavería, No. 6, 33006 Oviedo, Spain.
| |
Collapse
|
31
|
Blake R, Trounce IA. Mitochondrial dysfunction and complications associated with diabetes. Biochim Biophys Acta Gen Subj 2014; 1840:1404-12. [DOI: 10.1016/j.bbagen.2013.11.007] [Citation(s) in RCA: 105] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2013] [Revised: 10/18/2013] [Accepted: 11/06/2013] [Indexed: 02/06/2023]
|
32
|
Wakayama S, Haque A, Koide N, Kato Y, Odkhuu E, Bilegtsaikhan T, Naiki Y, Komatsu T, Yoshida T, Yokochi T. Lipopolysaccharide impairs insulin sensitivity via activation of phosphoinositide 3-kinase in adipocytes. Immunopharmacol Immunotoxicol 2014; 36:145-9. [PMID: 24506665 DOI: 10.3109/08923973.2014.887096] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
The effect of lipopolysaccharide (LPS) on insulin sensitivity in adipocytes were examined by using differentiated 3T3-L1 adipocytes. Insulin-mediated activation of insulin receptor substrate (IRS) 1/2 was inhibited in LPS-pretreated adipocytes and IRS1/2-mediated Akt activation was also attenuated in those cells. LPS inhibited activation of glycogen synthase kinase 3 as a negative regulator of glycogenesis and impaired the glycogen synthesis in response to insulin. LPS-induced activation of phosphoinositide 3-kinase (PI3K) in adipocytes. Involvement of suppressor of cytokine signaling 3 (SOCS3) in LPS-induced IRS1/2 inhibition was excluded. Considering that both insulin and LPS were able to activate the PI3K/Akt signaling pathway, LPS was suggested to impair insulin sensitivity of adipocytes through down-regulating insulin-mediated PI3K/Akt activation.
Collapse
|
33
|
Yogalakshmi B, Bhuvaneswari S, Sreeja S, Anuradha CV. Grape seed proanthocyanidins and metformin act by different mechanisms to promote insulin signaling in rats fed high calorie diet. J Cell Commun Signal 2013; 8:13-22. [PMID: 24026800 DOI: 10.1007/s12079-013-0210-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2013] [Accepted: 08/23/2013] [Indexed: 10/26/2022] Open
Abstract
Key pathways like insulin signaling, AMP activated kinase (AMPK) activation and inflammatory signaling are involved in the complex pathological network of hepatic insulin resistance. Our aim is to investigate whether grape seed proanthocyanidins (GSP) and metformin (MET) target any of these pathways in insulin resistant rat liver. Albino Wistar rats were rendered insulin resistant by feeding a high fat-fructose diet (HFFD). Either GSP (100 mg/kg b.w), MET(50 mg/kg b.w) or both were administered to insulin resistant rats as therapeutic options. HFFD-feeding caused hyperglycemia, hyperinsulinemia, increased gluconeogenesis, decreased tyrosine phosphorylation of insulin receptor-β(IR-β) and insulin receptor substrate-1 (IRS-1) and increased serine phosphorylation of IRS-1. The association of p85α subunit of phosphotidyl inositol 3 kinase(PI3K) with IRS-1 and subsequent Akt phosphorylation were reduced while the expression of mitogen activated protein kinases (MAPK) were increased in HFFD rats. Both MET and GSP reduced hyperglycemia and hyperinsulinemia and improved glycolysis, tyrosine phosphorylation of IR-β and IRS-1, IRS-1-PI3K association and Akt activation. However, activation of tumor necrosis factor-α, interleukin-6, leptin and suppressor of cytokine signaling-3 and reduction in adiponectin caused by chronic HFFD feeding were reversed by GSP better than by MET. Activation of AMPK by GSP was much less compared to that by MET. These findings suggest that GSP might activate PI3K pathway and promote insulin action by reducing serine kinase activation and cytokine signaling and MET by targeting AMPK. The beneficial effects were enhanced during combination therapy. Thus, combination therapy with MET and GSP may be considered for the management of metabolic syndrome.
Collapse
Affiliation(s)
- Baskaran Yogalakshmi
- Department of Biochemistry and Biotechnology, Annamalai University, Annamalai Nagar, 608 002, Tamil Nadu, India
| | | | | | | |
Collapse
|
34
|
Medhi B, Chakrabarty M. Insulin resistance: an emerging link in Alzheimer's disease. Neurol Sci 2013; 34:1719-25. [PMID: 23670236 DOI: 10.1007/s10072-013-1454-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2013] [Accepted: 05/06/2013] [Indexed: 12/18/2022]
Abstract
Relentless progression of Alzheimer's disease (AD) poses a grave situation for the biomedical community to tackle. Agents starting as hot favorites in clinical trials have failed in later stages and it is time we reconsidered our approaches to intervene the disease. Quite some interesting work in the last decade has introduced a new school of thought which factors in neuronal glycemic imbalance as a major component for the development of AD. Insulin resistance in the brain has brought forward subsequent sequelae which might work towards amyloid accretion and/or tau hyperphosphorylation. It is also pointed out that insulin works by distributing iron to neuronal tissue and an insulin resistant state throws it off gear leading to iron overloading of neurons which is ultimately detrimental. A relatively recent investigation finds the role of c-Jun-N-terminal kinase (JNK3) in AD which also seems to bear a link with insulin resistance.
Collapse
Affiliation(s)
- Bikash Medhi
- Department of Pharmacology, Postgraduate Institute of Medical Education and Research, Research Block B, 4th Floor, Room No. 4043, Chandigarh, 160012, India,
| | | |
Collapse
|
35
|
Yamamoto N, Matsubara T, Sobue K, Tanida M, Kasahara R, Naruse K, Taniura H, Sato T, Suzuki K. Brain insulin resistance accelerates Aβ fibrillogenesis by inducing GM1 ganglioside clustering in the presynaptic membranes. J Neurochem 2012; 121:619-28. [PMID: 22260232 DOI: 10.1111/j.1471-4159.2012.07668.x] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Type 2 diabetes mellitus is thought to be a significant risk factor for Alzheimer's disease. Insulin resistance also affects the central nervous system by regulating key processes, such as neuronal survival and longevity, learning and memory. However, the mechanisms underlying these effects remain uncertain. To investigate whether insulin resistance is associated with the assembly of amyloid β-protein (Aβ) at the cell surface of neurons, we inhibited insulin-signalling pathways of primary neurons. The treatments of insulin receptor (IR)-knockdown and a phosphatidylinositol 3-kinase inhibitor (LY294002), but not an extracellular signal-regulated kinase inhibitor, induced an increase in GM1 ganglioside (GM1) levels in detergent-resistant membrane microdomains of the neurons. The aged db/db mouse brain exhibited reduction in IR expression and phosphorylation of Akt, which later induced an increase in the high-density GM1-clusters on synaptosomes. Neurons treated with IR knockdown or LY294002, and synaptosomes of the aged db/db mouse brains markedly accelerated an assembly of Aβs. These results suggest that ageing and peripheral insulin resistance induce brain insulin resistance, which accelerates the assembly of Aβs by increasing and clustering of GM1 in detergent-resistant membrane microdomains of neuronal membranes.
Collapse
Affiliation(s)
- Naoki Yamamoto
- Laboratory of Neurochemistry, Department of Pharmacy, College of Pharmaceutical Sciences, Ritsumeikan University, Kusatsu, Shiga, Japan.
| | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Nyman E, Fagerholm S, Jullesson D, Strålfors P, Cedersund G. Mechanistic explanations for counter-intuitive phosphorylation dynamics of the insulin receptor and insulin receptor substrate-1 in response to insulin in murine adipocytes. FEBS J 2012; 279:987-99. [PMID: 22248283 DOI: 10.1111/j.1742-4658.2012.08488.x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Insulin signaling through insulin receptor (IR) and insulin receptor substrate-1 (IRS1) is important for insulin control of target cells. We have previously demonstrated a rapid and simultaneous overshoot behavior in the phosphorylation dynamics of IR and IRS1 in human adipocytes. Herein, we demonstrate that in murine adipocytes a similar overshoot behavior is not simultaneous for IR and IRS1. The peak of IRS1 phosphorylation, which is a direct consequence of the phosphorylation and the activation of IR, occurs earlier than the peak of IR phosphorylation. We used a conclusive modeling framework to unravel the mechanisms behind this counter-intuitive order of phosphorylation. Through a number of rejections, we demonstrate that two fundamentally different mechanisms may create the reversed order of peaks: (i) two pools of phosphorylated IR, where a large pool of internalized IR peaks late, but phosphorylation of IRS1 is governed by a small plasma membrane-localized pool of IR with an early peak, or (ii) inhibition of the IR-catalyzed phosphorylation of IRS1 by negative feedback. Although (i) may explain the reversed order, this two-pool hypothesis alone requires extensive internalization of IR, which is not supported by experimental data. However, with the additional assumption of limiting concentrations of IRS1, (i) can explain all data. Also, (ii) can explain all available data. Our findings illustrate how modeling can potentiate reasoning, to help draw nontrivial conclusions regarding competing mechanisms in signaling networks. Our work also reveals new differences between human and murine insulin signaling.
Collapse
Affiliation(s)
- Elin Nyman
- Department of Clinical and Experimental Medicine, Diabetes and Integrative Systems Biology, Linköping University, Sweden
| | | | | | | | | |
Collapse
|
37
|
Jablonka S, Holtmann B, Sendtner M, Metzger F. Therapeutic effects of PEGylated insulin-like growth factor I in the pmn mouse model of motoneuron disease. Exp Neurol 2011; 232:261-9. [DOI: 10.1016/j.expneurol.2011.09.015] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2011] [Revised: 08/31/2011] [Accepted: 09/09/2011] [Indexed: 02/08/2023]
|
38
|
A hepatocyte growth factor receptor (Met)-insulin receptor hybrid governs hepatic glucose metabolism. Nat Med 2011; 17:1577-84. [PMID: 22081023 PMCID: PMC3233634 DOI: 10.1038/nm.2531] [Citation(s) in RCA: 84] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2011] [Accepted: 09/20/2011] [Indexed: 01/01/2023]
Abstract
Met is the transmembrane tyrosine kinase cell surface receptor for hepatocyte growth factor (HGF) and is structurally related to the insulin receptor (INSR) tyrosine kinase. Here we report that the HGF-Met axis regulates metabolism by stimulating hepatic glucose uptake and suppressing hepatic glucose output. We show that Met is essential for an optimal hepatic insulin response by directly engaging INSR to form a Met-INSR hybrid complex, which culminates in a robust signal output. We also found that the HGF-Met system restores insulin responsiveness in a mouse model of insulin refractoriness. These results provide new insights into the molecular basis of hepatic insulin resistance and suggest that HGF may have therapeutic potential for type 2 diabetes in the clinical setting.
Collapse
|
39
|
Bosco D, Fava A, Plastino M, Montalcini T, Pujia A. Possible implications of insulin resistance and glucose metabolism in Alzheimer's disease pathogenesis. J Cell Mol Med 2011; 15:1807-21. [PMID: 21435176 PMCID: PMC3918038 DOI: 10.1111/j.1582-4934.2011.01318.x] [Citation(s) in RCA: 211] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2010] [Accepted: 03/17/2011] [Indexed: 01/21/2023] Open
Abstract
Type 2 diabetes mellitus (DM) appears to be a significant risk factor for Alzheimer disease (AD). Insulin and insulin-like growth factor-1 (IGF-1) also have intense effects in the central nervous system (CNS), regulating key processes such as neuronal survival and longevity, as well as learning and memory. Hyperglycaemia induces increased peripheral utilization of insulin, resulting in reduced insulin transport into the brain. Whereas the density of brain insulin receptor decreases during age, IGF-1 receptor increases, suggesting that specific insulin-mediated signals is involved in aging and possibly in cognitive decline. Molecular mechanisms that protect CNS neurons against β-amyloid-derived-diffusible ligands (ADDL), responsible for synaptic deterioration underlying AD memory failure, have been identified. The protection mechanism does not involve simple competition between ADDLs and insulin, but rather it is signalling dependent down-regulation of ADDL-binding sites. Defective insulin signalling make neurons energy deficient and vulnerable to oxidizing or other metabolic insults and impairs synaptic plasticity. In fact, destruction of mitochondria, by oxidation of a dynamic-like transporter protein, may cause synapse loss in AD. Moreover, interaction between Aβ and τ proteins could be cause of neuronal loss. Hyperinsulinaemia as well as complete lack of insulin result in increased τ phosphorylation, leading to an imbalance of insulin-regulated τ kinases and phosphatates. However, amyloid peptides accumulation is currently seen as a key step in the pathogenesis of AD. Inflammation interacts with processing and deposit of β-amyloid. Chronic hyperinsulinemia may exacerbate inflammatory responses and increase markers of oxidative stress. In addition, insulin appears to act as 'neuromodulator', influencing release and reuptake of neurotransmitters, and improving learning and memory. Thus, experimental and clinical evidence show that insulin action influences cerebral functions. In this paper, we reviewed several mechanisms by which insulin may affect pathophysiology in AD.
Collapse
Affiliation(s)
- Domenico Bosco
- Department of Neuroscience, 'S. Giovanni di Dio' Hospital, Via Largo Bologna, Crotone, Italy.
| | | | | | | | | |
Collapse
|
40
|
Nyman E, Brännmark C, Palmér R, Brugård J, Nyström FH, Strålfors P, Cedersund G. A hierarchical whole-body modeling approach elucidates the link between in Vitro insulin signaling and in Vivo glucose homeostasis. J Biol Chem 2011; 286:26028-41. [PMID: 21572040 PMCID: PMC3138269 DOI: 10.1074/jbc.m110.188987] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2010] [Revised: 03/08/2011] [Indexed: 01/15/2023] Open
Abstract
Type 2 diabetes is a metabolic disease that profoundly affects energy homeostasis. The disease involves failure at several levels and subsystems and is characterized by insulin resistance in target cells and tissues (i.e. by impaired intracellular insulin signaling). We have previously used an iterative experimental-theoretical approach to unravel the early insulin signaling events in primary human adipocytes. That study, like most insulin signaling studies, is based on in vitro experimental examination of cells, and the in vivo relevance of such studies for human beings has not been systematically examined. Herein, we develop a hierarchical model of the adipose tissue, which links intracellular insulin control of glucose transport in human primary adipocytes with whole-body glucose homeostasis. An iterative approach between experiments and minimal modeling allowed us to conclude that it is not possible to scale up the experimentally determined glucose uptake by the isolated adipocytes to match the glucose uptake profile of the adipose tissue in vivo. However, a model that additionally includes insulin effects on blood flow in the adipose tissue and GLUT4 translocation due to cell handling can explain all data, but neither of these additions is sufficient independently. We also extend the minimal model to include hierarchical dynamic links to more detailed models (both to our own models and to those by others), which act as submodules that can be turned on or off. The resulting multilevel hierarchical model can merge detailed results on different subsystems into a coherent understanding of whole-body glucose homeostasis. This hierarchical modeling can potentially create bridges between other experimental model systems and the in vivo human situation and offers a framework for systematic evaluation of the physiological relevance of in vitro obtained molecular/cellular experimental data.
Collapse
Affiliation(s)
- Elin Nyman
- From the Department of Clinical and Experimental Medicine, Diabetes and Integrative Systems Biology and
| | - Cecilia Brännmark
- From the Department of Clinical and Experimental Medicine, Diabetes and Integrative Systems Biology and
| | - Robert Palmér
- From the Department of Clinical and Experimental Medicine, Diabetes and Integrative Systems Biology and
| | - Jan Brugård
- MathCore Engineering AB, SE58330 Linköping, Sweden, and
| | - Fredrik H. Nyström
- the Department of Medical and Health Sciences, Linköping University, SE58185 Linköping, Sweden
| | - Peter Strålfors
- From the Department of Clinical and Experimental Medicine, Diabetes and Integrative Systems Biology and
| | - Gunnar Cedersund
- From the Department of Clinical and Experimental Medicine, Diabetes and Integrative Systems Biology and
- the School of Life Sciences, Freiburg Institute of Advanced Sciences, 79104 Freiburg, Germany
| |
Collapse
|
41
|
Correia SC, Santos RX, Perry G, Zhu X, Moreira PI, Smith MA. Insulin-resistant brain state: the culprit in sporadic Alzheimer's disease? Ageing Res Rev 2011; 10:264-73. [PMID: 21262392 PMCID: PMC3056939 DOI: 10.1016/j.arr.2011.01.001] [Citation(s) in RCA: 159] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2010] [Revised: 01/11/2011] [Accepted: 01/14/2011] [Indexed: 01/03/2023]
Abstract
Severe abnormalities in brain glucose/energy metabolism and insulin signaling have been documented to take a pivotal role in early sporadic Alzheimer's disease (sAD) pathology. Indeed, the "insulin-resistant brain state" has been hypothesized to form the core of the neurodegenerative events that occur in sAD. In this vein, intracerebroventricular administration of subdiabetogenic doses of streptozotocin (STZ) in rats can induce an insulin-resistant brain state, which is proposed as a suitable experimental model of sAD. This review highlights the involvement of disturbed brain insulin metabolism in sAD etiopathogenesis. Furthermore, current knowledge demonstrates that central STZ administration produces brain pathology and behavioral changes that resemble changes found in sAD patients. The STZ-intracerebroventricularly treated rat represents a promising experimental tool in this field by providing new insights concerning early brain alterations in sAD, which can be translated in novel etiopathogenic and therapeutic approaches in this disease.
Collapse
Affiliation(s)
- Sónia C. Correia
- Center for Neuroscience and Cell Biology of Coimbra, University of Coimbra, Coimbra, Portugal
- Faculty of Sciences and Technology, Department of Life Sciences, University of Coimbra, Coimbra, Portugal
| | - Renato X. Santos
- Center for Neuroscience and Cell Biology of Coimbra, University of Coimbra, Coimbra, Portugal
- Faculty of Sciences and Technology, Department of Life Sciences, University of Coimbra, Coimbra, Portugal
| | - George Perry
- Department of Pathology, Case Western Reserve University, Cleveland, Ohio, USA
- UTSA Neurosciences Institute and Department of Biology, University of Texas at San Antonio, San Antonio, Texas, USA
| | - Xiongwei Zhu
- Department of Pathology, Case Western Reserve University, Cleveland, Ohio, USA
| | - Paula I. Moreira
- Center for Neuroscience and Cell Biology of Coimbra, University of Coimbra, Coimbra, Portugal
- Faculty of Medicine, Institute of Physiology, University of Coimbra, Coimbra, Portugal
| | - Mark A. Smith
- Department of Pathology, Case Western Reserve University, Cleveland, Ohio, USA
| |
Collapse
|
42
|
Central insulin resistance and synaptic dysfunction in intracerebroventricular-streptozotocin injected rodents. Neurobiol Aging 2011; 33:430.e5-18. [PMID: 21256630 DOI: 10.1016/j.neurobiolaging.2010.12.002] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2010] [Revised: 11/22/2010] [Accepted: 12/07/2010] [Indexed: 11/24/2022]
Abstract
To better understand the role of insulin signaling in the development of Alzheimer's disease (AD), we utilized an animal model (intracerebroventricular injection of streptozotocin-ic-streptozotocin (STZ)) that displays insulin resistance only in the brain and exhibits AD pathology. In this model, deficits in hippocampal synaptic transmission and long-term potentiation (LTP) were observed. The decline in LTP correlated with decreased expression of NMDAR subunits NR2A and NR2B. The deficits in LTP were accompanied by changes in the expression and function of synaptic AMPARs. In ic-STZ animals, an alteration in integrin-linked kinase (ILK)-glycogen synthase kinase 3 beta (GSK-3-β) signaling was identified (p < 0.05). Similarly, there was decreased expression (p < 0.05) of brain derived neurotropic factor (BDNF) and stargazin, an AMPAR auxiliary subunit; both are required for driving AMPA receptors to the surface of the postsynaptic membrane. Our data illustrate that altered ILK-GSK-3β signaling due to impaired insulin signaling may decrease the trafficking and function of postsynaptic glutamate receptors; thereby, leading to synaptic deficits contributing to memory loss.
Collapse
|
43
|
Abstract
Ever since the discovery of insulin and its role in the regulation of glucose uptake and utilization, there has been great interest in insulin, its structure and the way in which it interacts with its receptor and effects signal transduction. As the 90th anniversary of the discovery of insulin approaches, it is timely to provide an overview of the landmark discoveries relating to the structure and function of this remarkable molecule and its receptor.
Collapse
Affiliation(s)
- Colin W. Ward
- Walter and Eliza Hall Institute of Medical ResearchParkville, VIC, Australia
| | - Michael C. Lawrence
- Walter and Eliza Hall Institute of Medical ResearchParkville, VIC, Australia
- Department of Medical Biology, University of MelbourneParkville, VIC, Australia
- *Correspondence: Michael C. Lawrence, Walter and Eliza Hall Institute, 1G Royal Parade, Parkville, VIC 3052, Australia. e-mail:
| |
Collapse
|
44
|
Boura-Halfon S, Shuster-Meiseles T, Beck A, Petrovich K, Gurevitch D, Ronen D, Zick Y. A novel domain mediates insulin-induced proteasomal degradation of insulin receptor substrate 1 (IRS-1). Mol Endocrinol 2010; 24:2179-92. [PMID: 20843941 PMCID: PMC5417385 DOI: 10.1210/me.2010-0072] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2010] [Accepted: 08/16/2010] [Indexed: 11/19/2022] Open
Abstract
Insulin receptor substrate-1 (IRS-1) plays a pivotal role in insulin signaling, therefore its degradation is exquisitely regulated. Here, we show that insulin-stimulated degradation of IRS-1 requires the presence of a highly conserved Ser/Thr-rich domain that we named domain involved in degradation of IRS-1 (DIDI). DIDI (amino acids 386-430 of IRS-1) was identified by comparing the intracellular degradation rate of several truncated forms of IRS-1 transfected into CHO cells. The isolated DIDI domain underwent insulin-stimulated Ser/Thr phosphorylation, suggesting that it serves as a target for IRS-1 kinases. The effects of deletion of DIDI were studied in Fao rat hepatoma and in CHO cells expressing Myc-IRS-1(WT) or Myc-IRS-1(Δ386-430). Deletion of DIDI maintained the ability of IRS-1(Δ386-434) to undergo ubiquitination while rendering it insensitive to insulin-induced proteasomal degradation, which affected IRS-1(WT) (80% at 8 h). Consequently, IRS-1(Δ386-434) mediated insulin signaling (activation of Akt and glycogen synthesis) better than IRS-1(WT). IRS-1(Δ386-434) exhibited a significant greater preference for nuclear localization, compared with IRS-1(WT). Higher nuclear localization was also observed when cells expressing IRS-1(WT) were incubated with the proteasome inhibitor MG-132. The sequence of DIDI is conserved more than 93% across species, from fish to mammals, as opposed to approximately 40% homology of the entire IRS-1. These findings implicate DIDI as a novel, highly conserved domain of IRS-1, which mediates its cellular localization, rate of degradation, and biological activity, with a direct impact on insulin signal transduction.
Collapse
Affiliation(s)
- Sigalit Boura-Halfon
- Department of Molecular Cell Biology, The Weizmann Institute of Science, Rehovot, 76100, Israel
| | | | | | | | | | | | | |
Collapse
|
45
|
Lipina C, Stretton C, Hastings S, Hundal JS, Mackie K, Irving AJ, Hundal HS. Regulation of MAP kinase-directed mitogenic and protein kinase B-mediated signaling by cannabinoid receptor type 1 in skeletal muscle cells. Diabetes 2010; 59:375-85. [PMID: 19933999 PMCID: PMC2809953 DOI: 10.2337/db09-0979] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2009] [Accepted: 11/06/2009] [Indexed: 12/18/2022]
Abstract
OBJECTIVE The endogenous cannabinoid (or endocannabinoid) system (ECS) is part of a central neuromodulatory system thought to play a key role in the regulation of feeding behavior and energy balance. However, increasing evidence suggests that modulation of the ECS may also act to regulate peripheral mechanisms involved in these processes, including lipogenesis in adipose tissue and liver, insulin release from pancreatic beta-cells, and glucose uptake into skeletal muscle. It was recently shown that cannabinoid receptor type 1 (CB1) and type 2 (CB2), both key components of the ECS, are expressed in human and rodent skeletal muscle. However, their role in modulating insulin sensitivity in this metabolically active tissue has yet to be determined. Our aim was to establish the role, if any, of these receptors in modulating insulin sensitivity in skeletal muscle cells. RESEARCH DESIGN AND METHODS Cultured skeletal muscle cells were exposed to CB1 and/or CB2 pharmacological agonists/antagonists/inverse agonists, and the resulting effects on insulin-regulated phosphatidylinositol 3 kinase (PI 3-kinase)-protein kinase B (PKB) and extracellular signal-related kinases 1/2 (ERK1/2)-directed signaling were determined. RESULTS Here, we report that modulating the activity of the ECS in skeletal muscle regulates both insulin-dependent mitogen-activated protein (MAP) kinase (ERK1/2) and the canonical PI 3-kinase/PKB signaling pathways. We show that pharmacological activation or inhibition of CB1 receptor activity exerts a differential effect with regard to MAP kinase- and PKB-directed signaling. CONCLUSIONS Our study provides evidence that signaling via cannabinoid receptors can significantly modulate mitogenic and metabolic signaling in skeletal muscle with important implications for muscle growth and differentiation as well as the regulation of glucose and lipid metabolism.
Collapse
Affiliation(s)
- Christopher Lipina
- Division of Molecular Physiology, James Black Centre, College of Life Sciences, University of Dundee, Dundee, Scotland
| | - Clare Stretton
- Division of Molecular Physiology, James Black Centre, College of Life Sciences, University of Dundee, Dundee, Scotland
| | - Simon Hastings
- Division of Molecular Physiology, James Black Centre, College of Life Sciences, University of Dundee, Dundee, Scotland
| | - Jonathan S. Hundal
- Division of Molecular Physiology, James Black Centre, College of Life Sciences, University of Dundee, Dundee, Scotland
| | - Ken Mackie
- Department of Psychological and Brain Sciences, Gill Center for Biomolecular Science, Indiana University, Bloomington, Indiana
| | - Andrew J. Irving
- Division of Medical Sciences, College of Medicine, Dentistry and Nursing, University of Dundee, Dundee, Scotland
| | - Harinder S. Hundal
- Division of Molecular Physiology, James Black Centre, College of Life Sciences, University of Dundee, Dundee, Scotland
| |
Collapse
|
46
|
Abstract
AbstractSporadic Alzheimer’s disease (sAD) is associated with decreased glucose/energy metabolism in the brain. The majority of glucose utilization in the brain appears to be mediated through glucose transporter protein 1 and 3 (GLUT1 and GLUT3). Deficiency of GLUT1 and GLUT3 in the brain has been found in sAD patients post mortem; however this is not unique to the disease as it is associated with different clinical syndromes as well. In line with recent findings that insulin resistant brain state precedes and may possibly cause sAD, an experimental sAD model based on the central application of the streptozotocin (STZ-icv rat model), which is a selective GLUT2 substrate, has drawn attention to the possible significance of the brain GLUT2 in sAD etiopathogenesis. Important steps in the GLUT2 and sAD interplay are reviewed and discussed. It is concluded that increased vulnerability of GLUT2 expressing neurons may be involved in development of sAD.
Collapse
|
47
|
Sato T, Gotoh N. The FRS2 family of docking/scaffolding adaptor proteins as therapeutic targets of cancer treatment. Expert Opin Ther Targets 2009; 13:689-700. [PMID: 19456272 DOI: 10.1517/14728220902942330] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
BACKGROUND There are two members--FRS2alpha and FRS2beta--in the fibroblast growth factor receptor substrate 2 (FRS2) family of docking/scaffolding adaptor proteins. These proteins function downstream of certain kinds of receptor tyrosine kinases (RTKs) that are important for tumorigenesis. FRS2alpha acts as a control centre for fibroblast growth factor receptor signalling and encourages tumorigenesis, while FRS2beta regulates EGFR signalling negatively, and might have a tumour suppressive role. Therefore, both proteins could be good therapeutic targets for the treatment of cancer. OBJECTIVE To examine the physiological and pathological roles of FRS2, especially in cancer, and describe their potential value as therapeutic targets. METHODS A review of relevant literature. RESULTS/CONCLUSIONS Although it is still difficult to develop small compounds to modify functions of FRS2 adaptor proteins, such compounds may be useful as the next generation of molecular targeting drugs. Combination therapy with RTK-targeting drugs and FRS2-targeting drugs may be useful for cancer treatment in the near future.
Collapse
Affiliation(s)
- Takuya Sato
- The University of Tokyo, Institute of Medical Science, Division of Systems Biomedical Technology, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan.
| | | |
Collapse
|
48
|
Lee YJ, Hsu TC, Du JY, Valentijn AJ, Wu TY, Cheng CF, Yang Z, Streuli CH. Extracellular matrix controls insulin signaling in mammary epithelial cells through the RhoA/Rok pathway. J Cell Physiol 2009; 220:476-84. [DOI: 10.1002/jcp.21793] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
|
49
|
Systems-level interactions between insulin-EGF networks amplify mitogenic signaling. Mol Syst Biol 2009; 5:256. [PMID: 19357636 PMCID: PMC2683723 DOI: 10.1038/msb.2009.19] [Citation(s) in RCA: 159] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2008] [Accepted: 02/23/2009] [Indexed: 01/01/2023] Open
Abstract
Crosstalk mechanisms have not been studied as thoroughly as individual signaling pathways. We exploit experimental and computational approaches to reveal how a concordant interplay between the insulin and epidermal growth factor (EGF) signaling networks can potentiate mitogenic signaling. In HEK293 cells, insulin is a poor activator of the Ras/ERK (extracellular signal-regulated kinase) cascade, yet it enhances ERK activation by low EGF doses. We find that major crosstalk mechanisms that amplify ERK signaling are localized upstream of Ras and at the Ras/Raf level. Computational modeling unveils how critical network nodes, the adaptor proteins GAB1 and insulin receptor substrate (IRS), Src kinase, and phosphatase SHP2, convert insulin-induced increase in the phosphatidylinositol-3,4,5-triphosphate (PIP3) concentration into enhanced Ras/ERK activity. The model predicts and experiments confirm that insulin-induced amplification of mitogenic signaling is abolished by disrupting PIP3-mediated positive feedback via GAB1 and IRS. We demonstrate that GAB1 behaves as a non-linear amplifier of mitogenic responses and insulin endows EGF signaling with robustness to GAB1 suppression. Our results show the feasibility of using computational models to identify key target combinations and predict complex cellular responses to a mixture of external cues.
Collapse
|
50
|
Calreticulin regulates insulin receptor expression and its downstream PI3 Kinase/Akt signalling pathway. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2008; 1783:2344-51. [DOI: 10.1016/j.bbamcr.2008.08.014] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/25/2008] [Revised: 08/27/2008] [Accepted: 08/28/2008] [Indexed: 01/09/2023]
|