1
|
Lin YC, Chang YJ, Gau SS, Lo CM, Yang RB. SCUBE2 regulates adherens junction dynamics and vascular barrier function during inflammation. Cardiovasc Res 2024; 120:1636-1649. [PMID: 38870316 DOI: 10.1093/cvr/cvae132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 01/22/2024] [Accepted: 05/03/2024] [Indexed: 06/15/2024] Open
Abstract
AIMS SCUBE2 (signal peptide-CUB-epidermal growth factor-like domain-containing protein 2) is a secreted or membrane-bound protein originally identified from endothelial cells (ECs). Our previous work showed that SCUBE2 forms a complex with E-cadherin and stabilizes epithelial adherens junctions (AJs) to promote epithelial phenotypes. However, it remains unclear whether SCUBE2 also interacts with vascular endothelial (VE)-cadherin and modulates EC barrier function. In this study, we investigated whether and how SCUBE2 in ECs regulates vascular barrier maintenance. METHODS AND RESULTS We showed that SCUBE2 colocalized and interacted with VE-cadherin and VE-protein tyrosine phosphatase (VE-PTP) within EC AJs. Furthermore, SCUBE2 knockdown disrupted EC AJs and increased EC permeability. Expression of EC SCUBE2 was suppressed at both mRNA and protein levels via the nuclear factor-κB signalling pathway in response to pro-inflammatory cytokines or permeability-inducing agents. In line with these findings, EC-specific deletion of Scube2 (EC-KO) in mice impaired baseline barrier function and worsened vascular leakiness of peripheral capillaries after local injection of histamine or vascular endothelial growth factor. EC-KO mice were also sensitive to pulmonary vascular hyperpermeability and leucocyte infiltration in response to acute endotoxin- or influenza virus-induced systemic inflammation. Meanwhile, EC-specific SCUBE2-overexpressing mice were protected from these effects. Molecular studies suggested that SCUBE2 acts as a scaffold molecule enabling VE-PTP to dephosphorylate VE-cadherin, which prevents VE-cadherin internalization and stabilizes EC AJs. As such, loss of SCUBE2 resulted in hyperphosphorylation of VE-cadherin at tyrosine 685, which led to its endocytosis, thus destabilizing EC AJs and reducing barrier function. All of these effects were exacerbated by inflammatory insults. CONCLUSION We found that SCUBE2 contributes to vascular integrity by recruiting VE-PTP to dephosphorylate VE-cadherin and stabilize AJs, thereby promoting EC barrier function. Moreover, our data suggest that genetic overexpression or pharmacological up-regulation of SCUBE2 may help to prevent vascular leakage and oedema in inflammatory diseases.
Collapse
Affiliation(s)
- Yuh-Charn Lin
- Department of Physiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110301, Taiwan
| | - Ya-Jen Chang
- Institute of Biomedical Sciences, Academia Sinica, 128 Academia Rd., Sec. 2, Taipei 115201, Taiwan
| | - Shiang-Shin Gau
- Institute of Biomedical Sciences, Academia Sinica, 128 Academia Rd., Sec. 2, Taipei 115201, Taiwan
- Graduate School of Biostudies, Kyoto University, Kyoto 6068501, Japan
- Institute for Integrated Cell-Material Sciences (WPI-iCeMS), Kyoto University, Kyoto 6068501, Japan
| | - Chun-Min Lo
- Department of Biomedical Engineering, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan
| | - Ruey-Bing Yang
- Institute of Biomedical Sciences, Academia Sinica, 128 Academia Rd., Sec. 2, Taipei 115201, Taiwan
- Biomedical Translation Research Center, Academia Sinica, 99, Ln. 130, Academia Rd., Sec. 1, Taipei 115201, Taiwan
- Ph.D. Program in Drug Discovery and Development Industry, College of Pharmacy, Taipei Medical University, 250 Wuxing St., Taipei 110301, Taiwan
| |
Collapse
|
2
|
Mazzoli A, Spagnuolo MS, De Palma F, Petecca N, Di Porzio A, Barrella V, Troise AD, Culurciello R, De Pascale S, Scaloni A, Mauriello G, Iossa S, Cigliano L. Limosilactobacillus reuteri DSM 17938 relieves inflammation, endoplasmic reticulum stress, and autophagy in hippocampus of western diet-fed rats by modulation of systemic inflammation. Biofactors 2024; 50:1236-1250. [PMID: 38801155 PMCID: PMC11627471 DOI: 10.1002/biof.2082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Accepted: 03/29/2024] [Indexed: 05/29/2024]
Abstract
The consumption of western diets, high in fats and sugars, is a crucial contributor to brain molecular alterations, cognitive dysfunction and neurodegenerative diseases. Therefore, a mandatory challenge is the individuation of strategies capable of preventing diet-induced impairment of brain physiology. A promising strategy might consist in the administration of probiotics that are known to influence brain function via the gut-brain axis. In this study, we explored whether Limosilactobacillus reuteri DSM 17938 (L. reuteri)-based approach can counteract diet-induced neuroinflammation, endoplasmic reticulum stress (ERS), and autophagy in hippocampus, an area involved in learning and memory, in rat fed a high fat and fructose diet. The western diet induced a microbiota reshaping, but L. reuteri neither modulated this change, nor the plasma levels of short-chain fatty acids. Interestingly, pro-inflammatory signaling pathway activation (increased NFkB phosphorylation, raised amounts of toll-like receptor-4, tumor necrosis factor-alpha, interleukin-6, GFAP, and Haptoglobin), as well as activation of ERS (increased PERK and eif2α phosphorylation, higher C/EBP-homologous protein amounts) and autophagy (increased beclin, P62-sequestosome-1, and LC3 II) was revealed in hippocampus of western diet fed rats. All these hippocampal alterations were prevented by L. reuteri administration, showing for the first time a neuroprotective role of this specific probiotic strain, mainly attributable to its ability to regulate western diet-induced metabolic endotoxemia and systemic inflammation, as decreased levels of lipopolysaccharide, plasma cytokines, and adipokines were also found. Therapeutic strategies based on the use of L. reuteri DSM17938 could be beneficial in reversing metabolic syndrome-mediated brain dysfunction and cognitive decline.
Collapse
Affiliation(s)
- Arianna Mazzoli
- Department of BiologyUniversity of Naples Federico II, Complesso Universitario Monte S. AngeloNaplesItaly
| | - Maria Stefania Spagnuolo
- Institute for the Animal Production System in the Mediterranean EnvironmentNational Research CouncilPorticiItaly
| | - Francesca De Palma
- Department of BiologyUniversity of Naples Federico II, Complesso Universitario Monte S. AngeloNaplesItaly
| | - Natasha Petecca
- Department of BiologyUniversity of Naples Federico II, Complesso Universitario Monte S. AngeloNaplesItaly
| | - Angela Di Porzio
- Department of BiologyUniversity of Naples Federico II, Complesso Universitario Monte S. AngeloNaplesItaly
| | - Valentina Barrella
- Department of BiologyUniversity of Naples Federico II, Complesso Universitario Monte S. AngeloNaplesItaly
| | - Antonio Dario Troise
- Institute for the Animal Production System in the Mediterranean EnvironmentNational Research CouncilPorticiItaly
| | - Rosanna Culurciello
- Department of BiologyUniversity of Naples Federico II, Complesso Universitario Monte S. AngeloNaplesItaly
| | - Sabrina De Pascale
- Institute for the Animal Production System in the Mediterranean EnvironmentNational Research CouncilPorticiItaly
| | - Andrea Scaloni
- Institute for the Animal Production System in the Mediterranean EnvironmentNational Research CouncilPorticiItaly
| | - Gianluigi Mauriello
- Department of Agricultural SciencesUniversity of Naples Federico IIPorticiItaly
| | - Susanna Iossa
- Department of BiologyUniversity of Naples Federico II, Complesso Universitario Monte S. AngeloNaplesItaly
- NBFC, National Biodiversity Future CenterPalermoItaly
- Task Force on Microbiome StudiesUniversity of Naples Federico IIPorticiItaly
| | - Luisa Cigliano
- Department of BiologyUniversity of Naples Federico II, Complesso Universitario Monte S. AngeloNaplesItaly
- Task Force on Microbiome StudiesUniversity of Naples Federico IIPorticiItaly
| |
Collapse
|
3
|
Yang H, Park M, Lee JH, Kim B, Moon CS, Bae S, Kim Y, Lee HJ, Park CY. New peripherally-restricted CB1 receptor antagonists, PMG-505-010 and -013 ameliorate obesity-associated NAFLD and fibrosis. Biomed Pharmacother 2024; 180:117501. [PMID: 39366030 DOI: 10.1016/j.biopha.2024.117501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Revised: 09/22/2024] [Accepted: 09/24/2024] [Indexed: 10/06/2024] Open
Abstract
The endocannabinoid system plays a crucial role in metabolic regulation, prompting the investigation of cannabinoid type 1 receptor (CB1R) antagonists for obesity and its complications like non-alcoholic fatty liver disease (NAFLD). Concerns over psychiatric side effects led to the development of peripheral CB1R antagonists that circumvent the blood-brain barrier (BBB). In this study, we synthesized PMG-505-010 and PMG-505-013 as peripherally restricted CB1 receptor antagonists by modifying rimonabant to minimize BBB penetration. Physicochemical analysis confirmed their reduced lipophilicity and increased polarity compared to rimonabant, indicating limited brain exposure. Molecular docking studies revealed similar binding modes to rimonabant at CB1R, characterized by robust hydrophobic interactions. Functionally, they acted as CB1R antagonists and inverse agonists, effectively reversing CP55,940-induced cAMP inhibition. In a murine model of obesity-related NAFLD, PMG-505-010 and -013 improved metabolic profiles, including fasting blood glucose levels and dyslipidemia. They also ameliorated hepatic injury, steatosis, and inflammation, evidenced by reduced liver enzymes, lipid peroxidation, hepatic lipid levels, and inflammatory cytokine levels. Notably, these compounds inhibited hepatic fibrosis by reducing extracellular matrix (ECM) deposition and altering fibrosis-related gene and protein expressions. In conclusion, PMG-505-010 and PMG-505-013 hold promise for treating obesity-related liver diseases, including NAFLD and fibrosis, through selective peripheral CB1R targeting, potentially avoiding CNS-related side effects seen with earlier CB1R antagonists.
Collapse
Affiliation(s)
- Hyekyung Yang
- Medical Research Institute, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul 03181, Republic of Korea.
| | - Miey Park
- Department of Food and Nutrition, Gachon University, Gyeonggi-do 13120, Republic of Korea; Institute for Aging and Clinical Nutrition Research, Gachon University, Gyeonggi-do 13120, Republic of Korea.
| | - Ji Hye Lee
- PharminoGen Inc., Yongin 16827, Republic of Korea.
| | - Bokyoung Kim
- PharminoGen Inc., Yongin 16827, Republic of Korea.
| | - Chang Sang Moon
- Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea.
| | - Suyeal Bae
- PharminoGen Inc., Yongin 16827, Republic of Korea.
| | | | - Hae-Jeung Lee
- Department of Food and Nutrition, Gachon University, Gyeonggi-do 13120, Republic of Korea; Institute for Aging and Clinical Nutrition Research, Gachon University, Gyeonggi-do 13120, Republic of Korea; Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon 21999, Republic of Korea.
| | - Cheol-Young Park
- Medical Research Institute, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul 03181, Republic of Korea; Division of Endocrinology and Metabolism, Department of Internal Medicine, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul 03181, Republic of Korea.
| |
Collapse
|
4
|
Chauhan A, Dubey S, Jain S. Association Between Type 2 Diabetes Mellitus and Alzheimer's Disease: Common Molecular Mechanism and Therapeutic Targets. Cell Biochem Funct 2024; 42:e4111. [PMID: 39228117 DOI: 10.1002/cbf.4111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 07/11/2024] [Accepted: 08/16/2024] [Indexed: 09/05/2024]
Abstract
Diabetes mellitus (DM) and Alzheimer's disease (AD) rates are rising, mirroring the global trend of an aging population. Numerous epidemiological studies have shown that those with Type 2 diabetes (T2DM) have an increased risk of developing dementia. These degenerative and progressive diseases share some risk factors. To a large extent, the amyloid cascade is responsible for AD development. Neurofibrillary tangles induce neurodegeneration and brain atrophy; this chain reaction begins with hyperphosphorylation of tau proteins caused by progressive amyloid beta (Aβ) accumulation. In addition to these processes, it seems that alterations in brain glucose metabolism and insulin signalling lead to cell death and reduced synaptic plasticity in AD, before the onset of symptoms, which may be years away. Due to the substantial evidence linking insulin resistance in the brain with AD, researchers have coined the name "Type 3 diabetes" to characterize the condition. We still know little about the processes involved, even though current animal models have helped illuminate the links between T2DM and AD. This brief overview discusses insulin and IGF-1 signalling disorders and the primary molecular pathways that may connect them. The presence of GSK-3β in AD is intriguing. These proteins' association with T2DM and pancreatic β-cell failure suggests they might be therapeutic targets for both disorders.
Collapse
Affiliation(s)
- Aparna Chauhan
- Department of Pharmacy, School of Chemical Sciences and Pharmacy, Central University of Rajasthan, Rajasthan, India
| | - Sachin Dubey
- Department of Pharmacy, School of Chemical Sciences and Pharmacy, Central University of Rajasthan, Rajasthan, India
| | - Smita Jain
- Department of Pharmacy, School of Chemical Sciences and Pharmacy, Central University of Rajasthan, Rajasthan, India
| |
Collapse
|
5
|
Shang G, Shao Q, Lv K, Xu W, Ji J, Fan S, Kang X, Cheng F, Wang X, Wang Q. Hypercholesterolemia and the Increased Risk of Vascular Dementia: a Cholesterol Perspective. Curr Atheroscler Rep 2024; 26:435-449. [PMID: 38814418 DOI: 10.1007/s11883-024-01217-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/17/2024] [Indexed: 05/31/2024]
Abstract
PURPOSE OF REVIEW Vascular dementia (VaD) is the second most prevalent type of dementia after Alzheimer's disease.Hypercholesterolemia may increase the risk of dementia, but the association between cholesterol and cognitive function is very complex. From the perspective of peripheral and brain cholesterol, we review the relationship between hypercholesterolemia and increased risk of VaD and how the use of lipid-lowering therapies affects cognition. RECENT FINDINGS Epidemiologic studies show since 1980, non-HDL-C levels of individuals has increased rapidly in Asian countries.The study has suggested that vascular risk factors increase the risk of VaD, such as disordered lipid metabolism. Dyslipidemia has been found to interact with chronic cerebral hypoperfusion to promote inflammation resulting in cognitive dysfunction in the brain.Hypercholesterolemia may be a risk factor for VaD. Inflammation could potentially serve as a link between hypercholesterolemia and VaD. Additionally, the potential impact of lipid-lowering therapy on cognitive function is also worth considering. Finding strategies to prevent and treat VaD is critical given the aging of the population to lessen the load on society. Currently, controlling underlying vascular risk factors is considered one of the most effective methods of preventing VaD. Understanding the relationship between abnormal cholesterol levels and VaD, as well as discovering potential serum biomarkers, is important for the early prevention and treatment of VaD.
Collapse
Affiliation(s)
- Guojiao Shang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, No.11 East Beisanhuan Road, Chaoyang District, Beijing, China
| | - Qi Shao
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, No.11 East Beisanhuan Road, Chaoyang District, Beijing, China
| | - Kai Lv
- Department of Geratology, The Third Affiliated Hospital of Beijing University of Traditional Chinese Medicine, No.51 Xiaoguan Street, Andingmenwai, Chaoyang District, Beijing, China
| | - Wenxiu Xu
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, No.11 East Beisanhuan Road, Chaoyang District, Beijing, China
| | - Jing Ji
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, No.11 East Beisanhuan Road, Chaoyang District, Beijing, China
| | - Shuning Fan
- Dongzhimen Hospital of Beijing University of Chinese Medicine, No.5 Haiyuncang, Dongcheng District, Beijing, China
| | - Xiangdong Kang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, No.11 East Beisanhuan Road, Chaoyang District, Beijing, China
| | - Fafeng Cheng
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, No.11 East Beisanhuan Road, Chaoyang District, Beijing, China.
| | - Xueqian Wang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, No.11 East Beisanhuan Road, Chaoyang District, Beijing, China.
| | - Qingguo Wang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, No.11 East Beisanhuan Road, Chaoyang District, Beijing, China.
| |
Collapse
|
6
|
Liu G, Shu W, Chen Y, Fu Y, Fang S, Zheng H, Cheng W, Lin Q, Hu Y, Jiang N, Yu B. Bone-derived PDGF-BB enhances hippocampal non-specific transcytosis through microglia-endothelial crosstalk in HFD-induced metabolic syndrome. J Neuroinflammation 2024; 21:111. [PMID: 38685040 PMCID: PMC11057146 DOI: 10.1186/s12974-024-03097-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Accepted: 04/10/2024] [Indexed: 05/02/2024] Open
Abstract
BACKGROUND It is well known that high-fat diet (HFD)-induced metabolic syndrome plays a crucial role in cognitive decline and brain-blood barrier (BBB) breakdown. However, whether the bone-brain axis participates in this pathological process remains unknown. Here, we report that platelet-derived growth factor-BB (PDGF-BB) secretion by preosteoclasts in the bone accelerates neuroinflammation. The expression of alkaline phosphatase (ALPL), a nonspecific transcytosis marker, was upregulated during HFD challenge. MAIN BODY Preosteoclast-specific Pdgfb transgenic mice with high PDGF-BB concentrations in the circulation recapitulated the HFD-induced neuroinflammation and transcytosis shift. Preosteoclast-specific Pdgfb knockout mice were partially rescued from hippocampal neuroinflammation and transcytosis shifts in HFD-challenged mice. HFD-induced PDGF-BB elevation aggravated microglia-associated neuroinflammation and interleukin-1β (IL-1β) secretion, which increased ALPL expression and transcytosis shift through enhancing protein 1 (SP1) translocation in endothelial cells. CONCLUSION Our findings confirm the role of bone-secreted PDGF-BB in neuroinflammation and the transcytosis shift in the hippocampal region during HFD challenge and identify a novel mechanism of microglia-endothelial crosstalk in HFD-induced metabolic syndrome.
Collapse
Affiliation(s)
- Guanqiao Liu
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Wen Shu
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Department of Trauma Orthopedics, Liuzhou People's Hospital, Liuzhou, China
| | - Yingqi Chen
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yong Fu
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Trauma Center, Department of Orthopaedic Trauma, The Second Affiliated Hospital of Hengyang Medical College, South China University, Hengyang, China
| | - Shuai Fang
- Trauma Center, Department of Orthopaedic Trauma, The Second Affiliated Hospital of Hengyang Medical College, South China University, Hengyang, China
| | - Haonan Zheng
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Weike Cheng
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Qingrong Lin
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yanjun Hu
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Nan Jiang
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, China.
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| | - Bin Yu
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, China.
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| |
Collapse
|
7
|
Feng Z, Fang C, Ma Y, Chang J. Obesity-induced blood-brain barrier dysfunction: phenotypes and mechanisms. J Neuroinflammation 2024; 21:110. [PMID: 38678254 PMCID: PMC11056074 DOI: 10.1186/s12974-024-03104-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 04/17/2024] [Indexed: 04/29/2024] Open
Abstract
Obesity, a burgeoning global health issue, is increasingly recognized for its detrimental effects on the central nervous system, particularly concerning the integrity of the blood-brain barrier (BBB). This manuscript delves into the intricate relationship between obesity and BBB dysfunction, elucidating the underlying phenotypes and molecular mechanisms. We commence with an overview of the BBB's critical role in maintaining cerebral homeostasis and the pathological alterations induced by obesity. By employing a comprehensive literature review, we examine the structural and functional modifications of the BBB in the context of obesity, including increased permeability, altered transport mechanisms, and inflammatory responses. The manuscript highlights how obesity-induced systemic inflammation and metabolic dysregulation contribute to BBB disruption, thereby predisposing individuals to various neurological disorders. We further explore the potential pathways, such as oxidative stress and endothelial cell dysfunction, that mediate these changes. Our discussion culminates in the summary of current findings and the identification of knowledge gaps, paving the way for future research directions. This review underscores the significance of understanding BBB dysfunction in obesity, not only for its implications in neurodegenerative diseases but also for developing targeted therapeutic strategies to mitigate these effects.
Collapse
Affiliation(s)
- Ziying Feng
- Key Laboratory of Biomedical Imaging Science, Shenzhen Institute of Advanced Technology, System of Chinese Academy of Sciences, Chinese Academy of Sciences, Shenzhen, Guangdong, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Cheng Fang
- Key Laboratory of Biomedical Imaging Science, Shenzhen Institute of Advanced Technology, System of Chinese Academy of Sciences, Chinese Academy of Sciences, Shenzhen, Guangdong, China
| | - Yinzhong Ma
- Key Laboratory of Biomedical Imaging Science, Shenzhen Institute of Advanced Technology, System of Chinese Academy of Sciences, Chinese Academy of Sciences, Shenzhen, Guangdong, China.
- Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Xueyuan Ave 1068, Nanshan, Shenzhen, 518055, Guangdong, China.
| | - Junlei Chang
- Key Laboratory of Biomedical Imaging Science, Shenzhen Institute of Advanced Technology, System of Chinese Academy of Sciences, Chinese Academy of Sciences, Shenzhen, Guangdong, China.
- University of Chinese Academy of Sciences, Beijing, China.
- Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Xueyuan Ave 1068, Nanshan, Shenzhen, 518055, Guangdong, China.
| |
Collapse
|
8
|
Mohammadi S, Moghadam MD, Nasiriasl M, Akhzari M, Barazesh M. Insights into the Therapeutic and Pharmacological Properties of Resveratrol as a Nutraceutical Antioxidant Polyphenol in Health Promotion and Disease Prevention. Curr Rev Clin Exp Pharmacol 2024; 19:327-354. [PMID: 38192151 DOI: 10.2174/0127724328268507231218051058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 11/20/2023] [Accepted: 11/22/2023] [Indexed: 01/10/2024]
Abstract
Resveratrol (3, 5, 4'-trihydroxystilbene) is a polyphenolic derivative with herbal origin. It has attracted considerable attention in recent decades. Many studies have revealed the benefits of Resveratrol over several human disease models, including heart and neurological diseases, nephroprotective, immune regulation, antidiabetic, anti-obesity, age-related diseases, antiviral, and anticancer in experimental and clinical conditions. Recently, the antioxidant and anti-inflammatory activities of Resveratrol have been observed, and it has been shown that Resveratrol reduces inflammatory biomarkers, such as tissue degradation factor, cyclooxygenase 2, nitric oxide synthase, and interleukins. All of these activities appear to be dependent on its structural properties, such as the number and position of the hydroxyl group, which regulates oxidative stress, cell death, and inflammation. Resveratrol is well tolerated and safe even at higher pharmacological doses and desirably affects cardiovascular, neurological, and diabetic diseases. Consequently, it is plausible that Resveratrol can be regarded as a beneficial nutritional additive and a complementary drug, particularly for therapeutic applications. The present review provides an overview of currently available investigations on preventive and therapeutic characteristics and the main molecular mechanisms of Resveratrol and its potent derivatives in various diseases. Thus, this review would enhance knowledge and information about Resveratrol and encourage researchers worldwide to consider it as a pharmaceutical drug to struggle with future health crises against different human disorders.
Collapse
Affiliation(s)
- Shiva Mohammadi
- Department of Medical Biotechnology, School of Medicine, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Maryam Dalaei Moghadam
- Razi Herbal Medicines Research Center, Department of Endodontic, Faculty of Dentistry, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Maryam Nasiriasl
- Radiology Department, Fasa University of Medical Sciences, Fasa, Iran
| | - Morteza Akhzari
- School of Nursing, Larestan University of Medical Sciences, Larestan, Iran
| | - Mahdi Barazesh
- School of Paramedical Sciences, Gerash University of Medical Sciences, Gerash, Iran
| |
Collapse
|
9
|
Wang X, Chen H, Song F, Zuo K, Chen X, Zhang X, Liang L, Ta Q, Zhang L, Li J. Resveratrol: a potential medication for the prevention and treatment of varicella zoster virus-induced ischemic stroke. Eur J Med Res 2023; 28:400. [PMID: 37794518 PMCID: PMC10552394 DOI: 10.1186/s40001-023-01291-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Accepted: 08/14/2023] [Indexed: 10/06/2023] Open
Abstract
BACKGROUND Infection rate of varicella zoster virus (VZV) is 95% in humans, and VZV infection is strongly associated with ischemic stroke (IS). However, the underlying molecular mechanisms of VZV-induced IS are still unclear, and there are no effective agents to treat and prevent VZV-induced IS. OBJECTIVE By integrating bioinformatics, this study explored the interactions between VZV and IS and potential medication to treat and prevent VZV-induced IS. METHODS In this study, the VZV and IS datasets from the GEO database were used to specify the common genes. Then, bioinformatics analysis including Gene Ontology, Kyoto Encyclopedia Genes Genomes and Protein-Protein Interaction network analysis was performed. Further, the hub genes, transcription factor (TF) gene interactions, TF-miRNA co-regulatory network and potential drug were obtained. Finally, validation was performed using molecular docking and molecular dynamics simulations. RESULTS The potential molecular mechanisms of VZV-induced IS were studied using multiple bioinformatics tools. Ten hub genes were COL1A2, DCN, PDGFRB, ACTA2, etc. TF genes and miRNAs included JUN, FOS, CREB, BRCA1, PPARG, STAT3, miR-29, etc. A series of mechanism may be involved, such as inflammation, oxidative stress, blood-brain barrier disruption, foam cell generation and among others. Finally, we proposed resveratrol as a potential therapeutic medicine for the prevention and treatment of VZV-induced IS. Molecular docking and molecular dynamics results showed that resveratrol and hub genes exhibited strong binding score. CONCLUSIONS Resveratrol could be an alternative for the prevention and treatment of VZV-IS. More in vivo and in vitro studies are needed in the future to fully explore the molecular mechanisms between VZV and IS and for medication development.
Collapse
Affiliation(s)
- Xu Wang
- School of Public Health, Jilin University, Changchun, 130021, Jilin, China
- College of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, 130117, Jilin, China
| | - Hao Chen
- Department of Neurovascular Surgery, First Hospital of Jilin University, Changchun, 130021, Jilin, China
| | - Feiyu Song
- Jilin Connell Pharmaceutical Co., Ltd, JilinJilin, 132013, China
| | - Kuiyang Zuo
- School of Public Health, Jilin University, Changchun, 130021, Jilin, China
| | - Xin Chen
- School of Public Health, Jilin University, Changchun, 130021, Jilin, China
| | - Xu Zhang
- School of Public Health, Jilin University, Changchun, 130021, Jilin, China
| | - Lanqian Liang
- School of Public Health, Jilin University, Changchun, 130021, Jilin, China
| | - Qiyi Ta
- College of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, 130117, Jilin, China
| | - Lin Zhang
- China-Japan Union Hospital of Jilin University, Changchun, 130021, Jilin, China.
| | - Jinhua Li
- School of Public Health, Jilin University, Changchun, 130021, Jilin, China.
| |
Collapse
|
10
|
Sun DP, Chen JT, Yang ST, Chen TH, Liu SH, Chen RM. Resveratrol triggers the ER stress-mediated intrinsic apoptosis of neuroblastoma cells coupled with suppression of Rho-dependent migration and consequently prolongs mouse survival. Chem Biol Interact 2023; 382:110645. [PMID: 37482209 DOI: 10.1016/j.cbi.2023.110645] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 07/15/2023] [Accepted: 07/21/2023] [Indexed: 07/25/2023]
Abstract
Neuroblastoma, the most common childhood tumor, are highly malignant and fatal because neuroblastoma cells extremely defend against apoptotic targeting. Traditional treatments for neuroblastomas are usually ineffective and lead to serious side effects and poor prognoses. In this study, we investigated the molecular mechanisms of resveratrol-induced insults to neuroblastoma cells and survival extension of nude mice with neuroblastomas, especially in the endoplasmic reticular (ER) stress-intracellular reactive oxygen species (iROS) axis-mediated signals. Resveratrol specifically killed neuroblastoma cells mainly via apoptosis and autophagy rather than necrosis. As to the mechanisms, resveratrol time-dependently triggered productions of Grp78 protein and iROS in neuroblastoma cells. Attenuating the ER stress-iROS signaling axis significantly suppressed resveratrol-induced autophagy, DNA damage, and cell apoptosis. Successively, resveratrol decreased phosphorylation of retinoblastoma protein and induced cell cycle arrest at the S phase, translocation of Bak protein to mitochondria, a reduction in the mitochondrial membrane potential, cascade activation of caspases-9, -3, and -6, and DNA fragmentation. Moreover, weakening the ER stress-iROS axis concomitantly overcome resveratrol-induced decreases in translocation of Rho protein to membranes and succeeding cell migration. Interestingly, administration of resveratrol did not cause significant side effects but could protect the neuroblastoma-bearing nude mice from body weight loss and consequently extended the animal survival. In parallel, resveratrol elevated levels of Grp78 and then induced cell apoptosis in neuroblastoma tissues. This study has shown that resveratrol could kill neuroblastoma cells and extend survival of animals with neuroblastomas by triggering the ER stress-iROS-involved intrinsic apoptosis and suppression of Rho-dependent cell migration. Our results imply the potential of resveratrol as a drug candidate for chemotherapy of neuroblastoma patients.
Collapse
Affiliation(s)
- Ding-Ping Sun
- Department of Surgery, Chi Mei Medical Center, Tainan, Taiwan; Cell Physiology and Molecular Image Research Center, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
| | - Jui-Tai Chen
- Cell Physiology and Molecular Image Research Center, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan; Department of Anesthesiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Shun-Tai Yang
- Division of Nephrology, Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
| | - Tso-Hsiao Chen
- Cell Physiology and Molecular Image Research Center, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
| | - Shing-Hwa Liu
- Cell Physiology and Molecular Image Research Center, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan; Institute of Toxicology, College of Medicine, National Taiwan University, Taipei, 100, Taiwan
| | - Ruei-Ming Chen
- Cell Physiology and Molecular Image Research Center, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan; Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan; Anesthesiology and Health Policy Research Center, Taipei Medical University Hospital, Taipei, Taiwan; International Ph.D. Program for Cell Therapy and Regeneration Medicine, College of Medicine, Taipei Medical University, Taipei, 11031, Taiwan.
| |
Collapse
|
11
|
Liu G, Wang J, Wei Z, Fang C, Shen K, Qian C, Qi C, Li T, Gao P, Wong PC, Lu H, Cao X, Wan M. Elevated PDGF-BB from Bone Impairs Hippocampal Vasculature by Inducing PDGFRβ Shedding from Pericytes. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2206938. [PMID: 37102631 PMCID: PMC10369301 DOI: 10.1002/advs.202206938] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 04/11/2023] [Indexed: 06/19/2023]
Abstract
Evidence suggests a unique association between bone aging and neurodegenerative/cerebrovascular disorders. However, the mechanisms underlying bone-brain interplay remain elusive. Here platelet-derived growth factor-BB (PDGF-BB) produced by preosteoclasts in bone is reported to promote age-associated hippocampal vascular impairment. Aberrantly elevated circulating PDGF-BB in aged mice and high-fat diet (HFD)-challenged mice correlates with capillary reduction, pericyte loss, and increased blood-brain barrier (BBB) permeability in their hippocampus. Preosteoclast-specific Pdgfb transgenic mice with markedly high plasma PDGF-BB concentration faithfully recapitulate the age-associated hippocampal BBB impairment and cognitive decline. Conversely, preosteoclast-specific Pdgfb knockout mice have attenuated hippocampal BBB impairment in aged mice or HFD-challenged mice. Persistent exposure of brain pericytes to high concentrations of PDGF-BB upregulates matrix metalloproteinase 14 (MMP14), which promotes ectodomain shedding of PDGF receptor β (PDGFRβ) from pericyte surface. MMP inhibitor treatment alleviates hippocampal pericyte loss and capillary reduction in the conditional Pdgfb transgenic mice and antagonizes BBB leakage in aged mice. The findings establish the role of bone-derived PDGF-BB in mediating hippocampal BBB disruption and identify the ligand-induced PDGFRβ shedding as a feedback mechanism for age-associated PDGFRβ downregulation and the consequent pericyte loss.
Collapse
Affiliation(s)
- Guanqiao Liu
- Department of Orthopaedic SurgeryJohns Hopkins University School of MedicineRoss Building, Room 232, 720 Rutland AvenueBaltimoreMD21205USA
| | - Jiekang Wang
- Department of Orthopaedic SurgeryJohns Hopkins University School of MedicineRoss Building, Room 232, 720 Rutland AvenueBaltimoreMD21205USA
| | - Zhiliang Wei
- The Russell H. Morgan Department of Radiology and Radiological ScienceThe Johns Hopkins University School of MedicineBaltimoreMD21205USA
| | - Ching‐Lien Fang
- Department of Orthopaedic SurgeryJohns Hopkins University School of MedicineRoss Building, Room 232, 720 Rutland AvenueBaltimoreMD21205USA
| | - Ke Shen
- Department of Orthopaedic SurgeryJohns Hopkins University School of MedicineRoss Building, Room 232, 720 Rutland AvenueBaltimoreMD21205USA
| | - Cheng Qian
- Department of Orthopaedic SurgeryJohns Hopkins University School of MedicineRoss Building, Room 232, 720 Rutland AvenueBaltimoreMD21205USA
| | - Cheng Qi
- Department of Orthopaedic SurgeryJohns Hopkins University School of MedicineRoss Building, Room 232, 720 Rutland AvenueBaltimoreMD21205USA
| | - Tong Li
- Department of PathologyJohns Hopkins University School of MedicineBaltimoreMD21205USA
| | - Peisong Gao
- Division of Allergy and Clinical ImmunologyJohns Hopkins University School of MedicineBaltimoreMD21224USA
| | - Philip C. Wong
- Department of PathologyJohns Hopkins University School of MedicineBaltimoreMD21205USA
- Department of NeuroscienceJohns Hopkins University School of MedicineBaltimoreMD21205USA
| | - Hanzhang Lu
- The Russell H. Morgan Department of Radiology and Radiological ScienceThe Johns Hopkins University School of MedicineBaltimoreMD21205USA
| | - Xu Cao
- Department of Orthopaedic SurgeryJohns Hopkins University School of MedicineRoss Building, Room 232, 720 Rutland AvenueBaltimoreMD21205USA
| | - Mei Wan
- Department of Orthopaedic SurgeryJohns Hopkins University School of MedicineRoss Building, Room 232, 720 Rutland AvenueBaltimoreMD21205USA
| |
Collapse
|
12
|
Li Q, Zheng Y, Sun Y, Xu G. Resveratrol attenuated fatty acid synthesis through MAPK-PPAR pathway in red tilapia. Comp Biochem Physiol C Toxicol Pharmacol 2023; 268:109598. [PMID: 36898469 DOI: 10.1016/j.cbpc.2023.109598] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Revised: 02/22/2023] [Accepted: 03/01/2023] [Indexed: 03/11/2023]
Abstract
High-fat (HF) diets have been shown to cause hepatic impairment in fish species, but the mode of action, especially the pathways involved, has not yet been determined. In this study, the effects of resveratrol (RES) supplementation on the hepatic structure and fat metabolism of red tilapia (Oreochromis niloticus) were determined. Based on transcriptome and proteomics results, RES was found to promote fatty acid β-oxidation in the blood, liver, and liver cells associated with apoptosis and the MAPK/PPAR signaling pathway. RES supplementation was found to alter the expression of genes related to apoptosis and fatty acid pathways like blood itga6a and armc5 which were upregulated and downregulated respectively by high-fat feeding while ggh and ensonig00000008711 increased and decreased, respectively, with RES addition. Relative to the PPAR signaling pathway, fabp10a and acbd7 showed a reverse U-shaped tendency, both in different treatments and at different times. Proteomics results demonstrated that MAPK/PPAR, carbon/glyoxylate, dicarboxylate/glycine serine, and threonine/drug-other enzymes/beta-alanine metabolism pathways in the RES group were significantly affected, and Fasn and Acox1 decreased and increased, respectively, with RES addition. Seven subgroups were obtained using scRNA-seq, and enrichment analysis showed that the PPAR signaling pathway was upregulated with RES supplementation. RES significantly increased the expression of the marked genes (pck1) ensonig00000037711, fbp10a, granulin, hbe1, and zgc:136461, which are liver cell-specific genes. In conclusion, RES resulted in significantly enriched DGEs associated with fat metabolism and synthesis via the MAPK-PPAR signaling pathway.
Collapse
Affiliation(s)
- Quanjie Li
- Key Laboratory of Freshwater Fisheries and Germplasm Resources Utilization, Ministry of Agriculture and Rural Affairs, Freshwater Fisheries Research Center (FFRC), Chinese Academy of Fishery Sciences (CAFS), Wuxi, Jiangsu 214081, China
| | - Yao Zheng
- Key Laboratory of Freshwater Fisheries and Germplasm Resources Utilization, Ministry of Agriculture and Rural Affairs, Freshwater Fisheries Research Center (FFRC), Chinese Academy of Fishery Sciences (CAFS), Wuxi, Jiangsu 214081, China
| | - Yi Sun
- Key Laboratory of Freshwater Fisheries and Germplasm Resources Utilization, Ministry of Agriculture and Rural Affairs, Freshwater Fisheries Research Center (FFRC), Chinese Academy of Fishery Sciences (CAFS), Wuxi, Jiangsu 214081, China
| | - Gangchun Xu
- Key Laboratory of Freshwater Fisheries and Germplasm Resources Utilization, Ministry of Agriculture and Rural Affairs, Freshwater Fisheries Research Center (FFRC), Chinese Academy of Fishery Sciences (CAFS), Wuxi, Jiangsu 214081, China.
| |
Collapse
|
13
|
Butler MJ, Sengupta S, Muscat SM, Amici SA, Biltz RG, Deems NP, Dravid P, Mackey-Alfonso S, Ijaz H, Bettes MN, Godbout JP, Kapoor A, Guerau-de-Arellano M, Barrientos RM. CD8 + T cells contribute to diet-induced memory deficits in aged male rats. Brain Behav Immun 2023; 109:235-250. [PMID: 36764399 PMCID: PMC10124165 DOI: 10.1016/j.bbi.2023.02.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 01/27/2023] [Accepted: 02/03/2023] [Indexed: 02/11/2023] Open
Abstract
We have previously shown that short-term (3-day) high fat diet (HFD) consumption induces a neuroinflammatory response and subsequent impairment of long-term memory in aged, but not young adult, male rats. However, the immune cell phenotypes driving this proinflammatory response are not well understood. Previously, we showed that microglia isolated from young and aged rats fed a HFD express similar levels of priming and proinflammatory transcripts, suggesting that additional factors may drive the exaggerated neuroinflammatory response selectively observed in aged HFD-fed rats. It is established that T cells infiltrate both the young and especially the aged central nervous system (CNS) and contribute to immune surveillance of the parenchyma. Thus, we investigated the modulating role of short-term HFD on T cell presence in the CNS in aged rats using bulk RNA sequencing and flow cytometry. RNA sequencing results indicate that aging and HFD altered the expression of genes and signaling pathways associated with T cell signaling, immune cell trafficking, and neuroinflammation. Moreover, flow cytometry data showed that aging alone increased CD4+ and CD8+ T cell presence in the brain and that CD8+, but not CD4+, T cells were further increased in aged rats fed a HFD. Based on these data, we selectively depleted circulating CD8+ T cells via an intravenous injection of an anti-CD8 antibody in aged rats prior to 3 days of HFD to infer the functional role these cells may be playing in long-term memory and neuroinflammation. Results indicate that peripheral depletion of CD8+ T cells lowered hippocampal cytokine levels and prevented the HFD-induced i) increase in brain CD8+ T cells, ii) memory impairment, and iii) alterations in pre- and post-synaptic structures in the hippocampus and amygdala. Together, these data indicate a substantial role for CD8+ T cells in mediating diet-induced memory impairments in aged male rats.
Collapse
Affiliation(s)
- Michael J Butler
- Institute for Behavioral Medicine Research, The Ohio State University, Columbus, OH, USA; Department of Psychiatry and Behavioral Health, The Ohio State University, Columbus, OH, USA.
| | - Shouvonik Sengupta
- Institute for Behavioral Medicine Research, The Ohio State University, Columbus, OH, USA; Biomedical Sciences Graduate Program, College of Medicine, The Ohio State University, Columbus, OH, USA
| | - Stephanie M Muscat
- Institute for Behavioral Medicine Research, The Ohio State University, Columbus, OH, USA; Biomedical Sciences Graduate Program, College of Medicine, The Ohio State University, Columbus, OH, USA
| | - Stephanie A Amici
- Institute for Behavioral Medicine Research, The Ohio State University, Columbus, OH, USA; School of Health and Rehabilitation Sciences, The Ohio State University, Columbus, OH, USA
| | - Rebecca G Biltz
- Institute for Behavioral Medicine Research, The Ohio State University, Columbus, OH, USA; Neuroscience Graduate Program, The Ohio State University, Columbus, OH, USA
| | - Nicholas P Deems
- Institute for Behavioral Medicine Research, The Ohio State University, Columbus, OH, USA; Neuroscience Graduate Program, The Ohio State University, Columbus, OH, USA
| | - Piyush Dravid
- College of Medicine, The Ohio State University, Columbus, OH 43210, USA; Center for Vaccines and Immunity, The Research Institute at Nationwide Children's Hospital, Columbus, OH 43205, USA
| | - Sabrina Mackey-Alfonso
- Institute for Behavioral Medicine Research, The Ohio State University, Columbus, OH, USA; Neuroscience Graduate Program, The Ohio State University, Columbus, OH, USA
| | - Haanya Ijaz
- Institute for Behavioral Medicine Research, The Ohio State University, Columbus, OH, USA
| | - Menaz N Bettes
- Institute for Behavioral Medicine Research, The Ohio State University, Columbus, OH, USA
| | - Jonathan P Godbout
- Institute for Behavioral Medicine Research, The Ohio State University, Columbus, OH, USA; Department of Neuroscience, The Ohio State University, Columbus, OH 43210, USA; Chronic Brain Injury Program, The Ohio State University, Columbus, OH 43210, USA
| | - Amit Kapoor
- College of Medicine, The Ohio State University, Columbus, OH 43210, USA; Center for Vaccines and Immunity, The Research Institute at Nationwide Children's Hospital, Columbus, OH 43205, USA
| | - Mireia Guerau-de-Arellano
- Institute for Behavioral Medicine Research, The Ohio State University, Columbus, OH, USA; School of Health and Rehabilitation Sciences, The Ohio State University, Columbus, OH, USA
| | - Ruth M Barrientos
- Institute for Behavioral Medicine Research, The Ohio State University, Columbus, OH, USA; Department of Psychiatry and Behavioral Health, The Ohio State University, Columbus, OH, USA; Department of Neuroscience, The Ohio State University, Columbus, OH 43210, USA; Chronic Brain Injury Program, The Ohio State University, Columbus, OH 43210, USA
| |
Collapse
|
14
|
Marcos JL, Olivares-Barraza R, Ceballo K, Wastavino M, Ortiz V, Riquelme J, Martínez-Pinto J, Muñoz P, Cruz G, Sotomayor-Zárate R. Obesogenic Diet-Induced Neuroinflammation: A Pathological Link between Hedonic and Homeostatic Control of Food Intake. Int J Mol Sci 2023; 24:ijms24021468. [PMID: 36674982 PMCID: PMC9866213 DOI: 10.3390/ijms24021468] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 12/22/2022] [Accepted: 12/26/2022] [Indexed: 01/13/2023] Open
Abstract
Obesity-induced neuroinflammation is a chronic aseptic central nervous system inflammation that presents systemic characteristics associated with increased pro-inflammatory cytokines such as interleukin 1 beta (IL-1β) and interleukin 18 (IL-18) and the presence of microglia and reactive astrogliosis as well as the activation of the NLRP3 inflammasome. The obesity pandemic is associated with lifestyle changes, including an excessive intake of obesogenic foods and decreased physical activity. Brain areas such as the lateral hypothalamus (LH), lateral septum (LS), ventral tegmental area (VTA), and nucleus accumbens (NAcc) have been implicated in the homeostatic and hedonic control of feeding in experimental models of diet-induced obesity. In this context, a chronic lipid intake triggers neuroinflammation in several brain regions such as the hypothalamus, hippocampus, and amygdala. This review aims to present the background defining the significant impact of neuroinflammation and how this, when induced by an obesogenic diet, can affect feeding control, triggering metabolic and neurological alterations.
Collapse
Affiliation(s)
- José Luis Marcos
- Centro de Neurobiología y Fisiopatología Integrativa (CENFI), Instituto de Fisiología, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile
- Escuela de Ciencias Agrícolas y Veterinarias, Universidad Viña del Mar, Viña del Mar 2572007, Chile
- Programa de Doctorado en Ciencias e Ingeniería para la Salud, Universidad de Valparaíso, Valparaíso 2360102, Chile
| | - Rossy Olivares-Barraza
- Centro de Neurobiología y Fisiopatología Integrativa (CENFI), Instituto de Fisiología, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile
- Programa de Doctorado en Ciencias Mención Neurociencias, Universidad de Valparaíso, Valparaíso 2360102, Chile
| | - Karina Ceballo
- Centro de Neurobiología y Fisiopatología Integrativa (CENFI), Instituto de Fisiología, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile
- Programa de Doctorado en Ciencias Mención Neurociencias, Universidad de Valparaíso, Valparaíso 2360102, Chile
| | - Melisa Wastavino
- Centro de Neurobiología y Fisiopatología Integrativa (CENFI), Instituto de Fisiología, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile
| | - Víctor Ortiz
- Centro de Neurobiología y Fisiopatología Integrativa (CENFI), Instituto de Fisiología, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile
| | - Julio Riquelme
- Escuela de Medicina y Centro de Neurología Traslacional (CENTRAS), Facultad de Medicina, Universidad de Valparaíso, Viña del Mar 2540064, Chile
| | - Jonathan Martínez-Pinto
- Centro de Neurobiología y Fisiopatología Integrativa (CENFI), Instituto de Fisiología, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile
| | - Pablo Muñoz
- Escuela de Medicina y Centro de Neurología Traslacional (CENTRAS), Facultad de Medicina, Universidad de Valparaíso, Viña del Mar 2540064, Chile
| | - Gonzalo Cruz
- Centro de Neurobiología y Fisiopatología Integrativa (CENFI), Instituto de Fisiología, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile
| | - Ramón Sotomayor-Zárate
- Centro de Neurobiología y Fisiopatología Integrativa (CENFI), Instituto de Fisiología, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile
- Correspondence: ; Tel.: +56-32-2508050
| |
Collapse
|
15
|
Singh T, Kwatra M, Kushwah P, Pant R, Bezbaruah BK, Jangra A. Binge alcohol consumption exacerbates high-fat diet-induced neurobehavioral anomalies: Possible underlying mechanisms. Chem Biol Interact 2022; 364:110039. [PMID: 35863473 DOI: 10.1016/j.cbi.2022.110039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 06/25/2022] [Accepted: 07/08/2022] [Indexed: 11/29/2022]
Abstract
The current study was aimed to validate the mice model of alcohol (ALC), high-fat diet (HFD), and HFD + ALC combination affecting neurobehavioral and neurochemical anomalies via inflammatory cascade, lowered neurogenesis, enhanced microgliosis, reactive astrogliosis, activated IDO-1 (indoleamine 2,3-dioxygenase), and reduce CHAT (choline acetyltransferase) signaling in the hippocampus (HIP). The adult male Swiss albino mice were provided with ALC (3-15%) and in-house prepared HFD for continuous 12 weeks. The HFD and HFD + ALC consumption impacted the liver and mediated HIP damage. The liver biomarkers (AST, ALT, γ-GT, TG, HDL-C, and LDL-C), oxidative stress, and proinflammatory cytokines (IL-1β and TNF-α) level were found significantly higher in the liver and HIP tissue of HFD + ALC. Furthermore, the neurobehavioral deficits that include cognitive dysfunction, depressive, and, anxiety-like behavior were found severely affected in HFD + ALC consumed mice. The overactivated HPA axis, intense oxidative insults, and increased AChE activity were seen in the HIP of HFD + ALC grouped mice. The gene and protein expression also confirmed disrupted NF-κB-mediated inflammatory and Nrf2-regulated antioxidant balance and dysregulated TrκB/BDNF signaling. Hence, our new findings explain the insight mechanism of chronic alcoholism in exacerbating the deleterious effect of chronic high-fat diet consumption on the HIP.
Collapse
Affiliation(s)
- Tavleen Singh
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Guwahati, Assam, India
| | - Mohit Kwatra
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Guwahati, Assam, India; Department of Pharmacy, Shri Jagdishprasad Jhabarmal Tibrewala (SJJT) University, Jhunjhunu, Churu Rd, Vidyanagari, Churela, Rajasthan, India; Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, The Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| | - Pawan Kushwah
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Guwahati, Assam, India
| | - Rajat Pant
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Guwahati, Assam, India
| | | | - Ashok Jangra
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Guwahati, Assam, India; Department of Pharmaceutical Sciences, Central University of Haryana, Mahendergarh, Haryana, India.
| |
Collapse
|
16
|
Galoppin M, Kari S, Soldati S, Pal A, Rival M, Engelhardt B, Astier A, Thouvenot E. Full spectrum of vitamin D immunomodulation in multiple sclerosis: mechanisms and therapeutic implications. Brain Commun 2022; 4:fcac171. [PMID: 35813882 PMCID: PMC9260308 DOI: 10.1093/braincomms/fcac171] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 05/03/2022] [Accepted: 06/28/2022] [Indexed: 11/17/2022] Open
Abstract
Vitamin D deficiency has been associated with the risk of multiple sclerosis, disease activity and progression. Results from in vitro experiments, animal models and analysis of human samples from randomized controlled trials provide comprehensive data illustrating the pleiotropic actions of Vitamin D on the immune system. They globally result in immunomodulation by decreasing differentiation of effector T and B cells while promoting regulatory subsets. Vitamin D also modulates innate immune cells such as macrophages, monocytes and dendritic cells, and acts at the level of the blood–brain barrier reducing immune cell trafficking. Vitamin D exerts additional activity within the central nervous system reducing microglial and astrocytic activation. The immunomodulatory role of Vitamin D detected in animal models of multiple sclerosis has suggested its potential therapeutic use for treating multiple sclerosis. In this review, we focus on recent published data describing the biological effects of Vitamin D in animal models of multiple sclerosis on immune cells, blood–brain barrier function, activation of glial cells and its potential neuroprotective effects. Based on the current knowledge, we also discuss optimization of therapeutic interventions with Vitamin D in patients with multiple sclerosis, as well as new technologies allowing in-depth analysis of immune cell regulations by vitamin D.
Collapse
Affiliation(s)
- Manon Galoppin
- IGF, University Montpellier, CNRS, INSERM , Montpellier , France
| | - Saniya Kari
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity), INSERM UMR1291 – CNRS UMR5051 – Université Toulouse III , 31024 Toulouse cedex 3 , France
| | - Sasha Soldati
- Theodor Kocher Institute, University of Bern , Bern , Switzerland
| | - Arindam Pal
- Theodor Kocher Institute, University of Bern , Bern , Switzerland
| | - Manon Rival
- IGF, University Montpellier, CNRS, INSERM , Montpellier , France
- Department of Neurology, Nîmes University Hospital, University Montpellier , Nîmes , France
| | | | - Anne Astier
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity), INSERM UMR1291 – CNRS UMR5051 – Université Toulouse III , 31024 Toulouse cedex 3 , France
| | - Eric Thouvenot
- IGF, University Montpellier, CNRS, INSERM , Montpellier , France
- Department of Neurology, Nîmes University Hospital, University Montpellier , Nîmes , France
| |
Collapse
|
17
|
Saiyasit N, Butlig EAR, Chaney SD, Traylor MK, Hawley NA, Randall RB, Bobinger HV, Frizell CA, Trimm F, Crook ED, Lin M, Hill BD, Keller JL, Nelson AR. Neurovascular Dysfunction in Diverse Communities With Health Disparities-Contributions to Dementia and Alzheimer's Disease. Front Neurosci 2022; 16:915405. [PMID: 35844216 PMCID: PMC9279126 DOI: 10.3389/fnins.2022.915405] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 05/31/2022] [Indexed: 12/13/2022] Open
Abstract
Alzheimer's disease and related dementias (ADRD) are an expanding worldwide crisis. In the absence of scientific breakthroughs, the global prevalence of ADRD will continue to increase as more people are living longer. Racial or ethnic minority groups have an increased risk and incidence of ADRD and have often been neglected by the scientific research community. There is mounting evidence that vascular insults in the brain can initiate a series of biological events leading to neurodegeneration, cognitive impairment, and ADRD. We are a group of researchers interested in developing and expanding ADRD research, with an emphasis on vascular contributions to dementia, to serve our local diverse community. Toward this goal, the primary objective of this review was to investigate and better understand health disparities in Alabama and the contributions of the social determinants of health to those disparities, particularly in the context of vascular dysfunction in ADRD. Here, we explain the neurovascular dysfunction associated with Alzheimer's disease (AD) as well as the intrinsic and extrinsic risk factors contributing to dysfunction of the neurovascular unit (NVU). Next, we ascertain ethnoregional health disparities of individuals living in Alabama, as well as relevant vascular risk factors linked to AD. We also discuss current pharmaceutical and non-pharmaceutical treatment options for neurovascular dysfunction, mild cognitive impairment (MCI) and AD, including relevant studies and ongoing clinical trials. Overall, individuals in Alabama are adversely affected by social and structural determinants of health leading to health disparities, driven by rurality, ethnic minority status, and lower socioeconomic status (SES). In general, these communities have limited access to healthcare and healthy food and other amenities resulting in decreased opportunities for early diagnosis of and pharmaceutical treatments for ADRD. Although this review is focused on the current state of health disparities of ADRD patients in Alabama, future studies must include diversity of race, ethnicity, and region to best be able to treat all individuals affected by ADRD.
Collapse
Affiliation(s)
- Napatsorn Saiyasit
- Department of Physiology and Cell Biology, College of Medicine, University of South Alabama, Mobile, AL, United States
| | - Evan-Angelo R. Butlig
- Department of Neurology, Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, Intellectual and Developmental Disabilities Research Center, University of California, Los Angeles, Los Angeles, CA, United States
| | - Samantha D. Chaney
- Department of Physiology and Cell Biology, College of Medicine, University of South Alabama, Mobile, AL, United States
| | - Miranda K. Traylor
- Department of Health, Kinesiology, and Sport, University of South Alabama, Mobile, AL, United States
| | - Nanako A. Hawley
- Department of Psychology, University of South Alabama, Mobile, AL, United States
| | - Ryleigh B. Randall
- Department of Physiology and Cell Biology, College of Medicine, University of South Alabama, Mobile, AL, United States
| | - Hanna V. Bobinger
- Department of Physiology and Cell Biology, College of Medicine, University of South Alabama, Mobile, AL, United States
| | - Carl A. Frizell
- Department of Physician Assistant Studies, University of South Alabama, Mobile, AL, United States
| | - Franklin Trimm
- College of Medicine, University of South Alabama, Mobile, AL, United States
| | - Errol D. Crook
- Department of Internal Medicine, College of Medicine, University of South Alabama, Mobile, AL, United States
| | - Mike Lin
- Department of Physiology and Cell Biology, College of Medicine, University of South Alabama, Mobile, AL, United States
| | - Benjamin D. Hill
- Department of Psychology, University of South Alabama, Mobile, AL, United States
| | - Joshua L. Keller
- Department of Health, Kinesiology, and Sport, University of South Alabama, Mobile, AL, United States
| | - Amy R. Nelson
- Department of Physiology and Cell Biology, College of Medicine, University of South Alabama, Mobile, AL, United States
| |
Collapse
|
18
|
Kim N, Lee J, Seon Song H, Joon Oh Y, Kwon MS, Yun M, Ki Lim S, Kyeong Park H, Seo Jang Y, Lee S, Choi SP, Woon Roh S, Choi HJ. Kimchi intake alleviates obesity-induced neuroinflammation by modulating the gut-brain axis. Food Res Int 2022; 158:111533. [DOI: 10.1016/j.foodres.2022.111533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Revised: 05/26/2022] [Accepted: 06/17/2022] [Indexed: 11/04/2022]
|
19
|
Vohra MS, Benchoula K, Serpell CJ, Hwa WE. AgRP/NPY and POMC neurons in the arcuate nucleus and their potential role in treatment of obesity. Eur J Pharmacol 2022; 915:174611. [PMID: 34798121 DOI: 10.1016/j.ejphar.2021.174611] [Citation(s) in RCA: 83] [Impact Index Per Article: 27.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2021] [Revised: 10/27/2021] [Accepted: 10/27/2021] [Indexed: 02/08/2023]
Abstract
Obesity is a major health crisis affecting over a third of the global population. This multifactorial disease is regulated via interoceptive neural circuits in the brain, whose alteration results in excessive body weight. Certain central neuronal populations in the brain are recognised as crucial nodes in energy homeostasis; in particular, the hypothalamic arcuate nucleus (ARC) region contains two peptide microcircuits that control energy balance with antagonistic functions: agouti-related peptide/neuropeptide-Y (AgRP/NPY) signals hunger and stimulates food intake; and pro-opiomelanocortin (POMC) signals satiety and reduces food intake. These neuronal peptides levels react to energy status and integrate signals from peripheral ghrelin, leptin, and insulin to regulate feeding and energy expenditure. To manage obesity comprehensively, it is crucial to understand cellular and molecular mechanisms of information processing in ARC neurons, since these regulate energy homeostasis. Importantly, a specific strategy focusing on ARC circuits needs to be devised to assist in treating obese patients and maintaining weight loss with minimal or no side effects. The aim of this review is to elucidate the recent developments in the study of AgRP-, NPY- and POMC-producing neurons, specific to their role in controlling metabolism. The impact of ghrelin, leptin, and insulin signalling via action of these neurons is also surveyed, since they also impact energy balance through this route. Lastly, we present key proteins, targeted genes, compounds, drugs, and therapies that actively work via these neurons and could potentially be used as therapeutic targets for treating obesity conditions.
Collapse
Affiliation(s)
- Muhammad Sufyan Vohra
- School of Medicine, Faculty of Health and Medical Sciences, Taylor's University Lakeside Campus, 47500, Subang Jaya, Selangor Darul Ehsan, Malaysia
| | - Khaled Benchoula
- School of Medicine, Faculty of Health and Medical Sciences, Taylor's University Lakeside Campus, 47500, Subang Jaya, Selangor Darul Ehsan, Malaysia
| | - Christopher J Serpell
- School of Physical Sciences, Ingram Building, University of Kent, Canterbury, Kent, CT2 7NH, United Kingdom
| | - Wong Eng Hwa
- School of Medicine, Faculty of Health and Medical Sciences, Taylor's University Lakeside Campus, 47500, Subang Jaya, Selangor Darul Ehsan, Malaysia.
| |
Collapse
|
20
|
Frank CJ, McNay EC. Breakdown of the blood-brain barrier: A mediator of increased Alzheimer's risk in patients with metabolic disorders? J Neuroendocrinol 2022; 34:e13074. [PMID: 34904299 PMCID: PMC8791015 DOI: 10.1111/jne.13074] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 11/12/2021] [Accepted: 11/26/2021] [Indexed: 01/03/2023]
Abstract
Metabolic disorders (MDs), including type 1 and 2 diabetes and chronic obesity, are among the faster growing diseases globally and are a primary risk factor for Alzheimer's disease (AD). The term "type-3 diabetes" has been proposed for AD due to the interrelated cellular, metabolic, and immune features shared by diabetes, insulin resistance (IR), and the cognitive impairment and neurodegeneration found in AD. Patients with MDs and/or AD commonly exhibit altered glucose homeostasis and IR; systemic chronic inflammation encompassing all of the periphery, blood-brain barrier (BBB), and central nervous system; pathological vascular remodeling; and increased BBB permeability that allows transfusion of neurotoxic molecules from the blood to the brain. This review summarizes the components of the BBB, mechanisms through which MDs alter BBB permeability via immune and metabolic pathways, the contribution of BBB dysfunction to the manifestation and progression of AD, and current avenues of therapeutic research that address BBB permeability. In addition, issues with the translational applicability of current animal models of AD regarding BBB dysfunction and proposals for future directions of research that address the relationship between MDs, BBB dysfunction, and AD are discussed.
Collapse
Affiliation(s)
- Corey J Frank
- Behavioral Neuroscience, University at Albany, SUNY, Albany, NY, USA
| | - Ewan C McNay
- Behavioral Neuroscience, University at Albany, SUNY, Albany, NY, USA
| |
Collapse
|
21
|
Fu X, Shah AP, Keighron J, Mou TCM, Ladenheim B, Alt J, Fukudome D, Niwa M, Tamashiro KL, Tanda G, Sawa A, Cadet JL, Rais R, Baraban JM. Elevated body fat increases amphetamine accumulation in brain: evidence from genetic and diet-induced forms of adiposity. Transl Psychiatry 2021; 11:427. [PMID: 34392304 PMCID: PMC8364554 DOI: 10.1038/s41398-021-01547-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 07/22/2021] [Accepted: 08/04/2021] [Indexed: 12/03/2022] Open
Abstract
Despite the high prevalence of obesity, little is known about its potential impact on the pharmacokinetics of psychotropic drugs. In the course of investigating the role of the microRNA system on neuronal signaling, we found that mice lacking the translin/trax microRNA-degrading enzyme display an exaggerated locomotor response to amphetamine. As these mice display robust adiposity in the context of normal body weight, we checked whether this phenotype might reflect elevated brain levels of amphetamine. To assess this hypothesis, we compared plasma and brain amphetamine levels of wild type and Tsn KO mice. Furthermore, we checked the effect of diet-induced increases in adiposity on plasma and brain amphetamine levels in wild type mice. Brain amphetamine levels were higher in Tsn KO mice than in wild type littermates and correlated with adiposity. Analysis of the effect of diet-induced increases in adiposity in wild type mice on brain amphetamine levels also demonstrated that brain amphetamine levels correlate with adiposity. Increased adiposity displayed by Tsn KO mice or by wild type mice fed a high-fat diet correlates with elevated brain amphetamine levels. As amphetamine and its analogues are widely used to treat attention deficit disorder, which is associated with obesity, further studies are warranted to assess the impact of adiposity on amphetamine levels in these patients.
Collapse
Affiliation(s)
- Xiuping Fu
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA
| | - Aparna P Shah
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA
| | - Jacqueline Keighron
- Medication Development Program, National Institute on Drug Abuse, Intramural Research Program, National Institutes of Health, 333 Cassell Drive, Baltimore, MD, 21224, USA
| | - Ta-Chung M Mou
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA
| | - Bruce Ladenheim
- Molecular Neuropsychiatry Research Branch, Intramural Research Program, NIDA/NIH/DHHS, 251 Bayview Boulevard, Baltimore, MD, 21224, USA
| | - Jesse Alt
- John Hopkins Drug Discovery, Department of Neurology, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA
| | - Daisuke Fukudome
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA
| | - Minae Niwa
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, AL, 35233, USA
| | - Kellie L Tamashiro
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA
| | - Gianluigi Tanda
- Medication Development Program, National Institute on Drug Abuse, Intramural Research Program, National Institutes of Health, 333 Cassell Drive, Baltimore, MD, 21224, USA
| | - Akira Sawa
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA
| | - Jean-Lud Cadet
- Molecular Neuropsychiatry Research Branch, Intramural Research Program, NIDA/NIH/DHHS, 251 Bayview Boulevard, Baltimore, MD, 21224, USA
| | - Rana Rais
- John Hopkins Drug Discovery, Department of Neurology, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA
| | - Jay M Baraban
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA.
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA.
| |
Collapse
|
22
|
Zimmerman B, Kundu P, Rooney WD, Raber J. The Effect of High Fat Diet on Cerebrovascular Health and Pathology: A Species Comparative Review. Molecules 2021; 26:3406. [PMID: 34199898 PMCID: PMC8200075 DOI: 10.3390/molecules26113406] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Revised: 05/19/2021] [Accepted: 06/01/2021] [Indexed: 02/08/2023] Open
Abstract
In both humans and animal models, consumption of a high-saturated-fat diet has been linked to vascular dysfunction and cognitive impairments. Laboratory animals provide excellent models for more invasive high-fat-diet-related research. However, the physiological differences between humans and common animal models in terms of how they react metabolically to high-fat diets need to be considered. Here, we review the factors that may affect the translatability of mechanistic research in animal models, paying special attention to the effects of a high-fat diet on vascular outcomes. We draw attention to the dissociation between metabolic syndrome and dyslipidemia in rodents, unlike the state in humans, where the two commonly occur. We also discuss the differential vulnerability between species to the metabolic and vascular effects of macronutrients in the diet. Findings from animal studies are better interpreted as modeling specific aspects of dysfunction. We conclude that the differences between species provide an opportunity to explore why some species are protected from the detrimental aspects of high-fat-diet-induced dysfunction, and to translate these findings into benefits for human health.
Collapse
Affiliation(s)
- Benjamin Zimmerman
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR 97239, USA; (B.Z.); (P.K.); (W.D.R.)
- Advanced Imaging Research Center, Oregon Health & Science University, Portland, OR 97239, USA
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Payel Kundu
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR 97239, USA; (B.Z.); (P.K.); (W.D.R.)
| | - William D. Rooney
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR 97239, USA; (B.Z.); (P.K.); (W.D.R.)
- Advanced Imaging Research Center, Oregon Health & Science University, Portland, OR 97239, USA
| | - Jacob Raber
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR 97239, USA; (B.Z.); (P.K.); (W.D.R.)
- Departments of Neurology and Radiation Medicine, Division of Neuroscience, ONPRC, Oregon Health & Science University, Portland, OR 97239, USA
| |
Collapse
|
23
|
Cruz AM, Partridge KM, Malekizadeh Y, Vlachaki Walker JM, Weightman Potter PG, Pye KR, Shaw SJ, Ellacott KLJ, Beall C. Brain Permeable AMP-Activated Protein Kinase Activator R481 Raises Glycaemia by Autonomic Nervous System Activation and Amplifies the Counterregulatory Response to Hypoglycaemia in Rats. Front Endocrinol (Lausanne) 2021; 12:697445. [PMID: 34975743 PMCID: PMC8718766 DOI: 10.3389/fendo.2021.697445] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Accepted: 11/15/2021] [Indexed: 11/13/2022] Open
Abstract
AIM We evaluated the efficacy of a novel brain permeable "metformin-like" AMP-activated protein kinase activator, R481, in regulating glucose homeostasis. MATERIALS AND METHODS We used glucose sensing hypothalamic GT1-7 neuronal cells and pancreatic αTC1.9 α-cells to examine the effect of R481 on AMPK pathway activation and cellular metabolism. Glucose tolerance tests and hyperinsulinemic-euglycemic and hypoglycemic clamps were used in Sprague-Dawley rats to assess insulin sensitivity and hypoglycemia counterregulation, respectively. RESULTS In vitro, we demonstrate that R481 increased AMPK phosphorylation in GT1-7 and αTC1.9 cells. In Sprague-Dawley rats, R481 increased peak glucose levels during a glucose tolerance test, without altering insulin levels or glucose clearance. The effect of R481 to raise peak glucose levels was attenuated by allosteric brain permeable AMPK inhibitor SBI-0206965. This effect was also completely abolished by blockade of the autonomic nervous system using hexamethonium. During hypoglycemic clamp studies, R481 treated animals had a significantly lower glucose infusion rate compared to vehicle treated controls. Peak plasma glucagon levels were significantly higher in R481 treated rats with no change to plasma adrenaline levels. In vitro, R481 did not alter glucagon release from αTC1.9 cells, but increased glycolysis. Non brain permeable AMPK activator R419 enhanced AMPK activity in vitro in neuronal cells but did not alter glucose excursion in vivo. CONCLUSIONS These data demonstrate that peripheral administration of the brain permeable "metformin-like" AMPK activator R481 increases blood glucose by activation of the autonomic nervous system and amplifies the glucagon response to hypoglycemia in rats. Taken together, our data suggest that R481 amplifies the counterregulatory response to hypoglycemia by a central rather than a direct effect on the pancreatic α-cell. These data provide proof-of-concept that central AMPK could be a target for future drug development for prevention of hypoglycemia in diabetes.
Collapse
Affiliation(s)
- Ana M Cruz
- Institute of Biomedical and Clinical Sciences, College of Medicine and Health, University of Exeter, Exeter, United Kingdom
| | - Katie M Partridge
- Institute of Biomedical and Clinical Sciences, College of Medicine and Health, University of Exeter, Exeter, United Kingdom
| | - Yasaman Malekizadeh
- Institute of Biomedical and Clinical Sciences, College of Medicine and Health, University of Exeter, Exeter, United Kingdom
| | - Julia M Vlachaki Walker
- Institute of Biomedical and Clinical Sciences, College of Medicine and Health, University of Exeter, Exeter, United Kingdom
| | - Paul G Weightman Potter
- Institute of Biomedical and Clinical Sciences, College of Medicine and Health, University of Exeter, Exeter, United Kingdom
| | - Katherine R Pye
- Institute of Biomedical and Clinical Sciences, College of Medicine and Health, University of Exeter, Exeter, United Kingdom
| | - Simon J Shaw
- Rigel Pharmaceuticals Inc., South San Francisco, CA, United States
| | - Kate L J Ellacott
- Institute of Biomedical and Clinical Sciences, College of Medicine and Health, University of Exeter, Exeter, United Kingdom
| | - Craig Beall
- Institute of Biomedical and Clinical Sciences, College of Medicine and Health, University of Exeter, Exeter, United Kingdom
| |
Collapse
|
24
|
Lee JY, Park T, Hong E, Amatya R, Park KA, Park YH, Min KA, Jin M, Lee S, Hwang S, Roh GS, Shin MC. Genetic engineering of novel super long-acting Exendin-4 chimeric protein for effective treatment of metabolic and cognitive complications of obesity. Biomaterials 2020; 257:120250. [PMID: 32736262 DOI: 10.1016/j.biomaterials.2020.120250] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2020] [Revised: 06/21/2020] [Accepted: 07/17/2020] [Indexed: 02/08/2023]
Abstract
A common bottleneck challenge for many therapeutic proteins lies in their short plasma half-lives, which often makes the treatment far less compliant or even disables achieving sufficient therapeutic efficacy. To address this problem, we introduce a novel drug delivery strategy based on the genetic fusion of an albumin binding domain (ABD) and an anti-neonatal Fc receptor (FcRn) affibody (AFF) to therapeutic proteins. This ABD-AFF fusion strategy can provide a synergistic effect on extending the plasma residence time by, on one hand, preventing the rapid glomerular filtration via ABD-mediated albumin binding and, on the other hand, increasing the efficiency of FcRn-mediated recycling by AFF-mediated high-affinity binding to the FcRn. In this research, we explored the feasibility of applying the ABD-AFF fusion strategy to exendin-4 (EX), a clinically available anti-diabetic peptide possessing a short plasma half-life. The EX-ABD-AFF produced from the E. coli displayed a remarkably (241-fold) longer plasma half-life than the SUMO tagged-EX (SUMO-EX) (0.7 h) in mice. Furthermore, in high-fat diet (HFD)-fed obese mice model, the EX-ABD-AFF could provide significant hypoglycemic effects for over 12 days, accompanied by a reduction of body weight. In the long-term study, the EX-ABD-AFF could significantly reverse the obesity-related metabolic complications (hyperglycemia, hyperlipidemia, and hepatic steatosis) and, moreover, improve cognitive deficits. Overall, this study demonstrated that the ABD-AFF fusion could be an effective strategy to greatly increase the plasma half-lives of therapeutic proteins and thus markedly improve their druggability.
Collapse
Affiliation(s)
- Jong Youl Lee
- Department of Anatomy and Convergence Medical Science, Bio Anti-Aging Medical Research Center, Institute of Health Sciences, College of Medicine, Gyeongsang National University, Jinju, Gyeongnam, 52727, Republic of Korea
| | - Taehoon Park
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Gyeongsang National University, 501 Jinju Daero, Jinju, Gyeongnam, 52828, Republic of Korea
| | - Eunmi Hong
- New Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation (DGMIF), Daegu, 41061, Republic of Korea
| | - Reeju Amatya
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Gyeongsang National University, 501 Jinju Daero, Jinju, Gyeongnam, 52828, Republic of Korea
| | - Kyung-Ah Park
- Department of Anatomy and Convergence Medical Science, Bio Anti-Aging Medical Research Center, Institute of Health Sciences, College of Medicine, Gyeongsang National University, Jinju, Gyeongnam, 52727, Republic of Korea
| | - Young-Hoon Park
- New Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation (DGMIF), Daegu, 41061, Republic of Korea
| | - Kyoung Ah Min
- College of Pharmacy and Inje Institute of Pharmaceutical Sciences and Research, Inje University, Gimhae, Gyeongnam, 50834, Republic of Korea
| | - Minki Jin
- College of Pharmacy and Inje Institute of Pharmaceutical Sciences and Research, Inje University, Gimhae, Gyeongnam, 50834, Republic of Korea
| | - Sumi Lee
- College of Pharmacy and Inje Institute of Pharmaceutical Sciences and Research, Inje University, Gimhae, Gyeongnam, 50834, Republic of Korea
| | - Seungmi Hwang
- College of Pharmacy and Inje Institute of Pharmaceutical Sciences and Research, Inje University, Gimhae, Gyeongnam, 50834, Republic of Korea
| | - Gu Seob Roh
- Department of Anatomy and Convergence Medical Science, Bio Anti-Aging Medical Research Center, Institute of Health Sciences, College of Medicine, Gyeongsang National University, Jinju, Gyeongnam, 52727, Republic of Korea.
| | - Meong Cheol Shin
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Gyeongsang National University, 501 Jinju Daero, Jinju, Gyeongnam, 52828, Republic of Korea.
| |
Collapse
|
25
|
Spagnuolo MS, Pallottini V, Mazzoli A, Iannotta L, Tonini C, Morone B, Ståhlman M, Crescenzo R, Strazzullo M, Iossa S, Cigliano L. A Short‐Term Western Diet Impairs Cholesterol Homeostasis and Key Players of Beta Amyloid Metabolism in Brain of Middle Aged Rats. Mol Nutr Food Res 2020; 64:e2000541. [DOI: 10.1002/mnfr.202000541] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Indexed: 12/28/2022]
Affiliation(s)
| | - Valentina Pallottini
- Department of ScienceBiomedical and Technology Science SectionUniversity Roma Tre Rome 00146 Italy
| | - Arianna Mazzoli
- Department of BiologyUniversity of Naples Federico II Naples 80126 Italy
| | - Lucia Iannotta
- Department of BiologyUniversity of Naples Federico II Naples 80126 Italy
| | - Claudia Tonini
- Department of ScienceBiomedical and Technology Science SectionUniversity Roma Tre Rome 00146 Italy
| | - Barbara Morone
- Institute of Genetics and Biophysics “A. Buzzati‐Traverso”National Research Council Naples 80131 Italy
| | - Marcus Ståhlman
- Wallenberg LaboratoryDepartment of Molecular and Clinical MedicineSahlgrenska AcademyUniversity of Gothenburg Gothenburg 413 45 Sweden
| | | | - Maria Strazzullo
- Institute of Genetics and Biophysics “A. Buzzati‐Traverso”National Research Council Naples 80131 Italy
| | - Susanna Iossa
- Department of BiologyUniversity of Naples Federico II Naples 80126 Italy
| | - Luisa Cigliano
- Department of BiologyUniversity of Naples Federico II Naples 80126 Italy
| |
Collapse
|
26
|
Pandey SN, Kwatra M, Dwivedi DK, Choubey P, Lahkar M, Jangra A. 7,8-Dihydroxyflavone alleviated the high-fat diet and alcohol-induced memory impairment: behavioral, biochemical and molecular evidence. Psychopharmacology (Berl) 2020; 237:1827-1840. [PMID: 32206827 DOI: 10.1007/s00213-020-05502-2] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2019] [Accepted: 03/05/2020] [Indexed: 12/12/2022]
Abstract
RATIONALE Alcoholism and obesity impart a deleterious impact on human health and affects the quality of life. Chronic consumption of alcohol and western diet has been reported to cause memory deficits. 7,8-dihydroxyflavone (7,8-DHF), a TrkB agonist, comprises antioxidant and anti-inflammatory properties in treating various neurological disorders. OBJECTIVES The current study was aimed to determine the protective effect and molecular mechanism of 7,8-DHF against alcohol and high-fat diet (HFD)-induced memory deficits in rats. METHODS The adult male Wistar rats were given alcohol (3-15%) and HFD ad libitum for 12 weeks in different experimental groups. 7,8-DHF (5 mg/kg) was intraperitoneally injected daily for the last 4 weeks (9th-12th week). RESULTS The alcohol and HFD administration caused cognitive impairment as evaluated through the Morris water maze (MWM) test in alcohol, HFD, and alcohol + HFD-fed animals. The last 4-week treatment of 7,8-DHF (5 mg/kg; i.p.) attenuated alcohol and HFD-induced memory loss. 7,8-DHF treatment also restored the glutathione (GSH) level along with attenuation of nitrite, malondialdehyde content (markers of oxidative and nitrosative stress), and reduction of the acetylcholinesterase activity in the hippocampus of alcohol and HFD-fed animals. Furthermore, the administration of 7,8-DHF caused downregulation of NF-κB, iNOS, and caspase-3 and upregulation of Nrf2, HO-1, and BDNF mRNA level in rat hippocampus. CONCLUSION 7,8-DHF administration conferred beneficial effects against alcohol and HFD-induced memory deficit via its unique antioxidant, anti-inflammatory, anti-apoptotic potential, along with the activation of TrkB/BDNF signaling pathway in the hippocampus.
Collapse
Affiliation(s)
- Surya Narayan Pandey
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Guwahati, Assam, India
| | - Mohit Kwatra
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Guwahati, Assam, India
| | - Durgesh Kumar Dwivedi
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Guwahati, Assam, India
| | - Priyansha Choubey
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Guwahati, Assam, India
| | - Mangala Lahkar
- Department of Pharmacology, Gauhati Medical College, Guwahati, Assam, India
| | - Ashok Jangra
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Guwahati, Assam, India.
- Department of Pharmacology, KIET School of Pharmacy, KIET Group of Institutions, Ghaziabad, Uttar Pradesh, India.
| |
Collapse
|
27
|
Gehrke N, Schattenberg JM. Metabolic Inflammation-A Role for Hepatic Inflammatory Pathways as Drivers of Comorbidities in Nonalcoholic Fatty Liver Disease? Gastroenterology 2020; 158:1929-1947.e6. [PMID: 32068022 DOI: 10.1053/j.gastro.2020.02.020] [Citation(s) in RCA: 122] [Impact Index Per Article: 24.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 02/05/2020] [Accepted: 02/11/2020] [Indexed: 02/06/2023]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is a global and growing health concern. Emerging evidence points toward metabolic inflammation as a key process in the fatty liver that contributes to multiorgan morbidity. Key extrahepatic comorbidities that are influenced by NAFLD are type 2 diabetes, cardiovascular disease, and impaired neurocognitive function. Importantly, the presence of nonalcoholic steatohepatitis and advanced hepatic fibrosis increase the risk for systemic comorbidity in NAFLD. Although the precise nature of the crosstalk between the liver and other organs has not yet been fully elucidated, there is emerging evidence that metabolic inflammation-in part, emanating from the fatty liver-is the engine that drives cellular dysfunction, cell death, and deleterious remodeling within various body tissues. This review describes several inflammatory pathways and mediators that have been implicated as links between NAFLD and type 2 diabetes, cardiovascular disease, and neurocognitive decline.
Collapse
Affiliation(s)
- Nadine Gehrke
- Metabolic Liver Research Program, I. Department of Medicine, University Medical Center, Mainz, Germany.
| | - Jörn M Schattenberg
- Metabolic Liver Research Program, I. Department of Medicine, University Medical Center, Mainz, Germany
| |
Collapse
|
28
|
De Paula GC, de Oliveira J, Engel DF, Lopes SC, Moreira ELG, Figueiredo CP, Prediger RD, Fabro de Bem A. Red wine consumption mitigates the cognitive impairments in low-density lipoprotein receptor knockout (LDLr -/-) mice. Nutr Neurosci 2020; 24:978-988. [PMID: 31910791 DOI: 10.1080/1028415x.2019.1704472] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Although the benefits of moderate intake of red wine in decreasing incidence of cardiovascular diseases associated to hypercholesterolemia are well recognized, there are still widespread misconceptions about its effects on the hypercholesterolemia-related cognitive impairments. Herein we investigated the putative benefits of regular red wine consumption on cognitive performance of low-density lipoprotein receptor knockout (LDLr-/-) mice, an animal model of familial hypercholesterolemia, which display cognitive impairments since early ages. The red wine was diluted into the drinking water to a final concentration of 6% ethanol and was available for 60 days for LDLr-/- mice fed a normal or high-cholesterol diet. The results indicated that moderate red wine consumption did not alter locomotor parameters and liver toxicity. Across multiple cognitive tasks evaluating spatial learning/reference memory and recognition/identification memory, hypercholesterolemic mice drinking red wine performed significantly better than water group, regardless of diet. Additionally, immunofluorescence assays indicated a reduction of astrocyte activation and lectin stain in the hippocampus of LDLr-/- mice under consumption of red wine. These findings demonstrate that the moderate consumption of red wine attenuates short- and long-term memory decline associated with hypercholesterolemia in mice and suggest that it could be through a neurovascular action.
Collapse
Affiliation(s)
| | - Jade de Oliveira
- Laboratory of Experimental Neurology, Extremo Sul Catarinense University, Criciúma, Brazil
| | - Daiane Fátima Engel
- Department of Biochemistry, Federal University of Santa Catarina, Florianópolis, Brazil
| | | | | | | | - Rui Daniel Prediger
- Department of Pharmacology, Federal University of Santa Catarina, Florianópolis, Brazil
| | - Andreza Fabro de Bem
- Department of Biochemistry, Federal University of Santa Catarina, Florianópolis, Brazil.,Department of Physiological Science, University of Brasilia, Brasilia, Brazil
| |
Collapse
|
29
|
Salameh TS, Mortell WG, Logsdon AF, Butterfield DA, Banks WA. Disruption of the hippocampal and hypothalamic blood-brain barrier in a diet-induced obese model of type II diabetes: prevention and treatment by the mitochondrial carbonic anhydrase inhibitor, topiramate. Fluids Barriers CNS 2019; 16:1. [PMID: 30616618 PMCID: PMC6323732 DOI: 10.1186/s12987-018-0121-6] [Citation(s) in RCA: 74] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Accepted: 12/18/2018] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Type II diabetes is a vascular risk factor for cognitive impairment and increased risk of dementia. Disruption of the blood-retinal barrier (BRB) and blood-brain barrier (BBB) are hallmarks of subsequent retinal edema and central nervous system dysfunction. However, the mechanisms by which diet or metabolic syndrome induces dysfunction are not understood. A proposed mechanism is an increase in reactive oxygen species (ROS) and oxidative stress. Inhibition of mitochondrial carbonic anhydrase (mCA) decreases ROS and oxidative stress. In this study, topiramate, a mCA inhibitor, was examined for its ability to protect the BRB and BBB in diet-induced obese type II diabetic mice. METHODS BBB and BRB permeability were assessed using 14C-sucrose and 99mTc-albumin in CD-1 mice fed a low-fat (control) or a high-fat diet. Topiramate administration was compared to saline controls in both preventative and efficacy arms examining BRB and BBB disruption. Body weight and blood glucose were measured weekly and body composition was assessed using EchoMRI. Metabolic activity was measured using a comprehensive laboratory animal monitoring system. Brain tissues collected from the mice were assessed for changes in oxidative stress and tight junction proteins. RESULTS High-fat feeding caused increased entry of 14C-sucrose and 99mTc-albumin into the brains of diet-induced obese type II diabetic mice. Increased permeability to 14C-sucrose was observed in the hypothalamus and hippocampus, and attenuated by topiramate treatment, while increased permeability to 99mTc-albumin occurred in the whole brain and was also attenuated by topiramate. Treatment with topiramate decreased measures of oxidative stress and increased expression of the tight junction proteins ZO-1 and claudin-12. In the retina, we observed increased entry of 99mTc-albumin simultaneously with increased entry into the whole brain during the preventative arm. This occurred prior to increased entry to the retina for 14C-sucrose which occurred during the efficacy arm. Treatment with topiramate had no effect on the retina. CONCLUSIONS Blood-brain barrier and blood-retinal barrier dysfunction were examined in a mouse model of diet-induced obese type II diabetes. These studies demonstrate that there are spatial and temporal differences in 14C-sucrose and 99mTc-albumin permeability in the brain and retina of diet-induced obese type II diabetic mice. Topiramate, a mitochondrial carbonic anhydrase inhibitor, is efficacious at both preventing and treating BBB disruption in this diet-induced obese type II diabetic mouse model.
Collapse
Affiliation(s)
- Therese S. Salameh
- Geriatrics Research, Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, 1660 S. Columbian Way, 810A/Bldg 1, Seattle, WA 98108 USA
- Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington, Seattle, WA USA
| | - William G. Mortell
- Geriatrics Research, Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, 1660 S. Columbian Way, 810A/Bldg 1, Seattle, WA 98108 USA
| | - Aric F. Logsdon
- Geriatrics Research, Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, 1660 S. Columbian Way, 810A/Bldg 1, Seattle, WA 98108 USA
- Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington, Seattle, WA USA
| | - D. Allan Butterfield
- Department of Chemistry and Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY USA
| | - William A. Banks
- Geriatrics Research, Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, 1660 S. Columbian Way, 810A/Bldg 1, Seattle, WA 98108 USA
- Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington, Seattle, WA USA
| |
Collapse
|
30
|
Wang W, Yang W, Shen Z, Wen S, Hu M. The Dose-Response Effect of Lycopene on Cerebral Vessel and Neuron Impairment Induced by Hyperlipidemia. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2018; 66:13173-13182. [PMID: 30474364 DOI: 10.1021/acs.jafc.8b05232] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
To study the dose-response effect of lycopene on vessel and neuron damage in the brain against hyperlipidemia, rats were fed with hypercholesterolemic feed and treated with lycopene orally by gavage at the dose of 5, 25, 45, 65, 85, and 105, 125 mg/kg/bw-1/d-1. At the end of the fourth week, lycopene doses and serum lycopene concentration showed an inverse U-shape curve. Serum lycopene concentration was negatively correlated with the levels of serum TC, TG, LDL-C, as well as the cerebral LDL-C, VEGF, and VCAM-1. Serum lycopene concentration was positively correlated with the expression of Claudin-5 and the number of neurons in hippocampal CA1 and CA3. Lycopene could also reduce the pathologic change of these areas. These results suggested an inverse U-shape relation between dose and serum concentration of lycopene, and intermediate doses were most effective to protect cerebral vessels and neurons from being damaged by hyperlipidemia.
Collapse
Affiliation(s)
- Wei Wang
- Department of Nutrition and Food Hygiene, Xiang Ya School of Public Health , Central South University , Changsha 410078 , China
| | - Weichun Yang
- Department of Nutrition and Food Hygiene, Xiang Ya School of Public Health , Central South University , Changsha 410078 , China
| | - Ziyi Shen
- Second Xiangya Hospital , Central South University , Changsha 410013 , China
| | - Sixian Wen
- Department of Nutrition and Food Hygiene, Xiang Ya School of Public Health , Central South University , Changsha 410078 , China
| | - Minyu Hu
- Department of Nutrition and Food Hygiene, Xiang Ya School of Public Health , Central South University , Changsha 410078 , China
| |
Collapse
|
31
|
Francisco A, Ronchi JA, Navarro CDC, Figueira TR, Castilho RF. Nicotinamide nucleotide transhydrogenase is required for brain mitochondrial redox balance under hampered energy substrate metabolism and high-fat diet. J Neurochem 2018; 147:663-677. [PMID: 30281804 DOI: 10.1111/jnc.14602] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Revised: 09/13/2018] [Accepted: 09/24/2018] [Indexed: 12/21/2022]
Abstract
Among mitochondrial NADP-reducing enzymes, nicotinamide nucleotide transhydrogenase (NNT) establishes an elevated matrix NADPH/NADP+ by catalyzing the reduction of NADP+ at the expense of NADH oxidation coupled to inward proton translocation across the inner mitochondrial membrane. Here, we characterize NNT activity and mitochondrial redox balance in the brain using a congenic mouse model carrying the mutated Nnt gene from the C57BL/6J strain. The absence of NNT activity resulted in lower total NADPH sources activity in the brain mitochondria of young mice, an effect that was partially compensated in aged mice. Nonsynaptic mitochondria showed higher NNT activity than synaptic mitochondria. In the absence of NNT, an increased release of H2 O2 from mitochondria was observed when the metabolism of respiratory substrates occurred with restricted flux through relevant mitochondrial NADPH sources or when respiratory complex I was inhibited. In accordance, mitochondria from Nnt-/- brains were unable to sustain NADP in its reduced state when energized in the absence of carbon substrates, an effect aggravated after H2 O2 bolus metabolism. These data indicate that the lack of NNT in brain mitochondria impairs peroxide detoxification, but peroxide detoxification can be partially counterbalanced by concurrent NADPH sources depending on substrate availability. Notably, only brain mitochondria from Nnt-/- mice chronically fed a high-fat diet exhibited lower activity of the redox-sensitive aconitase, suggesting that brain mitochondrial redox balance requires NNT under the metabolic stress of a high-fat diet. Overall, the role of NNT in the brain mitochondria redox balance especially comes into play under mitochondrial respiratory defects or high-fat diet.
Collapse
Affiliation(s)
- Annelise Francisco
- Faculty of Medical Sciences, Department of Clinical Pathology, University of Campinas (UNICAMP), Campinas, SP, Brazil
| | - Juliana A Ronchi
- Faculty of Medical Sciences, Department of Clinical Pathology, University of Campinas (UNICAMP), Campinas, SP, Brazil
| | - Claudia D C Navarro
- Faculty of Medical Sciences, Department of Clinical Pathology, University of Campinas (UNICAMP), Campinas, SP, Brazil
| | - Tiago R Figueira
- Faculty of Medical Sciences, Department of Clinical Pathology, University of Campinas (UNICAMP), Campinas, SP, Brazil.,School of Physical Education and Sport of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Roger F Castilho
- Faculty of Medical Sciences, Department of Clinical Pathology, University of Campinas (UNICAMP), Campinas, SP, Brazil
| |
Collapse
|
32
|
Wakabayashi C, Kunugi H. Involvement of IL-6 and GSK3β in impaired sensorimotor gating induced by high-fat diet. Neurosci Res 2018; 147:33-38. [PMID: 30326250 DOI: 10.1016/j.neures.2018.10.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Revised: 10/05/2018] [Accepted: 10/11/2018] [Indexed: 11/26/2022]
Abstract
Increased levels of proinflammatory cytokines have been implicated in schizophrenia; however, their pathophysiological roles in abnormal brain dysfunctions remain unclear. We evaluated the effect of proinflammatory cytokines on a high-fat diet (HFD)-induced prepulse inhibition (PPI) deficits in the acoustic startle response. Eight-week-old male C57BL/6J mice were fed a HFD for 3 weeks and then PPI was examined. HFD significantly induced PPI deficits and increased plasma IL-6, but not TNFα, levels. Interestingly, MR16-1 administration during the HFD period ameliorated PPI deficits. Further, in the striatum of HFD-fed mice, phosphorylation of GSK3β, but not GSK3α, was significantly increased; this increase was attenuated by MR16-1, although the protein levels of GSK3α and β were not altered. There were no significant differences in either phosphorylation or protein levels of GSK3α, β in the PFC during the HFD period. These results suggest that increased IL-6 levels during HFD may induce sensorimotor gating deficits, likely through the alteration of striatal GSK3β phosphorylation. MR16-1 might have a beneficial effect on such HFD-induced sensorimotor gating deficits.
Collapse
Affiliation(s)
- Chisato Wakabayashi
- Department of Mental Disorder Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, 4-1-1, Ogawa-higashi, Kodaira, Tokyo, 187-8502, Japan
| | - Hiroshi Kunugi
- Department of Mental Disorder Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, 4-1-1, Ogawa-higashi, Kodaira, Tokyo, 187-8502, Japan.
| |
Collapse
|
33
|
Resveratrol alleviates ethanol-induced neuroinflammation in vivo and in vitro: Involvement of TLR2-MyD88-NF-κB pathway. Int J Biochem Cell Biol 2018; 103:56-64. [PMID: 30107238 DOI: 10.1016/j.biocel.2018.07.007] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Revised: 07/17/2018] [Accepted: 07/19/2018] [Indexed: 02/03/2023]
Abstract
Excessive ethanol (EtOH) intake affects cognitive function and leads to permanent learning and memory deficits. EtOH-induced neuroinflammation plays an important role in EtOH neurotoxicity. Studies have shown that EtOH activates microglia and induces an inflammatory response. Resveratrol (Rsv) is a natural polyphenol found in a wide variety of plants and fruits, and produces the neuroprotective and anti-inflammatory effects in the central nervous system. However, effect of Rsv on EtOH-induced neuroinflammation is still unknown. We investigated the anti-inflammatory effect of Rsv in the context of EtOH-induced neurotoxicity and the molecular mechanisms potentially involved in the effect. The results showed that treatment of rats with Rsv prevented the deficits of spatial reference memory induced by EtOH and mitigated EtOH-induced neuroinflammation by inhibiting microglial activation and decreasing the levels of inflammatory cytokines including interleukin-1β, interleukin-6 and tumor necrosis factor α. The further studies indicated that Rsv reduced TLR2 expression in vivo and in vitro, and downregulated expression of myeloid differentiation primary response 88 (MyD88) and phosphorylation of nuclear factor κB (NF-κB). These data demonstrate that Rsv alleviates the ethanol-induced neuroinflammation via inhibition of TLR2-MyD88-NF-κB signal pathway.
Collapse
|
34
|
Liao WH, Suendermann C, Steuer AE, Pacheco Lopez G, Odermatt A, Faresse N, Henneberg M, Langhans W. Aldosterone deficiency in mice burdens respiration and accentuates diet-induced hyperinsulinemia and obesity. JCI Insight 2018; 3:99015. [PMID: 30046010 DOI: 10.1172/jci.insight.99015] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Accepted: 06/08/2018] [Indexed: 12/20/2022] Open
Abstract
Aldosterone synthase inhibitors (ASIs) should alleviate obesity-related cardiovascular and renal problems resulting partly from aldosterone excess, but their clinical use may have limitations. To improve knowledge for the use of ASIs, we investigated physiology in aldosterone synthase-knockout (ASKO) mice. On regular chow diet (CD), ASKO mice ate more and weighed less than WT mice, largely because they hyperventilated to eliminate acid as CO2. Replacing CD with high-fat diet (HFD) lessened the respiratory burden in ASKO mice, as did 12- to 15-hour fasting. The latter eliminated the genotype differences in respiratory workload and energy expenditure (EE). Thus, aldosterone deficiency burdened the organism more when the animals ate carbohydrate-rich chow than when they ate a HFD. Chronic HFD exposure further promoted hyperinsulinemia in ASKO mice that contributed to visceral fat accumulation accompanied by reduced lipolysis, thermogenic reprogramming, and the absence of weight-gain-related EE increases. Intracerebroventricular aldosterone supplementation in ASKO mice attenuated the HFD-induced hyperinsulinemia, but did not affect EE, suggesting that the presence of aldosterone increased the body's energetic efficiency, thus counteracting the EE-increasing effect of low insulin. ASIs may therefore cause acid-overload-induced respiratory burden and promote obesity. Their use in patients with preexisting renal and cardiopulmonary diseases might be contraindicated.
Collapse
Affiliation(s)
- Wan-Hui Liao
- Physiology and Behavior Laboratory, Institute of Food, Nutrition and Health, ETH Zurich, Schwerzenbach, Switzerland.,Institute of Anatomy, University of Zurich, Zurich, Switzerland.,Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, Zurich, Switzerland.,National Center of Competence in Research "Kidney.CH", Switzerland
| | | | - Andrea Eva Steuer
- Department of Forensic Pharmacology and Toxicology, Zurich Institute of Forensic Medicine, University of Zurich, Zurich, Switzerland
| | - Gustavo Pacheco Lopez
- Physiology and Behavior Laboratory, Institute of Food, Nutrition and Health, ETH Zurich, Schwerzenbach, Switzerland.,Department of Health Sciences, Division of Biological and Health Sciences, Metropolitan Autonomous University (UAM), Lerma, Mexico
| | - Alex Odermatt
- National Center of Competence in Research "Kidney.CH", Switzerland.,Division of Molecular and Systems Toxicology, Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland
| | - Nourdine Faresse
- Institute of Anatomy, University of Zurich, Zurich, Switzerland.,Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, Zurich, Switzerland.,National Center of Competence in Research "Kidney.CH", Switzerland
| | - Maciej Henneberg
- Institute of Evolutionary Medicine, University of Zurich, Zurich, Switzerland.,Biological Anthropology and Comparative Anatomy Unit, University of Adelaide, Australia
| | - Wolfgang Langhans
- Physiology and Behavior Laboratory, Institute of Food, Nutrition and Health, ETH Zurich, Schwerzenbach, Switzerland
| |
Collapse
|
35
|
Rijnsburger M, Unmehopa UA, Eggels L, Serlie MJ, la Fleur SE. One-week exposure to a free-choice high-fat high-sugar diet does not disrupt blood-brain barrier permeability in fed or overnight fasted rats. Nutr Neurosci 2017; 22:541-550. [PMID: 29284375 DOI: 10.1080/1028415x.2017.1418727] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Objectives: The hypothalamus lies adjacent to the third ventricle and is in close proximity with the median eminence (ME), a circumventricular organ with an incomplete blood-brain barrier (BBB) which controls direct entry of nutrients into the brain. The blood-CSF barrier of the hypothalamus shows dynamic changes upon neuroendocrine events and adjusts permeability with the tight junction (TJ) complex. It has been shown that chronic exposure to a high-fat diet (HFD) affects BBB permeability. HFD also induces leptin resistance and alters neuropeptide expression in the arcuate nucleus (Arc) of the hypothalamus starting early during overnutrition. We hypothesized altered integrity of the BBB to occur after exposing rats to a free-choice high-fat high-sugar (fcHFHS) diet for 1 week. Methods: We measured diffusion of Evans blue dye over the ME and assessed expression of the TJ proteins ZO-1, claudin-5, and occludin in the tanycytic wall of the third ventricle. Furthermore, we assessed protein expression of glucose transporter 1 (GLUT-1), which is highly expressed in the Arc-ME complex and facilitates glucose transport over the BBB. Results: fcHFHS-fed rats increased caloric intake compared to control, however, there was no effect of the fcHFHS diet on permeability of the BBB, nor changes in protein expression of tight TJ proteins or GLUT-1. Fasting acutely affects the BBB and we hypothesized that exposure to the fcHFHS diet affects the BBB differently compared to chow after fasting. We did not, however, find any differences in Evans blue diffusion nor protein expression between chow- and fcHFHS-fed rats when fasted overnight. Conclusions: We conclude that short-term consumption of a fcHFHS diet does not change permeability or diffusion in the hypothalamus barrier in ad libitum fed or fasted rats.
Collapse
Affiliation(s)
- M Rijnsburger
- a Department of Endocrinology & Metabolism , Academic Medical Center , Amsterdam , Netherlands.,b Laboratory of Endocrinology, Department of Clinical Chemistry , Academic Medical Center , Amsterdam , Netherlands
| | - U A Unmehopa
- a Department of Endocrinology & Metabolism , Academic Medical Center , Amsterdam , Netherlands.,b Laboratory of Endocrinology, Department of Clinical Chemistry , Academic Medical Center , Amsterdam , Netherlands
| | - L Eggels
- a Department of Endocrinology & Metabolism , Academic Medical Center , Amsterdam , Netherlands.,b Laboratory of Endocrinology, Department of Clinical Chemistry , Academic Medical Center , Amsterdam , Netherlands.,c Metabolism and Reward Group, Royal Netherlands Academy of Arts and Sciences , Netherlands Institute for Neuroscience , Amsterdam , Netherlands
| | - M J Serlie
- a Department of Endocrinology & Metabolism , Academic Medical Center , Amsterdam , Netherlands
| | - S E la Fleur
- a Department of Endocrinology & Metabolism , Academic Medical Center , Amsterdam , Netherlands.,b Laboratory of Endocrinology, Department of Clinical Chemistry , Academic Medical Center , Amsterdam , Netherlands.,c Metabolism and Reward Group, Royal Netherlands Academy of Arts and Sciences , Netherlands Institute for Neuroscience , Amsterdam , Netherlands
| |
Collapse
|
36
|
Bernier M, Wahl D, Ali A, Allard J, Faulkner S, Wnorowski A, Sanghvi M, Moaddel R, Alfaras I, Mattison JA, Tarantini S, Tucsek Z, Ungvari Z, Csiszar A, Pearson KJ, de Cabo R. Resveratrol supplementation confers neuroprotection in cortical brain tissue of nonhuman primates fed a high-fat/sucrose diet. Aging (Albany NY) 2017; 8:899-916. [PMID: 27070252 PMCID: PMC4931843 DOI: 10.18632/aging.100942] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2016] [Accepted: 03/30/2016] [Indexed: 01/19/2023]
Abstract
Previous studies have shown positive effects of long-term resveratrol (RSV) supplementation in preventing pancreatic beta cell dysfunction, arterial stiffening and metabolic decline induced by high-fat/high-sugar (HFS) diet in nonhuman primates. Here, the analysis was extended to examine whether RSV may reduce dietary stress toxicity in the cerebral cortex of the same cohort of treated animals. Middle-aged male rhesus monkeys were fed for 2 years with HFS alone or combined with RSV, after which whole-genome microarray analysis of cerebral cortex tissue was carried out along with ELISA, immunofluorescence, and biochemical analyses to examine markers of vascular health and inflammation in the cerebral cortices. A number of genes and pathways that were differentially modulated in these dietary interventions indicated an exacerbation of neuroinflammation (e.g., oxidative stress markers, apoptosis, NF-κB activation) in HFS-fed animals and protection by RSV treatment. The decreased expression of mitochondrial aldehyde dehydrogenase 2, dysregulation in endothelial nitric oxide synthase, and reduced capillary density induced by HFS stress were rescued by RSV supplementation. Our results suggest that long-term RSV treatment confers neuroprotection against cerebral vascular dysfunction during nutrient stress.
Collapse
Affiliation(s)
- Michel Bernier
- Translational Gerontology Branch, National Institute on Aging, NIH, Baltimore, MD 21224, USA
| | - Devin Wahl
- Translational Gerontology Branch, National Institute on Aging, NIH, Baltimore, MD 21224, USA
| | - Ahmed Ali
- Translational Gerontology Branch, National Institute on Aging, NIH, Baltimore, MD 21224, USA
| | - Joanne Allard
- Translational Gerontology Branch, National Institute on Aging, NIH, Baltimore, MD 21224, USA.,Department of Physiology and Biophysics, Howard University, College of Medicine, Washington, DC 20059, USA
| | - Shakeela Faulkner
- Translational Gerontology Branch, National Institute on Aging, NIH, Baltimore, MD 21224, USA
| | - Artur Wnorowski
- Department of Biopharmacy, Medical University of Lublin, 20-093 Lublin, Poland.,Laboratory of Clinical Investigation, National Institute on Aging, NIH, Baltimore, MD 21224, USA
| | - Mitesh Sanghvi
- Laboratory of Clinical Investigation, National Institute on Aging, NIH, Baltimore, MD 21224, USA
| | - Ruin Moaddel
- Laboratory of Clinical Investigation, National Institute on Aging, NIH, Baltimore, MD 21224, USA
| | - Irene Alfaras
- Translational Gerontology Branch, National Institute on Aging, NIH, Baltimore, MD 21224, USA
| | - Julie A Mattison
- Translational Gerontology Branch, National Institute on Aging, NIH, Baltimore, MD 21224, USA
| | - Stefano Tarantini
- University of Oklahoma Health Science Center, Oklahoma City, OK 73104, USA
| | - Zsuzsanna Tucsek
- University of Oklahoma Health Science Center, Oklahoma City, OK 73104, USA
| | - Zoltan Ungvari
- University of Oklahoma Health Science Center, Oklahoma City, OK 73104, USA
| | - Anna Csiszar
- University of Oklahoma Health Science Center, Oklahoma City, OK 73104, USA
| | - Kevin J Pearson
- Translational Gerontology Branch, National Institute on Aging, NIH, Baltimore, MD 21224, USA.,Graduate Center for Nutritional Sciences, University of Kentucky, Lexington, KY 40536, USA
| | - Rafael de Cabo
- Translational Gerontology Branch, National Institute on Aging, NIH, Baltimore, MD 21224, USA
| |
Collapse
|
37
|
Wu GJ, Chen JT, Tsai HC, Chen TL, Liu SH, Chen RM. Protection of Dexmedetomidine Against Ischemia/Reperfusion-Induced Apoptotic Insults to Neuronal Cells Occurs Via an Intrinsic Mitochondria-Dependent Pathway. J Cell Biochem 2017; 118:2635-2644. [DOI: 10.1002/jcb.25847] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2016] [Accepted: 12/15/2016] [Indexed: 01/26/2023]
Affiliation(s)
- Gong-Jhe Wu
- Department of Anesthesiology; Shin Kong Wu Ho-Su Memorial Hospital; Taipei Taiwan
- Comprehensive Cancer Center; Taipei Medical University; Taipei Taiwan
| | - Jui-Tai Chen
- Graduate Institute of Medical Sciences; College of Medicine; Taipei Medical University; Taipei Taiwan
| | - Hsiao-Chien Tsai
- Anesthesiology and Health Policy Research Center; Taipei Medical University; Taipei Taiwan
| | - Ta-Liang Chen
- Anesthesiology and Health Policy Research Center; Taipei Medical University; Taipei Taiwan
| | - Shing-Hwa Liu
- Institute of Toxicology, College of Medicine; National Taiwan University; Taipei Taiwan
| | - Ruei-Ming Chen
- Comprehensive Cancer Center; Taipei Medical University; Taipei Taiwan
- Graduate Institute of Medical Sciences; College of Medicine; Taipei Medical University; Taipei Taiwan
- Anesthesiology and Health Policy Research Center; Taipei Medical University; Taipei Taiwan
| |
Collapse
|
38
|
Contribution of thrombin-reactive brain pericytes to blood-brain barrier dysfunction in an in vivo mouse model of obesity-associated diabetes and an in vitro rat model. PLoS One 2017; 12:e0177447. [PMID: 28489922 PMCID: PMC5425209 DOI: 10.1371/journal.pone.0177447] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Accepted: 04/27/2017] [Indexed: 12/14/2022] Open
Abstract
Diabetic complications are characterized by the dysfunction of pericytes located around microvascular endothelial cells. The blood–brain barrier (BBB) exhibits hyperpermeability with progression of diabetes. Therefore, brain pericytes at the BBB may be involved in diabetic complications of the central nervous system (CNS). We hypothesized that brain pericytes respond to increased brain thrombin levels in diabetes, leading to BBB dysfunction and diabetic CNS complications. Mice were fed a high-fat diet (HFD) for 2 or 8 weeks to induce obesity. Transport of i.v.-administered sodium fluorescein and 125I-thrombin across the BBB were measured. We evaluated brain endothelial permeability and expression of tight junction proteins in the presence of thrombin–treated brain pericytes using a BBB model of co-cultured rat brain endothelial cells and pericytes. Mice fed a HFD for 8 weeks showed both increased weight gain and impaired glucose tolerance. In parallel, the brain influx rate of sodium fluorescein was significantly greater than that in mice fed a normal diet. HFD feeding inhibited the decline in brain thrombin levels occurring during 6 weeks of feeding. In the HFD fed mice, plasma thrombin levels were significantly increased, by up to 22%. 125I-thrombin was transported across the BBB in normal mice after i.v. injection, with uptake further enhanced by co-injection of unlabeled thrombin. Thrombin-treated brain pericytes increased brain endothelial permeability and caused decreased expression of zona occludens-1 (ZO-1) and occludin and morphological disorganization of ZO-1. Thrombin also increased mRNA expression of interleukin-1β and 6 and tumor necrosis factor-α in brain pericytes. Thrombin can be transported from circulating blood through the BBB, maintaining constant levels in the brain, where it can stimulate pericytes to induce BBB dysfunction. Thus, the brain pericyte–thrombin interaction may play a key role in causing BBB dysfunction in obesity-associated diabetes and represent a therapeutic target for its CNS complications.
Collapse
|
39
|
Ito S, Yanai M, Yamaguchi S, Couraud PO, Ohtsuki S. Regulation of Tight-Junction Integrity by Insulin in an In Vitro Model of Human Blood-Brain Barrier. J Pharm Sci 2017; 106:2599-2605. [PMID: 28456720 DOI: 10.1016/j.xphs.2017.04.036] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Revised: 04/14/2017] [Accepted: 04/17/2017] [Indexed: 12/30/2022]
Abstract
Although insulin receptor is expressed at the human blood-brain barrier (BBB), the physiological and pathologic roles of insulin signaling in biologic responses at the BBB remain unclear. Here, we investigate insulin signaling at the human BBB using human cerebral microvascular endothelial cell line (hCMEC/D3) as a well-established in vitro model. Western blot analysis showed that insulin induced phosphorylation of extracellular signal-regulated kinase and insulin receptor substrate-1 in hCMEC/D3 cells. Short-term insulin stimulation increased cell proliferation via the canonical phosphoinositide-3 kinase/protein kinase B and mitogen-activated protein kinase signaling pathways, suggesting that insulin signaling is involved in the regulation of biologic responses in the human BBB. We also found that insulin rapidly increased tight-junction integrity of hCMEC/D3 cells via the phosphoinositide-3 kinase/protein kinase B/glycogen synthase kinase-3 β signaling pathway. Inhibition of insulin/insulin-like growth factor-1 receptor kinase by AG1024 blocked the increase of tight-junction integrity. In addition, high-insulin/high-glucose treatment (as a model of hyperglycemia and hyperinsulinemia) synergistically reduced the tight-junction integrity in hCMEC/D3 cells, although either condition alone had little or no effect. Our findings suggest that, in addition to the established role of interactions of astrocytes and pericytes with brain capillary endothelial cells, insulin signaling from the blood side of the BBB contributes to maintenance of homeostasis by regulating cell proliferation and tight-junction integrity.
Collapse
Affiliation(s)
- Shingo Ito
- Department of Pharmaceutical Microbiology, Faculty of Life Sciences, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, Kumamoto 862-0973, Japan; Department of Pharmaceutical Microbiology, School of Pharmacy, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, Kumamoto 862-0973, Japan; Department of Pharmaceutical Microbiology, Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, Kumamoto 862-0973, Japan; AMED-CREST, Japan Agency for Medical Research and Development, 1-7-1 Otemachi, Chiyoda, Tokyo 100-0004, Japan
| | - Mari Yanai
- Department of Pharmaceutical Microbiology, School of Pharmacy, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, Kumamoto 862-0973, Japan
| | - Shunsuke Yamaguchi
- Department of Pharmaceutical Microbiology, Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, Kumamoto 862-0973, Japan
| | - Pierre-Olivier Couraud
- Institut Cochin, INSERM U1016, CNRS UMR8104, Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Sumio Ohtsuki
- Department of Pharmaceutical Microbiology, Faculty of Life Sciences, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, Kumamoto 862-0973, Japan; Department of Pharmaceutical Microbiology, School of Pharmacy, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, Kumamoto 862-0973, Japan; Department of Pharmaceutical Microbiology, Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, Kumamoto 862-0973, Japan; AMED-CREST, Japan Agency for Medical Research and Development, 1-7-1 Otemachi, Chiyoda, Tokyo 100-0004, Japan.
| |
Collapse
|
40
|
Guillemot-Legris O, Muccioli GG. Obesity-Induced Neuroinflammation: Beyond the Hypothalamus. Trends Neurosci 2017; 40:237-253. [PMID: 28318543 DOI: 10.1016/j.tins.2017.02.005] [Citation(s) in RCA: 370] [Impact Index Per Article: 46.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2016] [Revised: 02/15/2017] [Accepted: 02/17/2017] [Indexed: 12/21/2022]
|
41
|
Wang D, Wang X, Zhang P, Wang Y, Zhang R, Yan J, Zhou Z, Zhu W. The fate of technical-grade chlordane in mice fed a high-fat diet and its roles as a candidate obesogen. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2017; 222:532-542. [PMID: 28041837 DOI: 10.1016/j.envpol.2016.11.028] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Revised: 10/14/2016] [Accepted: 11/09/2016] [Indexed: 06/06/2023]
Abstract
Epidemiological studies indicate that exposure to persistent organic pollutants is positively associated with the prevalence of obesity. To delineate the potential role of technical-grade chlordane in obesity development, chlordane metabolism and chlordane-induced metabolic changes were investigated in mice fed high-fat diet (HFD) over a 6-week period. Gas chromatography-electron capture detector analysis showed that HFD induced more accumulation of technical chlordane in the liver, muscle and adipose tissue. The enantioselectivities of oxychlordane in selected tissues were also influenced by HFD. 1H NMR-based liver metabolome indicated that technical chlordane can enhance the metabolic alterations induced by HFD. Compared with the low-fat diet (LFD) group, no differences were observed in the LFD + chlordane group. However, as many as 16 metabolites were significantly different between the HFD group and HFD + chlordane group. Moreover, compared to the LFD + chlordane group, the abundances of 24 metabolites significantly increased or decreased in the HFD + chlordane group. Twenty metabolites were altered in the HFD group compared to the LFD group. Tryptophan profiling suggested that both chlordane and HFD can disturb tryptophan catabolism. These interactions between technical chlordane and HFD suggest that technical chlordane is a candidate obesogen.
Collapse
Affiliation(s)
- Dezhen Wang
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Department of Applied Chemistry, China Agricultural University, Yuanmingyuan West Road 2, Beijing 100193, PR China
| | - Xinru Wang
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Department of Applied Chemistry, China Agricultural University, Yuanmingyuan West Road 2, Beijing 100193, PR China
| | - Ping Zhang
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Department of Applied Chemistry, China Agricultural University, Yuanmingyuan West Road 2, Beijing 100193, PR China
| | - Yao Wang
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Department of Applied Chemistry, China Agricultural University, Yuanmingyuan West Road 2, Beijing 100193, PR China
| | - Renke Zhang
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Department of Applied Chemistry, China Agricultural University, Yuanmingyuan West Road 2, Beijing 100193, PR China
| | - Jin Yan
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Department of Applied Chemistry, China Agricultural University, Yuanmingyuan West Road 2, Beijing 100193, PR China
| | - Zhiqiang Zhou
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Department of Applied Chemistry, China Agricultural University, Yuanmingyuan West Road 2, Beijing 100193, PR China
| | - Wentao Zhu
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Department of Applied Chemistry, China Agricultural University, Yuanmingyuan West Road 2, Beijing 100193, PR China.
| |
Collapse
|
42
|
Abdanipour A, Noori-Zadeh A, Mesbah-Namin SA, Bakhtiyari S, Nejatbakhsh R, Anarkooli IJ. Di-(2-ethylhexyl) Phthalate-Induced Hippocampus-Derived Neural Stem Cells Proliferation. CELL JOURNAL 2016; 19:166-172. [PMID: 28367427 PMCID: PMC5241513 DOI: 10.22074/cellj.2016.4862] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/09/2016] [Accepted: 06/08/2016] [Indexed: 12/13/2022]
Abstract
The brain and spinal cord have a limited capacity for self-repair under damaged conditions. One of the best options to overcome these limitations involves the use of phytochemicals as potential therapeutic agents. In this study, we have aimed to investigate the
effects of di-(2-ethylhexyl) phthalate (DEHP) on hippocampus-derived neural stem cells
(NSCs) proliferation to search phytochemical candidates for possible treatment of neurological diseases using endogenous capacity.
In this experimental study, neonatal rat hippocampus-derived NSCs were cultured and
treated with various concentrations of DEHP (0, 100, 200, 400 and 600 µM) and Cirsium
vulgare (C. vulgare) hydroethanolic extract (0, 200, 400, 600, 800 and 1000 µg/ml) for 48
hours under in vitro conditions. Cell proliferation rates and quantitative Sox2 gene expression were evaluated using MTT assay and real-time reverse transcription polymerase
chain reaction (RT-PCR).
We observed the highest average growth rate in the 400 µM DEHP and 800 µg/ml C.
vulgare extract treated groups. Sox2 expression in the DEHP-treated NSCs significantly
increased compared to the control group. Gas chromatography/mass spectrometry (GC/
MS) results demonstrated that the active ingredients that naturally occurred in the C. vulgare hydroethanolic extract were 2-ethyl-1-hexanamine, n-heptacosane, 1-cyclopentanecarboxylic acid, 1-heptadecanamine, 2,6-octadien-1-ol,2,6,10,14,18,22-tetracosahexaene, and DEHP. DEHP profoundly stimulated NSCs proliferation through Sox2 gene
overexpression.
These results provide and opportunity for further use of the C. vulgure phytochemicals for
prevention and/or treatment of neurological diseases via phytochemical mediated-proliferation of endogenous adult NSCs.
Collapse
Affiliation(s)
- Alireza Abdanipour
- Department of Anatomy, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Ali Noori-Zadeh
- Department of Clinical Biochemistry, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Seyed Alireza Mesbah-Namin
- Department of Clinical Biochemistry, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Salar Bakhtiyari
- Student Research Committee Center, Ilam University of Medical Sciences, Ilam, Iran
| | - Reza Nejatbakhsh
- Department of Anatomy, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Iraj Jafari Anarkooli
- Department of Anatomy, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| |
Collapse
|
43
|
Lam V, Hackett M, Takechi R. Antioxidants and Dementia Risk: Consideration through a Cerebrovascular Perspective. Nutrients 2016; 8:nu8120828. [PMID: 27999412 PMCID: PMC5188481 DOI: 10.3390/nu8120828] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Revised: 12/16/2016] [Accepted: 12/16/2016] [Indexed: 12/16/2022] Open
Abstract
A number of natural and chemical compounds that exert anti-oxidative properties are demonstrated to be beneficial for brain and cognitive function, and some are reported to reduce the risk of dementia. However, the detailed mechanisms by which those anti-oxidative compounds show positive effects on cognition and dementia are still unclear. An emerging body of evidence suggests that the integrity of the cerebrovascular blood-brain barrier (BBB) is centrally involved in the onset and progression of cognitive impairment and dementia. While recent studies revealed that some anti-oxidative agents appear to be protective against the disruption of BBB integrity and structure, few studies considered the neuroprotective effects of antioxidants in the context of cerebrovascular integrity. Therefore, in this review, we examine the mechanistic insights of antioxidants as a pleiotropic agent for cognitive impairment and dementia through a cerebrovascular axis by primarily focusing on the current available data from physiological studies. Conclusively, there is a compelling body of evidence that suggest antioxidants may prevent cognitive decline and dementia by protecting the integrity and function of BBB and, indeed, further studies are needed to directly examine these effects in addition to underlying molecular mechanisms.
Collapse
Affiliation(s)
- Virginie Lam
- Curtin Health Innovation Research Institute, Curtin University, Perth WA 6845, Australia.
- School of Public Health, Faculty of Health Sciences, Curtin University, Perth WA 6845, Australia.
| | - Mark Hackett
- Curtin Health Innovation Research Institute, Curtin University, Perth WA 6845, Australia.
- Department of Chemistry, Faculty of Science and Engineering, Curtin University, Perth WA 6845, Australia.
| | - Ryusuke Takechi
- Curtin Health Innovation Research Institute, Curtin University, Perth WA 6845, Australia.
- School of Public Health, Faculty of Health Sciences, Curtin University, Perth WA 6845, Australia.
| |
Collapse
|
44
|
Jeong SI, Shin JA, Cho S, Kim HW, Lee JY, Kang JL, Park EM. Resveratrol attenuates peripheral and brain inflammation and reduces ischemic brain injury in aged female mice. Neurobiol Aging 2016; 44:74-84. [PMID: 27318135 DOI: 10.1016/j.neurobiolaging.2016.04.007] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Revised: 02/05/2016] [Accepted: 04/12/2016] [Indexed: 10/21/2022]
Abstract
Resveratrol is known to improve metabolic dysfunction associated with obesity. Visceral obesity is a sign of aging and is considered a risk factor for ischemic stroke. In this study, we investigated the effects of resveratrol on inflammation in visceral adipose tissue and the brain and its effects on ischemic brain injury in aged female mice. Mice treated with resveratrol (0.1 mg/kg, p.o.) for 10 days showed reduced levels of interleukin-1β and tumor necrosis factor-α, as well as a reduction in the size of adipocytes in visceral adipose tissue. Resveratrol also reduced interleukin-1β and tumor necrosis factor-α protein levels and immunoglobulin G extravasation in the brain. Mice treated with resveratrol demonstrated smaller infarct size, improved neurological function, and blunted peripheral inflammation at 3 days postischemic stroke. These results showed that resveratrol counteracted inflammation in visceral adipose tissue and in the brain and reduced stroke-induced brain injury and peripheral inflammation in aged female mice. Therefore, resveratrol administration can be a valuable strategy for the prevention of age-associated and disease-provoked inflammation in postmenopausal women.
Collapse
Affiliation(s)
- Sae Im Jeong
- Department of Pharmacology, School of Medicine, Ewha Womans University, Seoul, Republic of Korea; Tissue Injury Defense Research Center, School of Medicine, Ewha Womans University, Seoul, Republic of Korea
| | - Jin A Shin
- Department of Pharmacology, School of Medicine, Ewha Womans University, Seoul, Republic of Korea; Tissue Injury Defense Research Center, School of Medicine, Ewha Womans University, Seoul, Republic of Korea
| | - Sunghee Cho
- Department of Neurology/Neuroscience, Brain & Mind Research Institute, Weill Cornell Medical College at Burke Medical Research Institute, White Plains, NY, USA
| | - Hye Won Kim
- Department of Pharmacology, School of Medicine, Ewha Womans University, Seoul, Republic of Korea; Tissue Injury Defense Research Center, School of Medicine, Ewha Womans University, Seoul, Republic of Korea
| | - Ji Yoon Lee
- School of Medicine, Ewha Womans University, Seoul, Republic of Korea
| | - Jihee Lee Kang
- Tissue Injury Defense Research Center, School of Medicine, Ewha Womans University, Seoul, Republic of Korea; Department of Physiology, School of Medicine, Ewha Womans University, Seoul, Republic of Korea
| | - Eun-Mi Park
- Department of Pharmacology, School of Medicine, Ewha Womans University, Seoul, Republic of Korea; Tissue Injury Defense Research Center, School of Medicine, Ewha Womans University, Seoul, Republic of Korea.
| |
Collapse
|
45
|
Akbar M, Essa MM, Daradkeh G, Abdelmegeed MA, Choi Y, Mahmood L, Song BJ. Mitochondrial dysfunction and cell death in neurodegenerative diseases through nitroxidative stress. Brain Res 2016; 1637:34-55. [PMID: 26883165 PMCID: PMC4821765 DOI: 10.1016/j.brainres.2016.02.016] [Citation(s) in RCA: 121] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2015] [Revised: 02/02/2016] [Accepted: 02/05/2016] [Indexed: 12/12/2022]
Abstract
Mitochondria are important for providing cellular energy ATP through the oxidative phosphorylation pathway. They are also critical in regulating many cellular functions including the fatty acid oxidation, the metabolism of glutamate and urea, the anti-oxidant defense, and the apoptosis pathway. Mitochondria are an important source of reactive oxygen species leaked from the electron transport chain while they are susceptible to oxidative damage, leading to mitochondrial dysfunction and tissue injury. In fact, impaired mitochondrial function is commonly observed in many types of neurodegenerative diseases, including Alzheimer's disease, Parkinson's disease, Huntington's disease, alcoholic dementia, brain ischemia-reperfusion related injury, and others, although many of these neurological disorders have unique etiological factors. Mitochondrial dysfunction under many pathological conditions is likely to be promoted by increased nitroxidative stress, which can stimulate post-translational modifications (PTMs) of mitochondrial proteins and/or oxidative damage to mitochondrial DNA and lipids. Furthermore, recent studies have demonstrated that various antioxidants, including naturally occurring flavonoids and polyphenols as well as synthetic compounds, can block the formation of reactive oxygen and/or nitrogen species, and thus ultimately prevent the PTMs of many proteins with improved disease conditions. Therefore, the present review is aimed to describe the recent research developments in the molecular mechanisms for mitochondrial dysfunction and tissue injury in neurodegenerative diseases and discuss translational research opportunities.
Collapse
Affiliation(s)
- Mohammed Akbar
- Section of Molecular Pharmacology and Toxicology, Laboratory of Membrane Biochemistry and Biophysics, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, USA
| | - Musthafa Mohamed Essa
- Department of Food Science and Nutrition, College of Agriculture and Marine Sciences, Sultan Qaboos University, Oman; Ageing and Dementia Research Group, Sultan Qaboos University, Oman
| | - Ghazi Daradkeh
- Department of Food Science and Nutrition, College of Agriculture and Marine Sciences, Sultan Qaboos University, Oman
| | - Mohamed A Abdelmegeed
- Section of Molecular Pharmacology and Toxicology, Laboratory of Membrane Biochemistry and Biophysics, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, USA
| | - Youngshim Choi
- Section of Molecular Pharmacology and Toxicology, Laboratory of Membrane Biochemistry and Biophysics, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, USA
| | - Lubna Mahmood
- Department of Nutritional Sciences, Qatar University, Qatar
| | - Byoung-Joon Song
- Section of Molecular Pharmacology and Toxicology, Laboratory of Membrane Biochemistry and Biophysics, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
46
|
Rijnsburger M, Belegri E, Eggels L, Unmehopa UA, Boelen A, Serlie MJ, la Fleur SE. The effect of diet interventions on hypothalamic nutrient sensing pathways in rodents. Physiol Behav 2016; 162:61-8. [PMID: 27083123 DOI: 10.1016/j.physbeh.2016.04.011] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Revised: 03/25/2016] [Accepted: 04/07/2016] [Indexed: 12/13/2022]
Abstract
The hypothalamus plays a fundamental role in regulating homeostatic processes including regulation of food intake. Food intake is driven in part by energy balance, which is sensed by specific brain structures through signaling molecules such as nutrients and hormones. Both circulating glucose and fatty acids decrease food intake via a central mechanism involving the hypothalamus and brain stem. Besides playing a role in signaling energy status, glucose and fatty acids serve as fuel for neurons. This review focuses on the effects of glucose and fatty acids on hypothalamic pathways involved in regulation of energy metabolism as well as on the role of the family of peroxisome proliferator activated receptors (PPARs) which are implicated in regulation of central energy homeostasis. We further discuss the effects of different hypercaloric diets on these pathways.
Collapse
Affiliation(s)
- Merel Rijnsburger
- Department of Endocrinology & Metabolism, Academic Medical Center, Amsterdam, The Netherlands
| | - Evita Belegri
- Department of Endocrinology & Metabolism, Academic Medical Center, Amsterdam, The Netherlands
| | - Leslie Eggels
- Department of Endocrinology & Metabolism, Academic Medical Center, Amsterdam, The Netherlands
| | - Unga A Unmehopa
- Department of Endocrinology & Metabolism, Academic Medical Center, Amsterdam, The Netherlands
| | - Anita Boelen
- Department of Endocrinology & Metabolism, Academic Medical Center, Amsterdam, The Netherlands
| | - Mireille J Serlie
- Department of Endocrinology & Metabolism, Academic Medical Center, Amsterdam, The Netherlands
| | - Susanne E la Fleur
- Department of Endocrinology & Metabolism, Academic Medical Center, Amsterdam, The Netherlands.
| |
Collapse
|
47
|
Chen JT, Chen TG, Chang YC, Chen CY, Chen RM. Roles of NMDARs in maintenance of the mouse cerebrovascular endothelial cell-constructed tight junction barrier. Toxicology 2016; 339:40-50. [DOI: 10.1016/j.tox.2015.11.006] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Revised: 11/18/2015] [Accepted: 11/28/2015] [Indexed: 10/22/2022]
|
48
|
Chio CC, Wei L, Chen TG, Lin CM, Shieh JP, Yeh PS, Chen RM. Neuron-derived orphan receptor 1 transduces survival signals in neuronal cells in response to hypoxia-induced apoptotic insults. J Neurosurg 2015; 124:1654-64. [PMID: 26613168 DOI: 10.3171/2015.6.jns1535] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
OBJECT Hypoxia can induce cell death or trigger adaptive mechanisms to guarantee cell survival. Neuron-derived orphan receptor 1 (NOR-1) works as an early-response protein in response to a variety of environmental stresses. In this study, the authors evaluated the roles of NOR-1 in hypoxia-induced neuronal insults. METHODS Neuro-2a cells were exposed to oxygen/glucose deprivation (OGD). Cell viability, cell morphology, cas-pase-3 activity, DNA fragmentation, and cell apoptosis were assayed to determine the mechanisms of OGD-induced neuronal insults. RNA and protein analyses were carried out to evaluate the effects of OGD on expressions of NOR-1, cAMP response element-binding (CREB), and cellular inhibitor of apoptosis protein 2 (cIAP2) genes. Translations of these gene expressions were knocked down using RNA interference. Mice subjected to traumatic brain injury (TBI) and NOR-1 was immunodetected. RESULTS Exposure of neuro-2a cells to OGD decreased cell viability in a time-dependent manner. Additionally, OGD led to cell shrinkage, DNA fragmentation, and cell apoptosis. In parallel, treatment of neuro-2a cells with OGD time dependently increased cellular NOR-1 mRNA and protein expressions. Interestingly, administration of TBI also augmented NOR-1 levels in the impacted regions of mice. As to the mechanism, exposure to OGD increased nuclear levels of the transcription factor CREB protein. Downregulating CREB expression using RNA interference simultaneously inhibited OGD-induced NOR-1 mRNA expression. Also, levels of cIAP2 mRNA and protein in neuro-2a cells were augmented by OGD. After reducing cIAP2 translation, OGD-induced cell death was reduced. Sequentially, application of NOR-1 small interfering RNA to neuro-2a cells significantly inhibited OGD-induced cIAP2 mRNA expression and concurrently alleviated hypoxia-induced alterations in cell viability, caspase-3 activation, DNA damage, and cell apoptosis. CONCLUSIONS This study shows that NOR-1 can transduce survival signals in neuronal cells responsible for hypoxiainduced apoptotic insults through activation of a CREB/cIAP2-dependent mechanism.
Collapse
Affiliation(s)
| | - Li Wei
- Departments of 3 Neurosugery and
| | | | | | | | | | - Ruei-Ming Chen
- Comprehensive Cancer Center.,Graduate Institute of Medical Sciences, Taipei Medical University; ,Brain Disease Research Center, Wan-Fang Hospital, Taipei Medical University; and.,Anesthetics and Toxicology Research Center, Taipei Medical University Hospital, Taipei, Taiwan
| |
Collapse
|
49
|
Phytochemical Compounds and Protection from Cardiovascular Diseases: A State of the Art. BIOMED RESEARCH INTERNATIONAL 2015; 2015:918069. [PMID: 26504846 PMCID: PMC4609427 DOI: 10.1155/2015/918069] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/18/2015] [Accepted: 06/14/2015] [Indexed: 02/07/2023]
Abstract
Cardiovascular diseases represent a worldwide relevant socioeconomical problem. Cardiovascular disease prevention relies also on lifestyle changes, including dietary habits. The cardioprotective effects of several foods and dietary supplements in both animal models and in humans have been explored. It was found that beneficial effects are mainly dependent on antioxidant and anti-inflammatory properties, also involving modulation of mitochondrial function. Resveratrol is one of the most studied phytochemical compounds and it is provided with several benefits in cardiovascular diseases as well as in other pathological conditions (such as cancer). Other relevant compounds are Brassica oleracea, curcumin, and berberine, and they all exert beneficial effects in several diseases. In the attempt to provide a comprehensive reference tool for both researchers and clinicians, we summarized in the present paper the existing literature on both preclinical and clinical cardioprotective effects of each mentioned phytochemical. We structured the discussion of each compound by analyzing, first, its cellular molecular targets of action, subsequently focusing on results from applications in both ex vivo and in vivo models, finally discussing the relevance of the compound in the context of human diseases.
Collapse
|
50
|
Ho MH, Yao CJ, Liao MH, Lin PI, Liu SH, Chen RM. Chitosan nanofiber scaffold improves bone healing via stimulating trabecular bone production due to upregulation of the Runx2/osteocalcin/alkaline phosphatase signaling pathway. Int J Nanomedicine 2015; 10:5941-54. [PMID: 26451104 PMCID: PMC4590342 DOI: 10.2147/ijn.s90669] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Osteoblasts play critical roles in bone formation. Our previous study showed that chitosan nanofibers can stimulate osteoblast proliferation and maturation. This translational study used an animal model of bone defects to evaluate the effects of chitosan nanofiber scaffolds on bone healing and the possible mechanisms. In this study, we produced uniform chitosan nanofibers with fiber diameters of approximately 200 nm. A bone defect was surgically created in the proximal femurs of male C57LB/6 mice, and then the left femur was implanted with chitosan nanofiber scaffolds for 21 days and compared with the right femur, which served as a control. Histological analyses revealed that implantation of chitosan nanofiber scaffolds did not lead to hepatotoxicity or nephrotoxicity. Instead, imaging analyses by X-ray transmission and microcomputed tomography showed that implantation of chitosan nanofiber scaffolds improved bone healing compared with the control group. In parallel, microcomputed tomography and bone histomorphometric assays further demonstrated augmentation of the production of new trabecular bone in the chitosan nanofiber-treated group. Furthermore, implantation of chitosan nanofiber scaffolds led to a significant increase in the trabecular bone thickness but a reduction in the trabecular parameter factor. As to the mechanisms, analysis by confocal microscopy showed that implantation of chitosan nanofiber scaffolds increased levels of Runt-related transcription factor 2 (Runx2), a key transcription factor that regulates osteogenesis, in the bone defect sites. Successively, amounts of alkaline phosphatase and osteocalcin, two typical biomarkers that can simulate bone maturation, were augmented following implantation of chitosan nanofiber scaffolds. Taken together, this translational study showed a beneficial effect of chitosan nanofiber scaffolds on bone healing through stimulating trabecular bone production due to upregulation of Runx2-mediated alkaline phosphatase and osteocalcin gene expressions. Our results suggest the potential of chitosan nanofiber scaffolds for therapy of bone diseases, including bone defects and bone fractures.
Collapse
Affiliation(s)
- Ming-Hua Ho
- Department of Chemical Engineering, National Taiwan University of Science and Technology, Taipei, Taiwan ; Cell Physiology and Molecular Image Research Center, Taipei Medical University-Wan Fang Hospital, Taipei, Taiwan
| | - Chih-Jung Yao
- Department of Internal Medicine, School of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Mei-Hsiu Liao
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Pei-I Lin
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Shing-Hwa Liu
- Institute of Toxicology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Ruei-Ming Chen
- Cell Physiology and Molecular Image Research Center, Taipei Medical University-Wan Fang Hospital, Taipei, Taiwan ; Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan ; Anesthetics and Toxicology Research Center, Taipei Medical University Hospital, Taipei, Taiwan
| |
Collapse
|