1
|
Darwish R, Alcibahy Y, Bucheeri S, Albishtawi A, Tama M, Shetty J, Butler AE. The Role of Hypothalamic Microglia in the Onset of Insulin Resistance and Type 2 Diabetes: A Neuro-Immune Perspective. Int J Mol Sci 2024; 25:13169. [PMID: 39684879 DOI: 10.3390/ijms252313169] [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: 11/11/2024] [Revised: 12/05/2024] [Accepted: 12/05/2024] [Indexed: 12/18/2024] Open
Abstract
Historically, microglial activation has been associated with diseases of a neurodegenerative and neuroinflammatory nature. Some, like Alzheimer's disease, Parkinson's disease, and multiple system atrophy, have been explored extensively, while others pertaining to metabolism not so much. However, emerging evidence points to hypothalamic inflammation mediated by microglia as a driver of metabolic dysregulations, particularly insulin resistance and type 2 diabetes mellitus. Here, we explore this connection further and examine pathways that underlie this relationship, including the IKKβ/NF-κβ, IRS-1/PI3K/Akt, mTOR-S6 Kinase, JAK/STAT, and PPAR-γ signaling pathways. We also investigate the role of non-coding RNAs, namely microRNAs and long non-coding RNAs, in insulin resistance related to neuroinflammation and their diagnostic and therapeutic potential. Finally, we explore therapeutics further, searching for both pharmacological and non-pharmacological interventions that can help mitigate microglial activation.
Collapse
Affiliation(s)
- Radwan Darwish
- School of Medicine, Royal College of Surgeons in Ireland-Medical University of Bahrain (RCSI-MUB), Busaiteen 228, Bahrain
| | - Yasmine Alcibahy
- School of Medicine, Royal College of Surgeons in Ireland-Medical University of Bahrain (RCSI-MUB), Busaiteen 228, Bahrain
| | - Shahd Bucheeri
- School of Medicine, Royal College of Surgeons in Ireland-Medical University of Bahrain (RCSI-MUB), Busaiteen 228, Bahrain
| | - Ashraf Albishtawi
- School of Medicine, Royal College of Surgeons in Ireland-Medical University of Bahrain (RCSI-MUB), Busaiteen 228, Bahrain
| | - Maya Tama
- School of Medicine, Royal College of Surgeons in Ireland-Medical University of Bahrain (RCSI-MUB), Busaiteen 228, Bahrain
| | - Jeevan Shetty
- Department of Biochemistry, Royal College of Surgeons in Ireland-Medical University of Bahrain (RCSI-MUB), Busaiteen 228, Bahrain
| | - Alexandra E Butler
- School of Postgraduate Studies and Research, Royal College of Surgeons in Ireland-Medical University of Bahrain (RCSI-MUB), Busaiteen 228, Bahrain
| |
Collapse
|
2
|
Sharma N, An SSA, Kim SY. Medication Exposure and Risk of Dementia and Alzheimer's Disease. Int J Mol Sci 2024; 25:12850. [PMID: 39684561 DOI: 10.3390/ijms252312850] [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: 11/04/2024] [Revised: 11/19/2024] [Accepted: 11/26/2024] [Indexed: 12/18/2024] Open
Abstract
Alzheimer's disease (AD), a complex neurodegenerative disease (ND), is the most predominant cause of dementia among the elderly. Generally, elderly people have multiple chronic health conditions, like hypertension, arthritis, diabetes, insomnia, bowel problems, and depression. Although prescribed medications have beneficial therapeutic compositions, some may have side effects that could hinder cognitive function or worsen cognitive decline. Hence, we should evaluate those medications to guarantee their safety. In the present mechanistic review, we discussed frequently used categories of medication (analgesics, anticholinergics, benzodiazepines, proton pump inhibitors, and statins), concerning their possible involvement in increasing AD and dementia risks. This review summarized the results of various observational studies, meta-analyses, randomized case-control studies, and systematic reviews. As the results were contradictory, it was difficult to ascertain the clear associations between medication usage and increased risks of dementia or AD. The blood-based biomarkers (BBMs) offer a low-cost and accessible alternative for early diagnosis of AD. Systematic reviews combined with meta-analysis would be crucial tools for accurately assessing and summarizing the efficacy of health interventions, yet randomized clinical trials have always been the best way to help with clinical care decisions. Thus, an open discussion is necessary to help individuals determine whether the advantages of utilizing medications outweigh the possible drawbacks.
Collapse
Affiliation(s)
- Niti Sharma
- Bionano Research Institute, Gachon University, 1342 Seongnam-daero, Sujeong-gu, Seongnam-si 13120, Republic of Korea
| | - Seong Soo A An
- Bionano Research Institute, Gachon University, 1342 Seongnam-daero, Sujeong-gu, Seongnam-si 13120, Republic of Korea
| | - Sang Yun Kim
- Department of Neurology, Seoul National University Bundang Hospital & Seoul National University College of Medicine, 82, Gumi-ro 173 Beon-gil, Bundang-gu, Seongnam-si 13620, Republic of Korea
| |
Collapse
|
3
|
Fang M, Yu Q, Ou J, Lou J, Zhu J, Lin Z. The Neuroprotective Mechanisms of PPAR-γ: Inhibition of Microglia-Mediated Neuroinflammation and Oxidative Stress in a Neonatal Mouse Model of Hypoxic-Ischemic White Matter Injury. CNS Neurosci Ther 2024; 30:e70081. [PMID: 39496476 PMCID: PMC11534457 DOI: 10.1111/cns.70081] [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: 07/26/2024] [Revised: 09/12/2024] [Accepted: 09/22/2024] [Indexed: 11/06/2024] Open
Abstract
BACKGROUND Neuroinflammation and oxidative stress, mediated by microglial activation, hinder the development of oligodendrocytes (OLs) and delay myelination in preterm infants, leading to white matter injury (WMI) and long-term neurodevelopmental sequelae. Peroxisome proliferator-activated receptor gamma (PPAR-γ) has been reported to inhibit inflammation and oxidative stress via modulating microglial polarization in various central nervous system diseases. However, the relationship between PPAR-γ and microglial polarization in neonatal WMI is not well understood. Therefore, this study aimed to elucidate the role and mechanisms of PPAR-γ in preterm infants affected by WMI. METHODS In this study, an in vivo hypoxia-ischemia (HI) induced brain WMI neonatal mouse model was established. The mice were administered intraperitoneally with either RSGI or GW9662 to activate or inhibit PPAR-γ, respectively. Additionally, an in vitro oxygen-glucose deprivation (OGD) cell model was established and pretreated with pcDNA 3.1-PPAR-γ or si-PPAR-γ to overexpress or silence PPAR-γ, respectively. The neuroprotective effects of PPAR-γ were investigated in vivo. Firstly, open field test, novel object recognization test, and beam-walking test were employed to assess the effects of PPAR-γ on neurobehavioral recovery. Furthermore, assessment of OLs loss and OL-maturation disorder, the number of myelinated axons, myelin thickness, synaptic deficit, activation of microglia and astrocyte, and blood-brain barrier (BBB) were used to evaluate the effects of PPAR-γ on pathological repair. The mechanisms of PPAR-γ were explored both in vivo and in vitro. Assessment of microglia polarization, inflammatory mediators, reactive oxygen species (ROS), MDA, and antioxidant enzymes was used to evaluate the anti-inflammatory and antioxidative effects of PPAR-γ activation. An assessment of HMGB1/NF-κB and NRF2/KEAP1 signaling pathway was conducted to clarify the mechanisms by which PPAR-γ influences HI-induced WMI in neonatal mice. RESULTS Activation of PPAR-γ using RSGI significantly mitigated BBB disruption, promoted M2 polarization of microglia, inhibited activation of microglia and astrocytes, promoted OLs development, and enhanced myelination in HI-induced WMI. Conversely, inhibition of PPAR-γ using GW9662 further exacerbated the pathologic hallmark of WMI. Neurobehavioral tests revealed that neurological deficits were ameliorated by RSGI, while further aggravated by GW91662. In addition, activation of PPAR-γ significantly alleviated neuroinflammation and oxidative stress by suppressing HMGB1/NF-κB signaling pathway and activating NRF2 signaling pathway both in vivo and in vitro. Conversely, inhibition of PPAR-γ further exacerbated HI or OGD-induced neuroinflammation, oxidative stress via modulation of the same signaling pathway. CONCLUSIONS Our findings suggest that PPAR-γ regulates microglial activation/polarization as well as subsequent neuroinflammation/oxidative stress via the HMGB1/NF-κB and NRF2/KEAP1 signaling pathway, thereby contributing to neuroprotection and amelioration of HI-induced WMI in neonatal mice.
Collapse
Affiliation(s)
- Mingchu Fang
- Department of NeonatologyThe Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical UniversityWenzhouZhejiangChina
- The Second School of MedicineWenzhou Medical UniversityWenzhouZhejiangChina
- Key Laboratory of Perinatal Medicine of WenzhouWenzhouZhejiangChina
- Key Laboratory of Structural Malformations in Children of Zhejiang ProvinceWenzhouZhejiangChina
| | - Qianqian Yu
- Department of NeonatologyThe Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical UniversityWenzhouZhejiangChina
- The Second School of MedicineWenzhou Medical UniversityWenzhouZhejiangChina
| | - Jiahao Ou
- Department of NeonatologyThe Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical UniversityWenzhouZhejiangChina
- The Second School of MedicineWenzhou Medical UniversityWenzhouZhejiangChina
| | - Jia Lou
- Department of NeonatologyThe Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical UniversityWenzhouZhejiangChina
- The Second School of MedicineWenzhou Medical UniversityWenzhouZhejiangChina
| | - Jianghu Zhu
- Department of NeonatologyThe Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical UniversityWenzhouZhejiangChina
- The Second School of MedicineWenzhou Medical UniversityWenzhouZhejiangChina
- Key Laboratory of Perinatal Medicine of WenzhouWenzhouZhejiangChina
- Key Laboratory of Structural Malformations in Children of Zhejiang ProvinceWenzhouZhejiangChina
| | - Zhenlang Lin
- Department of NeonatologyThe Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical UniversityWenzhouZhejiangChina
- The Second School of MedicineWenzhou Medical UniversityWenzhouZhejiangChina
- Key Laboratory of Perinatal Medicine of WenzhouWenzhouZhejiangChina
- Key Laboratory of Structural Malformations in Children of Zhejiang ProvinceWenzhouZhejiangChina
| |
Collapse
|
4
|
Mishra SK, Mishra V. Saroglitazar Enhances Memory Functions and Adult Neurogenesis via Up-Regulation of Wnt/β Catenin Signaling in the Rat Model of Dementia. ACS Chem Neurosci 2024; 15:3449-3458. [PMID: 39265183 DOI: 10.1021/acschemneuro.4c00167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/14/2024] Open
Abstract
Peroxisome proliferator-activated receptors (PPARs) have emerged as a promising target for the treatment of various neurodegenerative disorders. Studies have shown that both PPAR α & γ individually modulate various pathophysiological events like neuroinflammation and insulin resistance, which are known to variedly affect neurogenesis. Our study aimed to evaluate the effect of saroglitazar (SGZR), a dual PPAR agonist, on adult neurogenesis and spatial learning and memory, in intracerebroventricular streptozotocin (ICV STZ)-induced dementia in rats. We have found that SGZR at the dose of 4 mg/kg per oral showed significant improvement in learning and memory compared to ICV STZ-treated rats. A substantial increase in neurogenesis was observed in the subventricular zone (SVZ) and the dentate gyrus (DG), as indicated by an increase in the number of 5-bromo-2'-deoxyuridine (BrdU)+ cells, BrdU+ nestin+ cells, and doublecortin (DCX)+cells. Treatment with SGZR also decreased the active form of glycogen synthase kinase 3β (GSK3β) and hence enhanced the nuclear translocation of the β-catenin. Enhanced expression of Wnt transcription factors and target genes indicates that the up-regulation of Wnt signaling might be involved in the observed increase in neurogenesis. Hence, it can be concluded that the SGZR enhances memory functions and adult neurogenesis via the upregulation of Wnt β-catenin signaling in ICV STZ-treated rats.
Collapse
Affiliation(s)
- Sandeep Kumar Mishra
- Division of Pharmacology, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh 226031, India
- Faculty of Pharmacy, Kalinga University, Raipur, Chhattisgarh 492101, India
| | - Vaibhav Mishra
- Amity Institute of Microbial Technology, Amity University, Noida, Uttar Pradesh 201313, India
| |
Collapse
|
5
|
Ríos JA, Bórquez JC, Godoy JA, Zolezzi JM, Furrianca MC, Inestrosa NC. Emerging role of Metformin in Alzheimer's disease: A translational view. Ageing Res Rev 2024; 100:102439. [PMID: 39074563 DOI: 10.1016/j.arr.2024.102439] [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: 05/09/2024] [Revised: 07/23/2024] [Accepted: 07/23/2024] [Indexed: 07/31/2024]
Abstract
Alzheimer's disease (AD) constitutes a major public-health issue of our time. Regrettably, despite our considerable understanding of the pathophysiological aspects of this disease, current interventions lead to poor outcomes. Furthermore, experimentally promising compounds have continuously failed when translated to clinical trials. Along with increased population ageing, Type 2 Diabetes Mellitus (T2DM) has become an extremely common condition, mainly due to unbalanced dietary habits. Substantial epidemiological evidence correlates T2DM with cognitive impairment as well. Considering that brain insulin resistance, mitochondrial dysfunction, oxidative stress, and amyloidogenesis are common phenomena, further approaching the common features among these pathological conditions. Metformin constitutes the first-choice drug to preclude insulin resistance in T2DM clinical management. Experimental evidence suggests that its functions might include neuroprotective effects, in addition to its hypoglycemic activity. This review aims to summarize and discuss current knowledge of experimental data on metformin on this path towards translational medicine. Finally, we discuss the controversial data of responses to metformin in vitro, and in vivo, animal models and human studies.
Collapse
Affiliation(s)
- Juvenal A Ríos
- Facultad de Medicina y Ciencia, Escuela de Medicina, Universidad San Sebastián, Santiago, Chile
| | - Juan Carlos Bórquez
- Centro de Excelencia en Biomedicina de Magallanes (CEBIMA), Escuela de Medicina, Universidad de Magallanes, Punta Arenas, Chile; Facultad de Ciencias de la Salud, Universidad de Magallanes, Punta Arenas, Chile
| | - Juan A Godoy
- Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Juan M Zolezzi
- Centro de Excelencia en Biomedicina de Magallanes (CEBIMA), Escuela de Medicina, Universidad de Magallanes, Punta Arenas, Chile
| | | | - Nibaldo C Inestrosa
- Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile; Centro de Excelencia en Biomedicina de Magallanes (CEBIMA), Escuela de Medicina, Universidad de Magallanes, Punta Arenas, Chile.
| |
Collapse
|
6
|
Baghcheghi Y, Razazpour F, Seyedi F, Arefinia N, Hedayati-Moghadam M. Exploring the molecular mechanisms of PPARγ agonists in modulating memory impairment in neurodegenerative disorders. Mol Biol Rep 2024; 51:945. [PMID: 39215798 DOI: 10.1007/s11033-024-09850-6] [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: 05/06/2024] [Accepted: 08/08/2024] [Indexed: 09/04/2024]
Abstract
Neurodegenerative diseases are characterized by progressive memory impairment and cognitive decline. This review aims to unravel the molecular mechanisms involved in the enhancement of memory function and mitigation of memory impairment through the activation of PPARγ agonists in neurodegenerative diseases. The findings suggest that PPARγ agonists modulate various molecular pathways involved in memory formation and maintenance. Activation of PPARγ enhances synaptic plasticity, promotes neuroprotection, suppresses neuroinflammation, attenuates oxidative stress, and regulates amyloid-beta metabolism. The comprehensive understanding of these molecular mechanisms would facilitate the development of novel therapeutic approaches targeting PPARγ to improve memory function and ultimately to alleviate the burden of neurodegenerative diseases. Further research, including clinical trials, is warranted to explore the efficacy, safety, and optimal use of specific PPARγ agonists as potential therapeutic agents in the treatment of memory impairments associated with neurodegenerative diseases.
Collapse
Affiliation(s)
- Yousef Baghcheghi
- Bio Environmental Health Hazards Research Center, Jiroft University of Medical Sciences, Jiroft, Iran
- Student Research Committee, Jiroft University of Medical Sciences, Jiroft, Iran
| | - Fateme Razazpour
- Student Research Committee, Jiroft University of Medical Sciences, Jiroft, Iran
| | - Fatemeh Seyedi
- Department of Anatomical Sciences, School of Medicine, Jiroft University of Medical Sciences, Jiroft, Iran
| | - Nasir Arefinia
- Bio Environmental Health Hazards Research Center, Jiroft University of Medical Sciences, Jiroft, Iran
- Student Research Committee, Jiroft University of Medical Sciences, Jiroft, Iran
| | - Mahdiyeh Hedayati-Moghadam
- Student Research Committee, Jiroft University of Medical Sciences, Jiroft, Iran.
- Department of Physiology, School of Medicine, Jiroft University of Medical Sciences, Jiroft, Iran.
| |
Collapse
|
7
|
Guo R, Shen X, Ealing J, Zhou J, Lu J, Ning Y. Efficacy and safety of acupuncture for cognitive impairment in Alzheimer's disease: a systematic review and meta-analysis. FRONTIERS IN DEMENTIA 2024; 3:1380221. [PMID: 39081600 PMCID: PMC11285646 DOI: 10.3389/frdem.2024.1380221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Accepted: 04/22/2024] [Indexed: 08/02/2024]
Abstract
Objective To systematically evaluate the efficacy of acupuncture in the treatment of cognitive impairment in Alzheimer's disease (AD) by meta-analysis, in order to provide evidence-based evidence for the application of acupuncture therapy in the clinical process of AD. Methods From the establishment of the database to December 31, 2022, China Biomedical Literature Database (CBM), China National Knowledge Network (CNKI), VIP database, WanFang Database, Pubmed, Embase and Cochrane Library Database were systematically searched. To collect published randomized controlled clinical trials (RCTS) of acupuncture in the treatment of cognitive impairment in AD. The subjects in the intervention group were given acupuncture alone or combined with other treatments the same as the control group; the control group received conventional Western medicine treatment. The main outcome indicators of the study were cognitive function assessment of subjects, including: Simple Mental State Examination Scale (MMSE), Assessment of daily Living Ability Scale (ADL), Alzheimer's Disease Cognitive Function Assessment Scale (ADAS-Cog), TCM syndrome score (SDSD), Montreal Cognitive Test (MoCA), Secondary outcome indicators were the occurrence of adverse reactions. Literature screening, data extraction, and quality evaluation of the included literature were performed independently by two researchers, according to bias risk assessment tools recommended in the Cochrane manual. Data were analyzed by RevMan5.3 software. Dichotomous variables were represented by risk ratio (OR) and 95% CI, and continuity variables were represented by mean difference (MD) and 95% CI. For heterogeneity analysis, when P > 0.1 and I 2 ≤ 50%, fixed effect model was applied. When P ≤ 0.1 and I 2 > 50%, the random effects model is applied. Results A total of 1,172 eligible subjects were included in 18 RCTS, including 595 in the intervention group and 577 in the control group. The results of meta-analysis are as follows: acupuncture intervention group improved MMSE [MD = 1.67, 95% CI (0.94, 2.41), P < 0.00001], ADL [MD = -1.18, 95% CI (-3.09, 0.72), P = 0.22], ADAS-Cog [MD = 3.31, 95% CI (5.84, 0.78), P = 0.01], SDSD [MD = 2.40, 95% CI (3.53, 1.26), P < 0.0001], MoCA [MD = 4.80, 95% CI (3.74, 5.86), P = 0.04] were better than the control group. No serious adverse reactions related to acupuncture were observed in the intervention group, and the incidence and severity of adverse reactions were lower than those in the control group, with statistical significance [OR = 0.17, 95% CI (0.04, 0.67), P = 0.01]. Conclusion Existing data show that acupuncture therapy has certain advantages in improving cognitive dysfunction and improving self-care ability of patients with Alzheimer's disease. However, due to the small number of RCTS and cases evaluating the efficacy of acupuncture, and the possibility of measurement bias and selectivity bias in included studies, it is still unable to conduct high-intensity demonstration on its effectiveness. Further large-scale, high-quality randomized, double-blind controlled trials are needed to evaluate its efficacy. Systematic Review Registration https://inplasy.com/inplasy-2021-12-0125/, identifier: INPLASY2021120125.
Collapse
Affiliation(s)
- Ruyue Guo
- Department of Encephalopathy, The First Affiliated Hospital of Henan University of Traditional Chinese Medicine, Zhengzhou, China
- The First Clinical Medical College of Henan University of Chinese Medicine, Zhengzhou, China
| | - Xiaoming Shen
- The First Clinical Medical College of Henan University of Chinese Medicine, Zhengzhou, China
| | - John Ealing
- Salford Royal Hospital, Salford, United Kingdom
| | - Jiao Zhou
- Department of Encephalopathy, The First Affiliated Hospital of Henan University of Traditional Chinese Medicine, Zhengzhou, China
- The First Clinical Medical College of Henan University of Chinese Medicine, Zhengzhou, China
| | - Jin Lu
- Department of Encephalopathy, The First Affiliated Hospital of Henan University of Traditional Chinese Medicine, Zhengzhou, China
- The First Clinical Medical College of Henan University of Chinese Medicine, Zhengzhou, China
| | - Yunfan Ning
- Department of Encephalopathy, The First Affiliated Hospital of Henan University of Traditional Chinese Medicine, Zhengzhou, China
- The First Clinical Medical College of Henan University of Chinese Medicine, Zhengzhou, China
| |
Collapse
|
8
|
AmeliMojarad M, AmeliMojarad M. The neuroinflammatory role of microglia in Alzheimer's disease and their associated therapeutic targets. CNS Neurosci Ther 2024; 30:e14856. [PMID: 39031970 PMCID: PMC11259573 DOI: 10.1111/cns.14856] [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: 04/30/2024] [Revised: 06/17/2024] [Accepted: 07/02/2024] [Indexed: 07/22/2024] Open
Abstract
INTRODUCTION Alzheimer's disease (AD), the main cause of dementia, is characterized by synaptic loss and neurodegeneration. Amyloid-β (Aβ) accumulation, hyperphosphorylation of tau protein, and neurofibrillary tangles (NFTs) in the brain are considered to be the initiating factors of AD. However, this hypothesis falls short of explaining many aspects of AD pathogenesis. Recently, there has been mounting evidence that neuroinflammation plays a key role in the pathophysiology of AD and causes neurodegeneration by over-activating microglia and releasing inflammatory mediators. METHODS PubMed, Web of Science, EMBASE, and MEDLINE were used for searching and summarizing all the recent publications related to inflammation and its association with Alzheimer's disease. RESULTS Our review shows how inflammatory dysregulation influences AD pathology as well as the roles of microglia in neuroinflammation, the possible microglia-associated therapeutic targets, top neuroinflammatory biomarkers, and anti-inflammatory drugs that combat inflammation. CONCLUSION In conclusion, microglial inflammatory reactions are important factors in AD pathogenesis and need to be discussed in more detail for promising therapeutic strategies.
Collapse
Affiliation(s)
- Melika AmeliMojarad
- Department of Bioprocess Engineering, Institute of Industrial and Environmental BiotechnologyNational Institute of Genetic Engineering and BiotechnologyTehranIran
| | - Mandana AmeliMojarad
- Department of Bioprocess Engineering, Institute of Industrial and Environmental BiotechnologyNational Institute of Genetic Engineering and BiotechnologyTehranIran
| |
Collapse
|
9
|
Steinke I, Singh M, Amin R. Dual PPAR delta/gamma agonists offer therapeutic potential for Alzheimer's disease. Neural Regen Res 2024; 19:1175-1176. [PMID: 37905851 PMCID: PMC11467939 DOI: 10.4103/1673-5374.386410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 08/15/2023] [Accepted: 08/26/2023] [Indexed: 11/02/2023] Open
Affiliation(s)
- Ian Steinke
- Department of Drug Discovery and Development, Auburn University, Auburn Alabama, AL, USA
| | - Meenakshi Singh
- Department of Drug Discovery and Development, Auburn University, Auburn Alabama, AL, USA
| | - Rajesh Amin
- Department of Drug Discovery and Development, Auburn University, Auburn Alabama, AL, USA
| |
Collapse
|
10
|
Abyadeh M, Kaya A. Application of Multiomics Approach to Investigate the Therapeutic Potentials of Stem Cell-derived Extracellular Vesicle Subpopulations for Alzheimer's Disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.10.593647. [PMID: 38798317 PMCID: PMC11118424 DOI: 10.1101/2024.05.10.593647] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Alzheimer's disease (AD) presents a complex interplay of molecular alterations, yet understanding its pathogenesis remains a challenge. In this study, we delved into the intricate landscape of proteome and transcriptome changes in AD brains compared to healthy controls, examining 788 brain samples revealing common alterations at both protein and mRNA levels. Moreover, our analysis revealed distinct protein-level changes in aberrant energy metabolism pathways in AD brains that were not evident at the mRNA level. This suggests that the changes in protein expression could provide a deeper molecular representation of AD pathogenesis. Subsequently, using a comparative proteomic approach, we explored the therapeutic potential of mesenchymal stem cell-derived extracellular vehicles (EVs), isolated through various methods, in mitigating AD-associated changes at the protein level. Our analysis revealed a particular EV-subtype that can be utilized for compensating dysregulated mitochondrial proteostasis in the AD brain. By using network biology approaches, we further revealed the potential regulators of key therapeutic proteins. Overall, our study illuminates the significance of proteome alterations in AD pathogenesis and identifies the therapeutic promise of a specific EV subpopulation with reduced pro-inflammatory protein cargo and enriched proteins to target mitochondrial proteostasis.
Collapse
Affiliation(s)
- Morteza Abyadeh
- Department of Biology, Virginia Commonwealth University, Richmond, VA 23284 USA
| | - Alaattin Kaya
- Department of Biology, Virginia Commonwealth University, Richmond, VA 23284 USA
- Department of Human and Molecular Genetics, Virginia Commonwealth University, Richmond, VA, 23284, USA
| |
Collapse
|
11
|
Alvariño R, Alfonso A, Tabudravu JN, González-Jartín J, Al Maqbali KS, Elhariry M, Vieytes MR, Botana LM. Psammaplin A and Its Analogs Attenuate Oxidative Stress in Neuronal Cells through Peroxisome Proliferator-Activated Receptor γ Activation. JOURNAL OF NATURAL PRODUCTS 2024; 87:1187-1196. [PMID: 38632902 PMCID: PMC11061836 DOI: 10.1021/acs.jnatprod.4c00153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 04/11/2024] [Accepted: 04/11/2024] [Indexed: 04/19/2024]
Abstract
Psammaplins are sulfur containing bromotyrosine alkaloids that have shown antitumor activity through the inhibition of class I histone deacetylases (HDACs). The cytotoxic properties of psammaplin A (1), the parent compound, are related to peroxisome proliferator-activated receptor γ (PPARγ) activation, but the mechanism of action of its analogs psammaplin K (2) and bisaprasin (3) has not been elucidated. In this study, the protective effects against oxidative stress of compounds 1-3, isolated from the sponge Aplysinella rhax, were evaluated in SH-SY5Y cells. The compounds improved cell survival, recovered glutathione (GSH) content, and reduced reactive oxygen species (ROS) release at nanomolar concentrations. Psammaplins restored mitochondrial membrane potential by blocking mitochondrial permeability transition pore opening and reducing cyclophilin D expression. This effect was mediated by the capacity of 1-3 to activate PPARγ, enhancing gene expression of the antioxidant enzymes catalase, nuclear factor E2-related factor 2 (Nrf2), and glutathione peroxidase. Finally, HDAC3 activity was reduced by 1-3 under oxidative stress conditions. This work is the first description of the neuroprotective activity of 1 at low concentrations and the mechanism of action of 2 and 3. Moreover, it links for the first time the previously described effects of 1 in HDAC3 and PPARγ signaling, opening a new research field for the therapeutic potential of this compound family.
Collapse
Affiliation(s)
- Rebeca Alvariño
- Departamento
de Fisiología, Facultad de Veterinaria, IDIS, Universidad de Santiago de Compostela, Lugo 27002, España
| | - Amparo Alfonso
- Departamento
de Farmacología, Facultad de Veterinaria, IDIS, Universidad de Santiago de Compostela, Lugo 27002, España
| | - Jioji N. Tabudravu
- School
of Pharmacy and Biomedical Sciences, University
of Central Lancashire, Preston, Lancashire PR1 2HE, United Kingdom
| | - Jesús González-Jartín
- Departamento
de Farmacología, Facultad de Veterinaria, IDIS, Universidad de Santiago de Compostela, Lugo 27002, España
| | - Khalid S. Al Maqbali
- School
of Pharmacy and Biomedical Sciences, University
of Central Lancashire, Preston, Lancashire PR1 2HE, United Kingdom
| | - Marwa Elhariry
- School
of Pharmacy and Biomedical Sciences, University
of Central Lancashire, Preston, Lancashire PR1 2HE, United Kingdom
| | - Mercedes R. Vieytes
- Departamento
de Fisiología, Facultad de Veterinaria, IDIS, Universidad de Santiago de Compostela, Lugo 27002, España
| | - Luis M. Botana
- Departamento
de Farmacología, Facultad de Veterinaria, IDIS, Universidad de Santiago de Compostela, Lugo 27002, España
| |
Collapse
|
12
|
Li Y, Pan Y, Zhao X, Wu S, Li F, Wang Y, Liu B, Zhang Y, Gao X, Wang Y, Zhou H. Peroxisome proliferator-activated receptors: A key link between lipid metabolism and cancer progression. Clin Nutr 2024; 43:332-345. [PMID: 38142478 DOI: 10.1016/j.clnu.2023.12.005] [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: 12/06/2023] [Accepted: 12/07/2023] [Indexed: 12/26/2023]
Abstract
Lipids represent the essential components of membranes, serve as fuels for high-energy processes, and play crucial roles in signaling and cellular function. One of the key hallmarks of cancer is the reprogramming of metabolic pathways, especially abnormal lipid metabolism. Alterations in lipid uptake, lipid desaturation, de novo lipogenesis, lipid droplets, and fatty acid oxidation in cancer cells all contribute to cell survival in a changing microenvironment by regulating feedforward oncogenic signals, key oncogenic functions, oxidative and other stresses, immune responses, or intercellular communication. Peroxisome proliferator-activated receptors (PPARs) are transcription factors activated by fatty acids and act as core lipid sensors involved in the regulation of lipid homeostasis and cell fate. In addition to regulating whole-body energy homeostasis in physiological states, PPARs play a key role in lipid metabolism in cancer, which is receiving increasing research attention, especially the fundamental molecular mechanisms and cancer therapies targeting PPARs. In this review, we discuss how cancer cells alter metabolic patterns and regulate lipid metabolism to promote their own survival and progression through PPARs. Finally, we discuss potential therapeutic strategies for targeting PPARs in cancer based on recent studies from the last five years.
Collapse
Affiliation(s)
- Yunkuo Li
- Department of Urology, The First Hospital of Jilin University, Changchun 130021, China
| | - Yujie Pan
- Department of Urology, The First Hospital of Jilin University, Changchun 130021, China
| | - Xiaodong Zhao
- Department of Urology, The First Hospital of Jilin University, Changchun 130021, China
| | - Shouwang Wu
- Department of Urology, The First Hospital of Jilin University, Changchun 130021, China
| | - Faping Li
- Department of Urology, The First Hospital of Jilin University, Changchun 130021, China
| | - Yuxiong Wang
- Department of Urology, The First Hospital of Jilin University, Changchun 130021, China
| | - Bin Liu
- Department of Urology, The First Hospital of Jilin University, Changchun 130021, China
| | - Yanghe Zhang
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun 130021, China
| | - Xin Gao
- Department of Urology, The First Hospital of Jilin University, Changchun 130021, China
| | - Yishu Wang
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun 130021, China.
| | - Honglan Zhou
- Department of Urology, The First Hospital of Jilin University, Changchun 130021, China.
| |
Collapse
|
13
|
Zhao T, Jia J. Polygalacic acid attenuates cognitive impairment by regulating inflammation through PPARγ/NF-κB signaling pathway. CNS Neurosci Ther 2024; 30:e14581. [PMID: 38421141 PMCID: PMC10851321 DOI: 10.1111/cns.14581] [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: 05/09/2023] [Revised: 11/06/2023] [Accepted: 12/13/2023] [Indexed: 03/02/2024] Open
Abstract
AIMS We aimed to explore the role and molecular mechanism of polygalacic acid (PA) extracted from traditional Chinese medicine Polygala tenuifolia in the treatment of Alzheimer's disease (AD). METHODS The network pharmacology analysis was used to predict the potential targets and pathways of PA. Molecular docking was applied to analyze the combination between PA and core targets. Aβ42 oligomer-induced AD mice model and microglia were used to detect the effect of PA on the release of pro-inflammatory mediators and its further mechanism. In addition, a co-culture system of microglia and neuronal cells was constructed to assess the effect of PA on activating microglia-mediated neuronal apoptosis. RESULTS We predict that PA might regulate inflammation by targeting PPARγ-mediated pathways by using network pharmacology. In vivo study, PA could attenuate cognitive deficits and inhibit the expression levels of inflammation-related factors. In vitro study, PA can also decrease the production of activated microglia-mediated inflammatory cytokines and reduce the apoptosis of N2a neuronal cells. PPARγ inhibitor GW9662 inversed the neuroprotective effect of PA. Both in vivo and in vitro studies showed PA might attenuate the inflammation through the PPARγ/NF-κB pathway. CONCLUSIONS PA is expected to provide a valuable candidate for new drug development for AD in the future.
Collapse
Affiliation(s)
- Tan Zhao
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu HospitalCapital Medical University, National Clinical Research Center for Geriatric DiseasesBeijingChina
| | - Jianping Jia
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu HospitalCapital Medical University, National Clinical Research Center for Geriatric DiseasesBeijingChina
- Beijing Key Laboratory of Geriatric Cognitive DisordersBeijingChina
- Clinical Center for Neurodegenerative Disease and Memory ImpairmentCapital Medical UniversityBeijingChina
- Center of Alzheimer's DiseaseBeijing Institute of Brain Disorders, Collaborative Innovation Center for Brain Disorders, Capital Medical UniversityBeijingChina
- Key Laboratory of Neurodegenerative Diseases, Ministry of EducationBeijingChina
| |
Collapse
|
14
|
Shah NZ, Khan A, Halim SA, Avula SK, Islam NU, Khan I, Karim N, Kifayatullah M, Khalid A, Alhazmi HA, Abdalla AN, Kashtoh H, Al-Harrasi A. Efficient microwave synthesis of flurbiprofen derivatives and their enhancement of efficacy in chronic inflammatory pain models and gastro-protective potential in post-operative model. J Biomol Struct Dyn 2024:1-16. [PMID: 38294707 DOI: 10.1080/07391102.2024.2309645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Accepted: 01/18/2024] [Indexed: 02/01/2024]
Abstract
Present research was designed to synthesize and characterize the flurbiprofen derivatives and to evaluate their analgesic, anti-inflammatory and gastro-protective activities in post-operative and chronic inflammatory pain models. Flurbiprofen derivatives were produced by using three-step processes involving esterification, hydrazide production, and schiff base, each of which modified a different carboxyl group. All the newly synthesized flurbiprofen derivatives (NS5-NS8) were characterized by 1H NMR,13C NMR,19F NMR and HR-ESI-MS, and the post-operative, inflammatory pain and ulcerogenic activities were determined in well-established in-vivo animal models. To evaluate post-operative and inflammatory pain, various doses of compounds [1, 3, 10, and 30 mg/kg (bwt)] were used, while their ulcerogenic potential was assessed at doses of 100 and 150 mg/kg (bwt). The incisional damage linked pain was significantly (p < 0.001) reduced by derivatives at different doses in both the acute and repeated tests with decreased response of phologistic agent-induced inflammation. The stomach histology and biochemical features demonstrate that the synthesized derivatives have no potential to cause ulcerogenicity as compared to aspirin and flurbiprofen. Furthermore, docking shows that the hydrazide moiety of these compounds is crucial in interacting within COX-2 binding site. Therefore, the synthesized compounds exhibit strong analgesic and anti-inflammatory effects and a low risk of causing ulcers. These attributes render them potentially valuable therapeutic agents for the treatment of pathological disorders associated with inflammation and pain.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Nisar Zamin Shah
- Department of Pharmacy, University of Malakand, Khyber Pakhtunkhwa, Pakistan
| | - Ajmal Khan
- Natural and Medical Sciences Research Center, University of Nizwa, Nizwa, Sultanate of Oman
| | - Sobia Ahsan Halim
- Natural and Medical Sciences Research Center, University of Nizwa, Nizwa, Sultanate of Oman
| | - Satya Kumar Avula
- Natural and Medical Sciences Research Center, University of Nizwa, Nizwa, Sultanate of Oman
| | - Nazar Ul Islam
- Department of Pharmacy, Sarhad University of Science and Information Technology, Peshawar, Pakistan
| | - Imran Khan
- Department of Pharmacy, University of Swabi, Swabi, KPK, Pakistan
| | - Nasiara Karim
- Department of Pharmacy, University of Peshawar, Khyber Pakhtunkhwa, Pakistan
| | | | - Asaad Khalid
- Substance Abuse and Toxicology Research Center, Jazan University, Jazan, Saudi Arabia
| | - Hassan A Alhazmi
- Substance Abuse and Toxicology Research Center, Jazan University, Jazan, Saudi Arabia
- Department of Pharmaceutical Chemistry, College of Pharmacy, Jazan University, Jazan, Saudi Arabia
| | - Ashraf N Abdalla
- Department of Pharmacology and Toxicology, College of Pharmacy, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Hamdy Kashtoh
- Department of Biotechnology, Yeungnam University, Gyeongsan, Gyeongbuk, Republic of Korea
| | - Ahmed Al-Harrasi
- Natural and Medical Sciences Research Center, University of Nizwa, Nizwa, Sultanate of Oman
| |
Collapse
|
15
|
Valencia-Olvera AC, Balu D, Bellur S, McNally T, Saleh Y, Pham D, Ghura S, York J, Johansson JO, LaDu MJ, Tai L. A novel apoE-mimetic increases brain apoE levels, reduces Aβ pathology and improves memory when treated before onset of pathology in male mice that express APOE3. Alzheimers Res Ther 2023; 15:216. [PMID: 38102668 PMCID: PMC10722727 DOI: 10.1186/s13195-023-01353-z] [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: 09/15/2023] [Accepted: 11/15/2023] [Indexed: 12/17/2023]
Abstract
BACKGROUND Alzheimer's disease (AD) is characterized by cognitive dysfunction and amyloid plaques composed of the amyloid-beta peptide (Aβ). APOE is the greatest genetic risk for AD with APOE4 increasing risk up to ~ 15-fold compared to APOE3. Evidence suggests that levels and lipidation of the apoE protein could regulate AD progression. In glia, apoE is lipidated via cholesterol efflux from intracellular pools, primarily by the ATP-binding cassette transporter A1 (ABCA1). Therefore, increasing ABCA1 activity is suggested to be a therapeutic approach for AD. CS-6253 (CS) is a novel apoE mimetic peptide that was developed to bind and stabilize ABCA1 and maintain its localization into the plasma membrane therefore promoting cholesterol efflux. The goal of this study was to determine whether CS could modulate apoE levels and lipidation, Aβ pathology, and behavior in a model that expresses human APOE and overproduce Aβ. METHODS In vitro, APOE3-glia or APOE4-glia were treated with CS. In vivo, male and female, E3FAD (5xFAD+/-/APOE3+/+) and E4FAD (5xFAD+/-/APOE4+/+) mice were treated with CS via intraperitoneal injection at early (from 4 to 8 months of age) and late ages (from 8 to 10 months of age). ApoE levels, ABCA1 levels and, apoE lipidation were measured by western blot and ELISA. Aβ and amyloid levels were assessed by histochemistry and ELISA. Learning and memory were tested by Morris Water Maze and synaptic proteins were measured by Western blot. RESULTS CS treatment increased apoE levels and cholesterol efflux in primary glial cultures. In young male E3FAD mice, CS treatment increased soluble apoE and lipid-associated apoE, reduced soluble oAβ and insoluble Aβ levels as well as Aβ and amyloid deposition, and improved memory and synaptic protein levels. CS treatment did not induce any therapeutic benefits in young female E3FAD and E4FAD mice or in any groups when treatment was started at later ages. CONCLUSIONS CS treatment reduced Aβ pathology and improved memory only in young male E3FAD, the cohort with the least AD pathology. Therefore, the degree of Aβ pathology or Aβ overproduction may impact the ability of targeting ABCA1 to be an effective AD therapeutic. This suggests that ABCA1-stabilizing treatment by CS-6253 works best in conditions of modest Aβ levels.
Collapse
Affiliation(s)
- Ana C Valencia-Olvera
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, USA
| | - Deebika Balu
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, USA
| | - Shreya Bellur
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, USA
| | - Thomas McNally
- Department of Surgery, University of Washington, Seattle, WA, USA
| | - Yaseen Saleh
- University of Miami/Jackson Healthcare System, Miami, FL, USA
| | - Don Pham
- Department of Dentistry, University of Illinois at Chicago, Chicago, IL, USA
| | - Shivesh Ghura
- Department of Pharmacology, University of Pennsylvania, Philadelphia, PA, USA
| | - Jason York
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, USA
| | | | - Mary Jo LaDu
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, USA
| | - Leon Tai
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, USA.
| |
Collapse
|
16
|
Fauzi A, Thoe ES, Quan TY, Yin ACY. Insights from insulin resistance pathways: Therapeutic approaches against Alzheimer associated diabetes mellitus. J Diabetes Complications 2023; 37:108629. [PMID: 37866274 DOI: 10.1016/j.jdiacomp.2023.108629] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 09/03/2023] [Accepted: 10/15/2023] [Indexed: 10/24/2023]
Abstract
Alzheimer Associated Diabetes Mellitus, commonly known as Type 3 Diabetes Mellitus (T3DM) is a distinct subtype of diabetes with a pronounced association with Alzheimer's disease (AD). Insulin resistance serves as a pivotal link between these two conditions, leading to diminished insulin sensitivity, hyperglycemia, and impaired glucose uptake. The brain, a vital organ in AD context, is also significantly impacted by insulin resistance, resulting in energy deficits and neuronal damage, which are hallmark features of the neurodegenerative disorder. To pave the way for potential therapeutic interventions targeting the insulin resistance pathway, it is crucial to comprehend the intricate pathophysiology of T3DM and identify the overlapped features between diabetes and AD. This comprehensive review article aims to explore various pathway such as AMPK, PPARγ, cAMP and P13K/Akt pathway as potential target for management of T3DM. Through the analysis of these complex mechanisms, our goal is to reveal their interdependencies and support the discovery of innovative therapeutic strategies. The review extensively discusses several promising pharmaceutical candidates that have demonstrated dual drug action mechanisms, addressing both peripheral and cerebral insulin resistance observed in T3DM. These candidates hold significant promise for restoring insulin function and mitigating the detrimental effects of insulin resistance on the brain. The exploration of these therapeutic options contributes to the development of innovative interventions that alleviate the burden of T3DM and enhance patient care.
Collapse
Affiliation(s)
- Ayesha Fauzi
- School of Biosciences, Faculty of Health & Medical Sciences, Taylor's University Lakeside Campus, 47500 Subang Jaya, Selangor Darul Ehsan, Malaysia
| | - Ewen Se Thoe
- School of Biosciences, Faculty of Health & Medical Sciences, Taylor's University Lakeside Campus, 47500 Subang Jaya, Selangor Darul Ehsan, Malaysia
| | - Tang Yin Quan
- School of Biosciences, Faculty of Health & Medical Sciences, Taylor's University Lakeside Campus, 47500 Subang Jaya, Selangor Darul Ehsan, Malaysia; Medical Advancement for Better Quality of Life Impact Lab, Taylor's University Lakeside Campus, 47500 Subang Jaya, Selangor Darul Ehsan, Malaysia
| | - Adeline Chia Yoke Yin
- School of Biosciences, Faculty of Health & Medical Sciences, Taylor's University Lakeside Campus, 47500 Subang Jaya, Selangor Darul Ehsan, Malaysia; Medical Advancement for Better Quality of Life Impact Lab, Taylor's University Lakeside Campus, 47500 Subang Jaya, Selangor Darul Ehsan, Malaysia.
| |
Collapse
|
17
|
Sandeep Ganesh G, Konduri P, Kolusu AS, Namburi SV, Chunduru BTC, Nemmani KVS, Samudrala PK. Neuroprotective Effect of Saroglitazar on Scopolamine-Induced Alzheimer's in Rats: Insights into the Underlying Mechanisms. ACS Chem Neurosci 2023; 14:3444-3459. [PMID: 37669120 DOI: 10.1021/acschemneuro.3c00320] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/07/2023] Open
Abstract
Alzheimer's disease (AD) is one of the most prevalent and progressive neurodegenerative disorders, hallmarked by increased amyloid-β deposition and enhanced oxidative load in the brain, ensuing cognitive decline. The present study is aimed at elucidating the neuroprotective effect of saroglitazar, a dual peroxisome-proliferator-activated receptor (PPARα/γ) agonist used in the treatment of diabetic dyslipidemia, against memory impairment induced by intraperitoneal scopolamine injection. 30 male Wistar rats were randomly divided into the following five groups: (A) Veh + Veh, (B) SGZ + Veh, (C) Veh + SCOP, (D) DPZ + SCOP, and (E) SGZ + SCOP. Rats of the respective groups were pretreated with saroglitazar (10 mg/kg, p.o.) and donepezil (3 mg/kg, p.o.) once daily for 16 days. During the final 9 days of the study, a daily injection of scopolamine (3 mg/kg, i.p.) was administered to the respective groups. Adjacent to the scopolamine injection, behavioral tests such as the open field, Y maze, novel object recognition test, and Morris water maze were conducted to assess learning and memory. Additionally, biochemical parameters such as acetylcholinesterase (AChE), butyrylcholinesterase (BuChE), nitric oxide (NO), malondialdehyde (MDA), reduced glutathione (GSH), superoxide dismutase (SOD), brain-derived neurotrophic factor (BDNF), β-amyloid levels, and NF-κB were measured in the hippocampus. The rats that received scopolamine injections showed significantly impaired short-term spatial and learning memory. This was associated with an increase in β-amyloid, iNOS, nitric oxide (NO), malondialdehyde, NF-κB, and TNF-α levels in the hippocampus of AD rats. On the other hand, saroglitazar has provided promising data on its protective role in cognition by protecting the BDNF, SOD, and GSH decline. As a result, saroglitazar was found to be a promising therapy in AD by upregulating the antioxidant status and cholinergic activity and preventing memory loss. Collectively, findings in the present study revealed that saroglitazar protected AD by suppressing scopolamine-mediated learning and memory deficits, oxidative stress, and cholinergic damage. Studying these mechanisms may conclude the protective role of saroglitazar against AD. However, further studies in transgenic animals will provide numerous insights into treatment mechanisms and contribute to developing a therapeutic intervention for AD.
Collapse
Affiliation(s)
- Grandhi Sandeep Ganesh
- Department of Pharmacology, Shri Vishnu College of Pharmacy, Bhimavaram, Andhra Pradesh 534202, India
| | - Prasad Konduri
- Department of Pharmacology, Shri Vishnu College of Pharmacy, Bhimavaram, Andhra Pradesh 534202, India
| | - Aravinda Sai Kolusu
- Department of Pharmacology, Shri Vishnu College of Pharmacy, Bhimavaram, Andhra Pradesh 534202, India
| | - Srihari Vandana Namburi
- Department of Pharmacology, Shri Vishnu College of Pharmacy, Bhimavaram, Andhra Pradesh 534202, India
| | - Bala Tejo Chandra Chunduru
- Clinical Data Manager, STATMINDS LLC, 501 Allendale Rd Suite 202, King of Prussia, Pennsylvania 19406, United States
| | - Kumar V S Nemmani
- Department of Pharmacology, Shri Vishnu College of Pharmacy, Bhimavaram, Andhra Pradesh 534202, India
| | - Pavan Kumar Samudrala
- Department of Pharmacology, Shri Vishnu College of Pharmacy, Bhimavaram, Andhra Pradesh 534202, India
| |
Collapse
|
18
|
Lutz MW, Chiba-Falek O. Bioinformatics pipeline to guide post-GWAS studies in Alzheimer's: A new catalogue of disease candidate short structural variants. Alzheimers Dement 2023; 19:4094-4109. [PMID: 37253165 PMCID: PMC10524333 DOI: 10.1002/alz.13168] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 04/27/2023] [Accepted: 05/08/2023] [Indexed: 06/01/2023]
Abstract
BACKGROUND Short structural variants (SSVs), including insertions/deletions (indels), are common in the human genome and impact disease risk. The role of SSVs in late-onset Alzheimer's disease (LOAD) has been understudied. In this study, we developed a bioinformatics pipeline of SSVs within LOAD-genome-wide association study (GWAS) regions to prioritize regulatory SSVs based on the strength of their predicted effect on transcription factor (TF) binding sites. METHODS The pipeline utilized publicly available functional genomics data sources including candidate cis-regulatory elements (cCREs) from ENCODE and single-nucleus (sn)RNA-seq data from LOAD patient samples. RESULTS We catalogued 1581 SSVs in candidate cCREs in LOAD GWAS regions that disrupted 737 TF sites. That included SSVs that disrupted the binding of RUNX3, SPI1, and SMAD3, within the APOE-TOMM40, SPI1, and MS4A6A LOAD regions. CONCLUSIONS The pipeline developed here prioritized non-coding SSVs in cCREs and characterized their putative effects on TF binding. The approach integrates multiomics datasets for validation experiments using disease models.
Collapse
Affiliation(s)
- Michael W. Lutz
- Division of Translational Brain Sciences, Department of Neurology, Duke University Medical Center, Durham, NC 27710, USA
| | - Ornit Chiba-Falek
- Division of Translational Brain Sciences, Department of Neurology, Duke University Medical Center, Durham, NC 27710, USA
- Center for Genomic and Computational Biology, Duke University Medical Center, Durham, NC 27710, USA
| |
Collapse
|
19
|
Chen C. Inhibiting degradation of 2-arachidonoylglycerol as a therapeutic strategy for neurodegenerative diseases. Pharmacol Ther 2023; 244:108394. [PMID: 36966972 PMCID: PMC10123871 DOI: 10.1016/j.pharmthera.2023.108394] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 03/17/2023] [Accepted: 03/21/2023] [Indexed: 03/29/2023]
Abstract
Endocannabinoids are endogenous lipid signaling mediators that participate in a variety of physiological and pathological processes. 2-Arachidonoylglycerol (2-AG) is the most abundant endocannabinoid and is a full agonist of G-protein-coupled cannabinoid receptors (CB1R and CB2R), which are targets of Δ9-tetrahydrocannabinol (Δ9-THC), the main psychoactive ingredient in cannabis. While 2-AG has been well recognized as a retrograde messenger modulating synaptic transmission and plasticity at both inhibitory GABAergic and excitatory glutamatergic synapses in the brain, growing evidence suggests that 2-AG also functions as an endogenous terminator of neuroinflammation in response to harmful insults, thus maintaining brain homeostasis. Monoacylglycerol lipase (MAGL) is the key enzyme that degrades 2-AG in the brain. The immediate metabolite of 2-AG is arachidonic acid (AA), a precursor of prostaglandins (PGs) and leukotrienes. Several lines of evidence indicate that pharmacological or genetic inactivation of MAGL, which boosts 2-AG levels and reduces its hydrolytic metabolites, resolves neuroinflammation, mitigates neuropathology, and improves synaptic and cognitive functions in animal models of neurodegenerative diseases, including Alzheimer's disease (AD), multiple sclerosis (MS), Parkinson's disease (PD), and traumatic brain injury (TBI)-induced neurodegenerative disease. Thus, it has been proposed that MAGL is a potential therapeutic target for treatment of neurodegenerative diseases. As the main enzyme hydrolyzing 2-AG, several MAGL inhibitors have been identified and developed. However, our understanding of the mechanisms by which inactivation of MAGL produces neuroprotective effects in neurodegenerative diseases remains limited. A recent finding that inhibition of 2-AG metabolism in astrocytes, but not in neurons, protects the brain from TBI-induced neuropathology might shed some light on this unsolved issue. This review provides an overview of MAGL as a potential therapeutic target for neurodegenerative diseases and discusses possible mechanisms underlying the neuroprotective effects of restraining degradation of 2-AG in the brain.
Collapse
|
20
|
Effects of Peroxisome Proliferator-Activated Receptor-Gamma Agonists on Cognitive Function: A Systematic Review and Meta-Analysis. Biomedicines 2023; 11:biomedicines11020246. [PMID: 36830783 PMCID: PMC9953157 DOI: 10.3390/biomedicines11020246] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 12/21/2022] [Accepted: 01/04/2023] [Indexed: 01/20/2023] Open
Abstract
Diabetes mellitus (DM) is known to be a risk factor for dementia, especially in the elderly population, and close associations between diabetes and Alzheimer disease (AD) have been determined. Peroxisome proliferator-activated receptor-gamma (PPAR-γ) agonists are insulin-sensitising drugs. In addition to their anti-diabetic properties, their effectiveness in preventing and decreasing cognitive impairment are the most recent characteristics that have been studied. For this study, we conducted a systematic review and meta-analysis to critically analyse and evaluate the existing data on the effects of PPAR-γ agonist therapy on the cognitive status of patients. For this purpose, we first analysed both early intervention and later treatment with PPAR-γ agonists, according to the disease status. The involved studies indicated that early PPAR-γ agonist intervention is beneficial for patients and that high-dose PPAR-γ therapy may have a better clinical effect, especially in reversing the effects of cognitive impairment. Furthermore, the efficacy of pioglitazone (PIO) seems to be promising, particularly for patients with comorbid diabetes. PIO presented a better clinical curative effect and safety, compared with rosiglitazone (RSG). Thus, PPAR-γ agonists play an important role in the inflammatory response of AD or DM patients, and clinical therapeutics should focus more on relevant metabolic indices.
Collapse
|
21
|
Xu L, Li L, Pan C, Song J, Zhang C, Wu X, Hu F, Liu X, Zhang Z, Zhang Z. Erythropoietin signaling in peripheral macrophages is required for systemic β-amyloid clearance. EMBO J 2022; 41:e111038. [PMID: 36215698 PMCID: PMC9670197 DOI: 10.15252/embj.2022111038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 09/10/2022] [Accepted: 09/14/2022] [Indexed: 01/13/2023] Open
Abstract
Impaired clearance of beta-amyloid (Aβ) is a primary cause of sporadic Alzheimer's disease (AD). Aβ clearance in the periphery contributes to reducing brain Aβ levels and preventing Alzheimer's disease pathogenesis. We show here that erythropoietin (EPO) increases phagocytic activity, levels of Aβ-degrading enzymes, and Aβ clearance in peripheral macrophages via PPARγ. Erythropoietin is also shown to suppress Aβ-induced inflammatory responses. Deletion of EPO receptor in peripheral macrophages leads to increased peripheral and brain Aβ levels and exacerbates Alzheimer's-associated brain pathologies and behavioral deficits in AD-model mice. Moreover, erythropoietin signaling is impaired in peripheral macrophages of old AD-model mice. Exogenous erythropoietin normalizes impaired EPO signaling and dysregulated functions of peripheral macrophages in old AD-model mice, promotes systemic Aβ clearance, and alleviates disease progression. Erythropoietin treatment may represent a potential therapeutic approach for Alzheimer's disease.
Collapse
Affiliation(s)
- Lu Xu
- School of Basic Medical SciencesNanjing Medical UniversityNanjingChina
- Key Laboratory of Antibody Technique of Ministry of HealthNanjing Medical UniversityNanjingChina
- Department of Neurology, Sir Run Run HospitalNanjing Medical UniversityNanjingChina
| | - Lei Li
- School of Basic Medical SciencesNanjing Medical UniversityNanjingChina
| | - Cai‐Long Pan
- School of Basic Medical SciencesNanjing Medical UniversityNanjingChina
- Key Laboratory of Antibody Technique of Ministry of HealthNanjing Medical UniversityNanjingChina
| | - Jing‐Jing Song
- School of Basic Medical SciencesNanjing Medical UniversityNanjingChina
| | - Chen‐Yang Zhang
- School of Basic Medical SciencesNanjing Medical UniversityNanjingChina
| | - Xiang‐Hui Wu
- School of Basic Medical SciencesNanjing Medical UniversityNanjingChina
| | - Fan Hu
- State Key Laboratory of Reproductive MedicineNanjing Medical UniversityNanjingChina
| | - Xue Liu
- School of Basic Medical SciencesNanjing Medical UniversityNanjingChina
| | - Zhiren Zhang
- Institute of ImmunologyArmy Medical UniversityChongqingChina
| | - Zhi‐Yuan Zhang
- School of Basic Medical SciencesNanjing Medical UniversityNanjingChina
- Key Laboratory of Antibody Technique of Ministry of HealthNanjing Medical UniversityNanjingChina
- Department of Neurology, Sir Run Run HospitalNanjing Medical UniversityNanjingChina
- Key Laboratory of Human Functional Genomics of Jiangsu ProvinceNanjing Medical UniversityNanjingChina
| |
Collapse
|
22
|
Cummings J, Ortiz A, Castellino J, Kinney J. Diabetes: Risk factor and translational therapeutic implications for Alzheimer's disease. Eur J Neurosci 2022; 56:5727-5757. [PMID: 35128745 PMCID: PMC9393901 DOI: 10.1111/ejn.15619] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 01/25/2022] [Accepted: 01/27/2022] [Indexed: 12/31/2022]
Abstract
Type 2 diabetes mellitus (T2DM) and Alzheimer's disease (AD) commonly co-occur. T2DM increases the risk for AD by approximately twofold. Animal models provide one means of interrogating the relationship of T2DM to AD and investigating brain insulin resistance in the pathophysiology of AD. Animal models show that persistent hyperglycaemia results in chronic low-grade inflammation that may contribute to the development of neuroinflammation and accelerate the pathobiology of AD. Epidemiological studies suggest that patients with T2DM who received treatment with specific anti-diabetic agents have a decreased risk for the occurrence of AD and all-cause dementia. Agents such as metformin ameliorate T2DM and may have other important systemic effects that lower the risk of AD. Glucagon-like peptide 1 (GLP-1) agonists have been associated with a decreased risk for AD in patients with T2DM. Both insulin and non-insulin anti-diabetic treatments have been evaluated for the treatment of AD in clinical trials. In most cases, patients included in the trials have clinical features of AD but do not have T2DM. Many of the trials were conducted prior to the use of diagnostic biomarkers for AD. Trials have had a wide range of durations and population sizes. Many of the agents used to treat T2DM do not cross the blood brain barrier, and the effects are posited to occur via lowering of peripheral hyperglycaemia and reduction of peripheral and central inflammation. Clinical trials of anti-diabetic agents to treat AD are ongoing and will provide insight into the therapeutic utility of these agents.
Collapse
Affiliation(s)
- Jeffrey Cummings
- Chambers‐Grundy Center for Transformative Neuroscience, Department of Brain Health, School of Integrated Health SciencesUniversity of Nevada Las Vegas (UNLV)Las VegasNevadaUSA
| | - Andrew Ortiz
- Department of Brain Health, School of Integrated Health SciencesUniversity of Nevada Las Vegas (UNLV)Las VegasNevadaUSA
| | | | - Jefferson Kinney
- Chambers‐Grundy Center for Transformative Neuroscience, Department of Brain Health, School of Integrated Health SciencesUniversity of Nevada Las Vegas (UNLV)Las VegasNevadaUSA,Department of Brain Health, School of Integrated Health SciencesUniversity of Nevada Las Vegas (UNLV)Las VegasNevadaUSA
| |
Collapse
|
23
|
Ballav S, Biswas B, Sahu VK, Ranjan A, Basu S. PPAR-γ Partial Agonists in Disease-Fate Decision with Special Reference to Cancer. Cells 2022; 11:3215. [PMID: 36291082 PMCID: PMC9601205 DOI: 10.3390/cells11203215] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2022] [Revised: 10/03/2022] [Accepted: 10/09/2022] [Indexed: 11/16/2023] Open
Abstract
Peroxisome proliferator-activated receptor-γ (PPAR-γ) has emerged as one of the most extensively studied transcription factors since its discovery in 1990, highlighting its importance in the etiology and treatment of numerous diseases involving various types of cancer, type 2 diabetes mellitus, autoimmune, dermatological and cardiovascular disorders. Ligands are regarded as the key determinant for the tissue-specific activation of PPAR-γ. However, the mechanism governing this process is merely a contradictory debate which is yet to be systematically researched. Either these receptors get weakly activated by endogenous or natural ligands or leads to a direct over-activation process by synthetic ligands, serving as complete full agonists. Therefore, fine-tuning on the action of PPAR-γ and more subtle modulation can be a rewarding approach which might open new avenues for the treatment of several diseases. In the recent era, researchers have sought to develop safer partial PPAR-γ agonists in order to dodge the toxicity induced by full agonists, akin to a balanced activation. With a particular reference to cancer, this review concentrates on the therapeutic role of partial agonists, especially in cancer treatment. Additionally, a timely examination of their efficacy on various other disease-fate decisions has been also discussed.
Collapse
Affiliation(s)
- Sangeeta Ballav
- Cancer and Translational Research Centre, Dr. D. Y. Patil Biotechnology and Bioinformatics Institute, Dr. D. Y. Patil Vidyapeeth, Tathawade, Pune 411033, India
| | - Bini Biswas
- Cancer and Translational Research Centre, Dr. D. Y. Patil Biotechnology and Bioinformatics Institute, Dr. D. Y. Patil Vidyapeeth, Tathawade, Pune 411033, India
| | - Vishal Kumar Sahu
- Cancer and Translational Research Centre, Dr. D. Y. Patil Biotechnology and Bioinformatics Institute, Dr. D. Y. Patil Vidyapeeth, Tathawade, Pune 411033, India
| | - Amit Ranjan
- Cancer and Translational Research Centre, Dr. D. Y. Patil Biotechnology and Bioinformatics Institute, Dr. D. Y. Patil Vidyapeeth, Tathawade, Pune 411033, India
| | - Soumya Basu
- Cancer and Translational Research Centre, Dr. D. Y. Patil Biotechnology and Bioinformatics Institute, Dr. D. Y. Patil Vidyapeeth, Tathawade, Pune 411033, India
| |
Collapse
|
24
|
Singla RK, Dhonchak K, Sodhi RK, Arockia Babu M, Madan J, Madaan R, Kumar S, Sharma R, Shen B. Bergenin ameliorates cognitive deficits and neuropathological alterations in sodium azide-induced experimental dementia. Front Pharmacol 2022; 13:994018. [PMID: 36249784 PMCID: PMC9556967 DOI: 10.3389/fphar.2022.994018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Accepted: 08/24/2022] [Indexed: 11/24/2022] Open
Abstract
Background: Bergenin, 4-O-methyl gallic acid glucoside, is a bioactive compound found in the cortex of Mallotus japonicus (L.f.) Müll.Arg. along with many other natural resources including that from Bergenia species. The present study delineates the neuroprotective potential of bergenin through the modulation of PPAR-γ receptors. Method: Dementia was induced in the Wistar rats by intraperitoneal (i.p.) administration of sodium azide (12.5 mg/kg for the first 5 days followed by 10 mg/kg for the next 9 days). The rats were then exposed to the Morris water maze test to assess the effect on cognitive abilities followed by a series of biochemical and histopathological evaluations. Results: Sodium azide-treated rats exhibited a severe deterioration of memory as suggested by poor performance in the spatial learning task in addition to the enhancement of brain acetylcholinesterase potential, oxidative stress, inflammation, and amyloid-β (Aβ) accumulation. Administration of bergenin to sodium azide-treated rats significantly recovered cognition and related biochemical variations. Further, co-administration of Bisphenol A diglycidyl ether (BADGE), a PPAR-γ antagonist with bergenin challenged its neuroprotective effects. Conclusions: The findings of our study exhibit that the cognitive restoration potential of bergenin may be attributed to its modulatory effects against cholinesterase, oxidative stress, and inflammatory markers, as well as its neuroprotective actions, thus aligning it as a possible therapy for Alzheimer's disease-related dementia. The study also fortifies the significance of PPAR-γ receptors in dementia.
Collapse
Affiliation(s)
- Rajeev K. Singla
- Institutes for Systems Genetics, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, India
| | - Konika Dhonchak
- Department of Pharmacology, Chandigarh College of Pharmacy, Mohali, India
| | - Rupinder K. Sodhi
- Department of Pharmacology, Chandigarh College of Pharmacy, Mohali, India
| | - M. Arockia Babu
- Department of Pharmacology, Chandigarh College of Pharmacy, Mohali, India
| | - Jitender Madan
- National Institute of Pharmaceutical Education and Research, Hyderabad, India
| | - Reecha Madaan
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | - Suresh Kumar
- Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, Punjab, India
| | - Rohit Sharma
- Department of Rasa Shastra and Bhaishajya Kalpana, Faculty of Ayurveda, Institute of Medical Sciences, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| | - Bairong Shen
- Institutes for Systems Genetics, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
25
|
Salidroside ameliorates orthopedic surgery-induced cognitive dysfunction by activating adenosine 5'-monophosphate-activated protein kinase signaling in mice. Eur J Pharmacol 2022; 929:175148. [PMID: 35834964 DOI: 10.1016/j.ejphar.2022.175148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 07/07/2022] [Accepted: 07/07/2022] [Indexed: 11/21/2022]
Abstract
Perioperative neurocognitive disorders (PND) are the most common postoperative complications with few therapeutic options. Salidroside, a plant-derived compound, has gained increased attention as a treatment for various neurological diseases and particularly as a modifier of microglia-mediated neuroinflammation. However, the effect of salidroside on orthopedic surgery-induced cognitive dysfunction and the underlying mechanisms are largely unknown. Here, we found that salidroside greatly attenuated cognitive impairment in mice after orthopedic surgery. Neuroinflammation in the mouse hippocampus was also attenuated by salidroside. Meanwhile, salidroside treatment induced a switch in microglial polarization to the anti-inflammatory phenotype. In vitro, salidroside suppressed the expression of proinflammatory cytokines and induced a switch in microglial phenotype to the anti-inflammatory phenotype. Mechanistically, molecular docking studies revealed the potential AMPK activation activity of salidroside. And salidroside did up-regulated the AMPK pathway proteins. Moreover, AMPK antagonist abolished the effects of salidroside in vivo and in vitro. Taken together, our results demonstrated that salidroside effectively suppressed PND by suppressing microglia-mediated neuroinflammation through activating AMPK pathway, and it might be a novel therapeutic approach for PND.
Collapse
|
26
|
Liu H, Zang C, Shang J, Zhang Z, Wang L, Yang H, Sheng C, Yuan F, Ju C, Li F, Yu Y, Yao X, Bao X, Zhang D. <em>Gardenia jasminoides</em> J. Ellis extract GJ-4 attenuates hyperlipidemic vascular dementia in rats via regulating PPAR-γ-mediated microglial polarization. Food Nutr Res 2022; 66:8101. [PMID: 35950104 PMCID: PMC9338452 DOI: 10.29219/fnr.v66.8101] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 10/13/2021] [Accepted: 10/28/2021] [Indexed: 11/20/2022] Open
Abstract
Background GJ-4 is extracted from Gardenia jasminoides J. Ellis (Fructus Gardenia) with crocin composition and has been demonstrated to improve memory deficits in several dementia models in our previous studies. Objective This study aimed to evaluate the effects of GJ-4 on hyperlipidemic vascular dementia (VD) and explore the underlying mechanisms. Design In the current study, we employed a chronic hyperlipidemic VD rat model by permanent bilateral common carotid arteries occlusion (2-VO) based on high-fat diet (HFD), which is an ideal model to mimic the clinical pathogenesis of human VD. Results Our results showed that GJ-4 could significantly reduce serum lipids level and improve cerebral blood flow in hyperlipidemic VD rats. Additionally, treatment with GJ-4 remarkedly ameliorated memory impairment and alleviated neuronal injury. Mechanistic investigation revealed that the neuroprotective effects of GJ-4 might be attributed to the inhibition of microglia-mediated neuro-inflammation via regulating the M1/M2 polarization. Our data further illustrated that GJ-4 could regulate the phenotype of microglia through activating the peroxisome proliferator-activated receptor-γ (PPAR-γ) and subsequently inhibited nuclear factor-κB (NF-κB) nuclear translocation and increased CCAAT/enhancer-binding protein β (C/EBPβ) expression. Conclusion Our results implied that GJ-4 might be a promising drug to improve VD through the regulation of microglial M1/M2 polarization and the subsequent inhibition of neuro-inflammation.
Collapse
Affiliation(s)
- Hui Liu
- State Key Laboratory of Bioactive Substrate and Function of Natural Medicine, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Caixia Zang
- State Key Laboratory of Bioactive Substrate and Function of Natural Medicine, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Junmei Shang
- State Key Laboratory of Bioactive Substrate and Function of Natural Medicine, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zihong Zhang
- State Key Laboratory of Bioactive Substrate and Function of Natural Medicine, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Lu Wang
- State Key Laboratory of Bioactive Substrate and Function of Natural Medicine, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Hanyu Yang
- State Key Laboratory of Bioactive Substrate and Function of Natural Medicine, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Chanjuan Sheng
- State Key Laboratory of Bioactive Substrate and Function of Natural Medicine, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Fangyu Yuan
- State Key Laboratory of Bioactive Substrate and Function of Natural Medicine, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Cheng Ju
- State Key Laboratory of Bioactive Substrate and Function of Natural Medicine, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Fangyuan Li
- State Key Laboratory of Bioactive Substrate and Function of Natural Medicine, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yang Yu
- Institute of TCM & Natural Products College of Pharmacy, Jinan University, Guangzhou, China
| | - Xinsheng Yao
- Institute of TCM & Natural Products College of Pharmacy, Jinan University, Guangzhou, China
| | - Xiuqi Bao
- State Key Laboratory of Bioactive Substrate and Function of Natural Medicine, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Xiuqi Bao and Dan Zhang, State Key Laboratory of Bioactive Substrate and Function of Natural Medicine, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, 1 Xian Nong Tan Street, Beijing 100050 China ;
| | - Dan Zhang
- State Key Laboratory of Bioactive Substrate and Function of Natural Medicine, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Xiuqi Bao and Dan Zhang, State Key Laboratory of Bioactive Substrate and Function of Natural Medicine, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, 1 Xian Nong Tan Street, Beijing 100050 China ;
| |
Collapse
|
27
|
Wang H, Chen M, Zhang T, Gao Z, Gong Y, Yu X, Wu H. Recombinant human erythropoietin upregulates PPARγ through the PI3K/Akt pathway to protect neurons in rats subjected to oxidative stress. Eur J Neurosci 2022; 56:4045-4059. [PMID: 35678781 DOI: 10.1111/ejn.15735] [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: 10/09/2021] [Revised: 05/01/2022] [Accepted: 06/03/2022] [Indexed: 11/26/2022]
Abstract
In vitro cell experiments have suggested that recombinant human erythropoietin (rhEPO) and peroxisome proliferator activated receptor γ (PPARγ) activation exert protective effects on neurons. This study observed the learning and memory ability, antioxidant capacity and the ratio of apoptotic cells after rhEPO intervention and investigated the relationship among rhEPO, PI3K/Akt and PPARγ in the anti-neural oxidative stress injury process in vivo. The results showed that rhEPO significantly improved the learning and memory abilities of rats subjected to oxidative stress, enhanced the antioxidant capacity of cells, and reduced neuronal apoptosis. Then, the PI3K/Akt and PPARγ pathways were inhibited, and TUNEL staining were used to observe the changes in the effect of rhEPO. After the PI3K/Akt and PPARγ pathways were inhibited, the effect of rhEPO on rats subjected to oxidative stress was significantly weakened, suggesting that both the PI3K/Akt and PPARγ pathways are involved in the process by which rhEPO protects neurons. Finally, Western blotting and immunofluorescence staining were used to observe the changes in PI3K/Akt and PPARγ signalling proteins in the neurons after the rhEPO intervention and to explore the relationship among the three. The results showed that rhEPO significantly increased the levels of the p-Akt and PPARγ proteins and the level of the PPARγ protein in the nucleus, indicating that the PI3K/Akt pathway was located upstream of and regulates PPARγ. In conclusion, this study suggested that rhEPO activates the PI3K/Akt to upregulate PPARγ, enhance the cellular antioxidant capacity, and protect neurons in rats subjected to oxidative stress.
Collapse
Affiliation(s)
- Huqing Wang
- Department of Neurology, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Ming Chen
- Department of Neurology, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Tao Zhang
- Department of Neurology, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Zhen Gao
- Department of Neurology, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yu Gong
- Department of Neurology, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Xiaorui Yu
- Department of Genetics and Molecular Biology, School of Medicine, Xi'an Jiaotong University, Xi'an, China
| | - Haiqin Wu
- Department of Neurology, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
28
|
rhEPO Upregulates the PPARγ Pathway in Long-term Cultured Primary Nerve Cells via PI3K/Akt to Delay Cell Senescence. J Mol Neurosci 2022; 72:1586-1597. [PMID: 35505269 DOI: 10.1007/s12031-022-01998-9] [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: 07/24/2021] [Accepted: 03/09/2022] [Indexed: 10/18/2022]
Abstract
Previous studies have confirmed that both recombinant human erythropoietin (rhEPO) and peroxisome proliferator-activated receptors γ (PPARγ) activator pioglitazone can protect senescent nerve cells, and their mechanisms involve enhancing cell antioxidant capacity and reducing cell apoptosis. However, whether the PPARγ pathway is involved in the rhEPO anti-aging process in neuronal cells is still unclear. In this study, to explore the relationship between rhEPO and the PPARγ pathway at the cellular level, primary nerve cells cultured for 22 days were used to simulate the natural aging process of nerve cells. Starting on the 11th day of culture, rhEPO, LY294002, and GW9662 were added for treatment. Immunochemical methods and SA-β-gal staining were used to observe the changes in cellular antioxidant capacity and the fraction of senescent cells. The results showed that PPARγ blockade retarded the effect of rhEPO on the cellular antioxidant capacity and altered the fraction of senescent cells. It was confirmed that PPARγ was involved in rhEPO's anti-aging process in neuronal cells. Real-time fluorescent quantitative RT-PCR, Western blotting, and immunofluorescence staining were used to observe the changes in PPARγ pathway-related factors in nerve cells after rhEPO treatment. The results showed that rhEPO significantly upregulated the expression of PPARγ coactivator-1α (PGC-1α), PPARγ, and nuclear PPARγ in cells but did not affect the level of phosphorylated PPARγ protein, confirming that rhEPO has the ability to upregulate the PPARγ pathway. PI3K/Akt and PPARγ pathway blockade experiments were used to explore the relationships among rhEPO, PI3K/Akt, and PPARγ. The results showed that after PPARγ blockade, rhEPO had no significant effect on the PI3K/Akt pathway-related factor p-Akt, while after PI3K/Akt blockade, rhEPO's effects on PPARγ-related factors (PGC-1α, PPARγ, and nuclear PPARγ) were significantly decreased. It is suggested that rhEPO delays the PI3K/Akt pathway in the process of neuronal senescence, which is located upstream of PPARγ regulation. In conclusion, this study confirmed that rhEPO can upregulate the expression of PGC-1α and PPARγ in cells and the level of PPARγ protein in the nucleus to enhance the antioxidant capacity of cells and delay the senescence of nerve cells through the PI3K/Akt pathway. These findings will provide ideas for finding new targets for neuroprotection research and will also provide a theoretical basis and experimental evidence for rhEPO anti-aging research in neural cells.
Collapse
|
29
|
Rai SN, Tiwari N, Singh P, Mishra D, Singh AK, Hooshmandi E, Vamanu E, Singh MP. Therapeutic Potential of Vital Transcription Factors in Alzheimer's and Parkinson's Disease With Particular Emphasis on Transcription Factor EB Mediated Autophagy. Front Neurosci 2022; 15:777347. [PMID: 34970114 PMCID: PMC8712758 DOI: 10.3389/fnins.2021.777347] [Citation(s) in RCA: 60] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Accepted: 11/22/2021] [Indexed: 12/12/2022] Open
Abstract
Autophagy is an important cellular self-digestion and recycling pathway that helps in maintaining cellular homeostasis. Dysregulation at various steps of the autophagic and endolysosomal pathway has been reported in several neurodegenerative disorders such as Alzheimer's disease (AD), Parkinson's disease (PD), and Huntington disease (HD) and is cited as a critically important feature for central nervous system (CNS) proteostasis. Recently, another molecular target, namely transcription factor EB (TFEB) has been explored globally to treat neurodegenerative disorders. This TFEB, is a key regulator of autophagy and lysosomal biogenesis pathway. Multiple research studies suggested therapeutic potential by targeting TFEB to treat human diseases involving autophagy-lysosomal dysfunction, especially neurodegenerative disorders. A common observation involving all neurodegenerative disorders is their poor efficacy in clearing and recycle toxic aggregated proteins and damaged cellular organelles due to impairment in the autophagy pathway. This dysfunction in autophagy characterized by the accumulation of toxic protein aggregates leads to a progressive loss in structural integrity/functionality of neurons and may even result in neuronal death. In recent years TFEB, a key regulator of autophagy and lysosomal biogenesis, has received considerable attention. It has emerged as a potential therapeutic target in numerous neurodegenerative disorders like AD and PD. In various neurobiology studies involving animal models, TFEB has been found to ameliorate neurotoxicity and rescue neurodegeneration. Since TFEB is a master transcriptional regulator of autophagy and lysosomal biogenesis pathway and plays a crucial role in defining autophagy activation. Studies have been done to understand the mechanisms for TFEB dysfunction, which may yield insights into how TFEB might be targeted and used for the therapeutic strategy to develop a treatment process with extensive application to neurodegenerative disorders. In this review, we explore the role of different transcription factor-based targeted therapy by some natural compounds for AD and PD with special emphasis on TFEB.
Collapse
Affiliation(s)
| | - Neeraj Tiwari
- Faculty of Biosciences, Institute of Biosciences and Technology, Shri Ramswaroop Memorial University, Barabanki, India
| | - Payal Singh
- Department of Zoology, Mahila Mahavidyalaya, Banaras Hindu University, Varanasi, India
| | - Divya Mishra
- Centre of Bioinformatics, University of Allahabad, Prayagraj, India
| | - Anurag Kumar Singh
- Centre of Experimental Medicine and Surgery, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Etrat Hooshmandi
- Clinical Neurology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Emanuel Vamanu
- Faculty of Biotechnology, University of Agronomic Science and Veterinary Medicine, Bucharest, Romania
| | - Mohan P Singh
- Centre of Biotechnology, University of Allahabad, Prayagraj, India
| |
Collapse
|
30
|
Hassan FE, Sakr HI, Mohie PM, Suliman HS, Mohamed AS, Attia MH, Eid DM. Pioglitazone improves skeletal muscle functions in reserpine-induced fibromyalgia rat model. Ann Med 2021; 53:1032-1040. [PMID: 34233552 PMCID: PMC8274527 DOI: 10.1080/07853890.2021.1916069] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 02/24/2021] [Accepted: 04/03/2021] [Indexed: 01/24/2023] Open
Abstract
BACKGROUND Fibromyalgia (FM) is characterized by musculoskeletal pain, fatigue, sleep and memory disturbance. There is no definitive cure yet for FM-related health problems. Peroxisome proliferator-activated receptor's (PPAR's) activation is associated with insulin sensitisation and improved glucose metabolism. PPAR-γ was reported to alleviate FM allodynia. Limited data are discussing its effect on motor disorders. OBJECTIVE To investigate the potential effect of PPAR-γ agonists (pioglitazone, as one member of thiazolidinediones (TZD)) on motor dysfunction in reserpine-induced FM in a rat model. MATERIALS AND METHODS Thirty-six male Wistar rats were divided into negative control (n = 9) and reserpine-induced FM (n = 27) groups. The latter was subdivided into three equal subgroups (n = 9), positive control (untreated FM model), pioglitazone-treated and GW9662-treated. We evaluated muscle functions and activity of chloramphenicol acetyltransferase, superoxide dismutase, malondialdehyde, and serum levels of interleukin-8 and monocyte chemoattractant protein-1. RESULTS Pioglitazone significantly relieved fatigue, improved muscle performance, reduced inflammatory cytokines and enhanced antioxidant's activity, while GW9662, a known PPAR-γ antagonist, aggravated the FM manifestations in the rat model. CONCLUSION PPAR-γ agonists show a promising role against FM-associated motor dysfunctions.
Collapse
Affiliation(s)
- Fatma E. Hassan
- Department of Medical Physiology, Faculty of Medicine, Cairo University, Egypt
| | - Hader I. Sakr
- Department of Medical Physiology, Faculty of Medicine, Cairo University, Egypt
- Department of Medical Physiology, Batterjee Medical College, Jeddah, Saudi Arabia
| | - Passant M. Mohie
- Department of Clinical Pharmacology, Faculty of Medicine, Alexandria University, Egypt
| | - Howayda Saeed Suliman
- Department of Medical Biochemistry, Faculty of Medicine, Alexandria University, Egypt
| | | | - Mohamed H. Attia
- Internal Medicine Department, Faculty of Medicine, Ain Shams University, Egypt
| | - Dalia M. Eid
- Department of Biochemistry, Faculty of Medicine, Ain Shams University, Egypt
| |
Collapse
|
31
|
Terzo S, Amato A, Mulè F. From obesity to Alzheimer's disease through insulin resistance. J Diabetes Complications 2021; 35:108026. [PMID: 34454830 DOI: 10.1016/j.jdiacomp.2021.108026] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2021] [Revised: 07/29/2021] [Accepted: 08/14/2021] [Indexed: 12/27/2022]
Abstract
Alzheimer's disease is one of the most frequent forms of dementia. It is a progressive neurodegenerative disease, characterized by presence of amyloid plaques and neurofibrillary tangles in the brain. Obesity is regarded as abnormal fat accumulation with deleterious impact on human health. There is full scientific evidence that obesity and the metabolic comorbidities (e.g., insulin resistance, hyperglycaemia, and type 2 diabetes) are related to Alzheimer's disease and likely in the causative pathway. Numerous studies have identified several overlapping neurodegenerative mechanisms, including oxidative stress, mitochondrial dysfunction, and inflammation. In this review, we present how obesity and the associated lipotoxicity as well as chronic inflammation initiate a state of insulin resistance that in turn, may have a role in causing the characteristic cerebral alterations of AD. In particular, we focus on the molecular mechanisms linking the obesity-induced impairment in insulin signalling to the upregulation of Aβ aggregation, tau hyper-phosphorylation, inflammation, oxidative stress and mitochondrial dysfunction.
Collapse
Affiliation(s)
- Simona Terzo
- Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche (STEBICEF), Università di Palermo, 90128, Italy.
| | - Antonella Amato
- Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche (STEBICEF), Università di Palermo, 90128, Italy.
| | - Flavia Mulè
- Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche (STEBICEF), Università di Palermo, 90128, Italy.
| |
Collapse
|
32
|
Prowse N, Hayley S. Microglia and BDNF at the crossroads of stressor related disorders: Towards a unique trophic phenotype. Neurosci Biobehav Rev 2021; 131:135-163. [PMID: 34537262 DOI: 10.1016/j.neubiorev.2021.09.018] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 09/08/2021] [Accepted: 09/08/2021] [Indexed: 12/16/2022]
Abstract
Stressors ranging from psychogenic/social to neurogenic/injury to systemic/microbial can impact microglial inflammatory processes, but less is known regarding their effects on trophic properties of microglia. Recent studies do suggest that microglia can modulate neuronal plasticity, possibly through brain derived neurotrophic factor (BDNF). This is particularly important given the link between BDNF and neuropsychiatric and neurodegenerative pathology. We posit that certain activated states of microglia play a role in maintaining the delicate balance of BDNF release onto neuronal synapses. This focused review will address how different "activators" influence the expression and release of microglial BDNF and address the question of tropomyosin receptor kinase B (TrkB) expression on microglia. We will then assess sex-based differences in microglial function and BDNF expression, and how microglia are involved in the stress response and related disorders such as depression. Drawing on research from a variety of other disorders, we will highlight challenges and opportunities for modulators that can shift microglia to a "trophic" phenotype with a view to potential therapeutics relevant for stressor-related disorders.
Collapse
Affiliation(s)
- Natalie Prowse
- Department of Neuroscience, Carleton University, Ottawa, ON K1S 5B6, Canada.
| | - Shawn Hayley
- Department of Neuroscience, Carleton University, Ottawa, ON K1S 5B6, Canada.
| |
Collapse
|
33
|
Wei Z, Koya J, Reznik SE. Insulin Resistance Exacerbates Alzheimer Disease via Multiple Mechanisms. Front Neurosci 2021; 15:687157. [PMID: 34349617 PMCID: PMC8326507 DOI: 10.3389/fnins.2021.687157] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 06/23/2021] [Indexed: 12/12/2022] Open
Abstract
Alzheimer disease (AD) is a chronic neurodegenerative disease that accounts for 60–70% of dementia and is the sixth leading cause of death in the United States. The pathogenesis of this debilitating disorder is still not completely understood. New insights into the pathogenesis of AD are needed in order to develop novel pharmacologic approaches. In recent years, numerous studies have shown that insulin resistance plays a significant role in the development of AD. Over 80% of patients with AD have type II diabetes (T2DM) or abnormal serum glucose, suggesting that the pathogenic mechanisms of insulin resistance and AD likely overlap. Insulin resistance increases neuroinflammation, which promotes both amyloid β-protein deposition and aberrant tau phosphorylation. By increasing production of reactive oxygen species, insulin resistance triggers amyloid β-protein accumulation. Oxidative stress associated with insulin resistance also dysregulates glycogen synthase kinase 3-β (GSK-3β), which leads to increased tau phosphorylation. Both insulin and amyloid β-protein are metabolized by insulin degrading enzyme (IDE). Defects in this enzyme are the basis for a strong association between T2DM and AD. This review highlights multiple pathogenic mechanisms induced by insulin resistance that are implicated in AD. Several pharmacologic approaches to AD associated with insulin resistance are presented.
Collapse
Affiliation(s)
- Zenghui Wei
- Department of Pharmaceutical Sciences, St. John's University, New York, NY, United States
| | - Jagadish Koya
- Department of Pharmaceutical Sciences, St. John's University, New York, NY, United States
| | - Sandra E Reznik
- Department of Pharmaceutical Sciences, St. John's University, New York, NY, United States.,Department of Pathology, Albert Einstein College of Medicine, New York, NY, United States.,Department of Obstetrics and Gynecology and Women's Health, Albert Einstein College of Medicine, New York, NY, United States
| |
Collapse
|
34
|
Wong LR, Tan EA, Lim MEJ, Shen W, Lian XL, Wang Y, Chen L, Ho PCL. Functional effects of berberine in modulating mitochondrial dysfunction and inflammatory response in the respective amyloidogenic cells and activated microglial cells - In vitro models simulating Alzheimer's disease pathology. Life Sci 2021; 282:119824. [PMID: 34265361 DOI: 10.1016/j.lfs.2021.119824] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 07/03/2021] [Accepted: 07/07/2021] [Indexed: 12/09/2022]
Abstract
AIM Berberine (BBR) is an alkaloid extracted from Coptidis Rhizoma, also known as Huang-Lian. Huang-Lian has been used extensively in traditional Chinese medicine for the treatment of various diseases, including diabetes and dementia. Because Alzheimer's disease (AD) is a complex disease that involves various pathophysiological changes, the diverse neuroprotective effects of BBR may be useful for improving the brain's energy state at an early stage of the disease. MAIN METHODS We performed extracellular flux and 1H NMR-based metabolic profiling analyses to investigate the effects of BBR on metabolic processes in these cells. Pioglitazone (PIO), a peroxisome proliferator-activated receptor-γ (PPARγ) agonist has been studied extensively for the treatment of AD. We explored the combination dosing effects of BBR and PIO in vitro, then leveraged computational methods to explain the experimental finding. KEY FINDINGS BBR demonstrates potential in modulating the mitochondrial bioenergetics and attenuating dysfunction of the primary energy and glutathione metabolism pathways in an AD cell model. It also suppresses basal respiration and reduces the production of pro-inflammatory cytokines in activated microglial cells. Both experimental and computational observations indicate that BBR and PIO have comparable binding affinities to the PPARγ protein, suggesting both drugs may have some overlapping effects for AD. SIGNIFICANCE BBR exerts beneficial effects on disrupted metabolic processes in amyloidogenic cells and activated microglial cells, which are important for preventing or delaying early-stage disease progression. The choice of BBR or PIO for AD treatment depends on their respective pharmacokinetic profiles, delivery, efficacy and safety, and warrants further study.
Collapse
Affiliation(s)
- Ling Rong Wong
- Department of Pharmacy, Faculty of Science, National University of Singapore, 18 Science Drive 4, Singapore 117559, Republic of Singapore
| | - Edwin Aik Tan
- Department of Pharmacy, Faculty of Science, National University of Singapore, 18 Science Drive 4, Singapore 117559, Republic of Singapore
| | - Ming En Joshua Lim
- Department of Pharmacy, Faculty of Science, National University of Singapore, 18 Science Drive 4, Singapore 117559, Republic of Singapore
| | - Wanxiang Shen
- Department of Pharmacy, Faculty of Science, National University of Singapore, 18 Science Drive 4, Singapore 117559, Republic of Singapore
| | - Xin Le Lian
- Department of Pharmacy, Faculty of Science, National University of Singapore, 18 Science Drive 4, Singapore 117559, Republic of Singapore
| | - Yali Wang
- Department of Pharmacy, Faculty of Science, National University of Singapore, 18 Science Drive 4, Singapore 117559, Republic of Singapore
| | - Lu Chen
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, 611137 Chengdu, Sichuan, People's Republic of China.
| | - Paul Chi-Lui Ho
- Department of Pharmacy, Faculty of Science, National University of Singapore, 18 Science Drive 4, Singapore 117559, Republic of Singapore.
| |
Collapse
|
35
|
Liu CH, Sung PS, Li YR, Huang WK, Lee TW, Huang CC, Lee TH, Chen TH, Wei YC. Telmisartan use and risk of dementia in type 2 diabetes patients with hypertension: A population-based cohort study. PLoS Med 2021; 18:e1003707. [PMID: 34280191 PMCID: PMC8289120 DOI: 10.1371/journal.pmed.1003707] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Accepted: 06/22/2021] [Indexed: 11/21/2022] Open
Abstract
BACKGROUND Angiotensin receptor blockers (ARBs) may have protective effects against dementia occurrence in patients with hypertension (HTN). However, whether telmisartan, an ARB with peroxisome proliferator-activated receptor γ (PPAR-γ)-modulating effects, has additional benefits compared to other ARBs remains unclear. METHODS AND FINDINGS Between 1997 and 2013, 2,166,944 type 2 diabetes mellitus (T2DM) patients were identified from the National Health Insurance Research Database of Taiwan. Patients with HTN using ARBs were included in the study. Patients with a history of stroke, traumatic brain injury, or dementia were excluded. Finally, 65,511 eligible patients were divided into 2 groups: the telmisartan group and the non-telmisartan ARB group. Propensity score matching (1:4) was used to balance the distribution of baseline characteristics and medications. The primary outcome was the diagnosis of dementia. The secondary outcomes included the diagnosis of Alzheimer disease and occurrence of symptomatic ischemic stroke (IS), any IS, and all-cause mortality. The risks between groups were compared using a Cox proportional hazard model. Statistical significance was set at p < 0.05. There were 2,280 and 9,120 patients in the telmisartan and non-telmisartan ARB groups, respectively. Patients in the telmisartan group had a lower risk of dementia diagnosis (telmisartan versus non-telmisartan ARBs: 2.19% versus 3.20%; HR, 0.72; 95% CI, 0.53 to 0.97; p = 0.030). They also had lower risk of dementia diagnosis with IS as a competing risk (subdistribution HR, 0.70; 95% CI, 0.51 to 0.95; p = 0.022) and with all-cause mortality as a competing risk (subdistribution HR, 0.71; 95% CI, 0.53 to 0.97; p = 0.029). In addition, the telmisartan users had a lower risk of any IS (6.84% versus 8.57%; HR, 0.79; 95% CI, 0.67 to 0.94; p = 0.008) during long-term follow-up. Study limitations included potential residual confounding by indication, interpretation of causal effects in an observational study, and bias caused by using diagnostic and medication codes to represent real clinical data. CONCLUSIONS The current study suggests that telmisartan use in hypertensive T2DM patients may be associated with a lower risk of dementia and any IS events in an East-Asian population.
Collapse
Affiliation(s)
- Chi-Hung Liu
- Department of Neurology, Linkou Chang Gung Memorial Hospital, Taoyuan, Taiwan
- College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Pi-Shan Sung
- Department of Neurology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Yan-Rong Li
- College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Linkou Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Wen-Kuan Huang
- College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Division of Hematology-Oncology, Department of Internal Medicine, Linkou Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Tay-Wey Lee
- Biostatistical Consultation Center, Keelung Chang Gung Memorial Hospital, Keelung, Taiwan
| | - Chin-Chang Huang
- Department of Neurology, Linkou Chang Gung Memorial Hospital, Taoyuan, Taiwan
- College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Tsong-Hai Lee
- Department of Neurology, Linkou Chang Gung Memorial Hospital, Taoyuan, Taiwan
- College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Tien-Hsing Chen
- Division of Cardiology, Department of Internal Medicine, Keelung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Keelung, Taiwan
- * E-mail: (THC); (YCW)
| | - Yi-Chia Wei
- Department of Neurology, Keelung Chang Gung Memorial Hospital, Keelung, Taiwan
- Institute of Neuroscience, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Community Medicine Research Center, Keelung Chang Gung Memorial Hospital, Keelung, Taiwan
- * E-mail: (THC); (YCW)
| |
Collapse
|
36
|
Xu Y, Kong J, Hu P. Computational Drug Repurposing for Alzheimer's Disease Using Risk Genes From GWAS and Single-Cell RNA Sequencing Studies. Front Pharmacol 2021; 12:617537. [PMID: 34276354 PMCID: PMC8277916 DOI: 10.3389/fphar.2021.617537] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Accepted: 06/15/2021] [Indexed: 01/14/2023] Open
Abstract
Background: Traditional therapeutics targeting Alzheimer's disease (AD)-related subpathologies have so far proved ineffective. Drug repurposing, a more effective strategy that aims to find new indications for existing drugs against other diseases, offers benefits in AD drug development. In this study, we aim to identify potential anti-AD agents through enrichment analysis of drug-induced transcriptional profiles of pathways based on AD-associated risk genes identified from genome-wide association analyses (GWAS) and single-cell transcriptomic studies. Methods: We systematically constructed four gene lists (972 risk genes) from GWAS and single-cell transcriptomic studies and performed functional and genes overlap analyses in Enrichr tool. We then used a comprehensive drug repurposing tool Gene2Drug by combining drug-induced transcriptional responses with the associated pathways to compute candidate drugs from each gene list. Prioritized potential candidates (eight drugs) were further assessed with literature review. Results: The genomic-based gene lists contain late-onset AD associated genes (BIN1, ABCA7, APOE, CLU, and PICALM) and clinical AD drug targets (TREM2, CD33, CHRNA2, PRSS8, ACE, TKT, APP, and GABRA1). Our analysis identified eight AD candidate drugs (ellipticine, alsterpaullone, tomelukast, ginkgolide A, chrysin, ouabain, sulindac sulfide and lorglumide), four of which (alsterpaullone, ginkgolide A, chrysin and ouabain) have shown repurposing potential for AD validated by their preclinical evidence and moderate toxicity profiles from literature. These support the value of pathway-based prioritization based on the disease risk genes from GWAS and scRNA-seq data analysis. Conclusion: Our analysis strategy identified some potential drug candidates for AD. Although the drugs still need further experimental validation, the approach may be applied to repurpose drugs for other neurological disorders using their genomic information identified from large-scale genomic studies.
Collapse
Affiliation(s)
- Yun Xu
- Department of Biochemistry and Medical Genetics, University of Manitoba, Winnipeg, MB, Canada
| | - Jiming Kong
- Department of Human Anatomy and Cell Science, University of Manitoba, Winnipeg, MB, Canada
| | - Pingzhao Hu
- Department of Biochemistry and Medical Genetics, University of Manitoba, Winnipeg, MB, Canada
| |
Collapse
|
37
|
Saunders AM, Burns DK, Gottschalk WK. Reassessment of Pioglitazone for Alzheimer's Disease. Front Neurosci 2021; 15:666958. [PMID: 34220427 PMCID: PMC8243371 DOI: 10.3389/fnins.2021.666958] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Accepted: 05/18/2021] [Indexed: 01/01/2023] Open
Abstract
Alzheimer's disease is a quintessential 'unmet medical need', accounting for ∼65% of progressive cognitive impairment among the elderly, and 700,000 deaths in the United States in 2020. In 2019, the cost of caring for Alzheimer's sufferers was $244B, not including the emotional and physical toll on caregivers. In spite of this dismal reality, no treatments are available that reduce the risk of developing AD or that offer prolonged mitiagation of its most devestating symptoms. This review summarizes key aspects of the biology and genetics of Alzheimer's disease, and we describe how pioglitazone improves many of the patholophysiological determinants of AD. We also summarize the results of pre-clinical experiments, longitudinal observational studies, and clinical trials. The results of animal testing suggest that pioglitazone can be corrective as well as protective, and that its efficacy is enhanced in a time- and dose-dependent manner, but the dose-effect relations are not monotonic or sigmoid. Longitudinal cohort studies suggests that it delays the onset of dementia in individuals with pre-existing type 2 diabetes mellitus, which small scale, unblinded pilot studies seem to confirm. However, the results of placebo-controlled, blinded clinical trials have not borne this out, and we discuss possible explanations for these discrepancies.
Collapse
Affiliation(s)
- Ann M. Saunders
- Zinfandel Pharmaceuticals, Inc., Chapel Hill, NC, United States
| | - Daniel K. Burns
- Zinfandel Pharmaceuticals, Inc., Chapel Hill, NC, United States
| | | |
Collapse
|
38
|
Muñoz-Jiménez M, Zaarkti A, García-Arnés JA, García-Casares N. Antidiabetic Drugs in Alzheimer's Disease and Mild Cognitive Impairment: A Systematic Review. Dement Geriatr Cogn Disord 2021; 49:423-434. [PMID: 33080602 DOI: 10.1159/000510677] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 07/30/2020] [Indexed: 11/19/2022] Open
Abstract
INTRODUCTION Considering that Alzheimer's disease (AD) and diabetes mellitus share pathophysiological features and AD remains with no cure, antidiabetic drugs like intranasal insulin, glitazones, metformin, and liraglutide are being tested as a potential treatment. OBJECTIVE The aim of this systematic review was to assess the efficacy of antidiabetic drugs in patients with AD, mild cognitive impairment (MCI), or subjective cognitive complaints (SCCs). Cognition was studied as the primary outcome and modulation of AD biomarkers, and imaging was also assessed as a secondary outcome. METHODS We conducted a search in the electronic databases PubMed/MEDLINE, EMBASE, and Scopus seeking clinical trials evaluating the effect on cognition of antidiabetic drugs in patients with AD, MCI, or SCCs. RESULTS A total of 23 articles were found eligible. Intranasal regular insulin improved verbal memory in most studies, especially in apoE4- patients, but results in other cognitive domains were unclear. Detemir improved cognition after 2 months of treatment, but it did not after 4 months. Pioglitazone improved cognition in diabetic patients with AD or MCI in 3 clinical trials, but it is controversial as 2 other studies did not show effect. Metformin and liraglutide showed promising results, but further research is needed as just 2 clinical trials involved each of these drugs. Almost all drugs tested were shown to modulate AD biomarkers and imaging. CONCLUSIONS Intranasal insulin, pioglitazone, metformin, and liraglutide are promising drugs that could be useful in the treatment of AD. However, many questions remain to be answered in future studies, so no particular antidiabetic drug can currently be recommended to treat AD.
Collapse
Affiliation(s)
- Mario Muñoz-Jiménez
- Department of Medicine, Faculty of Medicine, University of Málaga, Málaga, Spain
| | - Alí Zaarkti
- Department of Medicine, Faculty of Medicine, University of Málaga, Málaga, Spain
| | - Juan Antonio García-Arnés
- Department of Medicine, Faculty of Medicine, University of Málaga, Málaga, Spain.,Department of Pharmacology, Faculty of Medicine, University of Málaga, Málaga, Spain
| | - Natalia García-Casares
- Department of Medicine, Faculty of Medicine, University of Málaga, Málaga, Spain, .,Centro de Investigaciones, Médico-Sanitarias (C.I.M.ES), University of Málaga, Málaga, Spain, .,Instituto de Investigación, Biomédica de Málaga (I.B.I.M.A.), Málaga, Spain,
| |
Collapse
|
39
|
Andrés‐Benito P, Gelpi E, Jové M, Mota‐Martorell N, Obis È, Portero‐Otin M, Povedano M, Pujol A, Pamplona R, Ferrer I. Lipid alterations in human frontal cortex in ALS-FTLD-TDP43 proteinopathy spectrum are partly related to peroxisome impairment. Neuropathol Appl Neurobiol 2021; 47:544-563. [PMID: 33332650 PMCID: PMC8248144 DOI: 10.1111/nan.12681] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 10/26/2020] [Accepted: 12/13/2020] [Indexed: 01/08/2023]
Abstract
AIM Peroxisomes play a key role in lipid metabolism, and peroxisome defects have been associated with neurodegenerative diseases such as X-adrenoleukodystrophy and Alzheimer's disease. This study aims to elucidate the contribution of peroxisomes in lipid alterations of area 8 of the frontal cortex in the spectrum of TDP43-proteinopathies. Cases of frontotemporal lobar degeneration-TDP43 (FTLD-TDP), manifested as sporadic (sFTLD-TDP) or linked to mutations in various genes including expansions of the non-coding region of C9ORF72 (c9FTLD), and of sporadic amyotrophic lateral sclerosis (sALS) as the most common TDP43 proteinopathies, were analysed. METHODS We used transcriptomics and lipidomics methods to define the steady-state levels of gene expression and lipid profiles. RESULTS Our results show alterations in gene expression of some components of peroxisomes and related lipid pathways in frontal cortex area 8 in sALS, sFTLD-TDP and c9FTLD. Additionally, we identify a lipidomic pattern associated with the ALS-FTLD-TDP43 proteinopathy spectrum, notably characterised by down-regulation of ether lipids and acylcarnitine among other lipid species, as well as alterations in the lipidome of each phenotype of TDP43 proteinopathy, which reveals commonalities and disease-dependent differences in lipid composition. CONCLUSION Globally, lipid alterations in the human frontal cortex of the ALS-FTLD-TDP43 proteinopathy spectrum, which involve cell membrane composition and signalling, vulnerability against cellular stress and possible glucose metabolism, are partly related to peroxisome impairment.
Collapse
Affiliation(s)
- Pol Andrés‐Benito
- NeuropathologyBellvitge University Hospital‐Bellvitge Biomedical Research Institute (IDIBELLHospitalet de Llobregat, BarcelonaSpain
- Department of Pathology and Experimental TherapeuticsUniversity of BarcelonaBarcelonaSpain
- CIBERNED (Network Centre of Biomedical Research of Neurodegenerative DiseasesInstitute of Health Carlos IIIMinistry of Economy and CompetitivenessMadridSpain
- International Initiative for Treatment and Research Initiative to Cure ALS (TRICALSUtrechtThe Netherlands
| | - Ellen Gelpi
- Neurological Tissue Bank of the Biobanc‐Hospital Clínic‐Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPSBarcelonaSpain
- Institute of NeurologyMedical University of ViennaViennaAustria
| | - Mariona Jové
- Department of Experimental MedicineUniversity of Lleida ‐ Lleida Biomedical Research Institute (UdL‐IRBLleidaLleidaSpain
| | - Natalia Mota‐Martorell
- Department of Experimental MedicineUniversity of Lleida ‐ Lleida Biomedical Research Institute (UdL‐IRBLleidaLleidaSpain
| | - Èlia Obis
- Department of Experimental MedicineUniversity of Lleida ‐ Lleida Biomedical Research Institute (UdL‐IRBLleidaLleidaSpain
| | - Manuel Portero‐Otin
- Department of Experimental MedicineUniversity of Lleida ‐ Lleida Biomedical Research Institute (UdL‐IRBLleidaLleidaSpain
| | - Mònica Povedano
- International Initiative for Treatment and Research Initiative to Cure ALS (TRICALSUtrechtThe Netherlands
- Functional Unit of Amyotrophic Lateral Sclerosis (UFELAService of NeurologyBellvitge University HospitalHospitalet de LlobregatSpain
| | - Aurora Pujol
- Catalan Institution for Research and Advanced Studies (ICREABarcelonaSpain
- Neurometabolic Diseases LaboratoryBellvitge Biomedical Research InstituteHospital Duran i ReynalsHospitalet de Llobregat, BarcelonaSpain
- Center for Biomedical Research on Rare Diseases (CIBERERInstitute of Health Carlos IIIMadridSpain
| | - Reinald Pamplona
- Department of Experimental MedicineUniversity of Lleida ‐ Lleida Biomedical Research Institute (UdL‐IRBLleidaLleidaSpain
| | - Isidro Ferrer
- NeuropathologyBellvitge University Hospital‐Bellvitge Biomedical Research Institute (IDIBELLHospitalet de Llobregat, BarcelonaSpain
- Department of Pathology and Experimental TherapeuticsUniversity of BarcelonaBarcelonaSpain
- CIBERNED (Network Centre of Biomedical Research of Neurodegenerative DiseasesInstitute of Health Carlos IIIMinistry of Economy and CompetitivenessMadridSpain
- International Initiative for Treatment and Research Initiative to Cure ALS (TRICALSUtrechtThe Netherlands
- Institute of NeurosciencesUniversity of BarcelonaBarcelonaSpain
| |
Collapse
|
40
|
Revi N, Rengan AK. Impact of dietary polyphenols on neuroinflammation-associated disorders. Neurol Sci 2021; 42:3101-3119. [PMID: 33988799 DOI: 10.1007/s10072-021-05303-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2021] [Accepted: 05/05/2021] [Indexed: 12/21/2022]
Abstract
Neurodegenerative disorders like Alzheimer's, Parkinson's, and associated dementia typically originate with altered protein folding and aggregation of their β structures in the neurons. This self-aggregation leads to glial activation in the brain, causing neuroinflammation and leads to neuronal death. According to statistics provided by WHO, there are around 50 million people with dementia worldwide and every year, 10 million more cases are projected to increase. Also, around 5-8 percentage of people who are aged above 60 globally has dementia or associated disorders. Over 82 million in 2030 and 152 in 2050 are expected to have dementia. Most of these patients fall into low-middle-income countries which makes it even more essential to find an affordable and effective treatment method. Polyphenols of different origin are studied for their potential role as anti-neuro-inflammatory molecules. This review would summarize recent advances in three widely researched dietary polyphenols projected as potential therapeutic agents for disorders like Alzheimer's, Parkinson's, etc. They are Resveratrol, Catechins, and Tannins. The review would discuss the recent advances and challenges in using these polyphenols using specific examples as potential therapeutic agents against neuroinflammation associated disorders. An abstract of neuroinflammation-associated events and the effects by selected polyphenols.
Collapse
Affiliation(s)
- Neeraja Revi
- Department of Biotechnology, Indian Institute of Technology Hyderabad, Kandi, India
| | - Aravind Kumar Rengan
- Department of Biomedical Engineering, Indian Institute of Technology Hyderabad, Kandi, India.
| |
Collapse
|
41
|
Scheen AJ. Careful use to minimize adverse events of oral antidiabetic medications in the elderly. Expert Opin Pharmacother 2021; 22:2149-2165. [PMID: 33823723 DOI: 10.1080/14656566.2021.1912735] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
INTRODUCTION An increasing number of older patients has type 2 diabetes treated with different oral antidiabetic agents whose safety may raise concern considering some particularities of a heterogeneous elderly population. AREAS COVERED This article discusses some characteristics of older patients that could increase the risk of adverse events, with a focus on hypoglycemia. It describes the most frequent and/or severe complications reported in the elderly in both randomized controlled trials and observational studies with metformin, sulfonylureas, meglitinides, alpha-glucosidase inhibitors, thiazolidinediones, dipeptidyl peptidase-4 inhibitors (gliptins) and sodium-glucose cotransporter type 2 inhibitors (gliflozins). EXPERT OPINION Old patients may present comorbidities (renal impairment, vascular disease, heart failure, risk of dehydration, osteoporosis, cognitive dysfunction) that could increase the risk of severe adverse events. Sulfonylureas (and meglitinides) induce hypoglycemia, which may be associated with falls/fractures and cardiovascular events. Medications lacking hypoglycemia should be preferred. Gliptins appear to have the best tolerance/safety profile whereas gliflozins exert a cardiorenal protection. However, data are lacking in very old or frailty old patients so that caution and appropriate supervision of such patients are required. Taking advantage of a large choice of pharmacotherapies, personalized treatment is recommended based upon both drug safety profiles and old patient individual characteristics.
Collapse
Affiliation(s)
- André J Scheen
- Division of Clinical Pharmacology, Centre for Interdisciplinary Research on Medicines (CIRM), University of Liège, Liège, Belgium.,Division of Diabetes, Nutrition and Metabolic Disorders, Department of Medicine, CHU Liège, Liège, Belgium
| |
Collapse
|
42
|
Zhou YQ, Mei W, Tian XB, Tian YK, Liu DQ, Ye DW. The therapeutic potential of Nrf2 inducers in chronic pain: Evidence from preclinical studies. Pharmacol Ther 2021; 225:107846. [PMID: 33819559 DOI: 10.1016/j.pharmthera.2021.107846] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Accepted: 03/29/2021] [Indexed: 02/06/2023]
Abstract
Chronic pain remains an enormous health problem affecting approximatively 30% of the world's population. Opioids as the first line analgesics often leads to undesirable side effects when used long term. Therefore, novel therapeutic targets are urgently needed to the development of more efficacious analgesics. Substantial evidence indicates that excessive reactive oxygen species (ROS) are extremely important to the development of chronic pain. Nuclear factor erythroid 2-related factor 2 (Nrf2) is a master transcription factor regulating endogenous antioxidant defense. Emerging evidence suggests that Nrf2 and its downstream effectors are implicated in chronic inflammatory and neuropathic pain. Notably, controversial results have been reported regarding the expression of Nrf2 and its downstream targets in peripheral and central regions involved in pain transmission. However, our recent studies and results from other laboratories demonstrate that Nrf2 inducers exert potent analgesic effects in various murine models of chronic pain. In this review, we summarized and discussed the preclinical evidence demonstrating the therapeutic potential of Nrf2 inducers in chronic pain. These evidence indicates that Nrf2 activation are beneficial in chronic pain mostly by alleviating ROS-associated pathological processes. Overall, Nrf2-based therapy for chronic pain is an area with great promise, but more research regarding its detailed mechanisms is warranted.
Collapse
Affiliation(s)
- Ya-Qun Zhou
- Department of Anesthesiology and Pain Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Wei Mei
- Department of Anesthesiology and Pain Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Xue-Bi Tian
- Department of Anesthesiology and Pain Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yu-Ke Tian
- Department of Anesthesiology and Pain Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Dai-Qiang Liu
- Department of Anesthesiology and Pain Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| | - Da-Wei Ye
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Shanxi Medical University; Tongji Shanxi Hospital, Tongji Medical College, Huazhong University of Science and Technology, Taiyuan, 030032, China; Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
43
|
Role of insulin receptor substance-1 modulating PI3K/Akt insulin signaling pathway in Alzheimer's disease. 3 Biotech 2021; 11:179. [PMID: 33927970 DOI: 10.1007/s13205-021-02738-3] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Accepted: 03/10/2021] [Indexed: 12/12/2022] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disease, also regarded as "type 3 diabetes" for the last few years because of the brain insulin resistance (IR) and dysregulation of insulin signaling in the brain, which can further promote pathological progression of AD. IRS-1/PI3K/Akt insulin signaling pathway disorder and its downstream cascade reaction are responsible for cognitive decline in the brain. In recent years, a growing number of studies has documented that dysregulation of insulin signaling is a key feature of AD and has crucial correlations with serine/tyrosine (Ser/Tyr) phosphorylation of insulin receptor substance-1(IRS-1). Phosphorylation of this protein has been identified as an important molecule involved in the process of amyloid-β (Aβ) deposition into senile plaques (SPs) and tau hyperphosphorylation into neurofibrillary tangles (NFTs). In this paper, we review the links between IRS-1 and the PI3K/Akt insulin signaling pathway, and highlight phosphorylated IRS-1 which negatively regulated by downstream effector of Akt such as mTOR, S6K, and JNK, among others in AD. Furthermore, anti-diabetic drugs including metformin, thiazolidinediones, and glucagon-like peptide-1 (GLP-1) analogue could modulate IRS-1 phosphorylation, brain IR, PI3K/Akt insulin signaling pathway, and other pathologic processes of AD. The above suggest that anti-diabetic drugs may be promising strategies for AD disease-modifying treatments.
Collapse
|
44
|
Ashraf GM, Ebada MA, Suhail M, Ali A, Uddin MS, Bilgrami AL, Perveen A, Husain A, Tarique M, Hafeez A, Alexiou A, Ahmad A, Kumar R, Banu N, Najda A, Sayed AA, Albadrani GM, Abdel-Daim MM, Peluso I, Barreto GE. Dissecting Sex-Related Cognition between Alzheimer's Disease and Diabetes: From Molecular Mechanisms to Potential Therapeutic Strategies. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:4572471. [PMID: 33747345 PMCID: PMC7960032 DOI: 10.1155/2021/4572471] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 01/31/2021] [Accepted: 02/11/2021] [Indexed: 12/16/2022]
Abstract
The brain is a sexually dimorphic organ that implies different functions and structures depending on sex. Current pharmacological approaches against different neurological diseases act distinctly in male and female brains. In all neurodegenerative diseases, including Alzheimer's disease (AD), sex-related outcomes regarding pathogenesis, prevalence, and response to treatments indicate that sex differences are important for precise diagnosis and therapeutic strategy. Pathogenesis of AD includes vascular dementia, and in most cases, this is accompanied by metabolic complications with similar features as those assembled in diabetes. This review discusses how AD-associated dementia and diabetes affect cognition in relation to sex difference, as both diseases share similar pathological mechanisms. We highlight potential protective strategies to mitigate amyloid-beta (Aβ) pathogenesis, emphasizing how these drugs act in the male and female brains.
Collapse
Affiliation(s)
- Ghulam Md Ashraf
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Mahmoud Ahmed Ebada
- Faculty of Medicine, Zagazig University, Zagazig, El-Sharkia, Egypt
- National Hepatology and Tropical Medicine Research Institute, Cairo, Egypt
| | - Mohd Suhail
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Ashraf Ali
- Department of Sciences of Agriculture, Food, Natural Resources, and Engineering (DAFNE), University of Foggia, Via Napoli 25, 71122 Foggia, Italy
| | - Md. Sahab Uddin
- Department of Pharmacy, Southeast University, Dhaka, Bangladesh
- Pharmakon Neuroscience Research Network, Dhaka, Bangladesh
| | - Anwar L. Bilgrami
- Department of Entomology, Rutgers University, New Brunswick, NJ 018901, USA
- Deanship of Scientific Research, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Asma Perveen
- Glocal School of Life Sciences, Glocal University, Saharanpur, India
| | - Amjad Husain
- Glocal School of Life Sciences, Glocal University, Saharanpur, India
- Centre for Science and Society, IISER Bhopal, India
- Innovation and Incubation Centre for Entrepreneurship, IISER Bhopal, India
| | - Mohd Tarique
- Department of Child Health, University of Missouri, Columbia, MO 65201, USA
| | - Abdul Hafeez
- Glocal School of Pharmacy, Glocal University, Saharanpur, India
| | - Athanasios Alexiou
- Novel Global Community Educational Foundation, New South Wales, Australia
- AFNP Med Austria, Wien, Austria
| | - Ausaf Ahmad
- Amity Institute of Biotechnology, Amity University Uttar Pradesh Lucknow Campus, Uttar Pradesh, India
| | - Rajnish Kumar
- Amity Institute of Biotechnology, Amity University Uttar Pradesh Lucknow Campus, Uttar Pradesh, India
| | - Naheed Banu
- Department of Physical Therapy, College of Medical Rehabilitation, Qassim University, Buraidah, Qassim, Saudi Arabia
| | - Agnieszka Najda
- Laboratory of Quality of Vegetables and Medicinal Plants, Department of Vegetable Crops and Medicinal Plants, University of Life Sciences in Lublin, 15 Akademicka Street, 20-950 Lublin, Poland
| | - Amany A. Sayed
- Zoology Department, Faculty of Science, Cairo University, Giza 12613, Egypt
| | - Ghadeer M. Albadrani
- Department of Biology, College of Science, Princess Nourah bint Abdulrahman University, Riyadh 11474, Saudi Arabia
| | - Mohamed M. Abdel-Daim
- Pharmacology Department, Faculty of Veterinary Medicine, Suez Canal University, Ismailia 41522, Egypt
| | - Ilaria Peluso
- Research Centre for Food and Nutrition, Council for Agricultural Research and Economics (CREA-AN), 00142 Rome, Italy
| | - George E. Barreto
- Department of Biological Sciences, University of Limerick, Limerick, Ireland
- Health Research Institute, University of Limerick, Limerick, Ireland
| |
Collapse
|
45
|
Jamwal S, Blackburn JK, Elsworth JD. PPARγ/PGC1α signaling as a potential therapeutic target for mitochondrial biogenesis in neurodegenerative disorders. Pharmacol Ther 2021; 219:107705. [PMID: 33039420 PMCID: PMC7887032 DOI: 10.1016/j.pharmthera.2020.107705] [Citation(s) in RCA: 96] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Accepted: 10/05/2020] [Indexed: 12/13/2022]
Abstract
Neurodegenerative diseases represent some of the most devastating neurological disorders, characterized by progressive loss of the structure and function of neurons. Current therapy for neurodegenerative disorders is limited to symptomatic treatment rather than disease modifying interventions, emphasizing the desperate need for improved approaches. Abundant evidence indicates that impaired mitochondrial function plays a crucial role in pathogenesis of many neurodegenerative diseases and so biochemical factors in mitochondria are considered promising targets for pharmacological-based therapies. Peroxisome proliferator-activated receptors-γ (PPARγ) are ligand-inducible transcription factors involved in regulating various genes including peroxisome proliferator-activated receptor gamma co-activator-1 alpha (PGC1α). This review summarizes the evidence supporting the ability of PPARγ-PGC1α to coordinately up-regulate the expression of genes required for mitochondrial biogenesis in neurons and provide directions for future work to explore the potential benefit of targeting mitochondrial biogenesis in neurodegenerative disorders. We have highlighted key roles of NRF2, uncoupling protein-2 (UCP2), and paraoxonase-2 (PON2) signaling in mediating PGC1α-induced mitochondrial biogenesis. In addition, the status of PPARγ modulators being used in clinical trials for Parkinson's disease (PD), Alzheimer's disease (AD) and Huntington's disease (HD) has been compiled. The overall purpose of this review is to update and critique our understanding of the role of PPARγ-PGC1α-NRF2 in the induction of mitochondrial biogenesis together with suggestions for strategies to target PPARγ-PGC1α-NRF2 signaling in order to combat mitochondrial dysfunction in neurodegenerative disorders.
Collapse
Affiliation(s)
- Sumit Jamwal
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT 06511, USA
| | - Jennifer K Blackburn
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT 06511, USA
| | - John D Elsworth
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT 06511, USA.
| |
Collapse
|
46
|
Wang YZ, Meng L, Zhuang QS, Shen L. Screening Traditional Chinese Medicine Combination for Cotreatment of Alzheimer's Disease and Type 2 Diabetes Mellitus by Network Pharmacology. J Alzheimers Dis 2021; 80:787-797. [PMID: 33579846 DOI: 10.3233/jad-201336] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
BACKGROUND In recent years, the efficacy of type 2 diabetes mellitus (T2DM) drugs in the treatment of Alzheimer's disease (AD) has attracted extensive interest owing to the close associations between the two diseases. OBJECTIVE Here, we screened traditional Chinese medicine (TCM) and multi-target ingredients that may have potential therapeutic effects on both T2DM and AD from T2DM prescriptions. METHODS Network pharmacology and molecular docking were used. RESULTS Firstly, the top 10 frequently used herbs and corresponding 275 active ingredients were identified from 263 T2DM-related TCM prescriptions. Secondly, through the comparative analysis of 208 potential targets of ingredients, 1,740 T2DM-related targets, and 2,060 AD-related targets, 61 common targets were identified to be shared. Thirdly, by constructing pharmacological network, 26 key targets and 154 representative ingredients were identified. Further enrichment analysis showed that common targets were involved in regulating multiple pathways related to T2DM and AD, while network analysis also found that the combination of Danshen (Radix Salviae)-Gancao (Licorice)-Shanyao (Rhizoma Dioscoreae) contained the vast majority of the representative ingredients and might be potential for the cotreatment of the two diseases. Fourthly, MAPK1, PPARG, GSK3B, BACE1, and NR3C1 were selected as potential targets for virtual screening of multi-target ingredients. Further docking studies showed that multiple natural compounds, including salvianolic acid J, gancaonin H, gadelaidic acid, icos-5-enoic acid, and sigmoidin-B, exhibited high binding affinities with the five targets. CONCLUSION To summarize, the present study provides a potential TCM combination that might possess the potential advantage of cotreatment of AD and T2DM.
Collapse
Affiliation(s)
- Yi-Zhen Wang
- Institute of Biomedical Research, Shandong University of Technology, Zibo, Shandong, People's Republic of China.,Shandong Provincial Research Center for Bioinformatic Engineering and Technique, School of Life Sciences, Shandong University of Technology, Zibo, Shandong, People's Republic of China
| | - Lei Meng
- Institute of Biomedical Research, Shandong University of Technology, Zibo, Shandong, People's Republic of China.,Shandong Provincial Research Center for Bioinformatic Engineering and Technique, School of Life Sciences, Shandong University of Technology, Zibo, Shandong, People's Republic of China
| | - Qi-Shuai Zhuang
- Institute of Biomedical Research, Shandong University of Technology, Zibo, Shandong, People's Republic of China.,Shandong Provincial Research Center for Bioinformatic Engineering and Technique, School of Life Sciences, Shandong University of Technology, Zibo, Shandong, People's Republic of China
| | - Liang Shen
- Institute of Biomedical Research, Shandong University of Technology, Zibo, Shandong, People's Republic of China.,Shandong Provincial Research Center for Bioinformatic Engineering and Technique, School of Life Sciences, Shandong University of Technology, Zibo, Shandong, People's Republic of China
| |
Collapse
|
47
|
Yu CC, Du YJ, Wang SQ, Liu LB, Shen F, Wang L, Lin YF, Kong LH. Experimental Evidence of the Benefits of Acupuncture for Alzheimer's Disease: An Updated Review. Front Neurosci 2021; 14:549772. [PMID: 33408601 PMCID: PMC7779610 DOI: 10.3389/fnins.2020.549772] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Accepted: 11/18/2020] [Indexed: 12/15/2022] Open
Abstract
As the global population ages, the prevalence of Alzheimer's disease (AD), the most common form of dementia, is also increasing. At present, there are no widely recognized drugs able to ameliorate the cognitive dysfunction caused by AD. The failure of several promising clinical trials in recent years has highlighted the urgent need for novel strategies to both prevent and treat AD. Notably, a growing body of literature supports the efficacy of acupuncture for AD. In this review, we summarize the previously reported mechanisms of acupuncture's beneficial effects in AD, including the ability of acupuncture to modulate Aβ metabolism, tau phosphorylation, neurotransmitters, neurogenesis, synapse and neuron function, autophagy, neuronal apoptosis, neuroinflammation, cerebral glucose metabolism, and brain responses. Taken together, these findings suggest that acupuncture provides therapeutic effects for AD.
Collapse
Affiliation(s)
- Chao-Chao Yu
- Department of Tuina, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, China.,The Fourth Clinical College of Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Yan-Jun Du
- College of Acupuncture and Orthopedics, Hubei University of Chinese Medicine, Wuhan, China
| | - Shu-Qin Wang
- Department of Tuina, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, China.,The Fourth Clinical College of Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Le-Bin Liu
- Department of Rehabilitation Medicine, Hubei Rongjun Hospital, Wuhan, China
| | - Feng Shen
- College of Acupuncture and Orthopedics, Hubei University of Chinese Medicine, Wuhan, China
| | - Li Wang
- College of Acupuncture and Orthopedics, Hubei University of Chinese Medicine, Wuhan, China
| | - Yuan-Fang Lin
- Department of Tuina, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, China.,The Fourth Clinical College of Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Li-Hong Kong
- College of Acupuncture and Orthopedics, Hubei University of Chinese Medicine, Wuhan, China
| |
Collapse
|
48
|
Christofides A, Konstantinidou E, Jani C, Boussiotis VA. The role of peroxisome proliferator-activated receptors (PPAR) in immune responses. Metabolism 2021; 114:154338. [PMID: 32791172 PMCID: PMC7736084 DOI: 10.1016/j.metabol.2020.154338] [Citation(s) in RCA: 292] [Impact Index Per Article: 97.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 07/06/2020] [Accepted: 07/24/2020] [Indexed: 02/07/2023]
Abstract
Peroxisome proliferator-activated receptors (PPARs) are fatty acid-activated transcription factors of nuclear hormone receptor superfamily that regulate energy metabolism. Currently, three PPAR subtypes have been identified: PPARα, PPARγ, and PPARβ/δ. PPARα and PPARδ are highly expressed in oxidative tissues and regulate genes involved in substrate delivery and oxidative phosphorylation (OXPHOS) and regulation of energy homeostasis. In contrast, PPARγ is more important in lipogenesis and lipid synthesis, with highest expression levels in white adipose tissue (WAT). In addition to tissues regulating whole body energy homeostasis, PPARs are expressed in immune cells and have an emerging critical role in immune cell differentiation and fate commitment. In this review, we discuss the actions of PPARs in the function of the innate and the adaptive immune system and their implications in immune-mediated inflammatory conditions.
Collapse
Affiliation(s)
- Anthos Christofides
- Division of Hematology-Oncology, Harvard Medical School, Boston, MA 02215, United States of America; Department of Medicine, Harvard Medical School, Boston, MA 02215, United States of America; Cancer Center, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, United States of America
| | - Eirini Konstantinidou
- Division of Hematology-Oncology, Harvard Medical School, Boston, MA 02215, United States of America; Department of Medicine, Harvard Medical School, Boston, MA 02215, United States of America; Cancer Center, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, United States of America
| | - Chinmay Jani
- Department of Medicine, Harvard Medical School, Boston, MA 02215, United States of America; Department of Medicine, Mt. Auburn Hospital, Cambridge, MA 02138, United States of America
| | - Vassiliki A Boussiotis
- Division of Hematology-Oncology, Harvard Medical School, Boston, MA 02215, United States of America; Department of Medicine, Harvard Medical School, Boston, MA 02215, United States of America; Cancer Center, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, United States of America.
| |
Collapse
|
49
|
O’Bryant SE, Zhang F, Petersen M, Johnson L, Hall J, Rissman RA. A Precision Medicine Approach to Treating Alzheimer's Disease Using Rosiglitazone Therapy: A Biomarker Analysis of the REFLECT Trials. J Alzheimers Dis 2021; 81:557-568. [PMID: 33814447 PMCID: PMC8203239 DOI: 10.3233/jad-201610] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/01/2021] [Indexed: 12/14/2022]
Abstract
BACKGROUND The REFLECT trials were conducted to examine the treatment of mild-to-moderate Alzheimer's disease utilizing a peroxisome proliferator-activated receptor gamma agonist. OBJECTIVE To generate a predictive biomarker indicative of positive treatment response using samples from the previously conducted REFLECT trials. METHODS Data were analyzed on 360 participants spanning multiple negative REFLECT trials, which included treatment with rosiglitazone and rosiglitazone XR. Support vector machine analyses were conducted to generate a predictive biomarker profile. RESULTS A pre-defined 6-protein predictive biomarker (IL6, IL10, CRP, TNFα, FABP-3, and PPY) correctly classified treatment response with 100%accuracy across study arms for REFLECT Phase II trial (AVA100193) and multiple Phase III trials (AVA105640, AV102672, and AVA102670). When the data was combined across all rosiglitazone trial arms, a global RSG-predictive biomarker with the same 6-protein predictive biomarker was able to accurately classify 98%of treatment responders. CONCLUSION A predictive biomarker comprising of metabolic and inflammatory markers was highly accurate in identifying those patients most likely to experience positive treatment response across the REFLECT trials. This study provides additional proof-of-concept that a predictive biomarker can be utilized to help with screening and predicting treatment response, which holds tremendous benefit for clinical trials.
Collapse
Affiliation(s)
- Sid E. O’Bryant
- Institute for Translational Research, University of North Texas Health Science Center, Fort Worth, TX, USA
- Department of Pharmacology & Neuroscience, University of North Texas Health Science Center, Fort Worth, TX, USA
| | - Fan Zhang
- Institute for Translational Research, University of North Texas Health Science Center, Fort Worth, TX, USA
- Department of Family Medicine, University of North Texas Health Science Center, Fort Worth, TX, USA
| | - Melissa Petersen
- Institute for Translational Research, University of North Texas Health Science Center, Fort Worth, TX, USA
- Department of Family Medicine, University of North Texas Health Science Center, Fort Worth, TX, USA
| | - Leigh Johnson
- Institute for Translational Research, University of North Texas Health Science Center, Fort Worth, TX, USA
- Department of Pharmacology & Neuroscience, University of North Texas Health Science Center, Fort Worth, TX, USA
| | - James Hall
- Institute for Translational Research, University of North Texas Health Science Center, Fort Worth, TX, USA
- Department of Pharmacology & Neuroscience, University of North Texas Health Science Center, Fort Worth, TX, USA
| | - Robert A. Rissman
- Department of Family Medicine, University of North Texas Health Science Center, Fort Worth, TX, USA
- Department of Neurosciences, University of California San Diego, San Diego, CA, USA
| |
Collapse
|
50
|
The peroxisome proliferator-activated receptor gamma (PPARγ) agonist, rosiglitazone, ameliorates neurofunctional and neuroinflammatory abnormalities in a rat model of Gulf War Illness. PLoS One 2020; 15:e0242427. [PMID: 33186383 PMCID: PMC7665704 DOI: 10.1371/journal.pone.0242427] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Accepted: 11/03/2020] [Indexed: 12/12/2022] Open
Abstract
Background Gulf War (GW) Illness (GWI) is a debilitating condition with a complex constellation of immune, endocrine and neurological symptoms, including cognitive impairment, anxiety and depression. We studied a novel model of GWI based on 3 known common GW exposures (GWE): (i) intranasal lipopolysaccharide, to which personnel were exposed during desert sand storms; (ii) pyridostigmine bromide, used as prophylaxis against chemical warfare; and (iii) chronic unpredictable stress, an inescapable element of war. We used this model to evaluate prophylactic treatment with the PPARγ agonist, rosiglitazone (ROSI). Methods Rats were subjected to the three GWE for 33 days. In series 1 and 2, male and female GWE-rats were compared to naïve rats. In series 3, male rats with GWE were randomly assigned to prophylactic treatment with ROSI (GWE-ROSI) or vehicle. After the 33-day exposures, three neurofunctional domains were evaluated: cognition (novel object recognition), anxiety-like behaviors (elevated plus maze, open field) and depression-like behaviors (coat state, sucrose preference, splash test, tail suspension and forced swim). Brains were analyzed for astrocytic and microglial activation and neuroinflammation (GFAP, Iba1, tumor necrosis factor and translocator protein). Neurofunctional data from rats with similar exposures were pooled into 3 groups: naïve, GWE and GWE-ROSI. Results Compared to naïve rats, GWE-rats showed significant abnormalities in the three neurofunctional domains, along with significant neuroinflammation in amygdala and hippocampus. There were no differences between males and females with GWE. GWE-ROSI rats showed significant attenuation of neuroinflammation and of some of the neurofunctional abnormalities. Conclusion This novel GWI model recapitulates critical neurofunctional abnormalities reported by Veterans with GWI. Concurrent prophylactic treatment with ROSI was beneficial in this model.
Collapse
|