1
|
Rhea EM, Leclerc M, Yassine HN, Capuano AW, Tong H, Petyuk VA, Macauley SL, Fioramonti X, Carmichael O, Calon F, Arvanitakis Z. State of the Science on Brain Insulin Resistance and Cognitive Decline Due to Alzheimer's Disease. Aging Dis 2024; 15:1688-1725. [PMID: 37611907 PMCID: PMC11272209 DOI: 10.14336/ad.2023.0814] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Accepted: 08/14/2023] [Indexed: 08/25/2023] Open
Abstract
Type 2 diabetes mellitus (T2DM) is common and increasing in prevalence worldwide, with devastating public health consequences. While peripheral insulin resistance is a key feature of most forms of T2DM and has been investigated for over a century, research on brain insulin resistance (BIR) has more recently been developed, including in the context of T2DM and non-diabetes states. Recent data support the presence of BIR in the aging brain, even in non-diabetes states, and found that BIR may be a feature in Alzheimer's disease (AD) and contributes to cognitive impairment. Further, therapies used to treat T2DM are now being investigated in the context of AD treatment and prevention, including insulin. In this review, we offer a definition of BIR, and present evidence for BIR in AD; we discuss the expression, function, and activation of the insulin receptor (INSR) in the brain; how BIR could develop; tools to study BIR; how BIR correlates with current AD hallmarks; and regional/cellular involvement of BIR. We close with a discussion on resilience to both BIR and AD, how current tools can be improved to better understand BIR, and future avenues for research. Overall, this review and position paper highlights BIR as a plausible therapeutic target for the prevention of cognitive decline and dementia due to AD.
Collapse
Affiliation(s)
- Elizabeth M Rhea
- Geriatric Research Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA 98108, USA.
- Department of Medicine, Division of Gerontology and Geriatric Medicine, University of Washington, Seattle, WA 98195, USA.
| | - Manon Leclerc
- Faculty of Pharmacy, Laval University, Quebec, Quebec, Canada.
- Neuroscience Axis, CHU de Québec Research Center - Laval University, Quebec, Quebec, Canada.
| | - Hussein N Yassine
- Departments of Neurology and Medicine, University of Southern California, Los Angeles, CA 90033, USA.
| | - Ana W Capuano
- Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, IL 60612, USA.
| | - Han Tong
- Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, IL 60612, USA.
| | - Vladislav A Petyuk
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA 99352, USA.
| | - Shannon L Macauley
- Department of Physiology, University of Kentucky, Lexington, KY 40508, USA.
| | - Xavier Fioramonti
- International Associated Laboratory OptiNutriBrain, Bordeaux, France and Quebec, Canada.
- Univ. Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, F-33000 Bordeaux, France.
| | - Owen Carmichael
- Pennington Biomedical Research Center, Baton Rouge, LA 70808, USA.
| | - Frederic Calon
- Faculty of Pharmacy, Laval University, Quebec, Quebec, Canada.
- Neuroscience Axis, CHU de Québec Research Center - Laval University, Quebec, Quebec, Canada.
- International Associated Laboratory OptiNutriBrain, Bordeaux, France and Quebec, Canada.
| | - Zoe Arvanitakis
- Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, IL 60612, USA.
| |
Collapse
|
2
|
Zago AM, Carvalho FB, Rahmeier FL, Santin M, Guimarães GR, Gutierres JM, da C Fernandes M. Exendin-4 Prevents Memory Loss and Neuronal Death in Rats with Sporadic Alzheimer-Like Disease. Mol Neurobiol 2024; 61:2631-2652. [PMID: 37919602 DOI: 10.1007/s12035-023-03698-w] [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] [Received: 12/22/2022] [Accepted: 10/06/2023] [Indexed: 11/04/2023]
Abstract
This study investigated the neuroprotective effects of exendin-4 (EXE-4), an analog of the glucagon-like peptide 1 receptor (GLP-1R) on memory and on the neuronal populations that constitute the hippocampus of rats submitted to a sporadic dementia of Alzheimer's type (SDAT). Male Wistar rats received streptozotocin (STZ icv, 3 mg/kg diluted in aCFS, 5 µl/ventricle) and were treated for 21 days with EXE-4 (10 µg/kg, ip; saline as the vehicle). Four groups were formed: vehicle, EXE-4, STZ, and STZ + EXE-4. The groups were submitted to Y-Maze (YM), object recognition (ORT), and object displacement tasks (ODT) to assess learning and memory. The brains were used for immunohistochemical and immunofluorescent techniques with antibodies to NeuN, cleaved caspase-3 (CC3), PCNA, doublecortin (DCX), synaptophysin (SYP), and insulin receptor (IR). STZ worsened spatial memory in the YMT, as well as short-term (STM) and long-term (LTM) memories in the ORT and ODT, respectively. EXE-4 protected against memory impairment in STZ animals. STZ reduced mature neuron density (NeuN) and increased cell apoptosis (CC3) in the DG, CA1, and CA3. EXE-4 protected against neuronal death in all regions. EXE-4 increased PCNA+ cells in all regions of the hippocampus, and STZ attenuated this effect. STZ reduced neurogenesis in DG per se as well as synaptogenesis induced by EXE-4. EXE-4 increased immunoreactivity to IR in the CA1. From these findings, EXE-4 showed a beneficial effect on hippocampal pyramidal and granular neurons in the SDAT showing anti-apoptotic properties and promoting cell proliferation. In parallel, EXE-4 preserved the memory of SDAT rats. EXE-4 appears to enhance synapses at CA3 and DG. In conclusion, these data indicate that agonists to GLP-1R have a beneficial effect on hippocampal neurons in AD.
Collapse
Affiliation(s)
- Adriana M Zago
- Graduate Program in Pathology, Pathology Research Laboratory, Federal University of Health Sciences of Porto Alegre, Sarmento Leite, 245, Room 514, Building 3. CEP 90050-170, Porto Alegre, RS, Brazil
| | - Fabiano B Carvalho
- Graduate Program in Pathology, Pathology Research Laboratory, Federal University of Health Sciences of Porto Alegre, Sarmento Leite, 245, Room 514, Building 3. CEP 90050-170, Porto Alegre, RS, Brazil.
| | - Francine L Rahmeier
- Graduate Program in Pathology, Pathology Research Laboratory, Federal University of Health Sciences of Porto Alegre, Sarmento Leite, 245, Room 514, Building 3. CEP 90050-170, Porto Alegre, RS, Brazil
| | - Marta Santin
- Graduate Program in Pathology, Pathology Research Laboratory, Federal University of Health Sciences of Porto Alegre, Sarmento Leite, 245, Room 514, Building 3. CEP 90050-170, Porto Alegre, RS, Brazil
| | - Giuliano R Guimarães
- Graduate Program in Pathology, Pathology Research Laboratory, Federal University of Health Sciences of Porto Alegre, Sarmento Leite, 245, Room 514, Building 3. CEP 90050-170, Porto Alegre, RS, Brazil
| | - Jessié M Gutierres
- Graduate Program in Pathology, Pathology Research Laboratory, Federal University of Health Sciences of Porto Alegre, Sarmento Leite, 245, Room 514, Building 3. CEP 90050-170, Porto Alegre, RS, Brazil
| | - Marilda da C Fernandes
- Graduate Program in Pathology, Pathology Research Laboratory, Federal University of Health Sciences of Porto Alegre, Sarmento Leite, 245, Room 514, Building 3. CEP 90050-170, Porto Alegre, RS, Brazil.
| |
Collapse
|
3
|
Ulaganathan S, Pitchaimani A. Spontaneous and familial models of Alzheimer's disease: Challenges and advances in preclinical research. Life Sci 2023:121918. [PMID: 37422070 DOI: 10.1016/j.lfs.2023.121918] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 06/28/2023] [Accepted: 07/05/2023] [Indexed: 07/10/2023]
Abstract
Alzheimer's disease (AD) is a debilitating neurodegenerative disorder that is progressive and irreversible in nature. Even after decades of dedicated research and paradigm-shifting hypotheses of AD etiology, very few well-founded credible improvements have been foreseen in understanding the actual underlying mechanisms involved in the development of the disorder. As for any disease to be well-comprehended, AD also requires optimal modelling strategies, which will then pave way for effective therapeutic interventions. Most of the clinical trials and research towards better treatment of AD fail in translation, due to the inefficacy of explored animal models to mimic the actual AD pathology, precisely. The majority of the existing AD models are developed based on the mutations found in the familial form of AD (fAD) which accounts for less than 5 % of the incidence of AD. Further, the investigations also face more challenges due to the additional complexities and lacunae found in etiology of sporadic form of AD (sAD), which accounts for 95 % of total AD. This review illustrates the gaps found in different models of AD, both sporadic and familial variants with additional focus on recent avenues for accurate simulation of AD pathology using in vitro and chimeric AD models.
Collapse
Affiliation(s)
- Suryapriya Ulaganathan
- Precision Nanomedicine and Microfluidic Lab, Centre for Biomaterials, Cellular and Molecular Theranostics (CBCMT), Vellore Institute of Technology (VIT), Vellore 632014, TN, India; School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore 632014, TN, India
| | - Arunkumar Pitchaimani
- Precision Nanomedicine and Microfluidic Lab, Centre for Biomaterials, Cellular and Molecular Theranostics (CBCMT), Vellore Institute of Technology (VIT), Vellore 632014, TN, India; School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore 632014, TN, India.
| |
Collapse
|
4
|
Meng X, Zhang Y, Li Z, Ma G, Zhang X, Zhang D, Cao W, Wang S, Cai Q, Cui P, Huang G. Increasing brain glucose uptake by Gypenoside LXXV ameliorates cognitive deficits in a mouse model of diabetic Alzheimer's disease. Phytother Res 2023; 37:611-626. [PMID: 36325883 DOI: 10.1002/ptr.7639] [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: 06/01/2022] [Revised: 08/27/2022] [Accepted: 09/12/2022] [Indexed: 11/06/2022]
Abstract
We have previously reported that Gypenoside LXXV (GP-75), a novel natural PPARγ agonist isolated from Gynostemma pentaphyllum, ameliorated cognitive deficits in db/db mice. In this study, we further investigated the beneficial effects on cognitive impairment in APP/PS1 mice and a mouse model of diabetic AD (APP/PS1xdb/db mice). Interestingly, intragastric administration of GP-75 (40 mg/kg/day) for 3 months significantly attenuated cognitive deficits in APP/PS1 and APP/PS1xdb/db mice. GP-75 treatment markedly reduced the levels of glucose, HbA1c and insulin in serum and improved glucose tolerance and insulin sensitivity in APP/PS1xdb/db mice. Notably, GP-75 treatment decreased the β-amyloid (Aβ) burden, as measured by 11 C-PIB PET imaging. Importantly, GP-75 treatment increased brain glucose uptake as measured by 18 F-FDG PET imaging. Moreover, GP-75 treatment upregulated PPARγ and increased phosphorylation of Akt (Ser473) and GLUT4 expression levels but decreased phosphorylation of IRS-1 (Ser616) in the hippocampi of both APP/PS1 and APP/PS1xdb/db mice. Furthermore, GP-75-induced increases in GLUT4 membrane translocation in primary hippocampal neurons from APP/PS1xdb/db mice was abolished by cotreatment with the selective PPARγ antagonist GW9662 or the PI3K inhibitor LY294002. In summary, GP-75 ameliorated cognitive deficits in APP/PS1 and APP/PS1xdb/db mice by enhancing glucose uptake via activation of the PPARγ/Akt/GLUT4 signaling pathways.
Collapse
Affiliation(s)
- Xiangbao Meng
- College of Pharmacy, Jinan University, Guangzhou, China.,Department of Neurosurgery, Shenzhen Key Laboratory of Neurosurgery, Shenzhen Institute of Translational Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, China
| | - Yuan Zhang
- Department of Neurosurgery, Shenzhen Key Laboratory of Neurosurgery, Shenzhen Institute of Translational Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, China
| | - Zongyang Li
- Department of Neurosurgery, Shenzhen Key Laboratory of Neurosurgery, Shenzhen Institute of Translational Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, China
| | - Guoxu Ma
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiejun Zhang
- Department of Neurosurgery, Shenzhen Key Laboratory of Neurosurgery, Shenzhen Institute of Translational Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, China
| | - Di Zhang
- Department of Neurosurgery, Shenzhen Key Laboratory of Neurosurgery, Shenzhen Institute of Translational Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, China
| | - Weiwei Cao
- Department of Neurosurgery, Shenzhen Key Laboratory of Neurosurgery, Shenzhen Institute of Translational Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, China
| | - Sicen Wang
- School of Medicine, Xi'an Jiaotong University, Xi'an, China
| | - Qian Cai
- College of Pharmacy, Jinan University, Guangzhou, China
| | - Ping Cui
- Department of Pharmacy, Shenzhen Children's Hospital, Shenzhen, China
| | - Guodong Huang
- Department of Neurosurgery, Shenzhen Key Laboratory of Neurosurgery, Shenzhen Institute of Translational Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, China
| |
Collapse
|
5
|
Deng C, Meng Z, Chen H, Meng S. Tetramethylpyrazine ameliorates systemic streptozotocin-induced Alzheimer-like pathology. J Chem Neuroanat 2023; 127:102207. [PMID: 36470527 DOI: 10.1016/j.jchemneu.2022.102207] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 11/18/2022] [Accepted: 11/30/2022] [Indexed: 12/07/2022]
Abstract
Diabetes mellitus (DM) and its complications are the main threats to the global disease burden. DM-related cognitive dysfunction is a progressive neurodegenerative disease, similar to Alzheimer's disease (AD). The underlying pathophysiology remains unclear, and an effective treatment is unavailable. Tetramethylpyrazine (TMP) is a bioactive ingredient extracted from the plant Ligusticum wallichii, which has anti-diabetic and neuroprotective properties. In this study, streptozotocin (STZ) injection was used to establish a mouse STZ-AD model, and TMP was administered through the lateral ventricle (ICV) to evaluate the effects of TMP on cognitive ability and neurochemical changes and to explore the underlying cellular and molecular mechanisms. Using MWM and Y-maze behavioral paradigms, we observed that TMP protected against STZ-induced learning and memory impairment. STZ promoted the deposition of amyloid plaques, activation of glial cells, loss of neurons and synapses, and reduction of synaptic plasticity. In contrast, TMP restored these aberrations and improved cognitive deficits in STZ-induced diabetic animals. Moreover, TMP attenuated hippocampal mitochondrial dysfunction and oxidative stress through modulation of the SIRT1/Nrf2/ HO-1 pathway. This evidence shows that TMP exerts its therapeutic effects through multiple pathways. Our study provides new insights into the neuroprotective effects of TMP for the treatment of diabetes-related cognitive failure.
Collapse
Affiliation(s)
- Chujun Deng
- Department of Traditional Chinese Medicine, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Zeyu Meng
- The Second Clinical Medical College, Heilongjiang University of Chinese Medicine, Harbin 150040, China
| | - Huize Chen
- Department of Traditional Chinese Medicine, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Shengxi Meng
- Department of Traditional Chinese Medicine, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China.
| |
Collapse
|
6
|
Akhtar A, Gupta SM, Dwivedi S, Kumar D, Shaikh MF, Negi A. Preclinical Models for Alzheimer's Disease: Past, Present, and Future Approaches. ACS OMEGA 2022; 7:47504-47517. [PMID: 36591205 PMCID: PMC9798399 DOI: 10.1021/acsomega.2c05609] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 11/22/2022] [Indexed: 05/13/2023]
Abstract
A robust preclinical disease model is a primary requirement to understand the underlying mechanisms, signaling pathways, and drug screening for human diseases. Although various preclinical models are available for several diseases, clinical models for Alzheimer's disease (AD) remain underdeveloped and inaccurate. The pathophysiology of AD mainly includes the presence of amyloid plaques and neurofibrillary tangles (NFT). Furthermore, neuroinflammation and free radical generation also contribute to AD. Currently, there is a wide gap in scientific approaches to preventing AD progression. Most of the available drugs are limited to symptomatic relief and improve deteriorating cognitive functions. To mimic the pathogenesis of human AD, animal models like 3XTg-AD and 5XFAD are the primarily used mice models in AD therapeutics. Animal models for AD include intracerebroventricular-streptozotocin (ICV-STZ), amyloid beta-induced, colchicine-induced, etc., focusing on parameters such as cognitive decline and dementia. Unfortunately, the translational rate of the potential drug candidates in clinical trials is poor due to limitations in imitating human AD pathology in animal models. Therefore, the available preclinical models possess a gap in AD modeling. This paper presents an outline that critically assesses the applicability and limitations of the current approaches in disease modeling for AD. Also, we attempted to provide key suggestions for the best-fit model to evaluate potential therapies, which might improve therapy translation from preclinical studies to patients with AD.
Collapse
Affiliation(s)
- Ansab Akhtar
- Department
of Pharmaceutical Sciences, School of Health Sciences and Technology, UPES, Dehradun, Uttarakhand, Dehradun 248007, India
| | - Shraddha M. Gupta
- Department
of Pharmaceutical Sciences, School of Health Sciences and Technology, UPES, Dehradun, Uttarakhand, Dehradun 248007, India
| | - Shubham Dwivedi
- Department
of Pharmaceutical Sciences, School of Health Sciences and Technology, UPES, Dehradun, Uttarakhand, Dehradun 248007, India
| | - Devendra Kumar
- Faculty
of Pharmacy, DIT University, Uttarakhand, Dehradun 248009, India
| | - Mohd. Farooq Shaikh
- Neuropharmacology
Research Strength, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway, Selangor 47500, Malaysia
| | - Arvind Negi
- Department
of Bioproducts and Biosystems, Aalto University, FI-00076 Espoo, Finland
- E-mail:
| |
Collapse
|
7
|
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: 6.0] [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
|
8
|
Xiong J, Deng I, Kelliny S, Lin L, Bobrovskaya L, Zhou XF. Long term high fat diet induces metabolic disorders and aggravates behavioral disorders and cognitive deficits in MAPT P301L transgenic mice. Metab Brain Dis 2022; 37:1941-1957. [PMID: 35704147 DOI: 10.1007/s11011-022-01029-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Accepted: 06/07/2022] [Indexed: 01/18/2023]
Abstract
Most Alzheimer disease (AD) patients present as sporadic late onset AD, with metabolic factors playing an important role in the occurrence and development of AD. Given the link between peripheral insulin resistance and tau pathology in streptozotocin-injected and db/db mouse models of diabetes, we fed high fat diet (HFD) to pR5 mice expressing P301L mutant human tau, with the aim of developing a new model with characteristics of obesity, T2DM and AD to mimic AD patients exacerbated by obesity and T2DM, an increasing trend in modern society. In our study, pR5 and C57BL/6 (WT) mice were randomly allocated to a standard diet (STD) or HFD for 30 weeks starting at 8 weeks of age. Food intake was measured weekly, body weight and fasting glucose levels were measured fortnightly, and a comprehensive behavioral test battery was performed to assess anxiety, depression and cognitive dysfunction. Glucose and insulin tolerance tests were performed after 30 weeks of HFD. We also investigated the effect of long term HFD on tau pathology in the brains of WT and P301L mice by performing western blotting of whole brain homogenates for total tau, phosphorylated tau at Ser396 and Thr231. Our results show that pR5 mice fed with HFD are more vulnerable to diet induced obesity compared to WT, especially with increasing age. In addition, pR5 mice on HFD developed glucose intolerance and insulin resistance. It was identified that long term HFD significantly aggravates depression like behavior and impairs cognitive function in pR5 mice, and also induces anxiety like behavior in both pR5 and WT mice. Long term HFD was also shown to aggravate tau hyperphosphorylation in pR5 transgenic mice, and increase total and hyperphosphorylated tau in WT mice. These results indicate that diet induced obesity of pR5 transgenic mice expressing P301L mutant human tau generates T2DM, and aggravates tau phosphorylation, and is therefore a model useful for investigations that seek to understand the relationships between AD, T2DM and obesity, and the underlying biochemical changes and mechanisms associated with metabolic disorders and AD tauopathy.
Collapse
Affiliation(s)
- Jing Xiong
- School of Pharmacy and Medical Sciences, Division of Health Sciences, University of South Australia, Adelaide, SA, Australia.
- Department of Neurology, the Second Affiliated Hospital, Kunming Medical University, Kunming, Yunnan Province, China.
| | - Isaac Deng
- School of Pharmacy and Medical Sciences, Division of Health Sciences, University of South Australia, Adelaide, SA, Australia
| | - Sally Kelliny
- School of Pharmacy and Medical Sciences, Division of Health Sciences, University of South Australia, Adelaide, SA, Australia
- Faculty of Phamacy, Assiut University, Assiut, Egypt
| | - Liying Lin
- School of Pharmacy and Medical Sciences, Division of Health Sciences, University of South Australia, Adelaide, SA, Australia
| | - Larisa Bobrovskaya
- School of Pharmacy and Medical Sciences, Division of Health Sciences, University of South Australia, Adelaide, SA, Australia
| | - Xin-Fu Zhou
- School of Pharmacy and Medical Sciences, Division of Health Sciences, University of South Australia, Adelaide, SA, Australia.
| |
Collapse
|
9
|
Bilateral intracerebroventricular injection of streptozotocin induces AD-like behavioral impairments and neuropathological features in mice: Involved with the fundamental role of neuroinflammation. Biomed Pharmacother 2022; 153:113375. [PMID: 35834993 DOI: 10.1016/j.biopha.2022.113375] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2022] [Revised: 06/17/2022] [Accepted: 07/06/2022] [Indexed: 01/31/2023] Open
Abstract
OBJECTIVE To establish an Alzheimer's disease (AD) mouse model, investigate the behavioral performance changes and intracerebral molecular changes induced by bilateral intracerebroventricular injection of streptozotocin (STZ/I.C.V), and explore the potential pathogenesis of AD. METHODS An AD mouse model was established by STZ/I.C.V. The behavioral performance was observed via the open field test (OFT), novel object recognition test (NOR), and tail suspension test (TST). The mRNA and protein expressions of interleukin 1β (IL-1β), interleukin 6 (IL-6), and tumor necrosis factor α (TNF-α) in the hippocampus were measured via qPCR and Western blot. The expression of β-amyloid 1-42 (Aβ1-42), phosphorylated Tau protein (p-Tau (Ser396)), Tau5, β-site amyloid precursor protein (APP) cleaving enzyme (BACE), insulin receptor substrate 1 (IRS1), brain-derived neurotrophic factor (BDNF), Copine6, synaptotagmin-1 (Syt-1), synapsin-1, phosphoinositol 3 kinase (PI3K), serine/threonine kinase (Akt), phosphorylated serine/threonine kinase (p-Akt (Ser473)), triggering receptor expressed on myeloid cells-1/2 (TREM1/2) were detected using Western blot, and the expression of glial fibrillary acidic protein (GFAP), ionized calcium binding adapter molecule 1 (IBA1), Aβ1-42, p-Tau(Ser396), Syt-1, BDNF were measured via immunofluorescence staining. RESULTS STZ/I.C.V induced AD-like neuropsychiatric behaviors in mice, as indicated by the impairment of learning and memory, together with the reduced spontaneous movement and exploratory behavior. The expression of BACE, Aβ1-42, p-Tau(Ser396), and TREM2 were significantly increased in the hippocampus of model mice, while the expression of IRS1, BDNF, Copine6, Syt-1, synapsin-1, PI3K, p-Akt(Ser473), and TREM1 were decreased as compared with that of the controls. Furthermore, the model mice presented a hyperactivation of astrocytes and microglia in the hippocampus, accompanied by the increased mRNA and protein expressions of IL-1β, IL-6 and TNF-α. CONCLUSION STZ/I.C.V is an effective way to induce AD mice model, with not only AD-like neuropsychiatric behaviors, but also typic AD-like neuropathological features including neurofibrillary tangles, deposit of β-amyloid (Aβ), neuroinflammation, and imbalanced synaptic plasticity.
Collapse
|