1
|
Lemche E, Hortobágyi T, Kiecker C, Turkheimer F. Neuropathological links between T2DM and LOAD: systematic review and meta-analysis. Physiol Rev 2025; 105:1429-1486. [PMID: 40062731 DOI: 10.1152/physrev.00040.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 02/01/2025] [Accepted: 02/22/2025] [Indexed: 04/16/2025] Open
Abstract
Recent decades have described parallel neuropathological mechanisms increasing the risk for developing late-onset Alzheimer's dementia (LOAD) in type 2 diabetes mellitus (T2DM); however, still little is known of the role of diabetic encephalopathy and brain atrophy in LOAD. The aim of this systematic review is to provide a comprehensive view on diabetic encephalopathy/cerebral atrophy, taking into account neuroimaging data, neuropathology, metabolic and endocrine mechanisms, amyloid formation, brain perfusion impairments, neuroimmunology, and inflammasome activation. Key switches were identified, to further meta-analyze genomic candidate loci and epigenetic modifications. For the qualitative meta-analysis of genomic bases extracted, human linkage studies were examined; for epigenetic mechanisms, data from both human and animal studies are described. For the systematic review of pathophysiological mechanisms, 1,259 publications were evaluated and 93 gene loci extracted for candidate risk linkages. Sixty-six publications were evaluated for genomic association and descriptions of epigenomic modifications. Overall accumulated results highlight the insulin signaling system, vascular markers, inflammation and inflammasome pathways, amylin interactions, and glycosylation mechanisms. The protocol was registered with PROSPERO (ID: CRD42023440535).
Collapse
Affiliation(s)
- Erwin Lemche
- Section of Cognitive Neuropsychiatry, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, United Kingdom
- Centre for Neuroimaging Sciences, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, United Kingdom
| | - Tibor Hortobágyi
- Institute of Neuropathology, University Hospital Zurich, Zurich, Switzerland
- Department of Old Age Psychiatry, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, United Kingdom
- Department of Neurology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Clemens Kiecker
- Department for Developmental Neurobiology, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, United Kingdom
| | - Federico Turkheimer
- Centre for Neuroimaging Sciences, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, United Kingdom
| |
Collapse
|
2
|
Dahiya M, Yadav M, Goyal C, Kumar A. Insulin resistance in Alzheimer's disease: signalling mechanisms and therapeutics strategies. Inflammopharmacology 2025; 33:1817-1831. [PMID: 40064805 DOI: 10.1007/s10787-025-01704-2] [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: 01/26/2025] [Accepted: 02/14/2025] [Indexed: 04/13/2025]
Abstract
BACKGROUND Alzheimer's disease (AD), one of the most common neurodegenerative disorders, is characterised by hallmark abnormalities such as amyloid-β plaques and neurofibrillary tangles (NFTs). Emerging evidence suggests that faulty insulin signalling contributes to these pathological features, impairing critical cellular and metabolic processes. OBJECTIVE This review aims to elucidate the role of insulin signalling in the central nervous system (CNS) under normal and pathological conditions and to explore therapeutic approaches targeting insulin pathways in AD and other neurodegenerative diseases. METHODS We reviewed studies highlighting the involvement of insulin-signalling pathways in neuronal health, with a particular focus on the key components-IRS, PI3K, Akt, and GSK-3β-predominantly expressed in cortical and hippocampal regions. RESULTS Insulin, an essential growth factor, regulates numerous cellular functions, including glucose metabolism, mitochondrial activity, oxidative stress response, autophagy, synaptic plasticity, and cognitive processes. Altered phosphorylation of signalling molecules in insulin pathways contributes to oxidative stress, inflammation, and the formation of AD hallmarks. Indirect modulators such as NF-κB and caspases further exacerbate neuronal damage, linking impaired insulin signalling to neurodegeneration. CONCLUSION Insulin signalling plays a crucial role in maintaining neuronal health and mitigating neurodegenerative processes. Targeting insulin pathways and associated molecules offers promising therapeutic avenues for AD and other neurodegenerative disorders.
Collapse
Affiliation(s)
- Mini Dahiya
- University Institute of Pharmaceutical Sciences, UGC Centre of Advanced Studies (UGC-CAS), Panjab University, Chandigarh, 160014, India
| | - Monu Yadav
- Amity Institute of Pharmacy, Amity University, Haryana, Amity Education Valley Gurugram, Manesar, Panchgaon, Haryana, India
| | - Chetan Goyal
- University Institute of Pharmaceutical Sciences, UGC Centre of Advanced Studies (UGC-CAS), Panjab University, Chandigarh, 160014, India
| | - Anil Kumar
- University Institute of Pharmaceutical Sciences, UGC Centre of Advanced Studies (UGC-CAS), Panjab University, Chandigarh, 160014, India.
| |
Collapse
|
3
|
Yavari M, Kalupahana NS, Harris BN, Ramalingam L, Zu Y, Kahathuduwa CN, Moustaid-Moussa N. Mechanisms Linking Obesity, Insulin Resistance, and Alzheimer's Disease: Effects of Polyphenols and Omega-3 Polyunsaturated Fatty Acids. Nutrients 2025; 17:1203. [PMID: 40218960 PMCID: PMC11990358 DOI: 10.3390/nu17071203] [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: 03/05/2025] [Revised: 03/23/2025] [Accepted: 03/24/2025] [Indexed: 04/14/2025] Open
Abstract
Alzheimer's disease (AD) is a complex neurodegenerative disorder characterized by progressive cognitive decline, memory loss, and behavioral changes. It poses a significant global health challenge. AD is associated with the accumulation of amyloid-β (Aβ) plaques and neurofibrillary tangles (NFTs) in the brain, along with chronic inflammation, dysfunctional neurons, and synapse loss. While the prevalence of AD continues to rise, the current FDA-approved drugs offer only limited effectiveness. Emerging evidence suggests that obesity, insulin resistance (IR), and type 2 diabetes mellitus (T2DM) are also implicated in AD pathogenesis, with epidemiological studies and animal models confirming the impact of IR on Aβ accumulation, and high-fat diets also exacerbating Aβ accumulation. Since neuroinflammation activated by Aβ involves the nuclear factor kappa-light-chain-enhancer of the activated B cell (NF-κB) pathway, the inhibition of NF-κB and NLRP3 inflammasome activation are potential therapeutic strategies in AD. Bioactive compounds, including polyphenols (resveratrol, epigallocatechin-3-gallate, curcumin, and quercetin), and omega-3 polyunsaturated fatty acids, show promising results in animal studies and clinical trials for reducing Aβ levels, improving cognition and modulating the signaling pathways implicated in AD. This review explores the interplay between obesity, IR, inflammation, and AD pathology, emphasizing the potential of dietary compounds and their role in reducing inflammation, oxidative stress, and cognitive decline, as viable strategies for AD prevention and treatment. By integrating epidemiological findings, observational studies, and clinical trials, this review aims to provide a comprehensive understating of how metabolic dysfunctions and bioactive compounds influence AD progression.
Collapse
Affiliation(s)
- Mahsa Yavari
- Department of Nutritional Sciences, Texas Tech University, Lubbock, TX 79409, USA; (M.Y.); (L.R.); (Y.Z.)
- Obesity Research Institute, Office of Research & Innovation, Texas Tech University, Lubbock, TX 79409, USA; (B.N.H.); (C.N.K.)
| | - Nishan Sudheera Kalupahana
- Department of Nutrition and Health, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates;
| | - Breanna N. Harris
- Obesity Research Institute, Office of Research & Innovation, Texas Tech University, Lubbock, TX 79409, USA; (B.N.H.); (C.N.K.)
- Department of Biological Sciences, Texas Tech University, Lubbock, TX 79409, USA
- Institute for One Health Innovation, Offices of Research & Innovation, Texas Tech University, Texas Tech Health Sciences Center, Lubbock, TX 79409, USA
| | - Latha Ramalingam
- Department of Nutritional Sciences, Texas Tech University, Lubbock, TX 79409, USA; (M.Y.); (L.R.); (Y.Z.)
| | - Yujiao Zu
- Department of Nutritional Sciences, Texas Tech University, Lubbock, TX 79409, USA; (M.Y.); (L.R.); (Y.Z.)
- Obesity Research Institute, Office of Research & Innovation, Texas Tech University, Lubbock, TX 79409, USA; (B.N.H.); (C.N.K.)
| | - Chanaka Nadeeshan Kahathuduwa
- Obesity Research Institute, Office of Research & Innovation, Texas Tech University, Lubbock, TX 79409, USA; (B.N.H.); (C.N.K.)
- Department of Neurology, Texas Tech University Health Sciences Center, El Paso, TX 79409, USA
| | - Naima Moustaid-Moussa
- Department of Nutritional Sciences, Texas Tech University, Lubbock, TX 79409, USA; (M.Y.); (L.R.); (Y.Z.)
- Obesity Research Institute, Office of Research & Innovation, Texas Tech University, Lubbock, TX 79409, USA; (B.N.H.); (C.N.K.)
- Institute for One Health Innovation, Offices of Research & Innovation, Texas Tech University, Texas Tech Health Sciences Center, Lubbock, TX 79409, USA
| |
Collapse
|
4
|
Ficiarà E, Rabbito R, Roveta F, Rubino E, Rainero I, Guiot C, Boschi S. Iron Overload, Microbleeding and the Role of Bilirubin in Alzheimer's Disease Brain: Revisiting the Vascular Hypothesis. Int J Mol Sci 2025; 26:3060. [PMID: 40243777 PMCID: PMC11988816 DOI: 10.3390/ijms26073060] [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: 02/19/2025] [Revised: 03/20/2025] [Accepted: 03/23/2025] [Indexed: 04/18/2025] Open
Abstract
Alzheimer's disease (AD) and vascular dementia (VaD) are the two most prevalent forms of dementia, sharing overlapping clinical features yet distinct pathophysiological mechanisms. While AD is primarily driven by amyloid-beta (Aβ) plaques and tau neurofibrillary tangles, VaD results from cerebrovascular pathology, including ischemic lesions and chronic hypoperfusion. However, accumulating evidence suggests that vascular dysfunction is a crucial contributor to both conditions, bridging neurodegenerative and cerebrovascular pathologies. In this review, we explore the interplay between AD and VaD, focusing on shared pathways such as blood-brain barrier (BBB) breakdown, neuroinflammation, and microvascular damage. Notably, cerebral microbleeds have emerged as a common feature in both AD and VaD, further linking vascular pathology to neurodegeneration. Microbleeding contributes to BBB disruption, iron deposition, and exacerbated oxidative stress, creating a vicious cycle that accelerates cognitive decline. We highlight the role of iron dysregulation as a key driver in AD, exacerbating Aβ accumulation, tau hyperphosphorylation, and ferroptosis. Conversely, bilirubin emerges as a molecule with theranostic potential, acting as both a biomarker and a neuroprotective agent due to its antioxidant and anti-inflammatory properties. Despite its protective role, bilirubin's dysregulation under pathological conditions may contribute to oxidative damage and neurovascular dysfunction. In this context, the accumulation of iron from recurrent microbleeds may further disrupt bilirubin homeostasis, amplifying oxidative injury and inflammation. We propose a vascular hypothesis that integrates iron metabolism and bilirubin homeostasis, suggesting that their imbalance plays a central role in AD pathogenesis and worsening. Understanding the intricate molecular interplay between neurodegeneration and vascular dysfunction could provide novel insights into targeted interventions aimed at mitigating cognitive decline. Finally, we discuss the potential of bilirubin-based therapeutic strategies, including its role in counteracting oxidative stress and modulating neuroinflammatory pathways, offering promising avenues for future research and precision medicine in dementia.
Collapse
Affiliation(s)
- Eleonora Ficiarà
- School of Pharmacy, University of Camerino, 62032 Camerino, MC, Italy;
| | - Rosita Rabbito
- Department of Neurosciences, Università degli Studi di Torino, 10125 Torino, TO, Italy; (R.R.); (F.R.); (E.R.); (I.R.); (C.G.)
| | - Fausto Roveta
- Department of Neurosciences, Università degli Studi di Torino, 10125 Torino, TO, Italy; (R.R.); (F.R.); (E.R.); (I.R.); (C.G.)
| | - Elisa Rubino
- Department of Neurosciences, Università degli Studi di Torino, 10125 Torino, TO, Italy; (R.R.); (F.R.); (E.R.); (I.R.); (C.G.)
| | - Innocenzo Rainero
- Department of Neurosciences, Università degli Studi di Torino, 10125 Torino, TO, Italy; (R.R.); (F.R.); (E.R.); (I.R.); (C.G.)
| | - Caterina Guiot
- Department of Neurosciences, Università degli Studi di Torino, 10125 Torino, TO, Italy; (R.R.); (F.R.); (E.R.); (I.R.); (C.G.)
| | - Silvia Boschi
- Department of Neurosciences, Università degli Studi di Torino, 10125 Torino, TO, Italy; (R.R.); (F.R.); (E.R.); (I.R.); (C.G.)
| |
Collapse
|
5
|
Lu W, Huang X, Shen D, Wang K, Wang J, Diao Z, Qiu S. Potential compensatory mechanism for cognitive impairment in type 2 diabetes and prediabetes: altered structure-function coupling. Front Endocrinol (Lausanne) 2025; 16:1491377. [PMID: 40166679 PMCID: PMC11955491 DOI: 10.3389/fendo.2025.1491377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Accepted: 03/03/2025] [Indexed: 04/02/2025] Open
Abstract
Background Structure-function (SC-FC) coupling may be more sensitive to detecting changes in the brain than any single modality. The aim of this study was to investigate the effects of SC-FC coupling changes on cognition and their interactions in patients with prediabetes and type 2 diabetes mellitus (T2DM). Methods A total of 493 participants (119 with normal glucose metabolism (NGM), 125 with prediabetes, and 249 with T2DM) were included in the study. Diffusion-weighted MRI and resting state functional MRI data were used to quantify SC-FC coupling. General linear model and linear regression analysis were used to evaluate the relationship between glucose metabolism, SC-FC coupling, and cognition. Mediation models were used to evaluate the mediating role of regional SC-FC coupling between diabetes-related measures and cognition. Results The regional coupling strength of SC-FC varied greatly in different brain regions, but was strongest in the ventral attention and somatmotor network areas. Compared with NGM patients, T2DM patients had higher SC-FC coupling in the default mode network but lower SC-FC coupling in the limbic network. In addition, fasting glucose and HbA1c were associated with weaker SC-FC coupling in the limbic network, fasting insulin with higher SC-FC coupling in the limbic network, and HbA1c with higher SC-FC coupling in the dorsal attention network. Furthermore, through mediated models we found that SC-FC coupling in the limbic network suppressed the association between diabetes-related measures and cognition. Conclusion T2DM and diabetes-related measures were associated with abnormal SC-FC coupling of the limbic network. The recombination of SC-FC coupling relationships in the limbic network may indicate a potential compensatory mechanism for cognitive decline that begins in prediabetes.
Collapse
Affiliation(s)
- Weiye Lu
- The First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Xuan Huang
- The First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Die Shen
- The First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Kun Wang
- The First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Jiahe Wang
- The First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Ziyu Diao
- The First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Shijun Qiu
- Department of Radiology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
- State Key Laboratory of Traditional Chinese Medicine Syndrome, Guangzhou, Guangdong, China
| |
Collapse
|
6
|
Salem EA, Alqahtani SM, El-Shoura EAM, Zaghlool SS, Abdelzaher LA, Mohamed SAM, Alalhareth IS, Sheref AAM. Neuroprotective effects of semaglutide and metformin against rotenone-induced neurobehavioral changes in male diabetic rats. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-03920-7. [PMID: 40088335 DOI: 10.1007/s00210-025-03920-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/02/2024] [Accepted: 02/11/2025] [Indexed: 03/17/2025]
Abstract
Pre-existing diabetes raises the likelihood of Parkinson's disease (PD), according to epidemiological and animal research. Our study aimed to investigating the likely neuroprotective effect of metformin (Met) and/or semaglutide (Sem) in model of PD in male diabetic rats and the possible underlying mechanism. Type 2 diabetes (T2DM) was induced by giving high-fat diet (HFD) for 3 weeks followed by a single streptozotocin (STZ) injection (40 mg/kg, i.p., once dose) followed by injection of 9 doses of rotenone every 48 ± 2 h for induction of PD. Met and/or Sema were administered to DM+PD via gastric gavage once daily for 4 weeks. In comparison with the DM+PD group, Met and/or Sem significantly lowered blood glucose levels, HOMA-IR, HbA1C, cholesterol, triglycerides, and LDL with significantly increased insulin and HDL levels. In addition, there was enhanced brain antioxidant status with lower oxidative-inflammatory stress biomarkers associated with improved rat cognitive, locomotor, and olfactory functions. A significant downregulation of caspase 3 and GFAP with concomitant upregulation of NRF2 protein expressions were observed in treated groups. Overall, co-treatment with Met and Sem elicited more efficacy than that of the individual regimen. When combined, the results of this study have demonstrated for the first time that Met and Sem work in concert to create neuroprotection in PD model of male diabetic rats compared to when taken separately. The study's findings indicate that Met and/or Sem have a restorative effect on T2DM and PD-induced changes in neurobehavioral and biochemical/molecular indices ascribed to the improvement of endogenous antioxidant systems, decreased lipid peroxidation, suppression of oxidative/inflammatory stress, and-most importantly-regulation of Nrf2 and caspase 3.
Collapse
Affiliation(s)
- Esraa A Salem
- Department of Medical Physiology, Faculty of Medicine, Menoufia University, Shebeen ElKom, 32511, Egypt
| | - Saad Misfer Alqahtani
- Department of Pathology, College of Medicine, The University Hospital, Najran University, Najran, Saudi Arabia
| | - Ehab A M El-Shoura
- Department of Clinical Pharmacy, Faculty of Pharmacy, Al-Azhar University, Assiut Branch, Assiut, Egypt.
| | - Sameh S Zaghlool
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Modern University of Technology and Information (MTI), Mokattam, Cairo, 11571, Egypt
| | - Lobna A Abdelzaher
- Department of Pharmacology, Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Sally A M Mohamed
- Department of Histology and Cytology, Faculty of Veterinary Medicine, Zagazig University, Zagazig, Egypt
| | - Ibrahim S Alalhareth
- College of Pharmacy, The University Hospital, Najran University, Najran, Saudi Arabia
| | - Alzahraa A M Sheref
- Department of Medical Physiology, Faculty of Medicine, Menoufia University, Shebeen ElKom, 32511, Egypt
| |
Collapse
|
7
|
Ramakrishan P, Rajangam J, Mahinoor SS, Bisht S, Mekala S, Upadhyay DK, Solomon VR, Sabarees G, Pelluri R. Unveiling the mTOR pathway modulation by SGLT2 inhibitors: a novel approach to Alzheimer's disease in type 2 diabetes. Metab Brain Dis 2025; 40:132. [PMID: 40009301 DOI: 10.1007/s11011-025-01555-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Accepted: 02/07/2025] [Indexed: 02/27/2025]
Abstract
Alzheimer's disease (AD) is a neurological condition causing cognitive deterioration, leading to severe consequences. As the global prevalence of AD increases, new treatment approaches are needed to supplement current conventional therapies, as traditional treatments are not meeting the increasing demand for alternative treatments. It is increasingly evident that treating metabolic disorders like diabetes mellitus, obesity, and AD by blocking mechanistic target of rapamycin (mTOR) signalling is advantageous. Chronic mTOR activation may cause AD's metabolic, lysosomal, and mitochondrial dysfunction, tau hyperphosphorylation, amyloid plaque development, and disruption of the blood-brain barrier through endothelial cell malfunction. Chronic glucose loss through sodium-glucose transporter 2 (SGLT2) inhibitions can restore mTOR cycling, potentially halting or slowing AD pathogenesis. Chronic activation of mTOR is implicated in pathophysiological aspects of AD, such as metabolic dysfunction, tau hyperphosphorylation, amyloid plaque formation, and disruption of the blood-brain barrier. SGLT-2 inhibitors, commonly used in treating Type 2 Diabetes, have been shown to reduce mTOR activation and restore circadian regularity, a new finding in cognitive decline and metabolic disorders. Conversely, SGLT2 inhibitors decrease oxidative damage, inflammation, insulin signaling pathways, and proliferation of endothelial cells to enhance vascular tone, flexibility, and contractility. Along with reducing the formation of plaque containing amyloid and improving brain function, neural plasticity, acetylcholinesterase (AChE) activity, damage to the brain, and cognitive decline, they also regulate the mTOR pathway in the brain. Thus, repurposing SGLT-2 inhibitors, primarily used in diabetes treatment, presents a promising avenue for changing the way that AD is managed. The purpose of this review was to focus on the mTOR signalling cascade of SGLT 2 inhibitors to AD management in Type 2 Diabetes mellitus.
Collapse
Affiliation(s)
- Prakash Ramakrishan
- Crescent School of Pharmacy, B.S. Abdur Rahman Crescent Institute of Science & Technology-BSACIST University, Chennai, 600048, India
| | - Jayaraman Rajangam
- Shri Venkateshwara College of Pharmacy, Ariyur, Pondicherry, 605102, India.
| | - Shaheedha Shabudeen Mahinoor
- Crescent School of Pharmacy, B.S.Abdur Rahman Crescent Institute of Science & Technology-BSACIST University, Chennai, 600048, India
| | - Shradha Bisht
- College of Pharmacy, Shivalik Campus, Dehradun, Uttarakhand, 248197, India
| | - Sabareesh Mekala
- Department of Pharmaceutical Sciences, School of Biotechnology and Pharmaceutical Sciences, Vignan's Foundation for Science, Technology and Research, Vadlamudi, Guntur, 522213, India
| | - Dinesh Kumar Upadhyay
- School of Pharmaceutical Sciences, Jaipur National University, Jaipur, 302017, India
| | - Viswas Raja Solomon
- Medicinal Chemistry Research Laboratory, MNR College of Pharmacy, Sangareddy, 502294, India
| | | | - Ranakishor Pelluri
- Department of Pharmacy, KL College of Pharmacy, Koneru Lakshmaiah Education Foundation (Deemed to Be University), Vaddeswaram, Guntur, 522302, India
| |
Collapse
|
8
|
Parrotta ME, Colangeli L, Scipione V, Vitale C, Sbraccia P, Guglielmi V. Time Restricted Eating: A Valuable Alternative to Calorie Restriction for Addressing Obesity? Curr Obes Rep 2025; 14:17. [PMID: 39899119 PMCID: PMC11790783 DOI: 10.1007/s13679-025-00609-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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/20/2025] [Indexed: 02/04/2025]
Abstract
PURPOSE OF REVIEW In this review, we summarize the molecular effects of time-restricted eating (TRE) and its possible role in appetite regulation. We also discuss the potential clinical benefits of TRE in obesity. RECENT FINDINGS TRE is an emerging dietary approach consisting in limiting food intake to a specific window of time each day. The rationale behind this strategy is to restore the circadian misalignment, commonly seen in obesity. Preclinical studies have shown that restricting food intake only during the active phase of the day can positively influence several cellular functions including senescence, mitochondrial activity, inflammation, autophagy and nutrients' sensing pathways. Furthermore, TRE may play a role by modulating appetite and satiety hormones, though further research is needed to clarify its exact mechanisms. Clinical trials involving patients with obesity or type 2 diabetes suggest that TRE can be effective for weight loss, but its broader effects on improving other clinical outcomes, such as cardiovascular risk factors, remain less certain. The epidemic proportions of obesity cause urgency to find dietary, pharmacological and surgical interventions that can be effective in the medium and long term. According to its molecular effects, TRE can be an interesting alternative to caloric restriction in the treatment of obesity, but the considerable variability across clinical trials regarding population, intervention, and follow-up duration makes it difficult to reach definitive conclusions.
Collapse
Affiliation(s)
| | - Luca Colangeli
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
- Internal Medicine Unit - Obesity Center, University Hospital Policlinico Tor Vergata, Rome, Italy
| | - Valeria Scipione
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Carolina Vitale
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Paolo Sbraccia
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
- Internal Medicine Unit - Obesity Center, University Hospital Policlinico Tor Vergata, Rome, Italy
| | - Valeria Guglielmi
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy.
- Internal Medicine Unit - Obesity Center, University Hospital Policlinico Tor Vergata, Rome, Italy.
| |
Collapse
|
9
|
Nassar M, Nassar O, Abosheaishaa H, Misra A. Comparative outcomes of systemic diseases in people with type 2 diabetes, or obesity alone treated with and without GLP-1 receptor agonists: a retrospective cohort study from the Global Collaborative Network : Author list. J Endocrinol Invest 2025; 48:483-497. [PMID: 39302577 DOI: 10.1007/s40618-024-02466-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Accepted: 09/04/2024] [Indexed: 09/22/2024]
Abstract
BACKGROUND Glucagon-like peptide-1 receptor agonists (GLP-1RAs) are increasingly used to manage type 2 diabetes (T2D) and obesity. Despite their recognized benefits in glycemic control and weight management, their impact on broader systemic has been less explored. OBJECTIVE This study aimed to evaluate the impact of GLP-1RAs on a variety of systemic diseases in people with T2D or obesity. METHODS We conducted a retrospective cohort study using data from the Global Collaborative Network, accessed through the TriNetX analytics platform. The study comprised two primary groups: individuals with T2D and those with obesity. Each group was further divided into subgroups based on whether they received GLP-1RA treatment or not. Data were analyzed over more than a 5-year follow-up period, comparing incidences of systemic diseases; systemic lupus erythematosus (SLE), systemic sclerosis (SS), rheumatoid arthritis (RA), ulcerative colitis (UC), crohn's disease (CD), alzheimer's disease (AD), parkinson's disease (PD), dementia, bronchial asthma (BA), osteoporosis, and several cancers. RESULTS In the T2D cohorts, GLP-1RA treatment was associated with significantly lower incidences of several systemic and metabolic conditions as compared to those without GLP-1RA, specifically, dementia (Risk Difference (RD): -0.010, p < 0.001), AD (RD: -0.003, p < 0.001), PD (RD: -0.002, p < 0.001), and pancreatic cancer (RD: -0.003, p < 0.001). SLE and SS also saw statistically significant reductions, though the differences were minor in magnitude (RD: -0.001 and - 0.000 respectively, p < 0.001 for both). Conversely, BA a showed a slight increase in risk (RD: 0.002, p < 0.001). CONCLUSIONS GLP-1RAs demonstrate potential benefits in reducing the risk of several systemic conditions in people with T2D or obesity. Further prospective studies are needed to confirm these effects fully and understand the mechanisms.
Collapse
Affiliation(s)
- Mahmoud Nassar
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA.
| | - Omar Nassar
- Williamsville East High School, Buffalo, NY, USA
| | - Hazem Abosheaishaa
- Department of Internal Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Anoop Misra
- Fortis-C-DOC Centre of Excellence for Diabetes, Metabolic Diseases and Endocrinology, New Delhi, India
- National Diabetes, Obesity and Cholesterol Foundation (N-DOC), New Delhi, India
- Diabetes Foundation (India) (DFI) India, New Delhi, India
| |
Collapse
|
10
|
Zhu Y, Verkhratsky A, Chen H, Yi C. Understanding glucose metabolism and insulin action at the blood-brain barrier: Implications for brain health and neurodegenerative diseases. Acta Physiol (Oxf) 2025; 241:e14283. [PMID: 39822067 PMCID: PMC11737474 DOI: 10.1111/apha.14283] [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: 11/11/2024] [Revised: 12/09/2024] [Accepted: 01/01/2025] [Indexed: 01/19/2025]
Abstract
The blood-brain barrier (BBB) is a highly selective, semipermeable barrier critical for maintaining brain homeostasis. The BBB regulates the transport of essential nutrients, hormones, and signaling molecules between the bloodstream and the central nervous system (CNS), while simultaneously protecting the brain from potentially harmful substances and pathogens. This selective permeability ensures that the brain is nourished and shielded from toxins. An exception to this are brain regions, such as the hypothalamus and circumventricular organs, which are irrigated by fenestrated capillaries, allowing rapid and direct response to various blood components. We overview the metabolic functions of the BBB, with an emphasis on the impact of altered glucose metabolism and insulin signaling on BBB in the pathogenesis of neurodegenerative diseases. Notably, endothelial cells constituting the BBB exhibit distinct metabolic characteristics, primarily generating ATP through aerobic glycolysis. This occurs despite their direct exposure to the abundant oxygen in the bloodstream, which typically supports oxidative phosphorylation. The effects of insulin on astrocytes, which form the glial limitans component of the BBB, show a marked sexual dimorphism. BBB nutrient sensing in the hypothalamus, along with insulin signaling, regulates systemic metabolism. Insulin modifies BBB permeability by regulating the expression of tight junction proteins, angiogenesis, and vascular remodeling, as well as modulating blood flow in the brain. The disruptions in glucose and insulin signaling are particularly evident in neurodegenerative diseases, such as Alzheimer's disease and Parkinson's disease, where BBB breakdown accelerates cognitive decline. This review highlights the critical role of normal glucose metabolism and insulin signaling in maintaining BBB functionality and investigates how disruptions in these pathways contribute to the onset and progression of neurodegenerative diseases.
Collapse
Affiliation(s)
- Yiyi Zhu
- Research CenterThe Seventh Affiliated Hospital of Sun Yat‐Sen UniversityShenzhenChina
| | - Alexei Verkhratsky
- Faculty of Biology, Medicine and HealthThe University of ManchesterManchesterUK
- Department of NeurosciencesUniversity of the Basque Country, CIBERNEDLeioaBizkaiaSpain
- IKERBASQUE Basque Foundation for ScienceBilbaoSpain
- Department of Forensic Analytical Toxicology, School of Forensic MedicineChina Medical UniversityShenyangChina
| | - Hui Chen
- School of Life Sciences, Faculty of ScienceUniversity of Technology SydneyUltimoNew South WalesAustralia
| | - Chenju Yi
- Research CenterThe Seventh Affiliated Hospital of Sun Yat‐Sen UniversityShenzhenChina
- Guangdong Provincial Key Laboratory of Brain Function and DiseaseGuangzhouChina
- Shenzhen Key Laboratory of Chinese Medicine Active Substance Screening and Translational ResearchShenzhenChina
| |
Collapse
|
11
|
Buoso E, Masi M, Limosani RV, Oliviero C, Saeed S, Iulini M, Passoni FC, Racchi M, Corsini E. Endocrine Disrupting Toxicity of Bisphenol A and Its Analogs: Implications in the Neuro-Immune Milieu. J Xenobiot 2025; 15:13. [PMID: 39846545 PMCID: PMC11755641 DOI: 10.3390/jox15010013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 12/22/2024] [Accepted: 01/15/2025] [Indexed: 01/24/2025] Open
Abstract
Endocrine-disrupting chemicals (EDCs) are natural or synthetic substances that are able to interfere with hormonal systems and alter their physiological signaling. EDCs have been recognized as a public health issue due to their widespread use, environmental persistence and the potential levels of long-term exposure with implications in multiple pathological conditions. Their reported adverse effects pose critical concerns about their use, warranting their strict regulation. This is the case of bisphenol A (BPA), a well-known EDC whose tolerable daily intake (TDI) was re-evaluated in 2023 by the European Food Safety Authority (EFSA), and the immune system has been identified as the most sensitive to BPA exposure. Increasing scientific evidence indicates that EDCs can interfere with several hormone receptors, pathways and interacting proteins, resulting in a complex, cell context-dependent response that may differ among tissues. In this regard, the neuronal and immune systems are important targets of hormonal signaling and are now emerging as critical players in endocrine disruption. Here, we use BPA and its analogs as proof-of-concept EDCs to address their detrimental effects on the immune and nervous systems and to highlight complex interrelationships within the immune-neuroendocrine network (INEN). Finally, we propose that Receptor for Activated C Kinase 1 (RACK1), an important target for EDCs and a valuable screening tool, could serve as a central hub in our toxicology model to explain bisphenol-mediated adverse effects on the INEN.
Collapse
Affiliation(s)
- Erica Buoso
- Department of Drug Sciences, Pharmacology Section, University of Pavia, Via Taramelli 12/14, 27100 Pavia, Italy; (R.V.L.); (C.O.); (S.S.); (M.R.)
- Department of Pharmacology, Physiology & Biophysics, Chobanian & Avedisian School of Medicine, Boston University, Boston, MA 02215, USA
| | - Mirco Masi
- Computational and Chemical Biology, Italian Institute of Technology, Via Morego 30, 16163 Genova, Italy;
| | - Roberta Valeria Limosani
- Department of Drug Sciences, Pharmacology Section, University of Pavia, Via Taramelli 12/14, 27100 Pavia, Italy; (R.V.L.); (C.O.); (S.S.); (M.R.)
| | - Chiara Oliviero
- Department of Drug Sciences, Pharmacology Section, University of Pavia, Via Taramelli 12/14, 27100 Pavia, Italy; (R.V.L.); (C.O.); (S.S.); (M.R.)
| | - Sabrina Saeed
- Department of Drug Sciences, Pharmacology Section, University of Pavia, Via Taramelli 12/14, 27100 Pavia, Italy; (R.V.L.); (C.O.); (S.S.); (M.R.)
| | - Martina Iulini
- Laboratory of Toxicology, Department of Pharmacological and Biomolecular Science, University of Milan, Via Balzaretti 9, 20133 Milan, Italy; (M.I.); (F.C.P.); (E.C.)
| | - Francesca Carlotta Passoni
- Laboratory of Toxicology, Department of Pharmacological and Biomolecular Science, University of Milan, Via Balzaretti 9, 20133 Milan, Italy; (M.I.); (F.C.P.); (E.C.)
| | - Marco Racchi
- Department of Drug Sciences, Pharmacology Section, University of Pavia, Via Taramelli 12/14, 27100 Pavia, Italy; (R.V.L.); (C.O.); (S.S.); (M.R.)
| | - Emanuela Corsini
- Laboratory of Toxicology, Department of Pharmacological and Biomolecular Science, University of Milan, Via Balzaretti 9, 20133 Milan, Italy; (M.I.); (F.C.P.); (E.C.)
| |
Collapse
|
12
|
Martins Dos Santos K, Saunders SE, Antunes VR, Boychuk CR. Insulin activates parasympathetic hepatic-related neurons of the paraventricular nucleus of the hypothalamus through mTOR signaling. J Neurophysiol 2025; 133:320-332. [PMID: 39665212 PMCID: PMC11918334 DOI: 10.1152/jn.00284.2024] [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: 07/01/2024] [Revised: 11/05/2024] [Accepted: 11/21/2024] [Indexed: 12/13/2024] Open
Abstract
Integration of autonomic and metabolic regulation, including hepatic function, is a critical role played by the brain's hypothalamic region. Specifically, the paraventricular nucleus of the hypothalamus (PVN) regulates autonomic functions related to metabolism, such as hepatic glucose production. Although insulin can act directly on hepatic tissue to inhibit hepatic glucose production, recent evidence implicates that central actions of insulin within PVN also regulate glucose metabolism. However, specific central circuits responsible for insulin signaling with relation to hepatic regulation are poorly understood. As a heterogeneous nucleus essential to controlling parasympathetic motor output with notable expression of insulin receptors, PVN is an appealing target for insulin-dependent modulation of parasympathetic activity. Here, we tested the hypothesis that insulin activates hepatic-related PVN (PVNhepatic) neurons through a parasympathetic pathway. Using transsynaptic retrograde tracing, labeling within PVN was first identified 24 h after its expression in the dorsal motor nucleus of the vagus (DMV) and 72 h after hepatic injection. Critically, nearly all labeling in medial PVN was abolished after a left vagotomy, indicating that PVNhepatic neurons in this region are part of a central circuit innervating parasympathetic motor neurons. Insulin also significantly increased the firing frequency of PVNhepatic neurons in this subregion. Mechanistically, rapamycin pretreatment inhibited insulin-dependent activation of PVNhepatic neurons. Therefore, central insulin signaling can activate a subset of PVNhepatic neurons that are part of a unique parasympathetic network in control of hepatic function. Taken together, PVNhepatic neurons related to parasympathetic output regulation could serve as a key central network in insulin's ability to control hepatic functions.NEW & NOTEWORTHY Increased peripheral insulin concentrations are known to decrease hepatic glucose production through both direct actions on hepatocytes and central autonomic networks. Despite this understanding, how (and in which brain regions) insulin exerts its action is still obscure. Here, we demonstrate that insulin activates parasympathetic hepatic-related PVN neurons (PVNhepatic) and that this effect relies on mammalian target of rapamycin (mTOR) signaling, suggesting that insulin modulates hepatic function through autonomic pathways involving insulin receptor intracellular signaling cascades.
Collapse
Affiliation(s)
- Karoline Martins Dos Santos
- Department of Cellular and Integrative Physiology, Long School of Medicine, University of Texas Health San Antonio, San Antonio, Texas, United States
- Department of Physiology & Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Sandy E Saunders
- Dalton Cardiovascular Research Center, Department of Biomedical Sciences, College of Veterinary Medicine, University of Missouri, Columbia, Missouri, United States
| | - Vagner R Antunes
- Department of Physiology & Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Carie R Boychuk
- Department of Cellular and Integrative Physiology, Long School of Medicine, University of Texas Health San Antonio, San Antonio, Texas, United States
- Dalton Cardiovascular Research Center, Department of Biomedical Sciences, College of Veterinary Medicine, University of Missouri, Columbia, Missouri, United States
| |
Collapse
|
13
|
Yazawa K, Nakashima M, Nakagawa T, Yanase Y, Yoda Y, Ozawa K, Hosoi T. Pancreatic β cell-secreted factor FGF23 attenuates Alzheimer's disease-related amyloid β-induced neuronal death. PNAS NEXUS 2025; 4:pgae542. [PMID: 39876880 PMCID: PMC11773612 DOI: 10.1093/pnasnexus/pgae542] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Accepted: 11/18/2024] [Indexed: 01/31/2025]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder characterized by cognitive decline and memory impairment. The pathophysiology of AD may involve aggregated amyloid β (Aβ) accumulation, which may underlie the disease mechanism. Patients with diabetes exhibit an elevated risk of developing AD, indicating potential therapeutic implications upon elucidating the underlying mechanisms. We hypothesized that pancreatic β cell-secreted factors could protect neurons from Aβ-induced toxicity. Therefore, we established an experimental model to elucidate the communication between pancreatic β cells and neuronal cells. Notably, our findings demonstrate that pancreatic β cell culture supernatant effectively inhibits Aβ-induced neuronal cell death. Transcriptomic analysis showed significant up-regulation of multiple ribosomal protein genes in neuronal cells treated with pancreatic β cell culture supernatant. Fibroblast growth factor 23, a secreted factor from pancreatic β cells, significantly suppressed Aβ-induced neuronal cell death. Our findings suggest that pancreatic β cells may secrete previously unrecognized neuroprotective factors, thereby attenuating neuronal cell death in AD.
Collapse
Affiliation(s)
- Kyosuke Yazawa
- Department of Pharmacotherapy, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima 734-8551, Japan
| | - Mieko Nakashima
- Department of Pharmacotherapy, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima 734-8551, Japan
| | - Tadashi Nakagawa
- Department of Clinical Pharmacology, Faculty of Pharmaceutical Sciences, Sanyo-Onoda City University, 1-1-1 Daigaku-dori, Sanyo Onoda City, Yamaguchi 756-0884, Japan
| | - Yuhki Yanase
- Department of Pharmacotherapy, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima 734-8551, Japan
| | - Yukari Yoda
- Department of Pharmacotherapy, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima 734-8551, Japan
| | - Koichiro Ozawa
- Department of Pharmacotherapy, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima 734-8551, Japan
| | - Toru Hosoi
- Department of Clinical Pharmacology, Faculty of Pharmaceutical Sciences, Sanyo-Onoda City University, 1-1-1 Daigaku-dori, Sanyo Onoda City, Yamaguchi 756-0884, Japan
| |
Collapse
|
14
|
Roh JD, Bae M, Kim H, Yang Y, Lee Y, Cho Y, Lee S, Li Y, Yang E, Jang H, Kim H, Kim H, Kang H, Ellegood J, Lerch JP, Bae YC, Kim JY, Kim E. Lithium normalizes ASD-related neuronal, synaptic, and behavioral phenotypes in DYRK1A-knockin mice. Mol Psychiatry 2024:10.1038/s41380-024-02865-2. [PMID: 39633007 DOI: 10.1038/s41380-024-02865-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2024] [Accepted: 11/29/2024] [Indexed: 12/07/2024]
Abstract
Dyrk1A deficiency is linked to various neurodevelopmental disorders, including developmental delays, intellectual disability (ID) and autism spectrum disorders (ASD). Haploinsufficiency of Dyrk1a in mice reportedly leads to ASD-related phenotypes. However, the key pathological mechanisms remain unclear and human DYRK1A mutations remain uncharacterized in mice. Here, we generated and studied Dyrk1a-knockin mice carrying a human ASD patient mutation (Ile48LysfsX2; Dyrk1a-I48K mice). These mice display severe microcephaly, social and cognitive deficits, dendritic shrinkage, excitatory synaptic deficits, and altered phospho-proteomic patterns enriched for multiple signaling pathways and synaptic proteins. Early chronic lithium treatment of newborn mutant mice rescues the brain volume, behavior, dendritic, synaptic, and signaling/synapse phospho-proteomic phenotypes at juvenile and adult stages. These results suggest that signaling/synaptic alterations contribute to the phenotypic alterations seen in Dyrk1a-I48K mice, and that early correction of these alterations by lithium treatment has long-lasting effects in preventing juvenile and adult-stage phenotypes.
Collapse
Affiliation(s)
- Junyeop Daniel Roh
- Center for Synaptic Brain Dysfunctions, Institute for Basic Science (IBS), Daejeon, 34141, Korea
| | - Mihyun Bae
- Center for Synaptic Brain Dysfunctions, Institute for Basic Science (IBS), Daejeon, 34141, Korea
| | - Hyosang Kim
- Center for Synaptic Brain Dysfunctions, Institute for Basic Science (IBS), Daejeon, 34141, Korea
| | - Yeji Yang
- Department of Biological Sciences, Korea Advanced Institute for Science and Technology (KAIST), Daejeon, 34141, Korea
- Digital Omics Research Center, Korea Basic Science Institute, Cheongju, 28119, Korea
| | - Yeunkeum Lee
- Center for Synaptic Brain Dysfunctions, Institute for Basic Science (IBS), Daejeon, 34141, Korea
- Korea Institute of Drug Safety & Risk Management, Anyang, 14051, Korea
| | - Yisul Cho
- Department of Anatomy and Neurobiology, School of Dentistry, Kyungpook National University, Daegu, 41940, Korea
| | - Suho Lee
- Center for Synaptic Brain Dysfunctions, Institute for Basic Science (IBS), Daejeon, 34141, Korea
| | - Yan Li
- Center for Synaptic Brain Dysfunctions, Institute for Basic Science (IBS), Daejeon, 34141, Korea
| | - Esther Yang
- Department of Anatomy and BK21 Graduate Program, Biomedical Sciences, College of Medicine, Korea University, Seoul, 02841, Korea
| | | | | | - Hyun Kim
- Department of Anatomy and BK21 Graduate Program, Biomedical Sciences, College of Medicine, Korea University, Seoul, 02841, Korea
| | - Hyojin Kang
- Division of National Supercomputing, KISTI, Daejeon, 34141, Korea
| | - Jacob Ellegood
- Mouse Imaging Centre, Hospital for Sick Children, Toronto, Ontario, M5T 3H7, Canada
- Bloorview Research Institute, Holland Bloorview Kids Rehabilitation Hospital, Toronto, Ontario, M4G 1R8, Canada
| | - Jason P Lerch
- Mouse Imaging Centre, Hospital for Sick Children, Toronto, Ontario, M5T 3H7, Canada
- Wellcome Centre for Integrative Neuroimaging, University of Oxford, Oxford, Oxfordshire, OX39DU, UK
| | - Yong Chul Bae
- Department of Anatomy and Neurobiology, School of Dentistry, Kyungpook National University, Daegu, 41940, Korea
| | - Jin Young Kim
- Digital Omics Research Center, Korea Basic Science Institute, Cheongju, 28119, Korea
| | - Eunjoon Kim
- Center for Synaptic Brain Dysfunctions, Institute for Basic Science (IBS), Daejeon, 34141, Korea.
- Department of Biological Sciences, Korea Advanced Institute for Science and Technology (KAIST), Daejeon, 34141, Korea.
| |
Collapse
|
15
|
Singh R, Jain S, Paliwal V, Verma K, Paliwal S, Sharma S. Does Metabolic Manager Show Encouraging Outcomes in Alzheimer's?: Challenges and Opportunity for Protein Tyrosine Phosphatase 1b Inhibitors. Drug Dev Res 2024; 85:e70026. [PMID: 39655712 DOI: 10.1002/ddr.70026] [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: 05/20/2024] [Revised: 10/22/2024] [Accepted: 11/18/2024] [Indexed: 12/18/2024]
Abstract
Protein tyrosine phosphatase 1b (PTP1b) is a member of the protein tyrosine phosphatase (PTP) enzyme group and encoded as PTP1N gene. Studies have evidenced an overexpression of the PTP1b enzyme in metabolic syndrome, anxiety, schizophrenia, neurodegeneration, and neuroinflammation. PTP1b inhibitor negatively regulates insulin and leptin pathways and has been explored as an antidiabetic agent in various clinical trials. Notably, the preclinical studies have shown that recuperating metabolic dysfunction and dyshomeostasis can reverse cognition and could be a possible approach to mitigate multifaceted Alzheimer's disease (AD). PTP1b inhibitor thus has attracted attention in neuroscience, though the development is limited to the preclinical stage, and its exploration in large clinical trials is warranted. This review provides an insight on the development of PTP1b inhibitors from different sources in diabesity. The crosstalk between metabolic dysfunction and insulin insensitivity in AD and type-2 diabetes has also been highlighted. Furthermore, this review presents the significance of PTP1b inhibition in AD based on pathophysiological facets, and recent evidences from preclinical and clinical studies.
Collapse
Affiliation(s)
- Ritu Singh
- Department of Pharmacy, Banasthali Vidyapith, Banasthali, Rajasthan, India
| | - Smita Jain
- Department of Pharmacy, Banasthali Vidyapith, Banasthali, Rajasthan, India
| | - Vartika Paliwal
- Department of Pharmacy, Banasthali Vidyapith, Banasthali, Rajasthan, India
| | - Kanika Verma
- Department of Internal Medicine, Division of Cardiology, LSU Health Sciences Center Shreveport, Louisiana, USA
| | - Sarvesh Paliwal
- Department of Pharmacy, Banasthali Vidyapith, Banasthali, Rajasthan, India
| | - Swapnil Sharma
- Department of Pharmacy, Banasthali Vidyapith, Banasthali, Rajasthan, India
| |
Collapse
|
16
|
Perdaens O, van Pesch V. Should We Consider Neurodegeneration by Itself or in a Triangulation with Neuroinflammation and Demyelination? The Example of Multiple Sclerosis and Beyond. Int J Mol Sci 2024; 25:12637. [PMID: 39684351 PMCID: PMC11641818 DOI: 10.3390/ijms252312637] [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: 10/31/2024] [Revised: 11/20/2024] [Accepted: 11/20/2024] [Indexed: 12/18/2024] Open
Abstract
Neurodegeneration is preeminent in many neurological diseases, and still a major burden we fail to manage in patient's care. Its pathogenesis is complicated, intricate, and far from being completely understood. Taking multiple sclerosis as an example, we propose that neurodegeneration is neither a cause nor a consequence by itself. Mitochondrial dysfunction, leading to energy deficiency and ion imbalance, plays a key role in neurodegeneration, and is partly caused by the oxidative stress generated by microglia and astrocytes. Nodal and paranodal disruption, with or without myelin alteration, is further involved. Myelin loss exposes the axons directly to the inflammatory and oxidative environment. Moreover, oligodendrocytes provide a singular metabolic and trophic support to axons, but do not emerge unscathed from the pathological events, by primary myelin defects and cell apoptosis or secondary to neuroinflammation or axonal damage. Hereby, trophic failure might be an overlooked contributor to neurodegeneration. Thus, a complex interplay between neuroinflammation, demyelination, and neurodegeneration, wherein each is primarily and secondarily involved, might offer a more comprehensive understanding of the pathogenesis and help establishing novel therapeutic strategies for many neurological diseases and beyond.
Collapse
Affiliation(s)
- Océane Perdaens
- Neurochemistry Group, Institute of NeuroScience, Université Catholique de Louvain (UCLouvain), 1200 Brussels, Belgium;
| | - Vincent van Pesch
- Neurochemistry Group, Institute of NeuroScience, Université Catholique de Louvain (UCLouvain), 1200 Brussels, Belgium;
- Department of Neurology, Cliniques Universitaires Saint-Luc, Université Catholique de Louvain (UCLouvain), 1200 Brussels, Belgium
| |
Collapse
|
17
|
Kciuk M, Kruczkowska W, Gałęziewska J, Wanke K, Kałuzińska-Kołat Ż, Aleksandrowicz M, Kontek R. Alzheimer's Disease as Type 3 Diabetes: Understanding the Link and Implications. Int J Mol Sci 2024; 25:11955. [PMID: 39596023 PMCID: PMC11593477 DOI: 10.3390/ijms252211955] [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: 10/30/2024] [Revised: 11/04/2024] [Accepted: 11/05/2024] [Indexed: 11/28/2024] Open
Abstract
Alzheimer's disease (AD) and type 2 diabetes mellitus (T2DM) are two prevalent conditions that present considerable public health issue in aging populations worldwide. Recent research has proposed a novel conceptualization of AD as "type 3 diabetes", highlighting the critical roles of insulin resistance and impaired glucose metabolism in the pathogenesis of the disease. This article examines the implications of this association, exploring potential new avenues for treatment and preventive strategies for AD. Key evidence linking diabetes to AD emphasizes critical metabolic processes that contribute to neurodegeneration, including inflammation, oxidative stress, and alterations in insulin signaling pathways. By framing AD within this metabolic context, we can enhance our understanding of its etiology, which in turn may influence early diagnosis, treatment plans, and preventive measures. Understanding AD as a manifestation of diabetes opens up the possibility of employing novel therapeutic strategies that incorporate lifestyle modifications and the use of antidiabetic medications to mitigate cognitive decline. This integrated approach has the potential to improve patient outcomes and deepen our comprehension of the intricate relationship between neurodegenerative diseases and metabolic disorders.
Collapse
Affiliation(s)
- Mateusz Kciuk
- Department of Molecular Biotechnology and Genetics, Faculty of Biology and Environmental Protection, University of Lodz, Banacha Street 12/16, 90-237 Lodz, Poland; (K.W.); (R.K.)
| | - Weronika Kruczkowska
- Department of Functional Genomics, Medical University of Lodz, 90-752 Lodz, Poland; (W.K.); (J.G.); (Ż.K.-K.)
| | - Julia Gałęziewska
- Department of Functional Genomics, Medical University of Lodz, 90-752 Lodz, Poland; (W.K.); (J.G.); (Ż.K.-K.)
| | - Katarzyna Wanke
- Department of Molecular Biotechnology and Genetics, Faculty of Biology and Environmental Protection, University of Lodz, Banacha Street 12/16, 90-237 Lodz, Poland; (K.W.); (R.K.)
| | - Żaneta Kałuzińska-Kołat
- Department of Functional Genomics, Medical University of Lodz, 90-752 Lodz, Poland; (W.K.); (J.G.); (Ż.K.-K.)
- Department of Biomedicine and Experimental Surgery, Medical University of Lodz, 90-136 Lodz, Poland
| | - Marta Aleksandrowicz
- Laboratory of Preclinical Research and Environmental Agents, Mossakowski Medical Research Institute, Polish Academy of Sciences, 02-106 Warsaw, Poland;
| | - Renata Kontek
- Department of Molecular Biotechnology and Genetics, Faculty of Biology and Environmental Protection, University of Lodz, Banacha Street 12/16, 90-237 Lodz, Poland; (K.W.); (R.K.)
| |
Collapse
|
18
|
Ghondaghsaz E, Khalaji A, Mahalleh M, Masrour M, Mohammadi P, Cannavo A, Behnoush AH. Exploring the Association Between Cognitive Decline and Triglyceride-Glucose Index: A Systematic Review and Meta-Analysis. Brain Behav 2024; 14:e70131. [PMID: 39482852 PMCID: PMC11527841 DOI: 10.1002/brb3.70131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 09/21/2024] [Accepted: 10/18/2024] [Indexed: 11/03/2024] Open
Abstract
BACKGROUND Cognitive decline and dementia are debilitating conditions that compromise the quality of life and charge the healthcare system with a substantial socioeconomic burden. In this context, emerging evidence supports an association between the triglyceride-glucose index (TyG), a surrogate insulin resistance marker, and cognitive decline and dementia. Hence, we systematically reviewed the studies assessing the TyG index in patients with cognitive decline and their controls. METHODS Online international databases (PubMed, Scopus, Embase, and the Web of Science) were searched comprehensively for studies showing the TyG index in patients with cognitive decline/impairment. Random-effect meta-analyses were conducted to calculate the standardized mean difference (SMD), pooled odds ratio (OR), and pooled area under the curve (AUC), in addition to 95% confidence intervals (CIs) for the comparisons of groups. RESULTS Seventeen studies were included in our analysis. Then, we conducted a meta-analysis, demonstrating that patients with cognitive decline had significantly higher levels of TyG index than those without (SMD 0.83, 95% CI 0.16 to 1.50, p = 0.015). Moreover, our data showed that a 1-unit increase in the TyG index was associated with higher odds of cognitive decline (adjusted OR [aOR] 2.86, 95% CI 1.49 to 5.50, p = 0.002). Further, we observed that patients in the fourth TyG quartile with higher values of the TyG index than the first quartile presented with more increased cognitive decline (aOR 1.62, 95%CI 1.11 to 2.38, p = 0.013). Finally, pooled AUC data for the diagnostic performance of the TyG index resulted in an overall AUC value of 0.73 (95% CI 0.66 to 0.79). Sensitivity and specificity were also calculated as 0.695 and 0.687, respectively. CONCLUSION This study supports the clinical utility of the TyG index in patients with cognitive decline and solicits more focused studies to consolidate its usage in clinical settings and real-world practice.
Collapse
Affiliation(s)
- Elina Ghondaghsaz
- Undergraduate Program in NeuroscienceUniversity of British ColumbiaBritish ColumbiaVancouverCanada
| | - Amirmohammad Khalaji
- School of MedicineTehran University of Medical SciencesTehranIran
- Non‐Communicable Diseases Research CenterEndocrinology and Metabolism Population Sciences InstituteTehran University of Medical SciencesTehranIran
| | - Mehrdad Mahalleh
- School of MedicineTehran University of Medical SciencesTehranIran
- Rheumatology Research CenterTehran University of Medical SciencesTehranIran
| | - Mahdi Masrour
- School of MedicineTehran University of Medical SciencesTehranIran
| | - Parsa Mohammadi
- School of MedicineTehran University of Medical SciencesTehranIran
| | - Alessandro Cannavo
- Department of Translational Medicine SciencesFederico II University of NaplesNaplesItaly
| | - Amir Hossein Behnoush
- School of MedicineTehran University of Medical SciencesTehranIran
- Non‐Communicable Diseases Research CenterEndocrinology and Metabolism Population Sciences InstituteTehran University of Medical SciencesTehranIran
| |
Collapse
|
19
|
Weiner SP, Carr KD. Behavioral tests of the insulin-cholinergic-dopamine link in nucleus accumbens and inhibition by high fat-high sugar diet in male and female rats. Physiol Behav 2024; 284:114647. [PMID: 39067780 PMCID: PMC11323239 DOI: 10.1016/j.physbeh.2024.114647] [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: 05/13/2024] [Revised: 07/20/2024] [Accepted: 07/23/2024] [Indexed: 07/30/2024]
Abstract
It was previously shown in striatal slices obtained from male rats that insulin excites cholinergic interneurons and increases dopamine (DA) release via α4β2 nicotinic receptors on DA terminals. The effect of insulin on DA release was blocked either by maintaining rats on a high sugar-high fat (HS-HF) diet that induced hyperinsulinemia and nucleus accumbens (NAc) insulin receptor insensitivity, or applying the α4β2 antagonist DHβE. In vivo, NAc shell insulin inactivation decreased a glucose lick microstructure parameter indicative of hedonic impact in male and female rats, and prevented flavor-nutrient learning, tested only in males. The HS-HF diet decreased hedonic impact in males but not females, and prevented flavor-nutrient learning, tested only in males. The present study extends testing to more fully assess the translation of brain slice results to the behaving rat. Insulin inactivation by antibody microinjection in NAc shell was found to decrease the number of lick bursts emitted and average lick burst size, measures of incentive motivation and hedonic impact respectively, for a wide range of glucose concentrations in male and female rats. In contrast, the HS-HF diet decreased these lick parameters in males but not females. Follow-up two-bottle choice tests for 10 % versus 40 % glucose showed decreased intake of both concentrations by males but increased intake of 40 % glucose by females. In a further set of experiments, it was predicted that α4β2 receptor blockade would induce the same behavioral effects as insulin inactivation. In females, DHβE microinjection in NAc shell decreased both lick parameters for glucose as predicted, but in males only the number of lick bursts emitted was decreased. DHβE also decreased the number of lick bursts emitted for saccharin by females but not males. Finally, DHβE microinjection in NAc shell decreased flavor-nutrient learning in both sexes. The few discrepancies seen with regard to the hypothesized insulin-nicotinic-dopaminergic regulation of behavioral responses to nutritive sweetener, and its inhibition by HS-HF diet, are discussed with reference to sex differences in DA dynamics, female resistance to diet-induced metabolic morbidities, and extra-striatal cholinergic inputs to NAc.
Collapse
Affiliation(s)
- Sydney P Weiner
- Departments of Psychiatry, New York University Grossman School of Medicine, 435 East 30th Street, New York, NY 10016, USA
| | - Kenneth D Carr
- Departments of Psychiatry, New York University Grossman School of Medicine, 435 East 30th Street, New York, NY 10016, USA; Departments of Biochemistry and Molecular Pharmacology, New York University Grossman School of Medicine, 435 East 30th Street, New York, NY 10016, USA.
| |
Collapse
|
20
|
Arda DB, Tunç KC, Bozkurt MF, Bora ES, Çiğel A, Erbaş O. Intranasal Insulin Eases Autism in Rats via GDF-15 and Anti-Inflammatory Pathways. Curr Issues Mol Biol 2024; 46:10530-10544. [PMID: 39329976 PMCID: PMC11431515 DOI: 10.3390/cimb46090624] [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: 08/24/2024] [Revised: 09/16/2024] [Accepted: 09/18/2024] [Indexed: 09/28/2024] Open
Abstract
In rat models, it is well-documented that chronic administration of propionic acid (PPA) leads to autism-like behaviors. Although the intranasal (IN) insulin approach is predominantly recognized for its effects on food restriction, it has also been shown to enhance cognitive memory by influencing various proteins, modulating anti-inflammatory pathways in the brain, and reducing signaling molecules such as interleukins. This study seeks to explore the potential therapeutic benefits of IN insulin in a rat model of autism induced by PPA. Thirty male Wistar albino rats were categorized into three cohorts: the control group, the PPA-induced autism (250 mg/kg/day intraperitoneal PPA dosage for five days) group, treated with saline via IN, and the PPA-induced autism group, treated with 25 U/kg/day (250 µL/kg/day) insulin via IN. All treatments were administered for 15 days. After behavioral testing, all animals were euthanized, and brain tissue and blood samples were collected for histopathological and biochemical assessments. Following insulin administration, a substantial reduction in autism symptoms was observed in all three social behavior tests conducted on the rats. Moreover, insulin exhibited noteworthy capabilities in decreasing brain MDA, IL-2, IL-17, and TNF-α levels within autism models. Additionally, there is a notable elevation in the brain nerve growth factor level (p < 0.05) and GDF-15 (p < 0.05). The assessment of cell counts within the hippocampal region and cerebellum revealed that insulin displayed effects in decreasing glial cells and inducing a significant augmentation in cell types such as the Purkinje and Pyramidal cells. The administration of insulin via IN exhibits alleviating effects on autism-like behavioral, biochemical, and histopathological alterations induced by PPA in rats. Insulin-dependent protective effects show anti-inflammatory, anti-oxidative, and neuroprotective roles of insulin admitted nasally.
Collapse
Affiliation(s)
- Duygu Burcu Arda
- Department of Pediatrics, Taksim Research and Training Hospital, Istanbul 34365, Türkiye;
| | - Kerem Can Tunç
- Department of Biology, Faculty of Science, Adnan Menderes University, Aydın 09010, Türkiye;
| | - Mehmet Fatih Bozkurt
- Department of Pathology, Faculty of Veterinary Medicine, Afyon Kocatepe University, Afyon 03100, Türkiye;
| | - Ejder Saylav Bora
- Department of Emergency Medicine, Faculty of Medicine, Izmir Katip Çelebi University, Izmir 35150, Türkiye
| | - Ayşe Çiğel
- Department of Physiology, Faculty of Medicine, Izmir Democracy University, Izmir 35150, Türkiye;
| | - Oytun Erbaş
- Department of Physiology, Faculty of Medicine, Demiroğlu Bilim University, Istanbul 34381, Türkiye;
| |
Collapse
|
21
|
Karim A, Pathan AM, Warayo A, Ahmed I, Betanai A, Dropati FNU, Ahmed S, Sarhandi PR, Kumari P, Bansari RB, Mehmoodi A, Malik J. Intranasal Insulin for Treatment of Persistent Post-COVID-19 Olfactory Dysfunction: A Scoping Review. J Community Hosp Intern Med Perspect 2024; 14:55-62. [PMID: 39399204 PMCID: PMC11466328 DOI: 10.55729/2000-9666.1390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Revised: 06/02/2024] [Accepted: 06/18/2024] [Indexed: 10/15/2024] Open
Abstract
Olfactory dysfunction has emerged as a prominent symptom of COVID-19, persisting in a subset of patients even after recovery. This scoping review aims to explore the potential of intranasal insulin as a treatment modality for persistent post-COVID-19 olfactory dysfunction. A comprehensive literature search was conducted to gather relevant studies examining the role of intranasal insulin in treating olfactory dysfunction, particularly in post-COVID-19 cases. Studies were included investigating intranasal insulin's mechanisms, efficacy, safety, and clinical outcomes. The review synthesizes findings from various studies suggesting the therapeutic potential of intranasal insulin in improving olfactory function. Research highlights the influence of intranasal insulin on neuroprotection, neurogenesis, and synaptic plasticity within the olfactory system, providing insights into its mechanisms of action. Furthermore, preliminary clinical evidence suggests improvements in olfactory sensitivity and intensity following intranasal insulin administration in post-COVID-19 patients with persistent olfactory dysfunction. While initial findings are encouraging, further rigorous investigations, including clinical trials with larger cohorts, are essential to validate these observations, ascertain optimal dosage regimens, and establish the safety and efficacy of intranasal insulin. This review provides a foundation for future research directions aimed at harnessing the therapeutic potential of intranasal insulin in addressing olfactory dysfunction following COVID-19.
Collapse
Affiliation(s)
- Ali Karim
- Department of Medicine, Liaquat University of Medical and Health Sciences, Jamshoro,
Pakistan
| | - Arisha M.G. Pathan
- Department of Medicine, Liaquat University of Medical and Health Sciences, Jamshoro,
Pakistan
| | - Allah Warayo
- Department of Medicine, Liaquat University of Medical and Health Sciences, Jamshoro,
Pakistan
| | - Iftikhar Ahmed
- Department of Medicine, Liaquat University of Medical and Health Sciences, Jamshoro,
Pakistan
| | - Aminullah Betanai
- Department of Medicine, Liaquat University of Medical and Health Sciences, Jamshoro,
Pakistan
| | - FNU Dropati
- Department of Medicine, Liaquat University of Medical and Health Sciences, Jamshoro,
Pakistan
| | - Saqlain Ahmed
- Department of Medicine, Liaquat University of Medical and Health Sciences, Jamshoro,
Pakistan
| | - Pir R.J. Sarhandi
- Department of Medicine, Liaquat University of Medical and Health Sciences, Jamshoro,
Pakistan
| | - Poonam Kumari
- Department of Medicine, Liaquat University of Medical and Health Sciences, Jamshoro,
Pakistan
| | - Raveena B. Bansari
- epartment of Medicine, Peoples University Medical and Health Sciences for Women, Nawabshah,
Pakistan
| | - Amin Mehmoodi
- Department of Medicine, Ibn e Seena Hospital, Kabul,
Afghanistan
| | - Jahanzeb Malik
- Department of Cardiovascular Medicine, Cardiovascular Analytics Group, Islamabad,
Pakistan
| |
Collapse
|
22
|
Wen W, Huang SM, Zhang B. Mechanisms Underlying Obesity-induced Aβ Accumulation in Alzheimer's Disease: A Qualitative Review. J Integr Neurosci 2024; 23:163. [PMID: 39344225 DOI: 10.31083/j.jin2309163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 02/27/2024] [Accepted: 03/05/2024] [Indexed: 10/01/2024] Open
Abstract
Epidemiological studies show that individuals with obesity are more likely to develop Alzheimer's disease (AD) than those who do not have obesity. However, the mechanisms underlying the relationship between obesity and AD are not entirely unclear. Here, we have reviewed and analyzed relevant articles published in the literature and found that obesity has correlation or potential increase in the levels of β-amyloid (Aβ) protein, which may explain why people with obesity are more likely to suffer from AD. Additionally, the published findings point to the roles of obesity-related metabolic disorders, such as diabetes, inflammation, oxidative stress, and imbalance in gut microbiota in Aβ accumulation caused by obesity. Therefore, in-depth experimental and clinical studies on these mechanisms in the future may help shed light on appropriate prevention and treatment strategies for AD, such as dietary changes and regular exercise to reverse or prevent obesity and related metabolic disorders.
Collapse
Affiliation(s)
- Wei Wen
- Department of Pharmacology, College of Basic Medicine, Heilongjiang University of Chinese Medicine, 150040 Harbin, Heilongjiang, China
| | - Shu-Ming Huang
- Department of Neuroscience, Institute of Chinese Medicine, Heilongjiang University of Chinese Medicine, 150040 Harbin, Heilongjiang, China
| | - Bo Zhang
- Department of Neuroscience, Institute of Chinese Medicine, Heilongjiang University of Chinese Medicine, 150040 Harbin, Heilongjiang, China
| |
Collapse
|
23
|
Selenius JS, Silveira PP, von Bonsdorff M, Lahti J, Koistinen H, Koistinen R, Seppälä M, Eriksson JG, Wasenius NS. Biologically Informed Polygenic Scores for Brain Insulin Receptor Network Are Associated with Cardiometabolic Risk Markers and Diabetes in Women. Diabetes Metab J 2024; 48:960-970. [PMID: 38527457 PMCID: PMC11449818 DOI: 10.4093/dmj.2023.0039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Accepted: 11/25/2023] [Indexed: 03/27/2024] Open
Abstract
BACKGRUOUND To investigate associations between variations in the co-expression-based brain insulin receptor polygenic score and cardiometabolic risk factors and diabetes mellitus. METHODS This cross-sectional study included 1,573 participants from the Helsinki Birth Cohort Study. Biologically informed expression-based polygenic risk scores for the insulin receptor gene network were calculated for the hippocampal (hePRS-IR) and the mesocorticolimbic (mePRS-IR) regions. Cardiometabolic markers included body composition, waist circumference, circulating lipids, insulin-like growth factor 1 (IGF-1), and insulin-like growth factor-binding protein 1 and 3 (IGFBP-1 and -3). Glucose and insulin levels were measured during a standardized 2-hour 75 g oral glucose tolerance test and impaired glucose regulation status was defined by the World Health Organization 2019 criteria. Analyzes were adjusted for population stratification, age, smoking, alcohol consumption, socioeconomic status, chronic diseases, birth weight, and leisure-time physical activity. RESULTS Multinomial logistic regression indicated that one standard deviation increase in hePRS-IR was associated with increased risk of diabetes mellitus in all participants (adjusted relative risk ratio, 1.17; 95% confidence interval, 1.01 to 1.35). In women, higher hePRS-IR was associated with greater waist circumference and higher body fat percentage, levels of glucose, insulin, total cholesterol, low-density lipoprotein cholesterol, triglycerides, apolipoprotein B, insulin, and IGFBP-1 (all P≤0.02). The mePRS-IR was associated with decreased IGF-1 level in women (P=0.02). No associations were detected in men and studied outcomes. CONCLUSION hePRS-IR is associated with sex-specific differences in cardiometabolic risk factor profiles including impaired glucose regulation, abnormal metabolic markers, and unfavorable body composition in women.
Collapse
Affiliation(s)
- Jannica S. Selenius
- Folkhälsan Research Center, Helsinki, Finland
- Department of General Practice and Primary Health Care, Helsinki University Hospital, University of Helsinki, Helsinki, Finland
| | - Patricia P. Silveira
- Department of Psychiatry, Faculty of Medicine, McGill University, Verdun, QC, Canada
- Ludmer Center for Neuroinformatic and Mental Health, Douglas Mental Health University Institute, McGill University, Verdun, QC, Canada
| | - Mikaela von Bonsdorff
- Folkhälsan Research Center, Helsinki, Finland
- Gerontology Research Center and Faculty of Sport and Health Sciences, University of Jyväskylä, Jyväskylä, Finland
| | - Jari Lahti
- Department of Psychology and Logopedics, University of Helsinki, Helsinki, Finland
- Turku Institute for Advanced Studies, University of Turku, Turku, Finland
| | - Hannu Koistinen
- Department of Clinical Chemistry and Haematology, Helsinki University Hospital, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Riitta Koistinen
- Department of Clinical Chemistry and Haematology, Helsinki University Hospital, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Markku Seppälä
- Department of Clinical Chemistry and Obstetrics and Gynecology, Helsinki University Hospital, University of Helsinki, Helsinki, Finland
| | - Johan G. Eriksson
- Folkhälsan Research Center, Helsinki, Finland
- Department of General Practice and Primary Health Care, Helsinki University Hospital, University of Helsinki, Helsinki, Finland
- Department of Obstetrics & Gynecology and Human Potential Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Singapore Institute for Clinical Sciences (SICS), Agency for Science, Technology and Research (A*STAR), Singapore
| | - Niko S. Wasenius
- Folkhälsan Research Center, Helsinki, Finland
- Department of General Practice and Primary Health Care, Helsinki University Hospital, University of Helsinki, Helsinki, Finland
| |
Collapse
|
24
|
Di Domenico F, Lanzillotta C, Perluigi M. Redox imbalance and metabolic defects in the context of Alzheimer disease. FEBS Lett 2024; 598:2047-2066. [PMID: 38472147 DOI: 10.1002/1873-3468.14840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 02/12/2024] [Accepted: 02/12/2024] [Indexed: 03/14/2024]
Abstract
Redox reactions play a critical role for intracellular processes, including pathways involved in metabolism and signaling. Reactive oxygen species (ROS) act either as second messengers or generators of protein modifications, fundamental mechanisms for signal transduction. Disturbance of redox homeostasis is associated with many disorders. Among these, Alzheimer's disease is a neurodegenerative pathology that presents hallmarks of oxidative damage such as increased ROS production, decreased activity of antioxidant enzymes, oxidative modifications of macromolecules, and changes in mitochondrial homeostasis. Interestingly, alteration of redox homeostasis is closely associated with defects of energy metabolism, involving both carbohydrates and lipids, the major energy fuels for the cell. As the brain relies exclusively on glucose metabolism, defects of glucose utilization represent a harmful event for the brain. During aging, a progressive perturbation of energy metabolism occurs resulting in brain hypometabolism. This condition contributes to increase neuronal cell vulnerability ultimately resulting in cognitive impairment. The current review discusses the crosstalk between alteration of redox homeostasis and brain energy defects that seems to act in concert in promoting Alzheimer's neurodegeneration.
Collapse
Affiliation(s)
- Fabio Di Domenico
- Department of Biochemical Sciences "A. Rossi Fanelli", Sapienza University of Rome, Italy
| | - Chiara Lanzillotta
- Department of Biochemical Sciences "A. Rossi Fanelli", Sapienza University of Rome, Italy
| | - Marzia Perluigi
- Department of Biochemical Sciences "A. Rossi Fanelli", Sapienza University of Rome, Italy
- Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Rome, Italy
| |
Collapse
|
25
|
Latva-Rasku A, Rebelos E, Tuisku J, Aarnio R, Bhowmik A, Keskinen H, Laurila S, Lahesmaa-Hatting M, Pekkarinen L, Isackson H, Kirjavainen AK, Koffert J, Heurling K, Nummenmaa L, Ferrannini E, Oldgren J, Oscarsson J, Nuutila P. SGLT2 Inhibitor Dapagliflozin Increases Skeletal Muscle and Brain Fatty Acid Uptake in Individuals With Type 2 Diabetes: A Randomized Double-Blind Placebo-Controlled Positron Emission Tomography Study. Diabetes Care 2024; 47:1630-1637. [PMID: 38941156 DOI: 10.2337/dc24-0470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Accepted: 06/10/2024] [Indexed: 06/30/2024]
Abstract
OBJECTIVE The aim of this study was to investigate the impact of the sodium-glucose cotransporter 2 (SGLT2) inhibitor dapagliflozin on tissue fatty acid (FA) uptake in the skeletal muscle, brain, small intestine, and subcutaneous and visceral adipose tissue of individuals with type 2 diabetes by using positron emission tomography (PET). RESEARCH DESIGN AND METHODS In a 6-week randomized double-blind placebo-controlled trial, 53 patients with type 2 diabetes treated with metformin received either 10 mg dapagliflozin or placebo daily. Tissue FA uptake was quantified at baseline and end of treatment with PET and the long-chain FA analog radiotracer 14(R,S)-[18F]fluoro-6-thia-heptadecanoic acid. Treatment effects were assessed using ANCOVA, and the results are reported as least square means and 95% CIs for the difference between groups. RESULTS A total of 38 patients (dapagliflozin n = 21; placebo n = 17) completed the study. After 6 weeks, skeletal muscle FA uptake was increased by dapagliflozin compared with placebo (1.0 [0.07, 2.0] μmol ⋅ 100 g-1 ⋅ min-1; P = 0.032), whereas uptake was not significantly changed in the small intestine or visceral or subcutaneous adipose tissue. Dapagliflozin treatment significantly increased whole-brain FA uptake (0.10 [0.02, 0.17] μmol ⋅ 100 g-1 ⋅ min-1; P = 0.01), an effect observed in both gray and white matter regions. CONCLUSIONS Six weeks of treatment with dapagliflozin increases skeletal muscle and brain FA uptake, partly driven by a rise in free FA availability. This finding is in accordance with previous indirect measurements showing enhanced FA metabolism in response to SGLT2 inhibition and extends the notion of a shift toward increased FA use to muscle and brain.
Collapse
Affiliation(s)
- Aino Latva-Rasku
- Turku PET Centre, University of Turku, Turku, Finland
- Department of Endocrinology, Turku University Hospital, Turku, Finland
| | - Eleni Rebelos
- Turku PET Centre, University of Turku, Turku, Finland
| | - Jouni Tuisku
- Turku PET Centre, University of Turku, Turku, Finland
| | | | - Achol Bhowmik
- Turku PET Centre, University of Turku, Turku, Finland
| | | | - Sanna Laurila
- Turku PET Centre, University of Turku, Turku, Finland
- Heart Center, Turku University Hospital, Turku, Finland
| | | | - Laura Pekkarinen
- Turku PET Centre, University of Turku, Turku, Finland
- Department of Endocrinology, Turku University Hospital, Turku, Finland
| | - Henrik Isackson
- Clinical Physiology and Cardiology, Department of Medical Sciences, Uppsala University, Uppsala, Sweden
- Integrative Physiology, Medical Cell Biology, Uppsala University Hospital, Uppsala, Sweden
| | - Anna K Kirjavainen
- Radiopharmaceutical Chemistry Laboratory, Turku PET Centre, University of Turku, Turku, Finland
| | - Jukka Koffert
- Turku PET Centre, University of Turku, Turku, Finland
| | | | - Lauri Nummenmaa
- Turku PET Centre, University of Turku, Turku, Finland
- Department of Psychology, University of Turku, Turku, Finland
| | - Ele Ferrannini
- National Research Council Institute of Clinical Physiology, Pisa, Italy
| | - Jonas Oldgren
- Clinical Physiology and Cardiology, Department of Medical Sciences, Uppsala University, Uppsala, Sweden
- Uppsala Clinical Research Center, Uppsala University, Uppsala, Sweden
| | - Jan Oscarsson
- Late-Stage Development, Cardiovascular, Renal, and Metabolism, BioPharmaceuticals Research and Development, AstraZeneca, Gothenburg, Sweden
| | - Pirjo Nuutila
- Turku PET Centre, University of Turku, Turku, Finland
- Department of Endocrinology, Turku University Hospital, Turku, Finland
| |
Collapse
|
26
|
Presta M, Zoratto F, Mulder D, Ottomana AM, Pisa E, Arias Vásquez A, Slattery DA, Glennon JC, Macrì S. Hyperglycemia and cognitive impairments anticipate the onset of an overt type 2 diabetes-like phenotype in TALLYHO/JngJ mice. Psychoneuroendocrinology 2024; 167:107102. [PMID: 38896988 DOI: 10.1016/j.psyneuen.2024.107102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 04/30/2024] [Accepted: 06/08/2024] [Indexed: 06/21/2024]
Abstract
Type 2 Diabetes mellitus (T2DM) is a metabolic disorder characterized by chronic hyperglycemia, resulting from deficits in insulin secretion, insulin action, or both. Whilst the role of insulin in the peripheral nervous system has been ascertained in countless studies, its role in the central nervous system (CNS) is emerging only recently. Brain insulin has been lately associated with brain disorders like Alzheimer's disease, obsessive compulsive disorder, and attention deficit hyperactivity disorder. Thus, understanding the role of insulin as a common risk factor for mental and somatic comorbidities may disclose novel preventative and therapeutic approaches. We evaluated general metabolism (glucose tolerance, insulin sensitivity, energy expenditure, lipid metabolism, and polydipsia) and cognitive capabilities (attention, cognitive flexibility, and memory), in adolescent, young adult, and adult male and female TALLYHO/JngJ mice (TH, previously reported to constitute a valid experimental model of T2DM due to impaired insulin signaling). Adult TH mice have also been studied for alterations in gut microbiota diversity and composition. While TH mice exhibited profound deficits in cognitive flexibility and altered glucose metabolism, we observed that these alterations emerged either much earlier (males) or independent of (females) a comprehensive constellation of symptoms, isomorphic to an overt T2DM-like phenotype (insulin resistance, polydipsia, higher energy expenditure, and altered lipid metabolism). We also observed significant sex-dependent alterations in gut microbiota alpha diversity and taxonomy in adult TH mice. Deficits in insulin signaling may represent a common risk factor for both T2DM and CNS-related deficits, which may stem from (partly) independent mechanisms.
Collapse
Affiliation(s)
- Martina Presta
- Centre for Behavioural Sciences and Mental Health, Istituto Superiore di Sanità, Rome 00161, Italy; Department of Physiology and Pharmacology, Sapienza University of Rome, Rome 00185, Italy
| | - Francesca Zoratto
- Centre for Behavioural Sciences and Mental Health, Istituto Superiore di Sanità, Rome 00161, Italy
| | - Danique Mulder
- Donders Institute for Brain, Cognition and Behaviour, Departments of Psychiatry and Human Genetics, Radboud University Medical Centre, Nijmegen, the Netherlands
| | - Angela Maria Ottomana
- Centre for Behavioural Sciences and Mental Health, Istituto Superiore di Sanità, Rome 00161, Italy; Neuroscience Unit, Department of Medicine, University of Parma, Parma 43100, Italy
| | - Edoardo Pisa
- Centre for Behavioural Sciences and Mental Health, Istituto Superiore di Sanità, Rome 00161, Italy
| | - Alejandro Arias Vásquez
- Donders Institute for Brain, Cognition and Behaviour, Departments of Psychiatry and Human Genetics, Radboud University Medical Centre, Nijmegen, the Netherlands
| | - David A Slattery
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital Frankfurt, Frankfurt, Germany
| | - Jeffrey C Glennon
- Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Ireland
| | - Simone Macrì
- Centre for Behavioural Sciences and Mental Health, Istituto Superiore di Sanità, Rome 00161, Italy.
| |
Collapse
|
27
|
Mesarosova L, Scheper M, Iyer A, Anink JJ, Mills JD, Aronica E. miR-193b-3p/ PGC-1α pathway regulates an insulin dependent anti-inflammatory response in Parkinson's disease. Neurobiol Dis 2024; 199:106587. [PMID: 38950713 DOI: 10.1016/j.nbd.2024.106587] [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: 03/21/2024] [Revised: 06/18/2024] [Accepted: 06/28/2024] [Indexed: 07/03/2024] Open
Abstract
It has been shown that many miRNAs, including miR-193b-3p, are differentially expressed in Parkinson's disease (PD). Dysregulation of miR-193b-3p/PGC-1α axis may alter homeostasis in cells and can induce an inflammatory response commonly accompanied by metabolic disturbances. The aim of the present study is to investigate if dysregulation of the miR-193-3p/PGC-1α axis may contribute to the pathological changes observed in the PD brain. Brain tissue were obtained from middle frontal gyrus of non-demented controls and individuals with a PD diagnosis. RT-qPCR was used to determine the expression of miR-193b-3p and in situ hybridization (ISH) and immunological analysis were employed to establish the cellular distribution of miR-193b-3p. Functional assays were performed using SH-SY5Y cells, including transfection and knock-down of miR-193b-3p. We found significantly lower expression of miR-193b-3p in the early stages of PD (PD4) which increased throughout disease progression. Furthermore, altered expression of PGC-1α suggested a direct inhibitory effect of miR-193b-3p in the brain of individuals with PD. Moreover, we observed changes in expression of insulin after transfection of SH-SY5Y cells with miR-193b-3p, which led to dysregulation in the expression of several pro- or anti - inflammatory genes. Our findings indicate that the miR-193b-3p/PGC-1α axis is involved in the regulation of insulin signaling. This regulation is crucial, since insulin induced inflammatory response may serve as a protective mechanism during acute situations but potentially evolve into a pathological process in chronic conditions. This novel regulatory mechanism may represent an interesting therapeutic target with potential benefits for various neurodegenerative diseases.
Collapse
Affiliation(s)
- Lucia Mesarosova
- Amsterdam UMC location University of Amsterdam, Department of (Neuro)Pathology Amsterdam Neuroscience, Meibergdreef 9, Amsterdam, the Netherlands.
| | - Mirte Scheper
- Amsterdam UMC location University of Amsterdam, Department of (Neuro)Pathology Amsterdam Neuroscience, Meibergdreef 9, Amsterdam, the Netherlands
| | - Anand Iyer
- Amsterdam UMC location University of Amsterdam, Department of (Neuro)Pathology Amsterdam Neuroscience, Meibergdreef 9, Amsterdam, the Netherlands; Department of Internal Medicine, Erasmus MC, Rotterdam, the Netherlands
| | - Jasper J Anink
- Amsterdam UMC location University of Amsterdam, Department of (Neuro)Pathology Amsterdam Neuroscience, Meibergdreef 9, Amsterdam, the Netherlands
| | - James D Mills
- Amsterdam UMC location University of Amsterdam, Department of (Neuro)Pathology Amsterdam Neuroscience, Meibergdreef 9, Amsterdam, the Netherlands.
| | - Eleonora Aronica
- Amsterdam UMC location University of Amsterdam, Department of (Neuro)Pathology Amsterdam Neuroscience, Meibergdreef 9, Amsterdam, the Netherlands
| |
Collapse
|
28
|
Hurtado-Carneiro V, LeBaut-Ayuso Y, Velázquez E, Flores-Lamas C, Fernández-de la Rosa R, García-García L, Gómez-Oliver F, Ruiz-Albusac JM, Pozo MÁ. Effects of chronic treatment with metformin on brain glucose hypometabolism and central insulin actions in transgenic mice with tauopathy. Heliyon 2024; 10:e35752. [PMID: 39170185 PMCID: PMC11337050 DOI: 10.1016/j.heliyon.2024.e35752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 07/31/2024] [Accepted: 08/02/2024] [Indexed: 08/23/2024] Open
Abstract
Brain glucose hypometabolism and insulin alterations are common features of many neurological diseases. Herein we sought to corroborate the brain glucose hypometabolism that develops with ageing in 12-months old Tau-VLW transgenic mice, a model of tauopathy, as well as to determine whether this model showed signs of altered peripheral glucose metabolism. Our results demonstrated that 12-old months Tau mice exhibited brain glucose hypometabolism as well as basal hyperglycemia, impaired glucose tolerance, hyperinsulinemia, and signs of insulin resistance. Then, we further studied the effect of chronic metformin treatment (9 months) in Tau-VLW mice from 9 to 18 months of age. Longitudinal PET neuroimaging studies revealed that chronic metformin altered the temporal profile in the progression of brain glucose hypometabolism associated with ageing. Besides, metformin altered the content and/or phosphorylation of key components of the insulin signal transduction pathway in the frontal cortex leading to significant changes in the content of the active forms. Thus, metformin increased the expression of pAKT-Y474 while reducing pmTOR-S2448 and pGSK3β. These changes might be related, at least partially, to a slow progression of ageing, neurological damage, and cognitive decline. Metformin also improved the peripheral glucose tolerance and the ability of the Tau-VLW mice to maintain their body weight through ageing. Altogether our study shows that the tau-VLW mice could be a useful model to study the potential interrelationship between tauopathy and central and peripheral glucose metabolism alterations. More importantly our results suggest that chronic metformin treatment may have direct beneficial central effects by post-transcriptional modulation of key components of the insulin signal transduction pathway.
Collapse
Affiliation(s)
| | - Yannick LeBaut-Ayuso
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, Complutense University, Madrid, Spain
| | - Esther Velázquez
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, Complutense University, Madrid, Spain
| | - Cinthya Flores-Lamas
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, Complutense University, Madrid, Spain
| | | | - Luis García-García
- Pluridisciplinary Institute, Complutense University, IdISSC, Madrid, Spain
- Department of Pharmacology, Pharmacognosy and Botany, Faculty of Pharmacy, Complutense University, Madrid, Spain
| | - Francisca Gómez-Oliver
- Pluridisciplinary Institute, Complutense University, IdISSC, Madrid, Spain
- Department of Pharmacology, Pharmacognosy and Botany, Faculty of Pharmacy, Complutense University, Madrid, Spain
| | - Juan Miguel Ruiz-Albusac
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, Complutense University, Madrid, Spain
| | - Miguel Ángel Pozo
- Department of Physiology, Faculty of Medicine, Complutense University, Madrid, Spain
- Pluridisciplinary Institute, Complutense University, IdISSC, Madrid, Spain
| |
Collapse
|
29
|
Chaklai A, Rhea EM, O'Niel A, Babin A, Weaver R, Pemberton S, Banks WA, Raber J. Effects of a high-fat diet on cognition and brain distribution of intranasal insulin in E3 and E4 male and female mice. Sci Rep 2024; 14:18641. [PMID: 39128931 PMCID: PMC11317510 DOI: 10.1038/s41598-024-62053-8] [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: 03/05/2024] [Accepted: 05/08/2024] [Indexed: 08/13/2024] Open
Abstract
There are genetic and environmental risk factors that contribute to the development of cognitive decline in Alzheimer's disease (AD). Some of these include the genetic predisposition of the apolipoprotein E4 genotype, consuming a high-fat diet (HFD), and the female sex. Brain insulin receptor resistance and deficiency have also been shown to be associated with AD and cognitive impairment. Intranasal (INL) insulin enhances cognition in AD, but the response varies due to genotype, diet, and sex. We investigated here the combination of these risk factors in a humanized mouse model, expressing E3 or E4, following a HFD in males and females on cognitive performance and the brain distribution of insulin following INL delivery. The HFD had a negative effect on survival in male mice only, requiring sex to be collapsed. We found many genotype, diet, and genotype x diet effects in anxiety-related tasks. We further found beneficial effects of INL insulin in our memory tests, with the most important findings showing a beneficial effect of INL insulin in mice on a HFD. We found insulin distribution throughout the brain after INL delivery was largely unaffected by diet and genotype, indicating these susceptible groups can still receive adequate levels of insulin following INL delivery. Our findings support the involvement of brain insulin signaling in cognition and highlight continuing efforts investigating mechanisms resulting from treatment with INL insulin.
Collapse
Affiliation(s)
- Ariel Chaklai
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, 97239, USA
| | - Elizabeth M Rhea
- Geriatric Research Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA, 98108, USA
- Department of Medicine, University of Washington, Seattle, WA, 98195, USA
| | - Abigail O'Niel
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, 97239, USA
| | - Alice Babin
- Geriatric Research Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA, 98108, USA
| | - Riley Weaver
- Geriatric Research Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA, 98108, USA
| | - Sarah Pemberton
- Geriatric Research Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA, 98108, USA
| | - William A Banks
- Geriatric Research Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA, 98108, USA
- Department of Medicine, University of Washington, Seattle, WA, 98195, USA
| | - Jacob Raber
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, 97239, USA.
- Division of Neuroscience, Departments of Neurology and Radiation Medicine, ONPRC, Oregon Health & Science University, Portland, OR, 97239, USA.
| |
Collapse
|
30
|
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
|
31
|
Linsmayer D, Eckert GP, Reiff J, Braus DF. [Nutrition, metabolism, brain and mental health]. DER NERVENARZT 2024; 95:667-680. [PMID: 38884643 PMCID: PMC11222242 DOI: 10.1007/s00115-024-01678-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 05/08/2024] [Indexed: 06/18/2024]
Abstract
This review article explores the intricate relationship between nutrition, metabolism, brain function and mental health. It highlights two key complementary models: the energy balance model and the more comprehensive carbohydrate-insulin model, to understand the development of obesity and metabolic dysfunctions. It particularly focuses on the role of dopamine in dietary regulation and insulin in the brain, both of which are crucial in the pathogenesis of neurodegenerative and stress-associated mental disorders. Additionally, the significance of sleep and dietary habits, such as medically assisted calorie restriction for mental health and the concept of "brain food" are described. These findings emphasize the importance of nutritional medicine in psychiatry and psychotherapy and the consideration of metabolic states for the prevention and treatment of mental and neurodegenerative diseases.
Collapse
Affiliation(s)
- Denise Linsmayer
- Vitos Klinikum Rheingau, Kloster-Eberbach-Straße 4, 65346, Eltville, Deutschland
| | - Gunter P Eckert
- Institut für Ernährungswissenschaft, Justus-Liebig-Universität Gießen, Wilhelmstraße 20, 35392, Gießen, Deutschland
| | - Julia Reiff
- Vitos Klinikum Rheingau, Kloster-Eberbach-Straße 4, 65346, Eltville, Deutschland
| | - Dieter F Braus
- Vitos Klinikum Rheingau, Kloster-Eberbach-Straße 4, 65346, Eltville, Deutschland.
| |
Collapse
|
32
|
Gladding JM, Rafiei N, Mitchell CS, Begg DP. Excision of the endothelial blood-brain barrier insulin receptor does not alter spatial cognition in mice fed either a chow or high-fat diet. Neurobiol Learn Mem 2024; 212:107938. [PMID: 38772444 DOI: 10.1016/j.nlm.2024.107938] [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: 12/21/2023] [Revised: 05/02/2024] [Accepted: 05/15/2024] [Indexed: 05/23/2024]
Abstract
Insulin is transported across the blood-brain barrier (BBB) endothelium to regulate aspects of metabolism and cognition. Brain insulin resistance often results from high-fat diet (HFD) consumption and is thought to contribute to spatial cognition deficits. To target BBB insulin function, we used Cre-LoxP genetic excision of the insulin receptor (InsR) from endothelial cells in adult male mice. We hypothesized that this excision would impair spatial cognition, and that high-fat diet consumption would exacerbate these effects. Excision of the endothelial InsR did not impair performance in two spatial cognition tasks, the Y-Maze and Morris Water Maze, in tests held both before and after 14 weeks of access to high-fat (or chow control) diet. The HFD increased body weight gain and induced glucose intolerance but did not impair spatial cognition. Endothelial InsR excision tended to increase body weight and reduce sensitivity to peripheral insulin, but these metabolic effects were not associated with impairments to spatial cognition and did not interact with HFD exposure. Instead, all mice showed intact spatial cognitive performance regardless of whether they had been fed chow or a HFD, and whether the InsR had been excised or not. Overall, the results indicate that loss of the endothelial InsR does not impact spatial cognition, which is in line with pharmacological evidence that other mechanisms at the BBB facilitate insulin transport and allow it to exert its pro-cognitive effects.
Collapse
Affiliation(s)
- Joanne M Gladding
- School of Psychology, Faculty of Science, University of New South Wales, Australia.
| | - Neda Rafiei
- School of Psychology, Faculty of Science, University of New South Wales, Australia
| | - Caitlin S Mitchell
- School of Psychology, Faculty of Science, University of New South Wales, Australia
| | - Denovan P Begg
- School of Psychology, Faculty of Science, University of New South Wales, Australia
| |
Collapse
|
33
|
Li X, Yang Y, Bai X, Wang X, Tan H, Chen Y, Zhu Y, Liu Q, Wu MN, Li Y. A brain-derived insulin signal encodes protein satiety for nutrient-specific feeding inhibition. Cell Rep 2024; 43:114282. [PMID: 38795342 PMCID: PMC11220824 DOI: 10.1016/j.celrep.2024.114282] [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: 08/25/2023] [Revised: 04/08/2024] [Accepted: 05/10/2024] [Indexed: 05/27/2024] Open
Abstract
The suppressive effect of insulin on food intake has been documented for decades. However, whether insulin signals can encode a certain type of nutrients to regulate nutrient-specific feeding behavior remains elusive. Here, we show that in female Drosophila, a pair of dopaminergic neurons, tritocerebrum 1-dopaminergic neurons (T1-DANs), are directly activated by a protein-intake-induced insulin signal from insulin-producing cells (IPCs). Intriguingly, opto-activating IPCs elicits feeding inhibition for both protein and sugar, while silencing T1-DANs blocks this inhibition only for protein food. Elevating insulin signaling in T1-DANs or opto-activating these neurons is sufficient to mimic protein satiety. Furthermore, this signal is conveyed to local neurons of the protocerebral bridge (PB-LNs) and specifically suppresses protein intake. Therefore, our findings reveal that a brain-derived insulin signal encodes protein satiety and suppresses feeding behavior in a nutrient-specific manner, shedding light on the functional specificity of brain insulin signals in regulating behaviors.
Collapse
Affiliation(s)
- Xiaoyu Li
- Institute of Biophysics, State Key Laboratory of Brain and Cognitive Science, Center for Excellence in Biomacromolecules, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yang Yang
- Institute of Biophysics, State Key Laboratory of Brain and Cognitive Science, Center for Excellence in Biomacromolecules, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xiaobing Bai
- Institute of Biophysics, State Key Laboratory of Brain and Cognitive Science, Center for Excellence in Biomacromolecules, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China; Sino-Danish Center for Education and Research, Beijing 100190, China
| | - Xiaotong Wang
- Institute of Biophysics, State Key Laboratory of Brain and Cognitive Science, Center for Excellence in Biomacromolecules, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Houqi Tan
- Institute of Biophysics, State Key Laboratory of Brain and Cognitive Science, Center for Excellence in Biomacromolecules, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yanbo Chen
- Institute of Biophysics, State Key Laboratory of Brain and Cognitive Science, Center for Excellence in Biomacromolecules, Chinese Academy of Sciences, Beijing 100101, China
| | - Yan Zhu
- Institute of Biophysics, State Key Laboratory of Brain and Cognitive Science, Center for Excellence in Biomacromolecules, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China; Sino-Danish Center for Education and Research, Beijing 100190, China
| | - Qili Liu
- Department of Anatomy, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Mark N Wu
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Yan Li
- Institute of Biophysics, State Key Laboratory of Brain and Cognitive Science, Center for Excellence in Biomacromolecules, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China; Sino-Danish Center for Education and Research, Beijing 100190, China.
| |
Collapse
|
34
|
Singh MK, Shin Y, Ju S, Han S, Kim SS, Kang I. Comprehensive Overview of Alzheimer's Disease: Etiological Insights and Degradation Strategies. Int J Mol Sci 2024; 25:6901. [PMID: 39000011 PMCID: PMC11241648 DOI: 10.3390/ijms25136901] [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: 05/21/2024] [Revised: 06/19/2024] [Accepted: 06/21/2024] [Indexed: 07/14/2024] Open
Abstract
Alzheimer's disease (AD) is the most prevalent neurodegenerative disorder and affects millions of individuals globally. AD is associated with cognitive decline and memory loss that worsens with aging. A statistical report using U.S. data on AD estimates that approximately 6.9 million individuals suffer from AD, a number projected to surge to 13.8 million by 2060. Thus, there is a critical imperative to pinpoint and address AD and its hallmark tau protein aggregation early to prevent and manage its debilitating effects. Amyloid-β and tau proteins are primarily associated with the formation of plaques and neurofibril tangles in the brain. Current research efforts focus on degrading amyloid-β and tau or inhibiting their synthesis, particularly targeting APP processing and tau hyperphosphorylation, aiming to develop effective clinical interventions. However, navigating this intricate landscape requires ongoing studies and clinical trials to develop treatments that truly make a difference. Genome-wide association studies (GWASs) across various cohorts identified 40 loci and over 300 genes associated with AD. Despite this wealth of genetic data, much remains to be understood about the functions of these genes and their role in the disease process, prompting continued investigation. By delving deeper into these genetic associations, novel targets such as kinases, proteases, cytokines, and degradation pathways, offer new directions for drug discovery and therapeutic intervention in AD. This review delves into the intricate biological pathways disrupted in AD and identifies how genetic variations within these pathways could serve as potential targets for drug discovery and treatment strategies. Through a comprehensive understanding of the molecular underpinnings of AD, researchers aim to pave the way for more effective therapies that can alleviate the burden of this devastating disease.
Collapse
Affiliation(s)
- Manish Kumar Singh
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Yoonhwa Shin
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Songhyun Ju
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Sunhee Han
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Sung Soo Kim
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Insug Kang
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| |
Collapse
|
35
|
Shi Y, Zhang X, Feng Y, Yue Z. Association of metabolic syndrome and its components with Parkinson's disease: a cross-sectional study. BMC Endocr Disord 2024; 24:92. [PMID: 38890672 PMCID: PMC11186221 DOI: 10.1186/s12902-024-01623-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Accepted: 06/13/2024] [Indexed: 06/20/2024] Open
Abstract
BACKGROUND The interrelation between metabolic syndrome (MetS) and Parkinson's disease (PD) likely arises from shared pathological mechanisms. This study thus aims to examine the impact of MetS and its components on PD. METHODS This study utilized data extracted from the National Health and Nutrition Examination Survey database spanning 1999 to 2020. The random forest algorithm was applied to fill in the missing data. Propensity score optimal full matching was conducted. The data were adjusted by total weights derived from both sampling and matching weights. The weighted data were utilized to create multifactor logistic regression models. Odds ratios (ORs) and average marginal effects, along with their corresponding 95% confidence intervals (CIs), were calculated. RESULTS MetS did not significantly affect the risk of PD (OR: 1.01; 95% CI: 0.77, 1.34; P = 0.92). Hypertension elevated the risk of PD (OR: 1.33; 95% CI: 1.01, 1.76; P = 0.045), accompanied by a 0.26% increased probability of PD occurrence (95% CI: 0.01%, 0.52%; P = 0.04). Diabetes mellitus (DM) had a 1.38 times greater likelihood of developing PD (OR:1.38; 95% CI: 1.004, 1.89; P = 0.046), corresponding to a 0.32% increased probability of PD occurrence (95% CI: -0.03%, 0.67%; P = 0.07). Nevertheless, no correlation was observed between hyperlipidemia, waist circumference and PD. CONCLUSION MetS does not affect PD; however, hypertension and DM significantly increase the risk of PD.
Collapse
Affiliation(s)
- Yue Shi
- Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - XueYi Zhang
- Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yue Feng
- Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| | - ZongXiang Yue
- Meishan Hospital of Traditional Chinese Medicine, Meishan, China.
| |
Collapse
|
36
|
Lemche E, Killick R, Mitchell J, Caton PW, Choudhary P, Howard JK. Molecular mechanisms linking type 2 diabetes mellitus and late-onset Alzheimer's disease: A systematic review and qualitative meta-analysis. Neurobiol Dis 2024; 196:106485. [PMID: 38643861 DOI: 10.1016/j.nbd.2024.106485] [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: 06/30/2023] [Revised: 03/18/2024] [Accepted: 03/23/2024] [Indexed: 04/23/2024] Open
Abstract
Research evidence indicating common metabolic mechanisms through which type 2 diabetes mellitus (T2DM) increases risk of late-onset Alzheimer's dementia (LOAD) has accumulated over recent decades. The aim of this systematic review is to provide a comprehensive review of common mechanisms, which have hitherto been discussed in separate perspectives, and to assemble and evaluate candidate loci and epigenetic modifications contributing to polygenic risk linkages between T2DM and LOAD. For the systematic review on pathophysiological mechanisms, both human and animal studies up to December 2023 are included. For the qualitative meta-analysis of genomic bases, human association studies were examined; for epigenetic mechanisms, data from human studies and animal models were accepted. Papers describing pathophysiological studies were identified in databases, and further literature gathered from cited work. For genomic and epigenomic studies, literature mining was conducted by formalised search codes using Boolean operators in search engines, and augmented by GeneRif citations in Entrez Gene, and other sources (WikiGenes, etc.). For the systematic review of pathophysiological mechanisms, 923 publications were evaluated, and 138 gene loci extracted for testing candidate risk linkages. 3 57 publications were evaluated for genomic association and descriptions of epigenomic modifications. Overall accumulated results highlight insulin signalling, inflammation and inflammasome pathways, proteolysis, gluconeogenesis and glycolysis, glycosylation, lipoprotein metabolism and oxidation, cell cycle regulation or survival, autophagic-lysosomal pathways, and energy. Documented findings suggest interplay between brain insulin resistance, neuroinflammation, insult compensatory mechanisms, and peripheral metabolic dysregulation in T2DM and LOAD linkage. The results allow for more streamlined longitudinal studies of T2DM-LOAD risk linkages.
Collapse
Affiliation(s)
- Erwin Lemche
- Section of Cognitive Neuropsychiatry and Centre for Neuroimaging Sciences, Institute of Psychiatry, Psychology & Neuroscience, King's College London, De Crespigny Park, London SE5 8AF, United Kingdom.
| | - Richard Killick
- Section of Old Age Psychiatry, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology & Neuroscience, King's College London, De Crespigny Park, London SE5 8AF, United Kingdom
| | - Jackie Mitchell
- Department of Basic and Clinical Neurosciences, Maurice Wohl CIinical Neurosciences Institute, Institute of Psychiatry, Psychology & Neuroscience, King's College London, 125 Coldharbour Lane, London SE5 9NU, United Kingdom
| | - Paul W Caton
- Diabetes Research Group, School of Life Course Sciences, King's College London, Hodgkin Building, Guy's Campus, London SE1 1UL, United Kingdom
| | - Pratik Choudhary
- Diabetes Research Group, Weston Education Centre, King's College London, 10 Cutcombe Road, London SE5 9RJ, United Kingdom
| | - Jane K Howard
- School of Cardiovascular and Metabolic Medicine & Sciences, Hodgkin Building, Guy's Campus, King's College London, Great Maze Pond, London SE1 1UL, United Kingdom
| |
Collapse
|
37
|
Schwartz SS, Herman ME. Gluco-regulation & type 2 diabetes: entrenched misconceptions updated to new governing principles for gold standard management. Front Endocrinol (Lausanne) 2024; 15:1394805. [PMID: 38933821 PMCID: PMC11199379 DOI: 10.3389/fendo.2024.1394805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2024] [Accepted: 05/16/2024] [Indexed: 06/28/2024] Open
Abstract
Our understanding of type 2 diabetes (T2D) has evolved dramatically. Advances have upended entrenched dogmas pertaining to the onset and progression of T2D, beliefs that have prevailed from the early era of diabetes research-and continue to populate our medical textbooks and continuing medical education materials. This review article highlights key insights that lend new governing principles for gold standard management of T2D. From the historical context upon which old beliefs arose to new findings, this article outlines evidence and perspectives on beta cell function, the underlying defects in glucoregulation, the remediable nature of T2D, and, the rationale supporting the shift to complication-centric prescribing. Practical approaches translate this rectified understanding of T2D into strategies that fill gaps in current management practices of prediabetes through late type 2 diabetes.
Collapse
Affiliation(s)
- Stanley S. Schwartz
- Main Line Health, Wynnewood, PA, and University of Pennsylvania, Philadelphia, PA, United States
| | - Mary E. Herman
- Social Alchemy: Building Physician Competency Across the Globe, Sacatepéquez, Guatemala
| |
Collapse
|
38
|
Cai M, Wan J, Cai K, Li S, Du X, Song H, Sun W, Hu J. The mitochondrial quality control system: a new target for exercise therapeutic intervention in the treatment of brain insulin resistance-induced neurodegeneration in obesity. Int J Obes (Lond) 2024; 48:749-763. [PMID: 38379083 DOI: 10.1038/s41366-024-01490-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 01/30/2024] [Accepted: 02/01/2024] [Indexed: 02/22/2024]
Abstract
Obesity is a major global health concern because of its strong association with metabolic and neurodegenerative diseases such as diabetes, dementia, and Alzheimer's disease. Unfortunately, brain insulin resistance in obesity is likely to lead to neuroplasticity deficits. Since the evidence shows that insulin resistance in brain regions abundant in insulin receptors significantly alters mitochondrial efficiency and function, strategies targeting the mitochondrial quality control system may be of therapeutic and practical value in obesity-induced cognitive decline. Exercise is considered as a powerful stimulant of mitochondria that improves insulin sensitivity and enhances neuroplasticity. It has great potential as a non-pharmacological intervention against the onset and progression of obesity associated neurodegeneration. Here, we integrate the current knowledge of the mechanisms of neurodegenration in obesity and focus on brain insulin resistance to explain the relationship between the impairment of neuronal plasticity and mitochondrial dysfunction. This knowledge was synthesised to explore the exercise paradigm as a feasible intervention for obese neurodegenration in terms of improving brain insulin signals and regulating the mitochondrial quality control system.
Collapse
Affiliation(s)
- Ming Cai
- Jinshan District Central Hospital affiliated to Shanghai University of Medicine & Health Sciences, Shanghai, 201599, China
| | - Jian Wan
- Department of Emergency and Critical Care Medicine, Shanghai Pudong New Area People's Hospital, Shanghai, 201299, China
| | - Keren Cai
- College of Rehabilitation Sciences, Shanghai University of Medicine and Health Sciences, Shanghai, 201318, China
| | - Shuyao Li
- College of Rehabilitation Sciences, Shanghai University of Medicine and Health Sciences, Shanghai, 201318, China
| | - Xinlin Du
- College of Rehabilitation Sciences, Shanghai University of Medicine and Health Sciences, Shanghai, 201318, China
| | - Haihan Song
- Central Lab, Shanghai Key Laboratory of Pathogenic Fungi Medical Testing, Shanghai Pudong New Area People's Hospital, Shanghai, 201299, China
| | - Wanju Sun
- Central Lab, Shanghai Key Laboratory of Pathogenic Fungi Medical Testing, Shanghai Pudong New Area People's Hospital, Shanghai, 201299, China.
| | - Jingyun Hu
- Central Lab, Shanghai Key Laboratory of Pathogenic Fungi Medical Testing, Shanghai Pudong New Area People's Hospital, Shanghai, 201299, China.
| |
Collapse
|
39
|
Ohtsuki S. Insulin receptor at the blood-brain barrier: Transport and signaling. VITAMINS AND HORMONES 2024; 126:113-124. [PMID: 39029970 DOI: 10.1016/bs.vh.2024.05.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/21/2024]
Abstract
The blood-brain barrier (BBB) is a unique system of the brain microvasculature that limits the exchange between the blood and the brain. Brain microvascular endothelial cells form the BBB as part of the neurovascular unit and express insulin receptors. The insulin receptor at the BBB has been studied in two different functional aspects. These functions include (1) the supplying of blood insulin to the brain and (2) the modulation of BBB function via insulin signaling. The first function involves drug delivery to the brain, while the second function is related to the association between central nervous system diseases and type 2 diabetes through insulin resistance. This chapter summarizes recent progress in research on the function of insulin receptors at the BBB.
Collapse
Affiliation(s)
- Sumio Ohtsuki
- Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan.
| |
Collapse
|
40
|
Todorovic S, Simeunovic V, Prvulovic M, Dakic T, Jevdjovic T, Sokanovic S, Kanazir S, Mladenovic A. Dietary restriction alters insulin signaling pathway in the brain. Biofactors 2024; 50:450-466. [PMID: 37975613 DOI: 10.1002/biof.2018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Accepted: 09/07/2023] [Indexed: 11/19/2023]
Abstract
Insulin is known to be a key hormone in the regulation of peripheral glucose homeostasis, but beyond that, its effects on the brain are now undisputed. Impairments in insulin signaling in the brain, including changes in insulin levels, are thought to contribute significantly to declines in cognitive performance, especially during aging. As one of the most widely studied experimental interventions, dietary restriction (DR) is considered to delay the neurodegenerative processes associated with aging. Recently, however, data began to suggest that the onset and duration of a restrictive diet play a critical role in the putative beneficial outcome. Because the effects of DR on insulin signaling in the brain have been poorly studied, we decided to examine the effects of DR that differed in onset and duration: long-term DR (LTDR), medium-term DR (MTDR), and short-term DR (STDR) on the expression of proteins involved in insulin signaling in the hippocampus of 18- and 24-month-old male Wistar rats. We found that DR-induced changes in insulin levels in the brain may be independent of what happens in the periphery after restricted feeding. Significantly changed insulin content in the hippocampus, together with altered insulin signaling were found under the influence of DR, but the outcome was highly dependent on the onset and duration of DR.
Collapse
Affiliation(s)
- Smilja Todorovic
- Department for Neurobiology, Institute for Biological Research "Sinisa Stankovic", National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Valentina Simeunovic
- Department for Neurobiology, Institute for Biological Research "Sinisa Stankovic", National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Milica Prvulovic
- Department for Neurobiology, Institute for Biological Research "Sinisa Stankovic", National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Tamara Dakic
- Department for Comparative Physiology and Ecophysiology, Institute for Physiology and Biochemistry "Ivan Djaja", Faculty of Biology, University of Belgrade, Belgrade, Serbia
| | - Tanja Jevdjovic
- Department for Comparative Physiology and Ecophysiology, Institute for Physiology and Biochemistry "Ivan Djaja", Faculty of Biology, University of Belgrade, Belgrade, Serbia
| | - Srdjan Sokanovic
- Department for Neurobiology, Institute for Biological Research "Sinisa Stankovic", National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Selma Kanazir
- Department for Neurobiology, Institute for Biological Research "Sinisa Stankovic", National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Aleksandra Mladenovic
- Department for Neurobiology, Institute for Biological Research "Sinisa Stankovic", National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| |
Collapse
|
41
|
Albar NY, Hassaballa H, Shikh H, Albar Y, Ibrahim AS, Mousa AH, Alshanberi AM, Elgebaly A, Bahbah EI. The interaction between insulin resistance and Alzheimer's disease: a review article. Postgrad Med 2024; 136:377-395. [PMID: 38804907 DOI: 10.1080/00325481.2024.2360887] [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: 01/28/2024] [Accepted: 05/23/2024] [Indexed: 05/29/2024]
Abstract
Insulin serves multiple functions as a growth-promoting hormone in peripheral tissues. It manages glucose metabolism by promoting glucose uptake into cells and curbing the production of glucose in the liver. Beyond this, insulin fosters cell growth, drives differentiation, aids protein synthesis, and deters degradative processes like glycolysis, lipolysis, and proteolysis. Receptors for insulin and insulin-like growth factor-1 are widely expressed in the central nervous system. Their widespread presence in the brain underscores the varied and critical functions of insulin signaling there. Insulin aids in bolstering cognition, promoting neuron extension, adjusting the release and absorption of catecholamines, and controlling the expression and positioning of gamma-aminobutyric acid (GABA). Importantly, insulin can effortlessly traverse the blood-brain barrier. Furthermore, insulin resistance (IR)-induced alterations in insulin signaling might hasten brain aging, impacting its plasticity and potentially leading to neurodegeneration. Two primary pathways are responsible for insulin signal transmission: the phosphoinositide 3-kinase (PI3K)/protein kinase B (AKT) pathway, which oversees metabolic responses, and the mitogen-activated protein kinase (MAPK) pathway, which guides cell growth, survival, and gene transcription. This review aimed to explore the potential shared metabolic traits between Alzheimer's disease (AD) and IR disorders. It delves into the relationship between AD and IR disorders, their overlapping genetic markers, and shared metabolic indicators. Additionally, it addresses existing therapeutic interventions targeting these intersecting pathways.
Collapse
Affiliation(s)
- Nezar Y Albar
- Internal Medicine Department, Dr. Samir Abbas Hospital, Jeddah, Saudi Arabia
| | | | - Hamza Shikh
- Ibn Sina National College for Medical Studies, Jeddah, Saudi Arabia
| | - Yassin Albar
- Fakeeh College of Medical Sciences, Jeddah, Saudi Arabia
| | | | - Ahmed Hafez Mousa
- Department of Neurosurgery, Postgraduate Medical Education, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai, United Arab Emirates
- Department of Neurosurgery, Rashid Hospital, Dubai Academic Health Cooperation, Dubai, United Arab Emirates
| | - Asim Muhammed Alshanberi
- Department of Community Medicine and Pilgrims Health Care, Umm Alqura University, Makkah, Saudi Arabia
- Medicine Program, Batterjee Medical College, Jeddah, Saudi Arabia
| | - Ahmed Elgebaly
- Smart Health Academic Unit, University of East London, London, UK
| | - Eshak I Bahbah
- Faculty of Medicine, Al-Azhar University, Damietta, Egypt
| |
Collapse
|
42
|
Feng Z, Fang C, Ma Y, Chang J. Obesity-induced blood-brain barrier dysfunction: phenotypes and mechanisms. J Neuroinflammation 2024; 21:110. [PMID: 38678254 PMCID: PMC11056074 DOI: 10.1186/s12974-024-03104-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 04/17/2024] [Indexed: 04/29/2024] Open
Abstract
Obesity, a burgeoning global health issue, is increasingly recognized for its detrimental effects on the central nervous system, particularly concerning the integrity of the blood-brain barrier (BBB). This manuscript delves into the intricate relationship between obesity and BBB dysfunction, elucidating the underlying phenotypes and molecular mechanisms. We commence with an overview of the BBB's critical role in maintaining cerebral homeostasis and the pathological alterations induced by obesity. By employing a comprehensive literature review, we examine the structural and functional modifications of the BBB in the context of obesity, including increased permeability, altered transport mechanisms, and inflammatory responses. The manuscript highlights how obesity-induced systemic inflammation and metabolic dysregulation contribute to BBB disruption, thereby predisposing individuals to various neurological disorders. We further explore the potential pathways, such as oxidative stress and endothelial cell dysfunction, that mediate these changes. Our discussion culminates in the summary of current findings and the identification of knowledge gaps, paving the way for future research directions. This review underscores the significance of understanding BBB dysfunction in obesity, not only for its implications in neurodegenerative diseases but also for developing targeted therapeutic strategies to mitigate these effects.
Collapse
Affiliation(s)
- Ziying Feng
- Key Laboratory of Biomedical Imaging Science, Shenzhen Institute of Advanced Technology, System of Chinese Academy of Sciences, Chinese Academy of Sciences, Shenzhen, Guangdong, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Cheng Fang
- Key Laboratory of Biomedical Imaging Science, Shenzhen Institute of Advanced Technology, System of Chinese Academy of Sciences, Chinese Academy of Sciences, Shenzhen, Guangdong, China
| | - Yinzhong Ma
- Key Laboratory of Biomedical Imaging Science, Shenzhen Institute of Advanced Technology, System of Chinese Academy of Sciences, Chinese Academy of Sciences, Shenzhen, Guangdong, China.
- Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Xueyuan Ave 1068, Nanshan, Shenzhen, 518055, Guangdong, China.
| | - Junlei Chang
- Key Laboratory of Biomedical Imaging Science, Shenzhen Institute of Advanced Technology, System of Chinese Academy of Sciences, Chinese Academy of Sciences, Shenzhen, Guangdong, China.
- University of Chinese Academy of Sciences, Beijing, China.
- Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Xueyuan Ave 1068, Nanshan, Shenzhen, 518055, Guangdong, China.
| |
Collapse
|
43
|
Xu Y, Nie J, Lu C, Hu C, Chen Y, Ma Y, Huang Y, Lu L. Effects and mechanisms of bisphenols exposure on neurodegenerative diseases risk: A systemic review. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 919:170670. [PMID: 38325473 DOI: 10.1016/j.scitotenv.2024.170670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 01/12/2024] [Accepted: 02/01/2024] [Indexed: 02/09/2024]
Abstract
Environmental bisphenols (BPs) pose a global threat to human health because of their extensive use as additives in plastic products. BP residues are increasing in various environmental media (i.e., water, soil, and indoor dust) and biological and human samples (i.e., serum and brain). Both epidemiological and animal studies have determined an association between exposure to BPs and an increased risk of neurodegenerative diseases (e.g., Parkinson's disease, Alzheimer's disease, and amyotrophic lateral sclerosis), including cognitive abnormalities and behavioral disturbances. Hence, understanding the biological responses to different BPs is essential for prevention, and treatment. This study provides an overview of the underlying pathogenic molecular mechanisms as a valuable basis for understanding neurodegenerative disease responses to BPs, including accumulation of misfolded proteins, reduction of tyrosine hydroxylase and dopamine, abnormal hormone signaling, neuronal death, oxidative stress, calcium homeostasis, and inflammation. These findings provide new insights into the neurotoxic potential of BPs and ultimately contribute to a comprehensive health risk evaluation.
Collapse
Affiliation(s)
- Yeqing Xu
- School of Public Health, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
| | - Jun Nie
- School of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China; School of Engineering, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
| | - Chenghao Lu
- College of Mathematics and Computer Science, Zhejiang A & F University, Hangzhou 311300, China
| | - Chao Hu
- School of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China; School of Engineering, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
| | - Yunlu Chen
- School of Public Health, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
| | - Ying Ma
- School of Public Health, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
| | - Yuru Huang
- School of Public Health, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
| | - Liping Lu
- School of Public Health, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China.
| |
Collapse
|
44
|
Cukierman-Yaffe T, Ramasundarahettige C, Bosch J, Gerstein HC. Effect of basal insulin and omega 3 fatty acids on cognitive impairment in dysglycaemia: An exploratory analysis of the ORIGIN trial. Diabetes Obes Metab 2024; 26:1180-1187. [PMID: 38204215 DOI: 10.1111/dom.15412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 12/03/2023] [Accepted: 12/04/2023] [Indexed: 01/12/2024]
Abstract
AIM The outcomes reduction with an initial glargine intervention (ORIGIN) trial reported that, allocation to insulin glargine-mediated normoglycaemia versus standard care, and to omega 3 fatty acids versus placebo had a neutral effect on cognitive test scores when analysed as continuous variables. Analyses of these scores as standardized categorical variables using a previously validated strategy may yield different results. MATERIALS AND METHODS The ORIGIN trial recruited participants with dysglycaemia and additional cardiovascular risk factors from 573 sites in 40 countries. They completed a mini mental state examination and a subset completed the digit symbol substitution test at baseline and up to three subsequent visits. The effect of the interventions on country-standardized substantive cognitive impairment, defined as the first occurrence of a baseline-adjusted follow-up mini mental state examination or digit symbol substitution test score ≥1.5 standard deviations below the baseline mean score in each participant's country was assessed using Cox proportional hazards models. RESULTS During a median follow-up of 6.2 years, 2627 of 11 682 people (22.5%) developed country-standardized substantive cognitive impairment. The hazard of this outcome was reduced by 9% (hazard ratio 0.91, 95% confidence interval 0.85, 0.99; p = .023) in participants assigned to insulin glargine (21.6%) versus standard care (23.3%). Conversely, the hazard of this outcome was not affected by assignment to omega 3 fatty acid versus placebo (hazard ratio 0.93, 95% confidence interval 0.86, 1.01; p = .074). CONCLUSIONS In this post hoc exploratory analysis, insulin glargine-mediated normoglycaemia but not omega 3 fatty acids reduced the hazard of substantive cognitive impairment in people with dysglycaemia and additional cardiovascular risk factors.
Collapse
Affiliation(s)
- Tali Cukierman-Yaffe
- Division of Endocrinology & Metabolism, Sheba Medical Center, Ramat Gan, Israel
- Epidemiology Department, School of Public Health, Faculty of Medicine, Herczeg Institute of Aging, Tel-Aviv University, Tel Aviv, Israel
- Population Health Research Institute, McMaster University and Hamilton Health Sciences, Hamilton, Ontario, Canada
| | - Chinthanie Ramasundarahettige
- Population Health Research Institute, McMaster University and Hamilton Health Sciences, Hamilton, Ontario, Canada
- School of Rehabilitation Sciences, McMaster University, Hamilton, Ontario, Canada
| | - Jackie Bosch
- Population Health Research Institute, McMaster University and Hamilton Health Sciences, Hamilton, Ontario, Canada
- School of Rehabilitation Sciences, McMaster University, Hamilton, Ontario, Canada
| | - Hertzel C Gerstein
- Population Health Research Institute, McMaster University and Hamilton Health Sciences, Hamilton, Ontario, Canada
| |
Collapse
|
45
|
Rhea EM, Banks WA. Insulin and the blood-brain barrier. VITAMINS AND HORMONES 2024; 126:169-190. [PMID: 39029972 DOI: 10.1016/bs.vh.2024.02.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/21/2024]
Abstract
The blood-brain barrier (BBB) predominantly regulates insulin transport into and levels within the brain. The BBB is also an important site of insulin binding and mediator of insulin receptor (INSR) signaling. The insulin transporter is separate from the INSR, highlighting the important, unique role of each protein in this structure. After a brief introduction on the structure of insulin and the INSR, we discuss the importance of insulin interactions at the BBB, the properties of the insulin transporter and the role of the BBB insulin transporter in various physiological conditions. We go on to further describe insulin BBB signaling and the impact not only within brain endothelial cells but also the cascade into other cell types within the brain. We close with future considerations to advance our knowledge about the importance of insulin at the BBB.
Collapse
Affiliation(s)
- Elizabeth M Rhea
- Geriatric Research Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA, United States; Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington School of Medicine, Seattle, WA, United States.
| | - William A Banks
- Geriatric Research Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA, United States; Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington School of Medicine, Seattle, WA, United States
| |
Collapse
|
46
|
Anderson GM, Hill JW, Kaiser UB, Navarro VM, Ong KK, Perry JRB, Prevot V, Tena-Sempere M, Elias CF. Metabolic control of puberty: 60 years in the footsteps of Kennedy and Mitra's seminal work. Nat Rev Endocrinol 2024; 20:111-123. [PMID: 38049643 PMCID: PMC10843588 DOI: 10.1038/s41574-023-00919-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/19/2023] [Indexed: 12/06/2023]
Abstract
An individual's nutritional status has a powerful effect on sexual maturation. Puberty onset is delayed in response to chronic energy insufficiency and is advanced under energy abundance. The consequences of altered pubertal timing for human health are profound. Late puberty increases the chances of cardiometabolic, musculoskeletal and neurocognitive disorders, whereas early puberty is associated with increased risks of adult obesity, type 2 diabetes mellitus, cardiovascular diseases and various cancers, such as breast, endometrial and prostate cancer. Kennedy and Mitra's trailblazing studies, published in 1963 and using experimental models, were the first to demonstrate that nutrition is a key factor in puberty onset. Building on this work, the field has advanced substantially in the past decade, which is largely due to the impressive development of molecular tools for experimentation and population genetics. In this Review, we discuss the latest advances in basic and translational sciences underlying the nutritional and metabolic control of pubertal development, with a focus on perspectives and future directions.
Collapse
Affiliation(s)
- Greg M Anderson
- Centre for Neuroendocrinology, University of Otago, Dunedin, New Zealand
- Department of Anatomy, University of Otago, Dunedin, New Zealand
| | - Jennifer W Hill
- Department of Physiology and Pharmacology, University of Toledo, Toledo, OH, USA
- Center for Diabetes and Endocrine Research, University of Toledo, Toledo, OH, USA
| | - Ursula B Kaiser
- Division of Endocrinology, Diabetes, and Hypertension, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Victor M Navarro
- Division of Endocrinology, Diabetes, and Hypertension, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Ken K Ong
- Metabolic Research Laboratory, Wellcome-MRC Institute of Metabolic Science, University of Cambridge School of Clinical Medicine, Cambridge, UK
| | - John R B Perry
- Metabolic Research Laboratory, Wellcome-MRC Institute of Metabolic Science, University of Cambridge School of Clinical Medicine, Cambridge, UK
- MRC Epidemiology Unit, Wellcome-MRC Institute of Metabolic Science, University of Cambridge School of Clinical Medicine, Cambridge, UK
| | - Vincent Prevot
- University of Lille, Inserm, CHU Lille, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Lille Neuroscience & Cognition, UMR-S 1172, Lille, France
- European Genomic Institute for Diabetes (EGID), Lille, France
| | - Manuel Tena-Sempere
- Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Cordoba, Spain.
- Department of Cell Biology, Physiology and Immunology, University of Cordoba, Cordoba, Spain.
- CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Cordoba, Spain.
| | - Carol F Elias
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI, USA.
- Department of Obstetrics & Gynecology, University of Michigan, Ann Arbor, MI, USA.
- Caswell Diabetes Institute, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
47
|
Chaves JCS, Dando SJ, White AR, Oikari LE. Blood-brain barrier transporters: An overview of function, dysfunction in Alzheimer's disease and strategies for treatment. Biochim Biophys Acta Mol Basis Dis 2024; 1870:166967. [PMID: 38008230 DOI: 10.1016/j.bbadis.2023.166967] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 11/15/2023] [Accepted: 11/17/2023] [Indexed: 11/28/2023]
Abstract
The blood-brain-barrier (BBB) has a major function in maintaining brain homeostasis by regulating the entry of molecules from the blood to the brain. Key players in BBB function are BBB transporters which are highly expressed in brain endothelial cells (BECs) and critical in mediating the exchange of nutrients and waste products. BBB transporters can also influence drug delivery into the brain by inhibiting or facilitating the entry of brain targeting therapeutics for the treatment of brain disorders, such as Alzheimer's disease (AD). Recent studies have shown that AD is associated with a disrupted BBB and transporter dysfunction, although their roles in the development in AD are not fully understand. Modulation of BBB transporter activity may pose a novel approach to enhance the delivery of drugs to the brain for enhanced treatment of AD. In this review, we will give an overview of key functions of BBB transporters and known changes in AD. In addition, we will discuss current strategies for transporter modulation for enhanced drug delivery into the brain.
Collapse
Affiliation(s)
- Juliana C S Chaves
- Mental Health and Neuroscience Program, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia; School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, QUT, Brisbane, QLD, Australia
| | - Samantha J Dando
- Centre for Immunology and Infection Control, School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, Australia
| | - Anthony R White
- Mental Health and Neuroscience Program, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia; School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, QUT, Brisbane, QLD, Australia
| | - Lotta E Oikari
- Mental Health and Neuroscience Program, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia; School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, QUT, Brisbane, QLD, Australia.
| |
Collapse
|
48
|
Medina-Vera D, López-Gambero AJ, Navarro JA, Sanjuan C, Baixeras E, Decara J, de Fonseca FR. Novel insights into D-Pinitol based therapies: a link between tau hyperphosphorylation and insulin resistance. Neural Regen Res 2024; 19:289-295. [PMID: 37488880 PMCID: PMC10503604 DOI: 10.4103/1673-5374.379015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 04/21/2023] [Accepted: 05/18/2023] [Indexed: 07/26/2023] Open
Abstract
Alzheimer's disease is a neurodegenerative disorder characterized by the amyloid accumulation in the brains of patients with Alzheimer's disease. The pathogenesis of Alzheimer's disease is mainly mediated by the phosphorylation and aggregation of tau protein. Among the multiple causes of tau hyperphosphorylation, brain insulin resistance has generated much attention, and inositols as insulin sensitizers, are currently considered candidates for drug development. The present narrative review revises the interactions between these three elements: Alzheimer's disease-tau-inositols, which can eventually identify targets for new disease modifiers capable of bringing hope to the millions of people affected by this devastating disease.
Collapse
Affiliation(s)
- Dina Medina-Vera
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, UGC Salud Mental, Málaga, Spain
- Facultad de Ciencias, Universidad de Málaga, Málaga, Spain
- CIBER Enfermedades Cardiovasculares (CIBERCV), Hospital Universitario Virgen de la Victoria, UGC del Corazón, Málaga, Spain
| | - Antonio Jesús López-Gambero
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, UGC Salud Mental, Málaga, Spain
- Facultad de Ciencias, Universidad de Málaga, Málaga, Spain
- University of Bordeaux, INSERM, Neurocentre Magendie, Bordeaux, France
| | - Juan Antonio Navarro
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, UGC Salud Mental, Málaga, Spain
| | | | - Elena Baixeras
- Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Universidad de Málaga, Málaga, Spain
| | - Juan Decara
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, UGC Salud Mental, Málaga, Spain
| | - Fernando Rodríguez de Fonseca
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, UGC Salud Mental, Málaga, Spain
| |
Collapse
|
49
|
Zeng X, Wu C, Cao Y, Li H, Zhang X. Mdm2-mediated ubiquitination of PKCβII is responsible for insulin-induced heterologous desensitization of dopamine D 3 receptor. FEBS Lett 2024; 598:400-414. [PMID: 38302840 DOI: 10.1002/1873-3468.14815] [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: 11/13/2023] [Revised: 12/21/2023] [Accepted: 01/05/2024] [Indexed: 02/03/2024]
Abstract
The insulin and dopaminergic systems in the brain are associated with schizophrenia and Parkinson's disease with respect to etiology and treatment. The present study investigated the crosstalk between the insulin receptor (IR) and dopamine receptor and found that insulin stimulation selectively inhibits signaling of D3 R in a PKCβII-dependent manner. Upon insulin stimulation, E3 ligase enzyme Mdm2 moves out of the nucleus to ubiquitinate PKCβII. Subsequently, ubiquitinated PKCβII translocates to the cell membrane and interacts with D3 R in a phosphorylation-dependent manner at S229/257, resulting in the attenuation of D3 R signaling and initiating clathrin-mediated endocytosis and downregulation. Considering that both IR and D3 R are closely related to some neuropsychosis, this study could provide new molecular insight into the etiology of the disorder.
Collapse
Affiliation(s)
- Xingyue Zeng
- School of Pharmaceutical Sciences, Guizhou University, Guiyang, China
| | - ChengYan Wu
- School of Pharmaceutical Sciences, Guizhou University, Guiyang, China
| | - Yongkai Cao
- Department of Neurology, Institute of Translational Medicine, The First Affiliated Hospital of Shenzhen University, China
| | - Huijun Li
- Department of Pharmaceuticals, People's Hospital of Zunyi City Bo Zhou District, China
| | - Xiaohan Zhang
- School of Pharmaceutical Sciences, Guizhou University, Guiyang, China
| |
Collapse
|
50
|
Zamolodchikova TS, Tolpygo SM, Kotov AV. Insulin in the regulation of the renin-angiotensin system: a new perspective on the mechanism of insulin resistance and diabetic complications. Front Endocrinol (Lausanne) 2024; 15:1293221. [PMID: 38323106 PMCID: PMC10844507 DOI: 10.3389/fendo.2024.1293221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Accepted: 01/08/2024] [Indexed: 02/08/2024] Open
|