1
|
Xu F, Shi J. Insulin signaling and oxidative stress: Bridging the gap between type 2 diabetes mellitus and Alzheimer's disease. J Alzheimers Dis 2025:13872877241307404. [PMID: 39791373 DOI: 10.1177/13872877241307404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2025]
Abstract
BACKGROUND Type 2 diabetes mellitus (T2D) and Alzheimer's disease (AD) are two prevalent chronic diseases that pose significant global health challenges. Increasing evidence suggests a complex bidirectional relationship between these conditions, where T2D elevates the risk of AD, and AD exacerbates glucose metabolism abnormalities in T2D. OBJECTIVE This review explores the molecular mechanisms linking T2D and AD, focusing on the role of insulin signaling pathways and oxidative stress. METHODS A comprehensive literature search from PubMed, Web of Science, and other relevant databases was conducted and analyzed. RESULTS Insulin resistance in T2D leads to impaired insulin signaling in the brain, contributing to cognitive decline and the development of AD. Hyperglycemia-induced oxidative stress exacerbates neuronal damage, promoting the formation of amyloid-β plaques and neurofibrillary tangles characteristic of AD. Clinically antidiabetic drugs such as metformin show potential against AD in preclinical studies; Many natural products such as Dendrobium nobile Lindl. have anti-T2D efficacy and are also effective against AD in various in vivo and in vitro models. CONCLUSIONS Improving insulin resistance and reducing oxidative stress are important strategies in the treatment of T2D and AD. To understand the bridging role of insulin singling and oxidative stress in T2D and AD will provide insights and broader applications in alleviating T2D and AD.
Collapse
Affiliation(s)
- Fengqing Xu
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, China
- Department of Pharmacology, in School of Pharmacy, Zunyi Medical University, Zunyi, China
- Chinese Pharmacological Society-Guizhou Province Joint Laboratory for Pharmacology, Zunyi Medical University, Zunyi, China
| | - Jingshan Shi
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, China
- Department of Pharmacology, in School of Pharmacy, Zunyi Medical University, Zunyi, China
- Chinese Pharmacological Society-Guizhou Province Joint Laboratory for Pharmacology, Zunyi Medical University, Zunyi, China
| |
Collapse
|
2
|
Brossaud J, Barat P, Moisan MP. Cognitive Disorders in Type 1 Diabetes: Role of Brain Glucose Variation, Insulin Activity, and Glucocorticoid Exposure. Neuroendocrinology 2024:1-15. [PMID: 39401497 DOI: 10.1159/000541989] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Accepted: 10/09/2024] [Indexed: 11/19/2024]
Abstract
BACKGROUND The number of patients with type 2 diabetes (T2D) and type 1 diabetes (T1D) is on the rise, partly due to a global increase in new T1D cases among children. Beyond the well-documented microvascular and macrovascular complications, there is now substantial evidence indicating that diabetes also impacts the brain, leading to neuropsychological impairments. The risk of developing neuropsychiatric symptoms is notably higher in childhood due to the ongoing maturation of the brain, which makes it more susceptible to damage. Despite this awareness, the specific effects of diabetes on cognitive function remain poorly understood. SUMMARY This review synthesizes literature on the impact of diabetes on cognition and its relationship with brain structural changes. It presents data and hypotheses to explain how T1D contributes to cognitive dysfunction, with a particular focus on children and adolescents. The emphasis on the pediatric population is intentional, as young diabetic patients typically have fewer comorbidities, reducing confounding factors and simplifying the investigation of cognitive alterations. KEY MESSAGE We examine the roles of hypo- and hyperglycemia, as well as the emerging role of glucocorticoids in the development of neuropsychological disorders. When specific mechanisms related to T1D are available, they are highlighted; otherwise, data and hypotheses applicable to both T1D and T2D are discussed.
Collapse
Affiliation(s)
- Julie Brossaud
- Univ. Bordeaux, INRAE, Bordeaux INP, NutriNeurO, UMR 1286, Team NutriPsy, Bordeaux, France
- CHU Bordeaux, Nuclear Medicine, Pessac, France
| | - Pascal Barat
- Univ. Bordeaux, INRAE, Bordeaux INP, NutriNeurO, UMR 1286, Team NutriPsy, Bordeaux, France
- CHU Bordeaux, Pediatric Endocrinology and DiaBEA Unit, Hôpital des Enfants, Bordeaux, France
| | - Marie-Pierre Moisan
- Univ. Bordeaux, INRAE, Bordeaux INP, NutriNeurO, UMR 1286, Team NutriPsy, Bordeaux, France
| |
Collapse
|
3
|
Gómez-Guijarro MD, Cavero-Redondo I, Saz-Lara A, Pascual-Morena C, Álvarez-Bueno C, Martínez-García I. Intranasal insulin effect on cognitive and/or memory impairment: a systematic review and meta-analysis. Cogn Neurodyn 2024; 18:3059-3073. [PMID: 39555259 PMCID: PMC11564437 DOI: 10.1007/s11571-024-10138-5] [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: 08/30/2023] [Revised: 03/21/2024] [Accepted: 06/02/2024] [Indexed: 11/19/2024] Open
Abstract
Background: Cognitive impairment, characterized by deficits in cognitive functions and loss of delayed and immediate recall, disproportionately affects individuals aged 65 years and older, particularly those with comorbid cardiovascular conditions such as hypertension and diabetes mellitus. Objective: This study aimed to investigate the potential association between intranasal insulin and cognitive and/or memory impairment, with a specific focus on delayed and immediate recall, considering the rising prevalence of cognitive disorders in the aging population. Methodology: Employing a rigorous systematic approach, we conducted a thorough search of MEDLINE, Scopus, the Cochrane database, and Web of Science from inception to November 23, 2022, identifying relevant randomized clinical trials. Our analyses encompassed three key aspects: (i) assessing the impact of intranasal insulin on cognitive impairment, (ii) evaluating its effect on delayed recall, and (iii) examining its influence on immediate recall. Results: Five studies meeting the inclusion criteria were included. The results underscored a statistically significant effect of intranasal insulin on delayed memory (effect size: 1.37; 95% CI: 0.65 to 2.09) and overall cognition (effect size: 0.58; 95% CI: 0.08 to 1.08). However, no statistically significant effect was observed for immediate memory (effect size: 0.48; 95% CI: -0.00 to 0.96). Conclusions: This study provides compelling evidence supporting the significance and efficacy of intranasal insulin in enhancing delayed recall and overall cognition. The observed effects hold promise for potential therapeutic interventions in addressing cognitive deficits associated with aging and comorbid conditions. The findings emphasize the need for further research to elucidate the underlying mechanisms and optimize the application of intranasal insulin in cognitive enhancement strategies. Supplementary Information The online version contains supplementary material available at 10.1007/s11571-024-10138-5.
Collapse
Affiliation(s)
| | - Iván Cavero-Redondo
- Health and Social Research Center, Universidad de Castilla-La Mancha, Cuenca, Spain
- Facultad de Ciencias de la Salud, Universidad Autónoma de Chile, Talca, Chile
| | - Alicia Saz-Lara
- Health and Social Research Center, Universidad de Castilla-La Mancha, Cuenca, Spain
| | | | - Celia Álvarez-Bueno
- Health and Social Research Center, Universidad de Castilla-La Mancha, Cuenca, Spain
- Universidad Politécnica y Artística del Paraguay, Asunción, Paraguay
| | | |
Collapse
|
4
|
Rabiller G, Ip Z, Zarrabian S, Zhang H, Sato Y, Yazdan-Shahmorad A, Liu J. Type-2 Diabetes Alters Hippocampal Neural Oscillations and Disrupts Synchrony between the Hippocampus and Cortex. Aging Dis 2024; 15:2255-2270. [PMID: 38029397 PMCID: PMC11346393 DOI: 10.14336/ad.2023.1106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Accepted: 11/06/2023] [Indexed: 12/01/2023] Open
Abstract
Type 2 diabetes mellitus (T2DM) increases the risk of neurological diseases, yet how brain oscillations change as age and T2DM interact is not well characterized. To delineate the age and diabetic effect on neurophysiology, we recorded local field potentials with multichannel electrodes spanning the somatosensory cortex and hippocampus (HPC) under urethane anesthesia in diabetic and normoglycemic control mice, at 200 and 400 days of age. We analyzed the signal power of brain oscillations, brain state, sharp wave associate ripples (SPW-Rs), and functional connectivity between the cortex and HPC. We found that while both age and T2DM were correlated with a breakdown in long-range functional connectivity and reduced neurogenesis in the dentate gyrus and subventricular zone, T2DM further slowed brain oscillations and reduced theta-gamma coupling. Age and T2DM also prolonged the duration of SPW-Rs and increased gamma power during SPW-R phase. Our results have identified potential electrophysiological substrates of hippocampal changes associated with T2DM and age. The perturbed brain oscillation features and diminished neurogenesis may underlie T2DM-accelerated cognitive impairment.
Collapse
Affiliation(s)
- Gratianne Rabiller
- Department of Neurological Surgery, University of California at San Francisco, San Francisco, CA, USA
- San Francisco VA medical Center, San Francisco, CA, USA
| | - Zachary Ip
- Departments of Bioengineering, University of Washington, Seattle, WA, USA
| | - Shahram Zarrabian
- Department of Neurological Surgery, University of California at San Francisco, San Francisco, CA, USA
- San Francisco VA medical Center, San Francisco, CA, USA
| | - Hongxia Zhang
- Department of Neurological Surgery, University of California at San Francisco, San Francisco, CA, USA
- San Francisco VA medical Center, San Francisco, CA, USA
| | - Yoshimichi Sato
- Department of Neurological Surgery, University of California at San Francisco, San Francisco, CA, USA
- San Francisco VA medical Center, San Francisco, CA, USA
- Department of Neurosurgery, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Azadeh Yazdan-Shahmorad
- Departments of Bioengineering, University of Washington, Seattle, WA, USA
- Electrical and Computer Engineering, University of Washington, Seattle, WA, USA
| | - Jialing Liu
- Department of Neurological Surgery, University of California at San Francisco, San Francisco, CA, USA
- San Francisco VA medical Center, San Francisco, CA, USA
| |
Collapse
|
5
|
Tabassum A, Badulescu S, Singh E, Asoro R, McIntyre RS, Teopiz KM, Llach CD, Shah H, Mansur RB. Central effects of acute intranasal insulin on neuroimaging, cognitive, and behavioural outcomes: A systematic review. Neurosci Biobehav Rev 2024; 167:105907. [PMID: 39332547 DOI: 10.1016/j.neubiorev.2024.105907] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Revised: 08/10/2024] [Accepted: 09/24/2024] [Indexed: 09/29/2024]
Abstract
The distribution of insulin receptors throughout the brain implicates insulin in physiological functions and disease states, including cognition, appetite, mood, and metabolic disorders. Intranasally administered insulin offers a non-invasive approach for isolating and investigating brain insulin action. This systematic review synthesized the effects of acute intranasal insulin on neuroimaging, cognitive, and behavioural outcomes reported in 48 studies in adults. Age, sex, body mass index, and insulin resistance were found to moderate brain insulin action. Neuroimaging studies showed insulin affects brain activity, cerebral blood flow, and functional connectivity in regions like the hypothalamus, amygdala, and insula. Insulin also modified cognitive function, eating behaviour, and the stress response. Nonetheless, inconsistencies in study designs, dosages, and outcome measures necessitate standardized methodologies to better understand central insulin action. Taken together, insulin's ability to modify stress and fear, appetite and eating behaviour, and cognitive function in both healthy and diseased individuals highlight its potential in the therapeutic and mechanistic exploration of highly prevalent psychiatric, metabolic, and cognitive conditions like mood disorders, obesity, and Alzheimer's disease.
Collapse
Affiliation(s)
- Aniqa Tabassum
- Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada; Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, Ontario, Canada.
| | - Sebastian Badulescu
- Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada; Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, Ontario, Canada; Brain and Cognition Discovery Foundation, Toronto, Canada.
| | - Evanka Singh
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, Ontario, Canada.
| | - Renee Asoro
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, Ontario, Canada.
| | - Roger S McIntyre
- Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada; Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, Ontario, Canada; Department of Pharmacology & Toxicology, University of Toronto, Canada; Brain and Cognition Discovery Foundation, Toronto, Canada; Department of Psychiatry, University of Toronto, Canada.
| | - Kayla M Teopiz
- Brain and Cognition Discovery Foundation, Toronto, Canada.
| | - Cristian-Daniel Llach
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, Ontario, Canada.
| | - Hiya Shah
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, Ontario, Canada.
| | - Rodrigo B Mansur
- Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada; Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, Ontario, Canada.
| |
Collapse
|
6
|
Lanzillotta C, Tramutola A, Lanzillotta S, Greco V, Pagnotta S, Sanchini C, Di Angelantonio S, Forte E, Rinaldo S, Paone A, Cutruzzolà F, Cimini FA, Barchetta I, Cavallo MG, Urbani A, Butterfield DA, Di Domenico F, Paul BD, Perluigi M, Duarte JMN, Barone E. Biliverdin Reductase-A integrates insulin signaling with mitochondrial metabolism through phosphorylation of GSK3β. Redox Biol 2024; 73:103221. [PMID: 38843768 PMCID: PMC11190564 DOI: 10.1016/j.redox.2024.103221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Revised: 05/24/2024] [Accepted: 05/31/2024] [Indexed: 06/14/2024] Open
Abstract
Brain insulin resistance links the failure of energy metabolism with cognitive decline in both type 2 Diabetes Mellitus (T2D) and Alzheimer's disease (AD), although the molecular changes preceding overt brain insulin resistance remain unexplored. Abnormal biliverdin reductase-A (BVR-A) levels were observed in both T2D and AD and were associated with insulin resistance. Here, we demonstrate that reduced BVR-A levels alter insulin signaling and mitochondrial bioenergetics in the brain. Loss of BVR-A leads to IRS1 hyper-activation but dysregulates Akt-GSK3β complex in response to insulin, hindering the accumulation of pGSK3βS9 into the mitochondria. This event impairs oxidative phosphorylation and fosters the activation of the mitochondrial Unfolded Protein Response (UPRmt). Remarkably, we unveil that BVR-A is required to shuttle pGSK3βS9 into the mitochondria. Our data sheds light on the intricate interplay between insulin signaling and mitochondrial metabolism in the brain unraveling potential targets for mitigating the development of brain insulin resistance and neurodegeneration.
Collapse
Affiliation(s)
- Chiara Lanzillotta
- Department of Biochemical Sciences "A. Rossi-Fanelli", Sapienza University of Rome, Italy
| | - Antonella Tramutola
- Department of Biochemical Sciences "A. Rossi-Fanelli", Sapienza University of Rome, Italy
| | - Simona Lanzillotta
- Department of Biochemical Sciences "A. Rossi-Fanelli", Sapienza University of Rome, Italy
| | - Viviana Greco
- Department of Basic Biotechnology, Perioperative and Intensive Clinics, Faculty of Medicine and Surgery, Catholic University of the Sacred Heart, L.go F.Vito 1, 00168, Rome, Italy; Fondazione Policlinico Universitario A. Gemelli IRCCS, L.go A.Gemelli 8, 00168, Rome, Italy
| | - Sara Pagnotta
- Department of Biochemical Sciences "A. Rossi-Fanelli", Sapienza University of Rome, Italy
| | - Caterina Sanchini
- Center for Life Nano- & Neuro-Science, Istituto Italiano di Tecnologia, 00161, Rome, Italy
| | - Silvia Di Angelantonio
- Center for Life Nano- & Neuro-Science, Istituto Italiano di Tecnologia, 00161, Rome, Italy; Department of Physiology and Pharmacology, Sapienza University of Rome, Italy
| | - Elena Forte
- Department of Biochemical Sciences "A. Rossi-Fanelli", Sapienza University of Rome, Italy
| | - Serena Rinaldo
- Department of Biochemical Sciences "A. Rossi-Fanelli", Sapienza University of Rome, Italy
| | - Alessio Paone
- Department of Biochemical Sciences "A. Rossi-Fanelli", Sapienza University of Rome, Italy
| | - Francesca Cutruzzolà
- Department of Biochemical Sciences "A. Rossi-Fanelli", Sapienza University of Rome, Italy
| | | | - Ilaria Barchetta
- Department of Experimental Medicine, Sapienza University of Rome, Italy
| | | | - Andrea Urbani
- Department of Basic Biotechnology, Perioperative and Intensive Clinics, Faculty of Medicine and Surgery, Catholic University of the Sacred Heart, L.go F.Vito 1, 00168, Rome, Italy; Fondazione Policlinico Universitario A. Gemelli IRCCS, L.go A.Gemelli 8, 00168, Rome, Italy
| | - D Allan Butterfield
- Sanders-Brown Center on Aging, Department of Chemistry, University of Kentucky, Lexington, KY, USA
| | - Fabio Di Domenico
- Department of Biochemical Sciences "A. Rossi-Fanelli", Sapienza University of Rome, Italy
| | - Bindu D Paul
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Lieber Institute for Brain Development, Baltimore, MD, USA
| | - Marzia Perluigi
- Department of Biochemical Sciences "A. Rossi-Fanelli", Sapienza University of Rome, Italy
| | - Joao M N Duarte
- Department of Experimental Medical Science, Faculty of Medicine, Lund University, Sweden; Wallenberg Centre for Molecular Medicine, Lund University, Lund, Sweden
| | - Eugenio Barone
- Department of Biochemical Sciences "A. Rossi-Fanelli", Sapienza University of Rome, Italy.
| |
Collapse
|
7
|
Muleiro Alvarez M, Cano-Herrera G, Osorio Martínez MF, Vega Gonzales-Portillo J, Monroy GR, Murguiondo Pérez R, Torres-Ríos JA, van Tienhoven XA, Garibaldi Bernot EM, Esparza Salazar F, Ibarra A. A Comprehensive Approach to Parkinson's Disease: Addressing Its Molecular, Clinical, and Therapeutic Aspects. Int J Mol Sci 2024; 25:7183. [PMID: 39000288 PMCID: PMC11241043 DOI: 10.3390/ijms25137183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 06/24/2024] [Accepted: 06/27/2024] [Indexed: 07/16/2024] Open
Abstract
Parkinson's disease (PD) is a gradually worsening neurodegenerative disorder affecting the nervous system, marked by a slow progression and varied symptoms. It is the second most common neurodegenerative disease, affecting over six million people in the world. Its multifactorial etiology includes environmental, genomic, and epigenetic factors. Clinical symptoms consist of non-motor and motor symptoms, with motor symptoms being the classic presentation. Therapeutic approaches encompass pharmacological, non-pharmacological, and surgical interventions. Traditional pharmacological treatment consists of administering drugs (MAOIs, DA, and levodopa), while emerging evidence explores the potential of antidiabetic agents for neuroprotection and gene therapy for attenuating parkinsonian symptoms. Non-pharmacological treatments, such as exercise, a calcium-rich diet, and adequate vitamin D supplementation, aim to slow disease progression and prevent complications. For those patients who have medically induced side effects and/or refractory symptoms, surgery is a therapeutic option. Deep brain stimulation is the primary surgical option, associated with motor symptom improvement. Levodopa/carbidopa intestinal gel infusion through percutaneous endoscopic gastrojejunostomy and a portable infusion pump succeeded in reducing "off" time, where non-motor and motor symptoms occur, and increasing "on" time. This article aims to address the general aspects of PD and to provide a comparative comprehensive review of the conventional and the latest therapeutic advancements and emerging treatments for PD. Nevertheless, further studies are required to optimize treatment and provide suitable alternatives.
Collapse
Affiliation(s)
- Mauricio Muleiro Alvarez
- Centro de Investigación en Ciencias de la Salud (CICSA), Facultad de Ciencias de la Salud, Universidad Anáhuac Campus México Norte, Huixquilucan 52786, Mexico
| | - Gabriela Cano-Herrera
- Centro de Investigación en Ciencias de la Salud (CICSA), Facultad de Ciencias de la Salud, Universidad Anáhuac Campus México Norte, Huixquilucan 52786, Mexico
| | - María Fernanda Osorio Martínez
- Centro de Investigación en Ciencias de la Salud (CICSA), Facultad de Ciencias de la Salud, Universidad Anáhuac Campus México Norte, Huixquilucan 52786, Mexico
| | | | - Germán Rivera Monroy
- Centro de Investigación en Ciencias de la Salud (CICSA), Facultad de Ciencias de la Salud, Universidad Anáhuac Campus México Norte, Huixquilucan 52786, Mexico
| | - Renata Murguiondo Pérez
- Centro de Investigación en Ciencias de la Salud (CICSA), Facultad de Ciencias de la Salud, Universidad Anáhuac Campus México Norte, Huixquilucan 52786, Mexico
| | - Jorge Alejandro Torres-Ríos
- Centro de Investigación en Ciencias de la Salud (CICSA), Facultad de Ciencias de la Salud, Universidad Anáhuac Campus México Norte, Huixquilucan 52786, Mexico
| | - Ximena A. van Tienhoven
- Centro de Investigación en Ciencias de la Salud (CICSA), Facultad de Ciencias de la Salud, Universidad Anáhuac Campus México Norte, Huixquilucan 52786, Mexico
| | - Ernesto Marcelo Garibaldi Bernot
- Centro de Investigación en Ciencias de la Salud (CICSA), Facultad de Ciencias de la Salud, Universidad Anáhuac Campus México Norte, Huixquilucan 52786, Mexico
| | - Felipe Esparza Salazar
- Centro de Investigación en Ciencias de la Salud (CICSA), Facultad de Ciencias de la Salud, Universidad Anáhuac Campus México Norte, Huixquilucan 52786, Mexico
| | - Antonio Ibarra
- Centro de Investigación en Ciencias de la Salud (CICSA), Facultad de Ciencias de la Salud, Universidad Anáhuac Campus México Norte, Huixquilucan 52786, Mexico
- Secretaria de la Defensa Nacional, Escuela Militar de Graduados en Sanidad, Ciudad de México 11200, Mexico
| |
Collapse
|
8
|
Schatz S, Gutiérrez GR. Enhancing socio-communicative functions in an MCI patient with intra-nasal insulin: a case report. Front Psychiatry 2024; 15:1326702. [PMID: 39006824 PMCID: PMC11239438 DOI: 10.3389/fpsyt.2024.1326702] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2023] [Accepted: 05/28/2024] [Indexed: 07/16/2024] Open
Abstract
This report examines extended intra-nasal insulin treatment [INI] for an Insulin Resistant early Mild Cognitive Impairment [MCI] patient. Patient [EJ] also had medial temporal lobe [MTL] damage, poor short-term memory, significant irritability, and social and linguistic withdrawal at treatment start. Compared to baseline, nine months INI treatment increased grey matter volume, lowered beta-amyloid levels, and improved MCI and FAS scores. Patient also increased pragmatic capacities in social conversation and procedural memory. These findings align with results from prior clinical trials on INI and suggest that treatment can slow neurodegenerative disease progression in early MCI patients.
Collapse
Affiliation(s)
- Sara Schatz
- International Studies, The Ohio State University, Columbus, OH, United States
- Department of Evolution, Ecology, and Organismal Biology, The Ohio State University, Columbus, OH, United States
| | - Grace Rose Gutiérrez
- Department of Evolution, Ecology, and Organismal Biology, The Ohio State University, Columbus, OH, United States
| |
Collapse
|
9
|
Komici K, Pansini A, Bencivenga L, Rengo G, Pagano G, Guerra G. Frailty and Parkinson's disease: the role of diabetes mellitus. Front Med (Lausanne) 2024; 11:1377975. [PMID: 38882667 PMCID: PMC11177766 DOI: 10.3389/fmed.2024.1377975] [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: 01/28/2024] [Accepted: 05/02/2024] [Indexed: 06/18/2024] Open
Abstract
Parkinson's disease (PD) is a chronic neurodegenerative disease associated with a progressive loss of dopaminergic neurons, clinically characterized by motor and non-motor signs. Frailty is a clinical condition of increased vulnerability and negative health outcomes due to the loss of multiple physiological reserves. Chronic hyperglycemia and insulin resistance, which characterize diabetes mellitus (DM), have been reported to alter dopaminergic activity, increase the risk of PD, and influence the development of frailty. Even though diabetes may facilitate the development of frailty in patients with PD, this relationship is not established and a revision of the current knowledge is necessary. Furthermore, the synergy between DM, PD, and frailty may drive clinical complexity, worse outcomes, and under-representation of these populations in the research. In this review, we aimed to discuss the role of diabetes in the development of frailty among patients with PD. We summarized the clinical characteristics and outcomes of patients with concomitant DM, PD, and frailty. Finally, interventions to prevent frailty in this population are discussed.
Collapse
Affiliation(s)
- Klara Komici
- Department of Medicine and Health Sciences, University of Molise, Campobasso, Italy
| | | | - Leonardo Bencivenga
- Department of Translational Medical Sciences, University of Naples "Federico II", Naples, Italy
| | - Giuseppe Rengo
- Department of Translational Medical Sciences, University of Naples "Federico II", Naples, Italy
- Istituti Clinici Scientifici Maugeri IRCCS-Scientific Institute of Telese Terme, Telese Terme, BN, Italy
| | - Gennaro Pagano
- Roche Pharma Research and Early Development (pRED), Neuroscience and Rare Diseases Discovery and Translational Area, Roche Innovation Center, Basel, Switzerland
- University of Exeter Medical School, London, United Kingdom
| | - Germano Guerra
- Department of Medicine and Health Sciences, University of Molise, Campobasso, Italy
| |
Collapse
|
10
|
Rae CD, Baur JA, Borges K, Dienel G, Díaz-García CM, Douglass SR, Drew K, Duarte JMN, Duran J, Kann O, Kristian T, Lee-Liu D, Lindquist BE, McNay EC, Robinson MB, Rothman DL, Rowlands BD, Ryan TA, Scafidi J, Scafidi S, Shuttleworth CW, Swanson RA, Uruk G, Vardjan N, Zorec R, McKenna MC. Brain energy metabolism: A roadmap for future research. J Neurochem 2024; 168:910-954. [PMID: 38183680 PMCID: PMC11102343 DOI: 10.1111/jnc.16032] [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: 05/27/2023] [Revised: 11/29/2023] [Accepted: 12/05/2023] [Indexed: 01/08/2024]
Abstract
Although we have learned much about how the brain fuels its functions over the last decades, there remains much still to discover in an organ that is so complex. This article lays out major gaps in our knowledge of interrelationships between brain metabolism and brain function, including biochemical, cellular, and subcellular aspects of functional metabolism and its imaging in adult brain, as well as during development, aging, and disease. The focus is on unknowns in metabolism of major brain substrates and associated transporters, the roles of insulin and of lipid droplets, the emerging role of metabolism in microglia, mysteries about the major brain cofactor and signaling molecule NAD+, as well as unsolved problems underlying brain metabolism in pathologies such as traumatic brain injury, epilepsy, and metabolic downregulation during hibernation. It describes our current level of understanding of these facets of brain energy metabolism as well as a roadmap for future research.
Collapse
Affiliation(s)
- Caroline D. Rae
- School of Psychology, The University of New South Wales, NSW 2052 & Neuroscience Research Australia, Randwick, New South Wales, Australia
| | - Joseph A. Baur
- Department of Physiology and Institute for Diabetes, Obesity and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Karin Borges
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, St Lucia, QLD, Australia
| | - Gerald Dienel
- Department of Neurology, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
- Department of Cell Biology and Physiology, University of New Mexico School of Medicine, Albuquerque, New Mexico, USA
| | - Carlos Manlio Díaz-García
- Department of Biochemistry and Molecular Biology, Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| | | | - Kelly Drew
- Center for Transformative Research in Metabolism, Institute of Arctic Biology, University of Alaska Fairbanks, Fairbanks, Alaska, USA
| | - João M. N. Duarte
- Department of Experimental Medical Science, Faculty of Medicine, Lund University, Lund, & Wallenberg Centre for Molecular Medicine, Lund University, Lund, Sweden
| | - Jordi Duran
- Institut Químic de Sarrià (IQS), Universitat Ramon Llull (URL), Barcelona, Spain
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Oliver Kann
- Institute of Physiology and Pathophysiology, University of Heidelberg, D-69120; Interdisciplinary Center for Neurosciences (IZN), University of Heidelberg, Heidelberg, Germany
| | - Tibor Kristian
- Veterans Affairs Maryland Health Center System, Baltimore, Maryland, USA
- Department of Anesthesiology and the Center for Shock, Trauma, and Anesthesiology Research (S.T.A.R.), University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Dasfne Lee-Liu
- Facultad de Medicina y Ciencia, Universidad San Sebastián, Santiago, Región Metropolitana, Chile
| | - Britta E. Lindquist
- Department of Neurology, Division of Neurocritical Care, Gladstone Institute of Neurological Disease, University of California at San Francisco, San Francisco, California, USA
| | - Ewan C. McNay
- Behavioral Neuroscience, University at Albany, Albany, New York, USA
| | - Michael B. Robinson
- Departments of Pediatrics and System Pharmacology & Translational Therapeutics, Children’s Hospital of Philadelphia, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Douglas L. Rothman
- Magnetic Resonance Research Center and Departments of Radiology and Biomedical Engineering, Yale University, New Haven, Connecticut, USA
| | - Benjamin D. Rowlands
- School of Chemistry, Faculty of Science, The University of Sydney, Sydney, New South Wales, Australia
| | - Timothy A. Ryan
- Department of Biochemistry, Weill Cornell Medicine, New York, New York, USA
| | - Joseph Scafidi
- Department of Neurology, Kennedy Krieger Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Susanna Scafidi
- Anesthesiology & Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - C. William Shuttleworth
- Department of Neurosciences, University of New Mexico School of Medicine Albuquerque, Albuquerque, New Mexico, USA
| | - Raymond A. Swanson
- Department of Neurology, University of California, San Francisco, and San Francisco Veterans Affairs Medical Center, San Francisco, California, USA
| | - Gökhan Uruk
- Department of Neurology, University of California, San Francisco, and San Francisco Veterans Affairs Medical Center, San Francisco, California, USA
| | - Nina Vardjan
- Laboratory of Cell Engineering, Celica Biomedical, Ljubljana, Slovenia
- Laboratory of Neuroendocrinology—Molecular Cell Physiology, Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Robert Zorec
- Laboratory of Cell Engineering, Celica Biomedical, Ljubljana, Slovenia
- Laboratory of Neuroendocrinology—Molecular Cell Physiology, Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Mary C. McKenna
- Department of Pediatrics and Program in Neuroscience, University of Maryland School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
11
|
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
|
12
|
Kim OY, Song J. Important roles of linoleic acid and α-linolenic acid in regulating cognitive impairment and neuropsychiatric issues in metabolic-related dementia. Life Sci 2024; 337:122356. [PMID: 38123015 DOI: 10.1016/j.lfs.2023.122356] [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: 09/23/2023] [Revised: 12/02/2023] [Accepted: 12/13/2023] [Indexed: 12/23/2023]
Abstract
Metabolic syndrome (MetS), which is characterized by insulin resistance, high blood glucose, obesity, and dyslipidemia, is known to increase the risk of dementia accompanied by memory loss and depression. The direct pathways and specific mechanisms in the central nervous system (CNS) for addressing fatty acid imbalances in MetS have not yet been fully elucidated. Among polyunsaturated acids, linoleic acid (LA, n6-PUFA) and α-linolenic acid (ALA, n3-PUFA), which are two essential fatty acids that should be provided by food sources (e.g., vegetable oils and seeds), have been reported to regulate various cellular mechanisms including apoptosis, inflammatory responses, mitochondrial biogenesis, and insulin signaling. Furthermore, inadequate intake of LA and ALA is reported to be involved in neuropathology and neuropsychiatric diseases as well as imbalanced metabolic conditions. Herein, we review the roles of LA and ALA on metabolic-related dementia focusing on insulin resistance, dyslipidemia, synaptic plasticity, cognitive function, and neuropsychiatric issues. This review suggests that LA and ALA are important fatty acids for concurrent treatment of both MetS and neurological problems.
Collapse
Affiliation(s)
- Oh Yoen Kim
- Department of Food Science and Nutrition, Dong A University, Busan, Republic of Korea; Department of Health Sciences, Graduate School of Dong-A University, Busan, Republic of Korea.
| | - Juhyun Song
- Department of Anatomy, Chonnam National University Medical School, Seoul, Republic of Korea.
| |
Collapse
|
13
|
Mei M, Liu M, Mei Y, Zhao J, Li Y. Sphingolipid metabolism in brain insulin resistance and neurological diseases. Front Endocrinol (Lausanne) 2023; 14:1243132. [PMID: 37867511 PMCID: PMC10587683 DOI: 10.3389/fendo.2023.1243132] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Accepted: 09/22/2023] [Indexed: 10/24/2023] Open
Abstract
Sphingolipids, as members of the large lipid family, are important components of plasma membrane. Sphingolipids participate in biological signal transduction to regulate various important physiological processes such as cell growth, apoptosis, senescence, and differentiation. Numerous studies have demonstrated that sphingolipids are strongly associated with glucose metabolism and insulin resistance. Insulin resistance, including peripheral insulin resistance and brain insulin resistance, is closely related to the occurrence and development of many metabolic diseases. In addition to metabolic diseases, like type 2 diabetes, brain insulin resistance is also involved in the progression of neurodegenerative diseases including Alzheimer's disease and Parkinson's disease. However, the specific mechanism of sphingolipids in brain insulin resistance has not been systematically summarized. This article reviews the involvement of sphingolipids in brain insulin resistance, highlighting the role and molecular biological mechanism of sphingolipid metabolism in cognitive dysfunctions and neuropathological abnormalities of the brain.
Collapse
Affiliation(s)
- Meng Mei
- Department of Pharmacy, Wuhan Children’s Hospital (Wuhan Maternal and Child Healthcare Hospital), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Maochang Liu
- Department of Pharmacy, Wuhan Children’s Hospital (Wuhan Maternal and Child Healthcare Hospital), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yan Mei
- Department of Pharmacy, Wuhan Children’s Hospital (Wuhan Maternal and Child Healthcare Hospital), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jing Zhao
- Administrative Office, Beijing University of Chinese Medicine, Beijing, China
| | - Yang Li
- Department of Pharmacy, Wuhan Children’s Hospital (Wuhan Maternal and Child Healthcare Hospital), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
14
|
Monney M, Jornayvaz FR, Gariani K. GLP-1 receptor agonists effect on cognitive function in patients with and without type 2 diabetes. DIABETES & METABOLISM 2023; 49:101470. [PMID: 37657738 DOI: 10.1016/j.diabet.2023.101470] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Accepted: 07/24/2023] [Indexed: 09/03/2023]
Abstract
Glucagon-like peptide 1 (GLP-1) is a hormone of the incretin family, secreted in response to nutrient ingestion, and plays a role in metabolic homeostasis. GLP-1 receptor agonist has a peripheral and a central action, including stimulation of glucose-dependent insulin secretion and insulin biosynthesis, inhibition of glucagon secretion and gastric emptying, and inhibition of food intake. Through their mechanism, their use in the treatment of type 2 diabetes has been extended to the management of obesity, and numerous trials are being conducted to assess their cardiovascular effect. Type 2 diabetes appears to share common pathophysiological mechanisms with the development of cognitive disorders, such as Alzheimer's and Parkinson's disease, related to insulin resistance. In this review, we aim to examine the pathological features between type 2 diabetes and dementia, GLP-1 central effects, and analyze the relevant literature about the effect of GLP-1 analogs on cognitive function of patients with type 2 diabetes but also without. Results tends to show an improvement in some brain markers (e.g. hippocampal connections, cerebral glucose metabolism, hippocampal activation on functional magnetic resonance imaging), but without being able to demonstrate a strong correlation to cognitive scores. Some epidemiological studies suggest that GLP-1 receptor agonists may offer a protective effect, by delaying progression to dementia when diabetic patients are treated with GLP-1 receptor agonists. Ongoing trials are in progress and may provide disease-modifying care for Alzheimer's disease and Parkinson's disease patients in the future.
Collapse
Affiliation(s)
- Marine Monney
- Division of General Internal Medicine, Department of Medicine, Geneva University Hospitals, Geneva 1211, Switzerland.
| | - François R Jornayvaz
- Division of Endocrinology, Diabetes, Nutrition and Therapeutic Patient Education, Department of Medical Specialties, Geneva University Hospitals, Geneva 1211, Switzerland; Diabetes Center, Faculty of Medicine, University of Geneva, Geneva 1211, Switzerland; Department of Cell Physiology and Metabolism, Centre Medical Universitaire (CMU), Faculty of Medicine, University of Geneva, Geneva 1211, Switzerland
| | - Karim Gariani
- Division of Endocrinology, Diabetes, Nutrition and Therapeutic Patient Education, Department of Medical Specialties, Geneva University Hospitals, Geneva 1211, Switzerland; Diabetes Center, Faculty of Medicine, University of Geneva, Geneva 1211, Switzerland
| |
Collapse
|
15
|
Chung TWH, Zhang H, Wong FKC, Sridhar S, Lee TMC, Leung GKK, Chan KH, Lau KK, Tam AR, Ho DTY, Cheng VCC, Yuen KY, Hung IFN, Mak HKF. A Pilot Study of Short-Course Oral Vitamin A and Aerosolised Diffuser Olfactory Training for the Treatment of Smell Loss in Long COVID. Brain Sci 2023; 13:1014. [PMID: 37508945 PMCID: PMC10377650 DOI: 10.3390/brainsci13071014] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 06/20/2023] [Accepted: 06/27/2023] [Indexed: 07/30/2023] Open
Abstract
Background: Olfactory dysfunction (OD) is a common neurosensory manifestation in long COVID. An effective and safe treatment against COVID-19-related OD is needed. Methods: This pilot trial recruited long COVID patients with persistent OD. Participants were randomly assigned to receive short-course (14 days) oral vitamin A (VitA; 25,000 IU per day) and aerosolised diffuser olfactory training (OT) thrice daily (combination), OT alone (standard care), or observation (control) for 4 weeks. The primary outcome was differences in olfactory function by butanol threshold tests (BTT) between baseline and end-of-treatment. Secondary outcomes included smell identification tests (SIT), structural MRI brain, and serial seed-based functional connectivity (FC) analyses in the olfactory cortical network by resting-state functional MRI (rs-fMRI). Results: A total of 24 participants were randomly assigned to receive either combination treatment (n = 10), standard care (n = 9), or control (n = 5). Median OD duration was 157 days (IQR 127-175). Mean baseline BTT score was 2.3 (SD 1.1). At end-of-treatment, mean BTT scores were significantly higher for the combination group than control (p < 0.001, MD = 4.4, 95% CI 1.7 to 7.2) and standard care (p = 0.009) groups. Interval SIT scores increased significantly (p = 0.009) in the combination group. rs-fMRI showed significantly higher FC in the combination group when compared to other groups. At end-of-treatment, positive correlations were found in the increased FC at left inferior frontal gyrus and clinically significant improvements in measured BTT (r = 0.858, p < 0.001) and SIT (r = 0.548, p = 0.042) scores for the combination group. Conclusions: Short-course oral VitA and aerosolised diffuser OT was effective as a combination treatment for persistent OD in long COVID.
Collapse
Affiliation(s)
- Tom Wai-Hin Chung
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Hui Zhang
- Department of Rehabilitation Sciences, The Hong Kong Polytechnic University, Hong Kong, China
- Research Institute for Intelligent Wearable Systems, The Hong Kong Polytechnic University, Hong Kong, China
| | - Fergus Kai-Chuen Wong
- Department of Ear, Nose and Throat, Pamela Youde Nethersole Eastern Hospital, Hong Kong, China
| | - Siddharth Sridhar
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
- State Key Laboratory of Emerging Infectious Diseases, The University of Hong Kong, Hong Kong, China
- Carol Yu Centre for Infection, The University of Hong Kong, Hong Kong, China
| | - Tatia Mei-Chun Lee
- Department of Psychology, The University of Hong Kong, Hong Kong, China
- State Key Laboratory of Brain and Cognitive Sciences, The University of Hong Kong, Hong Kong, China
| | - Gilberto Ka-Kit Leung
- Department of Surgery, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Koon-Ho Chan
- Department of Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Kui-Kai Lau
- State Key Laboratory of Brain and Cognitive Sciences, The University of Hong Kong, Hong Kong, China
- Department of Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Anthony Raymond Tam
- Department of Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Deborah Tip-Yin Ho
- Department of Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Vincent Chi-Chung Cheng
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Kwok-Yung Yuen
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
- State Key Laboratory of Emerging Infectious Diseases, The University of Hong Kong, Hong Kong, China
- Carol Yu Centre for Infection, The University of Hong Kong, Hong Kong, China
- The Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The University of Hong Kong, Hong Kong, China
| | - Ivan Fan-Ngai Hung
- Carol Yu Centre for Infection, The University of Hong Kong, Hong Kong, China
- Department of Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
- The Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The University of Hong Kong, Hong Kong, China
| | - Henry Ka-Fung Mak
- State Key Laboratory of Brain and Cognitive Sciences, The University of Hong Kong, Hong Kong, China
- Department of Diagnostic Radiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
- Alzheimer's Disease Research Network, The University of Hong Kong, Hong Kong, China
| |
Collapse
|
16
|
Shrewsbury SB. The Upper Nasal Space: Option for Systemic Drug Delivery, Mucosal Vaccines and "Nose-to-Brain". Pharmaceutics 2023; 15:1720. [PMID: 37376168 PMCID: PMC10303426 DOI: 10.3390/pharmaceutics15061720] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 06/06/2023] [Accepted: 06/10/2023] [Indexed: 06/29/2023] Open
Abstract
Sino-nasal disease is appropriately treated with topical treatment, where the nasal mucosa acts as a barrier to systemic absorption. Non-invasive nasal delivery of drugs has produced some small molecule products with good bioavailability. With the recent COVID pandemic and the need for nasal mucosal immunity becoming more appreciated, more interest has become focused on the nasal cavity for vaccine delivery. In parallel, it has been recognized that drug delivery to different parts of the nose can have different results and for "nose-to-brain" delivery, deposition on the olfactory epithelium of the upper nasal space is desirable. Here the non-motile cilia and reduced mucociliary clearance lead to longer residence time that permits enhanced absorption, either into the systemic circulation or directly into the CNS. Many of the developments in nasal delivery have been to add bioadhesives and absorption/permeation enhancers, creating more complicated formulations and development pathways, but other projects have shown that the delivery device itself may allow more differential targeting of the upper nasal space without these additions and that could allow faster and more efficient programs to bring a wider range of drugs-and vaccines-to market.
Collapse
|
17
|
Rabiller G, Ip Z, Zarrabian S, Zhang H, Sato Y, Yazdan-Shahmorad A, Liu J. Type-2 diabetes alters hippocampal neural oscillations and disrupts synchrony between hippocampus and cortex. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.25.542288. [PMID: 37292743 PMCID: PMC10245872 DOI: 10.1101/2023.05.25.542288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Type 2 diabetes mellitus (T2DM) increases the risk of neurological diseases, yet how brain oscillations change as age and T2DM interact is not well characterized. To delineate the age and diabetic effect on neurophysiology, we recorded local field potentials with multichannel electrodes spanning the somatosensory cortex and hippocampus (HPC) under urethane anesthesia in diabetic and normoglycemic control mice, at 200 and 400 days of age. We analyzed the signal power of brain oscillations, brain state, sharp wave associate ripples (SPW-Rs), and functional connectivity between the cortex and HPC. We found that while both age and T2DM were correlated with a breakdown in long-range functional connectivity and reduced neurogenesis in the dentate gyrus and subventricular zone, T2DM further slowed brain oscillations and reduced theta-gamma coupling. Age and T2DM also prolonged the duration of SPW-Rs and increased gamma power during SPW-R phase. Our results have identified potential electrophysiological substrates of hippocampal changes associated with T2DM and age. The perturbed brain oscillation features and diminished neurogenesis may underlie T2DM-accelerated cognitive impairment.
Collapse
|
18
|
Shpakov AO, Zorina II, Derkach KV. Hot Spots for the Use of Intranasal Insulin: Cerebral Ischemia, Brain Injury, Diabetes Mellitus, Endocrine Disorders and Postoperative Delirium. Int J Mol Sci 2023; 24:3278. [PMID: 36834685 PMCID: PMC9962062 DOI: 10.3390/ijms24043278] [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] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 01/27/2023] [Accepted: 01/31/2023] [Indexed: 02/11/2023] Open
Abstract
A decrease in the activity of the insulin signaling system of the brain, due to both central insulin resistance and insulin deficiency, leads to neurodegeneration and impaired regulation of appetite, metabolism, endocrine functions. This is due to the neuroprotective properties of brain insulin and its leading role in maintaining glucose homeostasis in the brain, as well as in the regulation of the brain signaling network responsible for the functioning of the nervous, endocrine, and other systems. One of the approaches to restore the activity of the insulin system of the brain is the use of intranasally administered insulin (INI). Currently, INI is being considered as a promising drug to treat Alzheimer's disease and mild cognitive impairment. The clinical application of INI is being developed for the treatment of other neurodegenerative diseases and improve cognitive abilities in stress, overwork, and depression. At the same time, much attention has recently been paid to the prospects of using INI for the treatment of cerebral ischemia, traumatic brain injuries, and postoperative delirium (after anesthesia), as well as diabetes mellitus and its complications, including dysfunctions in the gonadal and thyroid axes. This review is devoted to the prospects and current trends in the use of INI for the treatment of these diseases, which, although differing in etiology and pathogenesis, are characterized by impaired insulin signaling in the brain.
Collapse
Affiliation(s)
- Alexander O. Shpakov
- Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences, 194223 St. Petersburg, Russia
| | | | | |
Collapse
|
19
|
Cullinane PW, de Pablo Fernandez E, König A, Outeiro TF, Jaunmuktane Z, Warner TT. Type 2 Diabetes and Parkinson's Disease: A Focused Review of Current Concepts. Mov Disord 2023; 38:162-177. [PMID: 36567671 DOI: 10.1002/mds.29298] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 10/25/2022] [Accepted: 11/15/2022] [Indexed: 12/27/2022] Open
Abstract
Highly reproducible epidemiological evidence shows that type 2 diabetes (T2D) increases the risk and rate of progression of Parkinson's disease (PD), and crucially, the repurposing of certain antidiabetic medications for the treatment of PD has shown early promise in clinical trials, suggesting that the effects of T2D on PD pathogenesis may be modifiable. The high prevalence of T2D means that a significant proportion of patients with PD may benefit from personalized antidiabetic treatment approaches that also confer neuroprotective benefits. Therefore, there is an immediate need to better understand the mechanistic relation between these conditions and the specific molecular pathways affected by T2D in the brain. Although there is considerable evidence that processes such as insulin signaling, mitochondrial function, autophagy, and inflammation are involved in the pathogenesis of both PD and T2D, the primary aim of this review is to highlight the evidence showing that T2D-associated dysregulation of these pathways occurs not only in the periphery but also in the brain and how this may facilitate neurodegeneration in PD. We also discuss the challenges involved in disentangling the complex relationship between T2D, insulin resistance, and PD, as well as important questions for further research. © 2022 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Patrick W Cullinane
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London, United Kingdom.,Reta Lila Weston Institute of Neurological Studies and Queen Square Brain Bank for Neurological Disorders, UCL Queen Square Institute of Neurology, University College London, London, United Kingdom
| | - Eduardo de Pablo Fernandez
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London, United Kingdom.,Reta Lila Weston Institute of Neurological Studies and Queen Square Brain Bank for Neurological Disorders, UCL Queen Square Institute of Neurology, University College London, London, United Kingdom
| | - Annekatrin König
- Department of Experimental Neurodegeneration, Center for Biostructural Imaging of Neurodegeneration, University Medical Center Göttingen, Göttingen, Germany
| | - Tiago Fleming Outeiro
- Department of Experimental Neurodegeneration, Center for Biostructural Imaging of Neurodegeneration, University Medical Center Göttingen, Göttingen, Germany.,Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany.,Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle Upon Tyne, United Kingdom.,Scientific Employee with an Honorary Contract at Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Göttingen, Germany
| | - Zane Jaunmuktane
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London, United Kingdom.,Reta Lila Weston Institute of Neurological Studies and Queen Square Brain Bank for Neurological Disorders, UCL Queen Square Institute of Neurology, University College London, London, United Kingdom.,Division of Neuropathology, National Hospital for Neurology and Neurosurgery, University College London NHS Foundation Trust, London, United Kingdom.,Queen Square Movement Disorders Centre, UCL Queen Square Institute of Neurology, London, United Kingdom
| | - Thomas T Warner
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London, United Kingdom.,Reta Lila Weston Institute of Neurological Studies and Queen Square Brain Bank for Neurological Disorders, UCL Queen Square Institute of Neurology, University College London, London, United Kingdom.,Queen Square Movement Disorders Centre, UCL Queen Square Institute of Neurology, London, United Kingdom
| |
Collapse
|
20
|
Zhang G, Liu T, Wei W, Zhang R, Wang H, Wang M. Evaluation of altered brain activity in type 2 diabetes using various indices of brain function: A resting-state functional magnetic resonance imaging study. Front Hum Neurosci 2023; 16:1032264. [PMID: 36699964 PMCID: PMC9870028 DOI: 10.3389/fnhum.2022.1032264] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 12/05/2022] [Indexed: 01/11/2023] Open
Abstract
Background Type 2 diabetes mellitus (T2DM) has been identified as a risk factor that increases the rate of cognitive decline. Previous studies showed that patients with T2DM had brain function alterations based on a single index of resting-state functional magnetic resonance imaging (rs-fMRI). The present study aimed to explore spontaneous brain activity in patients with T2DM by comparing various rs-fMRI indices, and to determine the relationship between these changes and cognitive dysfunction. Methods A total of 52 patients with T2DM and age- and sex-matched control participants were included in this study. The amplitude of low-frequency fluctuation (ALFF), regional homogeneity (ReHo), and voxel-mirrored homotopic connectivity (VMHC) values were calculated to represent the status of spontaneous neural activity. The Montreal Cognitive Assessment (MoCA) was used for the rapid evaluation of cognition in all subjects. Pearson correlation and mediation analyses were conducted to investigate the relationship between rs-fMRI indices and clinical parameters such as fasting glucose, disease duration, and MoCA. Results Patients with T2DM had alterations of concordant spontaneous brain activity in brain areas including the bilateral cerebellum posterior lobe, the left inferior temporal gyrus (ITG.L), the parahippocampal gyrus, and the left supplementary motor area (SMA.L). The indices were significantly correlated to each other in most of the detected brain areas. Positive correlations were observed between fasting glucose and neural activity in the surrounding areas of the left insula and the inferior frontal gyrus. MoCA scores were negatively correlated with the ReHo values extracted from the left anterior occipital lobe and the superior cerebellar cortex and were positively correlated with VMHC values extracted from the left caudate and the precentral gyrus (PreCG). No significant mediation effect of abnormal brain activity was found in the relationship between clinical parameters and MoCA scores. Conclusion The current study demonstrated the functional concordance of abnormal brain activities in patients with T2DM by comparing ALFF, ReHo, and VMHC measurements. Widespread abnormalities mainly involved in motor and sensory processing functions may provide insight into examining T2DM-related neurological pathophysiology.
Collapse
Affiliation(s)
- Ge Zhang
- Department of Radiology, Henan Provincial People's Hospital, Zhengzhou, China,Department of Radiology, Bethune International Peace Hospital, Shijiazhuang, China
| | - Taiyuan Liu
- Department of Radiology, Henan Provincial People's Hospital, Zhengzhou, China
| | - Wei Wei
- Department of Radiology, Henan Provincial People's Hospital, Zhengzhou, China
| | - Rui Zhang
- Department of Radiology, Henan Provincial People's Hospital, Zhengzhou, China
| | - Huilin Wang
- Department of Radiology, Bethune International Peace Hospital, Shijiazhuang, China,*Correspondence: Huilin Wang ✉
| | - Meiyun Wang
- Department of Radiology, Henan Provincial People's Hospital, Zhengzhou, China,Laboratory of Brian Science and Brain-Like Intelligence Technology, Institute for Integrated Medical Science and Engineering, Henan Academy of Sciences, Zhengzhou, China,Meiyun Wang ✉
| |
Collapse
|
21
|
Aranyi SC, Képes Z, Nagy M, Opposits G, Garai I, Káplár M, Emri M. Topological dissimilarities of hierarchical resting networks in type 2 diabetes mellitus and obesity. J Comput Neurosci 2023; 51:71-86. [PMID: 36056275 PMCID: PMC9840595 DOI: 10.1007/s10827-022-00833-9] [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: 07/30/2021] [Revised: 07/29/2022] [Accepted: 08/23/2022] [Indexed: 01/18/2023]
Abstract
Type 2 diabetes mellitus (T2DM) is reported to cause widespread changes in brain function, leading to cognitive impairments. Research using resting-state functional magnetic resonance imaging data already aims to understand functional changes in complex brain connectivity systems. However, no previous studies with dynamic causal modelling (DCM) tried to investigate large-scale effective connectivity in diabetes. We aimed to examine the differences in large-scale resting state networks in diabetic and obese patients using combined DCM and graph theory methodologies. With the participation of 70 subjects (43 diabetics, 27 obese), we used cross-spectra DCM to estimate connectivity between 36 regions, subdivided into seven resting networks (RSN) commonly recognized in the literature. We assessed group-wise connectivity of T2DM and obesity, as well as group differences, with parametric empirical Bayes and Bayesian model reduction techniques. We analyzed network connectivity globally, between RSNs, and regionally. We found that average connection strength was higher in T2DM globally and between RSNs, as well. On the network level, the salience network shows stronger total within-network connectivity in diabetes (8.07) than in the obese group (4.02). Regionally, we measured the most significant average decrease in the right middle temporal gyrus (-0.013 Hz) and the right inferior parietal lobule (-0.01 Hz) relative to the obese group. In comparison, connectivity increased most notably in the left anterior prefrontal cortex (0.01 Hz) and the medial dorsal thalamus (0.009 Hz). In conclusion, we find the usage of complex analysis of large-scale networks suitable for diabetes instead of focusing on specific changes in brain function.
Collapse
Affiliation(s)
- Sándor Csaba Aranyi
- Division of Nuclear Medicine and Translational Imaging, Department of Medical Imaging, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Zita Képes
- Division of Nuclear Medicine and Translational Imaging, Department of Medical Imaging, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Marianna Nagy
- Division of Radiology and Imaging Science, Department of Medical Imaging, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Gábor Opposits
- Division of Nuclear Medicine and Translational Imaging, Department of Medical Imaging, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Ildikó Garai
- Division of Nuclear Medicine and Translational Imaging, Department of Medical Imaging, Faculty of Medicine, University of Debrecen, Debrecen, Hungary ,Translational Research Centre, ScanoMed Ltd., Debrecen, Hungary
| | - Miklós Káplár
- Department of Internal Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Miklós Emri
- Division of Nuclear Medicine and Translational Imaging, Department of Medical Imaging, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| |
Collapse
|
22
|
Cui Y, Tang TY, Lu CQ, Ju S. Insulin Resistance and Cognitive Impairment: Evidence From Neuroimaging. J Magn Reson Imaging 2022; 56:1621-1649. [PMID: 35852470 DOI: 10.1002/jmri.28358] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 07/02/2022] [Accepted: 07/05/2022] [Indexed: 01/04/2023] Open
Abstract
Insulin is a peptide well known for its role in regulating glucose metabolism in peripheral tissues. Emerging evidence from human and animal studies indicate the multifactorial role of insulin in the brain, such as neuronal and glial metabolism, glucose regulation, and cognitive processes. Insulin resistance (IR), defined as reduced sensitivity to the action of insulin, has been consistently proposed as an important risk factor for developing neurodegeneration and cognitive impairment. Although the exact mechanism of IR-related cognitive impairment still awaits further elucidation, neuroimaging offers a versatile set of novel contrasts to reveal the subtle cerebral abnormalities in IR. These imaging contrasts, including but not limited to brain volume, white matter (WM) microstructure, neural function and brain metabolism, are expected to unravel the nature of the link between IR, cognitive decline, and brain abnormalities, and their changes over time. This review summarizes the current neuroimaging studies with multiparametric techniques, focusing on the cerebral abnormalities related to IR and therapeutic effects of IR-targeting treatments. According to the results, brain regions associated with IR pathophysiology include the medial temporal lobe, hippocampus, prefrontal lobe, cingulate cortex, precuneus, occipital lobe, and the WM tracts across the globe. Of these, alterations in the temporal lobe are highly reproducible across different imaging modalities. These structures have been known to be vulnerable to Alzheimer's disease (AD) pathology and are critical in cognitive processes such as memory and executive functioning. Comparing to asymptomatic subjects, results are more mixed in patients with metabolic disorders such as type 2 diabetes and obesity, which might be attributed to a multifactorial mechanism. Taken together, neuroimaging, especially MRI, is beneficial to reveal early abnormalities in cerebral structure and function in insulin-resistant brain, providing important evidence to unravel the underlying neuronal substrate that reflects the cognitive decline in IR. EVIDENCE LEVEL: 5 TECHNICAL EFFICACY: Stage 2.
Collapse
Affiliation(s)
- Ying Cui
- Department of Radiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Tian-Yu Tang
- Jiangsu Key Laboratory of Molecular and Functional Imaging, Department of Radiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Chun-Qiang Lu
- Department of Radiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Shenghong Ju
- Department of Radiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| |
Collapse
|
23
|
Malin SK, Stewart NR, Ude AA, Alderman BL. Brain insulin resistance and cognitive function: influence of exercise. J Appl Physiol (1985) 2022; 133:1368-1380. [PMID: 36269295 PMCID: PMC9744647 DOI: 10.1152/japplphysiol.00375.2022] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 10/12/2022] [Accepted: 10/12/2022] [Indexed: 12/15/2022] Open
Abstract
Exercise has systemic health benefits in people, in part, through improving whole body insulin sensitivity. The brain is an insulin-sensitive organ that is often underdiscussed relative to skeletal muscle, liver, and adipose tissue. Although brain insulin action may have only subtle impacts on peripheral regulation of systemic glucose homeostasis, it is important for weight regulation as well as mental health. In fact, brain insulin signaling is also involved in processes that support healthy cognition. Furthermore, brain insulin resistance has been associated with age-related declines in memory and executive function as well as Alzheimer's disease pathology. Herein, we provide an overview of brain insulin sensitivity in relation to cognitive function from animal and human studies, with particular emphasis placed on the impact exercise may have on brain insulin sensitivity. Mechanisms discussed include mitochondrial function, brain growth factors, and neurogenesis, which collectively help combat obesity-related metabolic disease and Alzheimer's dementia.
Collapse
Affiliation(s)
- Steven K Malin
- Department of Kinesiology & Health, Rutgers University, New Brunswick, New Jersey
- Division of Endocrinology, Metabolism & Nutrition, Rutgers University, New Brunswick, New Jersey
- New Jersey Institute for Food, Nutrition and Health, Rutgers University, New Brunswick, New Jersey
- Institute of Translational Medicine and Science, Rutgers University, New Brunswick, New Jersey
| | - Nathan R Stewart
- Department of Kinesiology & Health, Rutgers University, New Brunswick, New Jersey
| | - Andrew A Ude
- Department of Kinesiology & Health, Rutgers University, New Brunswick, New Jersey
| | - Brandon L Alderman
- Department of Kinesiology & Health, Rutgers University, New Brunswick, New Jersey
- Center of Alcohol and Substance Use Studies, Rutgers University, New Brunswick, New Jersey
| |
Collapse
|
24
|
Zhu Y, Li S, Lai H, Mo L, Tan C, Liu X, Deng F, Chen L. Effects of Anti-Parkinsonian Drugs on Verbal Fluency in Patients with Parkinson's Disease: A Network Meta-Analysis. Brain Sci 2022; 12:1496. [PMID: 36358422 PMCID: PMC9688928 DOI: 10.3390/brainsci12111496] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 10/24/2022] [Accepted: 11/02/2022] [Indexed: 06/06/2024] Open
Abstract
Verbal fluency impairment is common in patients with Parkinson's disease (PD), but the effect of drugs on verbal fluency in PD patients has not been comprehensively evaluated. We conducted a network meta-analysis based on four online databases to compare the effect of drugs on verbal fluency in PD patients. This study was performed and reported according to PRISMA-NMA guidelines. In total, 6 out of 3707 articles (three RCTS and three cross-sectional studies) covering eight drug regimens were included (five for letter fluency, five for semantic fluency). In terms of letter fluency, the ranking of the overall efficacy of included drug regimens was: levodopa, levodopa combined with pramipexole, rotigotine, cabergoline, pramipexole, pergolide, but no drug regimen presented a significant advantage over the others. In terms of semantic fluency, the ranking of the overall efficacy of included drug regimens was: rotigotine, levodopa, cabergoline, pergolide, pramipexole, among which, levodopa alone (SMD = 0.93, 95%CI: 0.28-1.59) and rotigotine alone (SMD = 1.18, 95%CI: 0.28-2.09) were statistically superior to pramipexole, while no significant difference was identified between all the other drug regimens. Levodopa and rotigotine seem to be more appropriate choices for PD patients with verbal fluency impairment. Further study is needed to illustrate the efficacy of drugs on verbal fluency in PD patients.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Fen Deng
- Department of Neurology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | | |
Collapse
|
25
|
Kang S, Chen Y, Wu J, Liang Y, Rao Y, Yue X, Lyu W, Li Y, Tan X, Huang H, Qiu S. Altered cortical thickness, degree centrality, and functional connectivity in middle-age type 2 diabetes mellitus. Front Neurol 2022; 13:939318. [PMID: 36408505 PMCID: PMC9672081 DOI: 10.3389/fneur.2022.939318] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Accepted: 10/12/2022] [Indexed: 05/01/2024] Open
Abstract
PURPOSE This study aimed to investigate the changes in brain structure and function in middle-aged patients with type 2 diabetes mellitus (T2DM) using morphometry and blood oxygen level-dependent functional magnetic resonance imaging (BOLD-fMRI). METHODS A total of 44 middle-aged patients with T2DM and 45 matched healthy controls (HCs) were recruited. Surface-based morphometry (SBM) was used to evaluate the changes in brain morphology. Degree centrality (DC) and functional connectivity (FC) were used to evaluate the changes in brain function. RESULTS Compared with HCs, middle-aged patients with T2DM exhibited cortical thickness reductions in the left pars opercularis, left transverse temporal, and right superior temporal gyri. Decreased DC values were observed in the cuneus and precuneus in T2DM. Hub-based FC analysis of these regions revealed lower connectivity in the bilateral hippocampus and parahippocampal gyrus, left precuneus, as well as left frontal sup. CONCLUSION Cortical thickness, degree centrality, as well as functional connectivity were found to have significant changes in middle-aged patients with T2DM. Our observations provide potential evidence from neuroimaging for analysis to examine diabetes-related brain damage.
Collapse
Affiliation(s)
- Shangyu Kang
- The First School of Clinical Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yuna Chen
- Department of Endocrinology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jinjian Wu
- The First School of Clinical Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yi Liang
- Department of Radiology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yawen Rao
- The First School of Clinical Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xiaomei Yue
- The First School of Clinical Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Wenjiao Lyu
- The First School of Clinical Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yifan Li
- The First School of Clinical Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xin Tan
- Department of Radiology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Haoming Huang
- Department of Radiology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Shijun Qiu
- Department of Radiology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
26
|
Savelieff MG, Chen KS, Elzinga SE, Feldman EL. Diabetes and dementia: Clinical perspective, innovation, knowledge gaps. J Diabetes Complications 2022; 36:108333. [PMID: 36240668 PMCID: PMC10076101 DOI: 10.1016/j.jdiacomp.2022.108333] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Accepted: 09/30/2022] [Indexed: 10/31/2022]
Abstract
The world faces a pandemic-level prevalence of type 2 diabetes. In parallel with this massive burden of metabolic disease is the growing prevalence of dementia as the population ages. The two health issues are intertwined. The Lancet Commission on dementia prevention, intervention, and care was convened to tackle the growing global concern of dementia by identifying risk factors. It concluded, along with other studies, that diabetes as well as obesity and the metabolic syndrome more broadly, which are frequently comorbid, raise the risk of developing dementia. Type 2 diabetes is a modifiable risk factor; however, it is uncertain whether anti-diabetic drugs mitigate risk of developing dementia. Reasons are manifold but constitute a critical knowledge gap in the field. This review outlines studies of type 2 diabetes on risk of dementia, illustrating key concepts. Moreover, it identifies knowledge gaps, reviews strategies to help fill these gaps, and concludes with a series of recommendations to mitigate risk and advance understanding of type 2 diabetes and dementia.
Collapse
Affiliation(s)
- Masha G Savelieff
- NeuroNetwork for Emerging Therapies, University of Michigan, Ann Arbor, MI 48109, USA
| | - Kevin S Chen
- NeuroNetwork for Emerging Therapies, University of Michigan, Ann Arbor, MI 48109, USA; Department of Neurology, University of Michigan, Ann Arbor, MI 48109, USA; Department of Neurosurgery, University of Michigan, Ann Arbor, MI 48109, USA.
| | - Sarah E Elzinga
- NeuroNetwork for Emerging Therapies, University of Michigan, Ann Arbor, MI 48109, USA; Department of Neurology, University of Michigan, Ann Arbor, MI 48109, USA.
| | - Eva L Feldman
- NeuroNetwork for Emerging Therapies, University of Michigan, Ann Arbor, MI 48109, USA; Department of Neurology, University of Michigan, Ann Arbor, MI 48109, USA.
| |
Collapse
|
27
|
Kullmann S, Goj T, Veit R, Fritsche L, Wagner L, Schneeweiss P, Hoene M, Hoffmann C, Machann J, Niess A, Preissl H, Birkenfeld AL, Peter A, Häring HU, Fritsche A, Moller A, Weigert C, Heni M. Exercise restores brain insulin sensitivity in sedentary adults who are overweight and obese. JCI Insight 2022; 7:161498. [PMID: 36134657 DOI: 10.1172/jci.insight.161498] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 08/04/2022] [Indexed: 11/17/2022] Open
Abstract
BACKGROUNDInsulin resistance of the brain can unfavorably affect long-term weight maintenance and body fat distribution. Little is known if and how brain insulin sensitivity can be restored in humans. We aimed to evaluate the effects of an exercise intervention on insulin sensitivity of the brain and how this relates to exercise-induced changes in whole-body metabolism and behavior.METHODSIn this clinical trial, sedentary participants who were overweight and obese underwent an 8-week supervised aerobic training intervention. Brain insulin sensitivity was assessed in 21 participants (14 women, 7 men; age range 21-59 years; BMI range 27.5-45.5 kg/m2) using functional MRI, combined with intranasal administration of insulin, before and after the intervention.RESULTSThe exercise program resulted in enhanced brain insulin action to the level of a person of healthy weight, demonstrated by increased insulin-induced striatal activity and strengthened hippocampal functional connectivity. Improved brain insulin action correlated with increased mitochondrial respiration in skeletal muscle, reductions in visceral fat and hunger, as well as improved cognition. Mediation analyses suggest that improved brain insulin responsiveness helps mediate the peripheral exercise effects leading to healthier body fat distribution and reduced perception of hunger.CONCLUSIONOur study demonstrates that an 8-week exercise intervention in sedentary individuals can restore insulin action in the brain. Hence, the ameliorating benefits of exercise toward brain insulin resistance may provide an objective therapeutic target in humans in the challenge to reduce diabetes risk factors.TRIAL REGISTRATIONClinicalTrials.gov (NCT03151590).FUNDINGBMBF/DZD 01GI0925.
Collapse
Affiliation(s)
- Stephanie Kullmann
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the University of Tübingen, Tübingen, Germany.,German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany.,Department of Internal Medicine, Division of Endocrinology, Diabetology and Nephrology, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Thomas Goj
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the University of Tübingen, Tübingen, Germany.,German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany.,Institute for Clinical Chemistry and Pathobiochemistry and
| | - Ralf Veit
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the University of Tübingen, Tübingen, Germany.,German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany
| | - Louise Fritsche
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the University of Tübingen, Tübingen, Germany.,German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany
| | - Lore Wagner
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the University of Tübingen, Tübingen, Germany.,German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany
| | - Patrick Schneeweiss
- Department of Sports Medicine, University Hospital Tübingen, Germany.,Interfaculty Research Institute for Sport and Physical Activity, University of Tübingen, Tübingen, Germany
| | - Miriam Hoene
- Institute for Clinical Chemistry and Pathobiochemistry and
| | | | - Jürgen Machann
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the University of Tübingen, Tübingen, Germany.,German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany.,Department of Radiology, Section on Experimental Radiology, University Hospital Tübingen, Germany
| | - Andreas Niess
- Department of Sports Medicine, University Hospital Tübingen, Germany.,Interfaculty Research Institute for Sport and Physical Activity, University of Tübingen, Tübingen, Germany
| | - Hubert Preissl
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the University of Tübingen, Tübingen, Germany.,German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany.,Department of Internal Medicine, Division of Endocrinology, Diabetology and Nephrology, Eberhard Karls University Tübingen, Tübingen, Germany.,Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Neuherberg, Germany
| | - Andreas L Birkenfeld
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the University of Tübingen, Tübingen, Germany.,German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany.,Department of Internal Medicine, Division of Endocrinology, Diabetology and Nephrology, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Andreas Peter
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the University of Tübingen, Tübingen, Germany.,German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany.,Institute for Clinical Chemistry and Pathobiochemistry and
| | - Hans-Ulrich Häring
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the University of Tübingen, Tübingen, Germany.,German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany.,Department of Internal Medicine, Division of Endocrinology, Diabetology and Nephrology, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Andreas Fritsche
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the University of Tübingen, Tübingen, Germany.,German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany.,Department of Internal Medicine, Division of Endocrinology, Diabetology and Nephrology, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Anja Moller
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the University of Tübingen, Tübingen, Germany.,German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany.,Department of Internal Medicine, Division of Endocrinology, Diabetology and Nephrology, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Cora Weigert
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the University of Tübingen, Tübingen, Germany.,German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany.,Institute for Clinical Chemistry and Pathobiochemistry and
| | - Martin Heni
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the University of Tübingen, Tübingen, Germany.,German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany.,Department of Internal Medicine, Division of Endocrinology, Diabetology and Nephrology, Eberhard Karls University Tübingen, Tübingen, Germany.,Institute for Clinical Chemistry and Pathobiochemistry and.,Division of Endocrinology and Diabetology, Department of Internal Medicine I, Ulm University Hospital, Ulm, Germany
| |
Collapse
|
28
|
Nose-to-Brain Delivery of Therapeutic Peptides as Nasal Aerosols. Pharmaceutics 2022; 14:pharmaceutics14091870. [PMID: 36145618 PMCID: PMC9502087 DOI: 10.3390/pharmaceutics14091870] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 08/27/2022] [Accepted: 08/31/2022] [Indexed: 11/22/2022] Open
Abstract
Central nervous system (CNS) disorders, such as psychiatric disorders, neurodegeneration, chronic pain, stroke, brain tumor, spinal cord injury, and many other CNS diseases, would hugely benefit from specific and potent peptide pharmaceuticals and their low inherent toxicity. The delivery of peptides to the brain is challenging due to their low metabolic stability, which decreases their duration of action, poor penetration of the blood-brain barrier (BBB), and their incompatibility with oral administration, typically resulting in the need for parenteral administration. These challenges limit peptides’ clinical application and explain the interest in alternative routes of peptide administration, particularly nose-to-brain (N-to-B) delivery, which allows protein and peptide drugs to reach the brain noninvasively. N-to-B delivery can be a convenient method for rapidly targeting the CNS, bypassing the BBB, and minimizing systemic exposure; the olfactory and trigeminal nerves provide a unique pathway to the brain and the external environment. This review highlights the intranasal delivery of drugs, focusing on peptide delivery, illustrating various clinical applications, nasal delivery devices, and the scope and limitations of this approach.
Collapse
|
29
|
Novak V, Mantzoros CS, Novak P, McGlinchey R, Dai W, Lioutas V, Buss S, Fortier CB, Khan F, Aponte Becerra L, Ngo LH. MemAID: Memory advancement with intranasal insulin vs. placebo in type 2 diabetes and control participants: a randomized clinical trial. J Neurol 2022; 269:4817-4835. [PMID: 35482079 PMCID: PMC9046533 DOI: 10.1007/s00415-022-11119-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 03/29/2022] [Accepted: 03/30/2022] [Indexed: 01/21/2023]
Abstract
BACKGROUND This study aimed at assessing the long-term effects of intranasal insulin (INI) on cognition and gait in older people with and without type 2 diabetes mellitus (T2DM). METHODS Phase 2 randomized, double-blinded trial consisted of 24 week treatment with 40 IU of INI (Novolin® R, off-label use) or placebo (sterile saline) once daily and 24 week follow-up. Primary outcomes were cognition, normal (NW), and dual-task (DTW) walking speeds. Of 244 randomized, 223 completed baseline (51 DM-INI, 55 DM-Placebo, 58 Control-INI, 59 Control-Placebo; 109 female, 65.8 ± 9.1; 50-85 years old); 174 completed treatment (84 DM, 90 Controls); 156 completed follow-up (69 DM). RESULTS DM-INI had faster NW (~ 7 cm/s; p = 0.025) and DTW on-treatment (p = 0.007; p = 0.812 adjusted for baseline difference) than DM-Placebo. Control-INI had better executive functioning on-treatment (p = 0.008) and post-treatment (p = 0.007) and verbal memory post-treatment (p = 0.004) than Control-Placebo. DM-INI increased cerebral blood flow in medio-prefrontal cortex (p < 0.001) on MRI. Better vasoreactivity was associated with faster DTW (p < 0.008). In DM-INI, plasma insulin (p = 0.006) and HOMA-IR (p < 0.013) decreased post-treatment. Overall INI effect demonstrated faster walking (p = 0.002) and better executive function (p = 0.002) and verbal memory (p = 0.02) (combined DM-INI and Control-INI cohort, hemoglobin A1c-adjusted). INI was not associated with serious adverse events, hypoglycemic episodes, or weight gain. CONCLUSION There is evidence for positive INI effects on cognition and gait. INI-treated T2DM participants walked faster, showed increased cerebral blood flow and decreased plasma insulin, while controls improved executive functioning and verbal memory. The MemAID trial provides proof-of-concept for preliminary safety and efficacy and supports future evaluation of INI role to treat T2DM and age-related functional decline.
Collapse
Affiliation(s)
- Vera Novak
- Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, 185 Pilgrim Rd, Boston, MA, 02215, USA.
| | - Christos S Mantzoros
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
- Department of Medicine, Boston VA Healthcare System, Boston, MA, USA
| | - Peter Novak
- Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Regina McGlinchey
- Translational Research Center for TBI and Stress Disorders (TRACTS) and Geriatric Research Educational and Clinical Research Center (GRECC), VA Boston Healthcare System, Boston, MA, USA
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA
| | - Weiying Dai
- Department of Computer Science, State University of New York (SUNY), Binghamton, NY, USA
| | - Vasileios Lioutas
- Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, 185 Pilgrim Rd, Boston, MA, 02215, USA
| | - Stephanie Buss
- Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, 185 Pilgrim Rd, Boston, MA, 02215, USA
| | - Catherine B Fortier
- Translational Research Center for TBI and Stress Disorders (TRACTS) and Geriatric Research Educational and Clinical Research Center (GRECC), VA Boston Healthcare System, Boston, MA, USA
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA
| | - Faizan Khan
- Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, 185 Pilgrim Rd, Boston, MA, 02215, USA
| | - Laura Aponte Becerra
- Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, 185 Pilgrim Rd, Boston, MA, 02215, USA
| | - Long H Ngo
- Department of Medicine, Beth Israel Deaconess Medical Center and School of Public Health, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
30
|
Li ZY, Ma T, Yu Y, Hu B, Han Y, Xie H, Ni MH, Chen ZH, Zhang YM, Huang YX, Li WH, Wang W, Yan LF, Cui GB. Changes of brain function in patients with type 2 diabetes mellitus measured by different analysis methods: A new coordinate-based meta-analysis of neuroimaging. Front Neurol 2022; 13:923310. [PMID: 36090859 PMCID: PMC9449648 DOI: 10.3389/fneur.2022.923310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Accepted: 07/11/2022] [Indexed: 11/22/2022] Open
Abstract
Objective Neuroimaging meta-analysis identified abnormal neural activity alterations in patients with type 2 diabetes mellitus (T2DM), but there was no consistency or heterogeneity analysis between different brain imaging processing strategies. The aim of this meta-analysis was to determine consistent changes of regional brain functions in T2DM via the indicators obtained by using different post-processing methods. Methods Since the indicators obtained using varied post-processing methods reflect different neurophysiological and pathological characteristics, we further conducted a coordinate-based meta-analysis (CBMA) of the two categories of neuroimaging literature, which were grouped according to similar data processing methods: one group included regional homogeneity (ReHo), independent component analysis (ICA), and degree centrality (DC) studies, while the other group summarized the literature on amplitude of low-frequency fluctuation (ALFF) and cerebral blood flow (CBF). Results The final meta-analysis included 23 eligible trials with 27 data sets. Compared with the healthy control group, when neuroimaging studies were combined with ReHo, ICA, and DC measurements, the brain activity of the right Rolandic operculum, right supramarginal gyrus, and right superior temporal gyrus in T2DM patients decreased significantly. When neuroimaging studies were combined with ALFF and CBF measurements, there was no clear evidence of differences in the brain function between T2DM and HCs. Conclusion T2DM patients have a series of spontaneous abnormal brain activities, mainly involving brain regions related to learning, memory, and emotion, which provide early biomarkers for clarifying the mechanism of cognitive impairment and neuropsychiatric disorders in diabetes. Systematic review registration https://www.crd.york.ac.uk/prospero/display_record.php?RecordID=247071, PROSPERO [CRD42021247071].
Collapse
Affiliation(s)
- Ze-Yang Li
- Department of Radiology, Functional and Molecular Imaging Key Lab of Shaanxi Province, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Teng Ma
- Department of Radiology, Functional and Molecular Imaging Key Lab of Shaanxi Province, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Ying Yu
- Department of Radiology, Functional and Molecular Imaging Key Lab of Shaanxi Province, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Bo Hu
- Department of Radiology, Functional and Molecular Imaging Key Lab of Shaanxi Province, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Yu Han
- Department of Radiology, Functional and Molecular Imaging Key Lab of Shaanxi Province, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Hao Xie
- Department of Radiology, Functional and Molecular Imaging Key Lab of Shaanxi Province, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Min-Hua Ni
- Department of Radiology, Functional and Molecular Imaging Key Lab of Shaanxi Province, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
- Faculty of Medical Technology, Shaanxi University of Chinese Medicine, Xianyang, China
| | - Zhu-Hong Chen
- Department of Radiology, Functional and Molecular Imaging Key Lab of Shaanxi Province, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Yang-Ming Zhang
- Battalion of the Second Regiment of Cadets of Basic Medicine, Fourth Military Medical University, Xi'an, China
| | - Yu-Xiang Huang
- Battalion of the Second Regiment of Cadets of Basic Medicine, Fourth Military Medical University, Xi'an, China
| | - Wen-Hua Li
- Battalion of the Second Regiment of Cadets of Basic Medicine, Fourth Military Medical University, Xi'an, China
| | - Wen Wang
- Department of Radiology, Functional and Molecular Imaging Key Lab of Shaanxi Province, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
- *Correspondence: Guang-Bin Cui ;
| | - Lin-Feng Yan
- Department of Radiology, Functional and Molecular Imaging Key Lab of Shaanxi Province, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
- Lin-Feng Yan
| | - Guang-Bin Cui
- Department of Radiology, Functional and Molecular Imaging Key Lab of Shaanxi Province, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
- Wen Wang
| |
Collapse
|
31
|
Aponte Becerra L, Galindo Mendez B, Khan F, Lioutas V, Novak P, Mantzoros CS, Ngo LH, Novak V. Safety of Intranasal Insulin in Type 2 Diabetes on Systemic Insulin: A Double-Blinded Placebo-Controlled Sub-Study of Memaid Trial. ARCHIVES OF DIABETES & OBESITY 2022; 4:403-415. [PMID: 35903156 PMCID: PMC9328174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
AIMS To determine safety of intranasal insulin (INI) in MemAID trial participants with diabetes treated with systemic insulins. MATERIALS AND METHODS This randomized, double-blinded trial consisted of 24-week INI or placebo treatment once daily and 24-week follow-up. Safety outcomes were: 1) Short-term effects on glycemic variability, hypoglycemic episodes on continuous glucose monitoring (CGM) at baseline and on-treatment. 2) Long-term effects on glucose metabolism and weight on INI/placebo treatment and post-treatment follow-up. Of 86 screened subjects, 14 were randomized, 9 (5 INI, 4 Placebo) completed CGM at baseline and on-treatment, and 5 (2 INI, 3 Placebo) completed treatment and follow-up. RESULTS INI was safe and was not associated with serious adverse events, hypoglycemic episodes or weight gain. INI administration did not acutely affect capillary glucose. Glycemic variability on CGM decreased with INI, compared to baseline. On INI treatment, there was a long-term trend toward lower HbA1c, plasma glucose and insulin. No interactions with subcutaneous insulins were observed. CONCLUSIONS INI is safe in older people with diabetes treated with systemic insulins, and it is not associated with adverse events, hypoglycemia or weight gain. Future studies are needed to determine whether INI administration can reduce glycemic variability, improve insulin sensitivity and thus potentially lessen diabetes burden in this population.
Collapse
Affiliation(s)
- L Aponte Becerra
- Department of Neurology, SAFE Laboratory, Beth Israel Deaconess Medical Center, Harvard Medical School, USA
- Department of Internal Medicine, Jackson Memorial Hospital, University of Miami, Miami, FL, USA
| | - B Galindo Mendez
- Department of Neurology, SAFE Laboratory, Beth Israel Deaconess Medical Center, Harvard Medical School, USA
| | - F Khan
- Department of Neurology, SAFE Laboratory, Beth Israel Deaconess Medical Center, Harvard Medical School, USA
| | - V Lioutas
- Department of Neurology, SAFE Laboratory, Beth Israel Deaconess Medical Center, Harvard Medical School, USA
| | - P Novak
- Department of Neurology, Brigham and Women's Faulkner Hospital, Harvard Medical School, Boston, MA, USA
| | - C S Mantzoros
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston MA and Department of Medicine, Boston VA Healthcare System, Harvard Medical School, Boston, MA, USA
| | - L H Ngo
- Department of Medicine, Beth Israel Deaconess Medical Center and School of Public Health, Harvard Medical School, Boston, MA, USA
| | - V Novak
- Department of Neurology, SAFE Laboratory, Beth Israel Deaconess Medical Center, Harvard Medical School, USA
| |
Collapse
|
32
|
Wagner L, Veit R, Fritsche L, Häring HU, Fritsche A, Birkenfeld AL, Heni M, Preissl H, Kullmann S. Sex differences in central insulin action: Effect of intranasal insulin on neural food cue reactivity in adults with normal weight and overweight. Int J Obes (Lond) 2022; 46:1662-1670. [PMID: 35715625 PMCID: PMC9395264 DOI: 10.1038/s41366-022-01167-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 05/30/2022] [Accepted: 06/07/2022] [Indexed: 01/17/2023]
Abstract
Background/Objectives Central insulin action influences cognitive processes, peripheral metabolism, and eating behavior. However, the contribution of obesity and sex on central insulin-mediated neural food cue processing still remains unclear. Subjects/Methods In a randomized within-participant design, including two visits, 60 participants (30 women, BMI 18–32 kg/m2, age 21–69 years) underwent a functional MRI task measuring blood oxygen level-dependent (BOLD) signal in response to visual food cues after intranasal insulin or placebo spray administration. Central insulin action was defined as the neural BOLD response to food cues after insulin compared to placebo administration. Afterwards, participants were asked to rate the food cues for desire to eat (i.e., wanting rating). For statistical analyses, participants were grouped according to BMI and sex. Results Food cue reactivity in the amygdala showed higher BOLD activation in response to central insulin compared to placebo. Furthermore, women with overweight and obesity and men of normal weight showed higher BOLD neural food cue responsivity to central insulin compared to placebo. Higher central insulin action in the insular cortex was associated with better peripheral insulin sensitivity and higher cognitive control. Moreover, central insulin action in the dorsolateral prefrontal cortex (DLPFC) revealed significant sex differences. In response to central insulin compared to placebo, men showed lower DLPFC BOLD activity, whereas women showed higher DLPFC activity in response to highly desired food cues. On behavioral level, central insulin action significantly reduced hunger, whereas the desire to eat, especially for low caloric food cues was significantly higher with central insulin than with placebo. Conclusions Obesity and sex influenced the central insulin-mediated neural BOLD activity to visual food cues in brain regions implicated in reward and cognitive control. These findings show that central insulin action regulates food response differentially in men and women, which may have consequences for metabolism and eating behavior.
Collapse
Affiliation(s)
- Lore Wagner
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the University of Tübingen, Tübingen, Germany. .,German Center for Diabetes Research (DZD e.V.), Tübingen, Germany.
| | - Ralf Veit
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the University of Tübingen, Tübingen, Germany.,German Center for Diabetes Research (DZD e.V.), Tübingen, Germany
| | - Louise Fritsche
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the University of Tübingen, Tübingen, Germany.,German Center for Diabetes Research (DZD e.V.), Tübingen, Germany
| | - Hans-Ulrich Häring
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the University of Tübingen, Tübingen, Germany.,German Center for Diabetes Research (DZD e.V.), Tübingen, Germany.,Department of Internal Medicine, Division of Endocrinology, Diabetology and Nephrology, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Andreas Fritsche
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the University of Tübingen, Tübingen, Germany.,German Center for Diabetes Research (DZD e.V.), Tübingen, Germany.,Department of Internal Medicine, Division of Endocrinology, Diabetology and Nephrology, Eberhard Karls University Tübingen, Tübingen, Germany.,Nutritional and Preventive Medicine, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Andreas L Birkenfeld
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the University of Tübingen, Tübingen, Germany.,German Center for Diabetes Research (DZD e.V.), Tübingen, Germany.,Department of Internal Medicine, Division of Endocrinology, Diabetology and Nephrology, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Martin Heni
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the University of Tübingen, Tübingen, Germany.,German Center for Diabetes Research (DZD e.V.), Tübingen, Germany.,Department of Internal Medicine, Division of Endocrinology, Diabetology and Nephrology, Eberhard Karls University Tübingen, Tübingen, Germany.,Institute for Clinical Chemistry and Pathobiochemistry, University Hospital Tübingen, Tübingen, Germany.,Department of Internal Medicine I, Division of Endocrinology and Diabetology, Ulm University Hospital, Ulm, Germany
| | - Hubert Preissl
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the University of Tübingen, Tübingen, Germany.,German Center for Diabetes Research (DZD e.V.), Tübingen, Germany.,Department of Internal Medicine, Division of Endocrinology, Diabetology and Nephrology, Eberhard Karls University Tübingen, Tübingen, Germany.,Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Center Munich, German Research Center for Environmental Health (GmbH), Neuherberg, Germany
| | - Stephanie Kullmann
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the University of Tübingen, Tübingen, Germany.,German Center for Diabetes Research (DZD e.V.), Tübingen, Germany.,Department of Internal Medicine, Division of Endocrinology, Diabetology and Nephrology, Eberhard Karls University Tübingen, Tübingen, Germany
| |
Collapse
|
33
|
Luo A, Xie Z, Wang Y, Wang X, Li S, Yan J, Zhan G, Zhou Z, Zhao Y, Li S. Type 2 diabetes mellitus-associated cognitive dysfunction: Advances in potential mechanisms and therapies. Neurosci Biobehav Rev 2022; 137:104642. [PMID: 35367221 DOI: 10.1016/j.neubiorev.2022.104642] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2021] [Revised: 03/24/2022] [Accepted: 03/27/2022] [Indexed: 12/22/2022]
Abstract
Type 2 diabetes (T2D) and its target organ injuries cause distressing impacts on personal health and put an enormous burden on the healthcare system, and increasing attention has been paid to T2D-associated cognitive dysfunction (TDACD). TDACD is characterized by cognitive dysfunction, delayed executive ability, and impeded information-processing speed. Brain imaging data suggest that extensive brain regions are affected in patients with T2D. Based on current findings, a wide spectrum of non-specific neurodegenerative mechanisms that partially overlap with the mechanisms of neurodegenerative diseases is hypothesized to be associated with TDACD. However, it remains unclear whether TDACD is a consequence of T2D or a complication that co-occurs with T2D. Theoretically, anti-diabetes methods are promising neuromodulatory approaches to reduce brain injury in patients with T2D. In this review, we summarize potential mechanisms underlying TDACD and promising neurotropic effects of anti-diabetes methods and some neuroprotective natural compounds. Constructing screening or diagnostic tools and developing targeted treatment and preventive strategies would be expected to reduce the burden of TDACD.
Collapse
Affiliation(s)
- Ailin Luo
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology.
| | - Zheng Xie
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology.
| | - Yue Wang
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology.
| | - Xuan Wang
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology.
| | - Shan Li
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology.
| | - Jing Yan
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology.
| | - Gaofeng Zhan
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology.
| | - Zhiqiang Zhou
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology.
| | - Yilin Zhao
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology.
| | - Shiyong Li
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology.
| |
Collapse
|
34
|
Wu CY, Shapiro L, Ouk M, MacIntosh BJ, Black SE, Shah BR, Swardfager W. Glucose-lowering drugs, cognition, and dementia: The clinical evidence. Neurosci Biobehav Rev 2022; 137:104654. [PMID: 35398114 DOI: 10.1016/j.neubiorev.2022.104654] [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: 02/01/2022] [Revised: 04/01/2022] [Accepted: 04/03/2022] [Indexed: 11/19/2022]
Abstract
Type 2 diabetes mellitus (T2DM) is an important risk factor for dementia. The possibility to mitigate this risk by controlling T2DM is compelling; however, different glucose-lowering drugs have different effects on the brain by virtue of their different mechanisms of action. The clinical and epidemiological data appear mixed, warranting careful critical evaluation of the human studies. Here we examine the evidence in the context of dementia prevention and treatment, both for people with and without T2DM. We discuss the evidence on this scaffold of research directions, identifying methodological complexities in the extant literature (e.g. comparator discrepancies, changes in the therapeutic landscape), and the implications of different outcome measures (e.g. neuropsychological). We consider possible implications of cerebrovascular protection vs. effects on progression of neurodegenerative proteinopathy, and we present a research roadmap for glucose-lowering drugs in cognitive neurology, including neuroimaging, and fluid biomarkers. We conclude that there is great potential to advance personalized strategies to prevent and treat dementia with glucose-lowering drugs.
Collapse
Affiliation(s)
- Che-Yuan Wu
- Department of Pharmacology & Toxicology, University of Toronto, Toronto, Ontario, Canada; Sandra Black Centre for Brain Resilience and Recovery, Hurvitz Brain Sciences Program, Sunnybrook Research Institute, Toronto, Ontario, Canada
| | - Lila Shapiro
- Department of Pharmacology & Toxicology, University of Toronto, Toronto, Ontario, Canada; Sandra Black Centre for Brain Resilience and Recovery, Hurvitz Brain Sciences Program, Sunnybrook Research Institute, Toronto, Ontario, Canada
| | - Michael Ouk
- Department of Pharmacology & Toxicology, University of Toronto, Toronto, Ontario, Canada; Sandra Black Centre for Brain Resilience and Recovery, Hurvitz Brain Sciences Program, Sunnybrook Research Institute, Toronto, Ontario, Canada
| | - Bradley J MacIntosh
- Sandra Black Centre for Brain Resilience and Recovery, Hurvitz Brain Sciences Program, Sunnybrook Research Institute, Toronto, Ontario, Canada; Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada; Canadian Partnership for Stroke Recovery, Sunnybrook Research Institute, Toronto, Ontario, Canada
| | - Sandra E Black
- Sandra Black Centre for Brain Resilience and Recovery, Hurvitz Brain Sciences Program, Sunnybrook Research Institute, Toronto, Ontario, Canada; Canadian Partnership for Stroke Recovery, Sunnybrook Research Institute, Toronto, Ontario, Canada; Department of Medicine, Division of Neurology, University of Toronto, Toronto, Ontario, Canada; Toronto Dementia Research Alliance, Toronto, Ontario, Canada
| | - Baiju R Shah
- ICES, Toronto, Ontario, Canada; Divisions of Endocrinology and Obstetric Medicine, Department of Medicine, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada
| | - Walter Swardfager
- Department of Pharmacology & Toxicology, University of Toronto, Toronto, Ontario, Canada; Sandra Black Centre for Brain Resilience and Recovery, Hurvitz Brain Sciences Program, Sunnybrook Research Institute, Toronto, Ontario, Canada; Canadian Partnership for Stroke Recovery, Sunnybrook Research Institute, Toronto, Ontario, Canada; KITE UHN Toronto Rehabilitation Institute, Toronto, Ontario, Canada
| |
Collapse
|
35
|
Tadevosyan NE, Khachunts AS, Gohargani M, Sahakyan AA, Tumanyan AA. Voluntary Attention and Quality of Life in Patients With Type 1 and Type 2 Diabetes Mellitus: Differences in Changes Depending on Disease Type and Duration. J EVOL BIOCHEM PHYS+ 2022. [DOI: 10.1134/s0022093022020247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
36
|
Haas SS, Myoraku A, Watson K, Robakis T, Frangou S, Abbasi F, Rasgon N. Lower functional hippocampal connectivity in healthy adults is jointly associated with higher levels of leptin and insulin resistance. Eur Psychiatry 2022; 65:e29. [PMID: 35492025 PMCID: PMC9158395 DOI: 10.1192/j.eurpsy.2022.21] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 02/25/2022] [Accepted: 04/22/2022] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND Metabolic dysregulation is currently considered a major risk factor for hippocampal pathology. The aim of the present study was to characterize the influence of key metabolic drivers on functional connectivity of the hippocampus in healthy adults. METHODS Insulin resistance was directly quantified by measuring steady-state plasma glucose (SSPG) concentration during the insulin suppression test and fasting levels of insulin, glucose, leptin, and cortisol, and measurements of body mass index and waist circumference were obtained in a sample of healthy cognitively intact adults (n = 104). Resting-state neuroimaging data were also acquired for the quantification of hippocampal functional cohesiveness and integration with the major resting-state networks (RSNs). Data-driven analysis using unsupervised machine learning (k-means clustering) was then employed to identify clusters of individuals based on their metabolic and functional connectivity profiles. RESULTS K-means clustering identified two clusters of increasing metabolic deviance evidenced by cluster differences in the plasma levels of leptin (40.36 (29.97) vs. 27.59 (25.58) μg/L) and the degree of insulin resistance (SSPG concentration: 161.63 (65.27) vs. 125.72 (66.81) mg/dL). Individuals in the cluster with higher metabolic deviance showed lower functional cohesiveness within each hippocampus and lower integration of posterior and anterior components of the left and right hippocampus with the major RSNs. The two clusters did not differ in general intellectual ability or episodic memory. CONCLUSIONS We identified two clusters of individuals differentiated by abnormalities in insulin resistance, leptin levels, and hippocampal connectivity, with one of the clusters showing greater deviance. These findings support the link between metabolic dysregulation and hippocampal function even in nonclinical samples.
Collapse
Affiliation(s)
- Shalaila S. Haas
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Alison Myoraku
- Department of Psychiatry, Stanford University School of Medicine, Palo Alto, California, USA
| | - Kathleen Watson
- Department of Psychiatry, Stanford University School of Medicine, Palo Alto, California, USA
| | - Thalia Robakis
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Sophia Frangou
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Department of Psychiatry, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, British Columbia, Canada
| | - Fahim Abbasi
- Division of Cardiovascular Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, California, USA
| | - Natalie Rasgon
- Department of Psychiatry, Stanford University School of Medicine, Palo Alto, California, USA
| |
Collapse
|
37
|
Chau ACM, Smith AE, Hordacre B, Kumar S, Cheung EYW, Mak HKF. A scoping review of resting-state brain functional alterations in Type 2 diabetes. Front Neuroendocrinol 2022; 65:100970. [PMID: 34922997 DOI: 10.1016/j.yfrne.2021.100970] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 11/18/2021] [Accepted: 12/07/2021] [Indexed: 11/28/2022]
Abstract
Resting-state functional magnetic resonance imaging (rs-fMRI) has been actively used in the last decade to investigate brain functional connectivity alterations in Type 2 Diabetes Mellitus (T2DM) to understand the neuropathophysiology of T2DM in cognitive degeneration. Given the emergence of new analysis techniques, this scoping review aims to map the rs-fMRI analysis techniques that have been applied in the literature and reports the latest rs-fMRI findings that have not been covered in previous reviews. Graph theory, the contemporary rs-fMRI analysis, has been used to demonstrate altered brain topological organisations in people with T2DM, which included altered degree centrality, functional connectivity strength, the small-world architecture and network-based statistics. These alterations were correlated with T2DM patients' cognitive performances. Graph theory also contributes to identify unbiased seeds for seed-based analysis. The expanding rs-fMRI analytical approaches continue to provide new evidence that helps to understand the mechanisms of T2DM-related cognitive degeneration.
Collapse
Affiliation(s)
- Anson C M Chau
- Medical Imaging, Medical Radiation Science, Allied Health and Human Performance, University of South Australia, Adelaide, Australia; Alliance for Research in Exercise, Nutrition and Activity (ARENA), Allied Health and Human Performance, University of South Australia, Adelaide, Australia.
| | - Ashleigh E Smith
- Alliance for Research in Exercise, Nutrition and Activity (ARENA), Allied Health and Human Performance, University of South Australia, Adelaide, Australia.
| | - Brenton Hordacre
- IIMPACT in Health, Allied Health and Human Performance, University of South Australia, Adelaide, Australia.
| | - Saravana Kumar
- IIMPACT in Health, Allied Health and Human Performance, University of South Australia, Adelaide, Australia; Allied Health and Human Performance, University of South Australia, Adelaide, Australia.
| | - Eva Y W Cheung
- School of Medical and Health Sciences, Tung Wah College, Hong Kong.
| | - Henry K F Mak
- Department of Diagnostic Radiology, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong; Alzheimer's Disease Research Network, The University of Hong Kong, Hong Kong; State Key Laboratory for Brain and Cognitive Sciences, The University of Hong Kong, Hong Kong.
| |
Collapse
|
38
|
Del Moro L, Rota E, Pirovano E, Rainero I. Migraine, Brain Glucose Metabolism and the "Neuroenergetic" Hypothesis: A Scoping Review. THE JOURNAL OF PAIN 2022; 23:1294-1317. [PMID: 35296423 DOI: 10.1016/j.jpain.2022.02.006] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 02/04/2022] [Accepted: 02/14/2022] [Indexed: 02/06/2023]
Abstract
Increasing evidence suggests that migraine may be the result of an impaired brain glucose metabolism. Several studies have reported brain mitochondrial dysfunction, impaired brain glucose metabolism and gray matter volume reduction in specific brain areas of migraineurs. Furthermore, peripheral insulin resistance, a condition demonstrated in several studies, may extend to the brain, leading to brain insulin resistance. This condition has been proven to downregulate insulin receptors, both in astrocytes and neurons, triggering a reduction in glucose uptake and glycogen synthesis, mainly during high metabolic demand. This scoping review examines the clinical, epidemiologic and pathophysiologic data supporting the hypothesis that abnormalities in brain glucose metabolism may generate a mismatch between the brain's energy reserve and metabolic expenditure, triggering migraine attacks. Moreover, alteration in glucose homeostasis could generate a chronic brain energy deficit promoting migraine chronification. Lastly, insulin resistance may link migraine with its comorbidities, like obesity, depression, cognitive impairment and cerebrovascular diseases. PERSPECTIVE: Although additional experimental studies are needed to support this novel "neuroenergetic" hypothesis, brain insulin resistance in migraineurs may unravel the pathophysiological mechanisms of the disease, explaining the migraine chronification and connecting migraine with comorbidities. Therefore, this hypothesis could elucidate novel potential approaches for migraine treatment.
Collapse
Affiliation(s)
- Lorenzo Del Moro
- Foundation Allineare Sanità and Salute, Scientific Committee, Milan, Italy; LUMEN APS, European Salus Network, Scientific Committee, San Pietro in Cerro (PC), Italy.
| | - Eugenia Rota
- Neurology Unit, ASL AL, San Giacomo Hospital, Novi Ligure, Italy
| | - Elenamaria Pirovano
- Foundation Allineare Sanità and Salute, Scientific Committee, Milan, Italy; LUMEN APS, European Salus Network, Scientific Committee, San Pietro in Cerro (PC), Italy
| | - Innocenzo Rainero
- Headache Center, Department of Neuroscience "Rita Levi Montalcini", University of Torino, Italy
| |
Collapse
|
39
|
Lei H, Hu R, Luo G, Yang T, Shen H, Deng H, Chen C, Zhao H, Liu J. Altered Structural and Functional MRI Connectivity in Type 2 Diabetes Mellitus Related Cognitive Impairment: A Review. Front Hum Neurosci 2022; 15:755017. [PMID: 35069149 PMCID: PMC8770326 DOI: 10.3389/fnhum.2021.755017] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Accepted: 12/13/2021] [Indexed: 12/16/2022] Open
Abstract
Type 2 diabetes mellitus (T2DM) is associated with cognitive impairment in many domains. There are several pieces of evidence that changes in neuronal neuropathies and metabolism have been observed in T2DM. Structural and functional MRI shows that abnormal connections and synchronization occur in T2DM brain circuits and related networks. Neuroplasticity and energy metabolism appear to be principal effector systems, which may be related to amyloid beta (Aβ) deposition, although there is no unified explanation that includes the complex etiology of T2DM with cognitive impairment. Herein, we assume that cognitive impairment in diabetes may lead to abnormalities in neuroplasticity and energy metabolism in the brain, and those reflected to MRI structural connectivity and functional connectivity, respectively.
Collapse
|
40
|
Fang F, Gong YJ, Luo Q, Ge RB, Kang M, Ma MM, Zhang L, Mu D, Yin DZ, Wang YF. Cognitive Dysfunction in Type 2 Diabetes Is Not a One-Way Process: Evidence From a Longitudinal Brain Connectivity Study. Front Endocrinol (Lausanne) 2022; 13:874538. [PMID: 35573998 PMCID: PMC9095898 DOI: 10.3389/fendo.2022.874538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Accepted: 03/29/2022] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Cognitive dysfunction is an important comorbidity of diabetes characterized by brain functional hypo-connectivity. However, our recent study demonstrated an adaptive hyper-connectivity in young type 2 diabetes with cognitive decrements. This longitudinal study aimed to further explore the changes in functional connectivity and cognitive outcomes after regular glycemic control. METHODS At 18 months after recruitment, participants underwent a second cognitive assessment and magnetic resonance imaging. Three enhanced functional connectivities previously identified at baseline were followed up. Linear mixed-effects models were performed to compare the longitudinal changes of cognition and functional connectivity in patients with type 2 diabetes and non-diabetic controls. A linear regression model was used to investigate the association between changes in functional connectivity and changes in cognitive performance. RESULTS Improvements in multiple cognitive domains were observed in diabetes; however, the enhanced functional connectivity at baseline decreased significantly. Moreover, the decrease in hippocampal connectivity was correlated with an increase in the accuracy of Stroop task and the decrease in posterior cingulate cortex connectivity was correlated with an increase in Montreal Cognitive Assessment in diabetes. CONCLUSION This study suggests diabetes-related cognitive dysfunction is not a one-way process and the early-stage enhancement of brain connectivity was a potential "window period" for cognitive reversal.
Collapse
Affiliation(s)
- Fang Fang
- Department of Endocrinology and Metabolism, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Yu-Jia Gong
- Department of Endocrinology and Metabolism, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Qian Luo
- Department of Radiology, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Ren-Bin Ge
- Department of Radiology, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Mei Kang
- Clinical Research Center, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Ming-Ming Ma
- Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Lei Zhang
- Department of Radiology, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Di Mu
- Department of Anesthesiology, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Da-Zhi Yin
- Shanghai Key Laboratory of Brain Functional Genomics (Ministry of Education), School of Psychology and Cognitive Science, East China Normal University, Shanghai, China
- Shanghai Changning Mental Health Center, Shanghai, China
- *Correspondence: Yu-Fan Wang, ; Da-Zhi Yin,
| | - Yu-Fan Wang
- Department of Endocrinology and Metabolism, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai, China
- *Correspondence: Yu-Fan Wang, ; Da-Zhi Yin,
| |
Collapse
|
41
|
Teng Z, Feng J, Dong Y, Xu J, Jiang X, Chen H, Qi Q, Li R, Chen W, Lv P. Triglyceride glucose index is associated with cerebral small vessel disease burden and cognitive impairment in elderly patients with type 2 diabetes mellitus. Front Endocrinol (Lausanne) 2022; 13:970122. [PMID: 35992100 PMCID: PMC9390881 DOI: 10.3389/fendo.2022.970122] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Accepted: 07/20/2022] [Indexed: 11/13/2022] Open
Abstract
OBJECTIVE To investigate the relations of Triglyceride glucose (TyG) index with cerebral small vessel disease (CSVD) burden and cognitive function in aged patients with type 2 diabetes mellitus (T2DM). METHODS A total of 308 elderly patients with T2DM were included in this retrospective study. The standardized Chinese version of Mini-Mental State Examination was used to assess cognitive function. The total CSVD burden score was assessed by combining four imaging markers of CSVD, including the presence of white matter hyperintensity, cerebral microbleeds in the deep, lacunes and enlarged perivascular spaces in the basal ganglia. The TyG index was calculated as the formula of ln [fasting triglyceride (mg/dl) × fasting plasma glucose (mg/dl)/2]. We used logistic regression analysis and mediation analysis to investigate the relations of TyG index with CSVD and cognitive function. RESULTS Multivariate binary logistic regression analysis showed that increased TyG index (OR: 2.241; 95% Confidence Interval(CI): 1.439 to 3.490; P <0.001), or severe CSVD burden (OR: 2.198; 95% CI: 1.283 to 3.763; P = 0.004) was associated with an increased risk of cognitive impairment in elderly patients with T2DM after adjusting for potential confounders. In addition, TyG index was an independent risk factor of severe CSVD burden (OR: 1.472; 95% CI: 1.003 to 2.160; P = 0.048) after controlling for potential confounders. Compared with the lowest TyG index tertile, the multivariable-adjusted OR of the highest tertile was 3.298 (95% CI: 1.685 to 6.452; P for trend <0.001) for cognitive impairment, 1.933 (95% CI: 1.010 to 3.698; P for trend = 0.047) for severe CSVD burden. Mediation analysis found a significant moderating effect of the severe CSVD burden on the association between higher TyG index levels and cognitive impairment. CONCLUSIONS The increased TyG index is an independent risk factor for cognitive impairment and severe CSVD burden in clinical practice. A proportion of the effect of increased TyG index on cognitive impairment may be due to the aggravation of CSVD burden.
Collapse
Affiliation(s)
- Zhenjie Teng
- Department of Neurology, Hebei Medical University, Shijiazhuang, China
- Department of Neurology, Hebei General Hospital, Shijiazhuang, China
| | - Jing Feng
- Department of Endocrinology, Hebei General Hospital, Shijiazhuang, China
| | - Yanhong Dong
- Department of Neurology, Hebei General Hospital, Shijiazhuang, China
| | - Jing Xu
- Department of Neurology, Hebei General Hospital, Shijiazhuang, China
| | - Xin Jiang
- Department of Neurology, Hebei General Hospital, Shijiazhuang, China
| | - Huifang Chen
- Department of Neurology, Hebei General Hospital, Shijiazhuang, China
| | - Qianqian Qi
- Department of Neurology, Hebei Medical University, Shijiazhuang, China
- Department of Neurology, Hebei General Hospital, Shijiazhuang, China
| | - Rui Li
- Department of Neurology, Hebei General Hospital, Shijiazhuang, China
| | - Weihong Chen
- Department of Neurology, Hebei General Hospital, Shijiazhuang, China
| | - Peiyuan Lv
- Department of Neurology, Hebei Medical University, Shijiazhuang, China
- Department of Neurology, Hebei General Hospital, Shijiazhuang, China
- *Correspondence: Peiyuan Lv,
| |
Collapse
|
42
|
Barone E, Di Domenico F, Perluigi M, Butterfield DA. The interplay among oxidative stress, brain insulin resistance and AMPK dysfunction contribute to neurodegeneration in type 2 diabetes and Alzheimer disease. Free Radic Biol Med 2021; 176:16-33. [PMID: 34530075 PMCID: PMC8595768 DOI: 10.1016/j.freeradbiomed.2021.09.006] [Citation(s) in RCA: 73] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 08/31/2021] [Accepted: 09/09/2021] [Indexed: 02/08/2023]
Abstract
Alzheimer's disease (AD) is the most common form of dementia in the elderly followed by vascular dementia. In addition to clinically diagnosed dementia, cognitive dysfunction has been reported in diabetic patients. Recent studies are now beginning to recognize type 2 diabetes mellitus (T2DM), characterized by chronic hyperglycemia and insulin resistance, as a risk factor for AD and other cognitive disorders. While studies on insulin action have remained traditionally in the domain of peripheral tissues, the detrimental effects of insulin resistance in the central nervous system on cognitive dysfunction are increasingly being reported in recent clinical and preclinical studies. Brain functions require continuous supply of glucose and oxygen and a tight regulation of metabolic processes. Loss of this metabolic regulation has been proposed to be a contributor to memory dysfunction associated with neurodegeneration. Within the above scenario, this review will focus on the interplay among oxidative stress (OS), insulin resistance and AMPK dysfunctions in the brain by highlighting how these neurotoxic events contribute to neurodegeneration. We provide an overview on the detrimental effects of OS on proteins regulating insulin signaling and how these alterations impact cell metabolic dysfunctions through AMPK dysregulation. Such processes, we assert, are critically involved in the molecular pathways that underlie AD.
Collapse
Affiliation(s)
- Eugenio Barone
- Department of Biochemical Sciences "A. Rossi-Fanelli", Sapienza University of Rome, Piazzale A. Moro 5, 00185, Roma, Italy
| | - Fabio Di Domenico
- Department of Biochemical Sciences "A. Rossi-Fanelli", Sapienza University of Rome, Piazzale A. Moro 5, 00185, Roma, Italy
| | - Marzia Perluigi
- Department of Biochemical Sciences "A. Rossi-Fanelli", Sapienza University of Rome, Piazzale A. Moro 5, 00185, Roma, Italy
| | - D Allan Butterfield
- Department of Chemistry and Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, 40506-0055, USA.
| |
Collapse
|
43
|
Du X, Zhang Y, Zhao Q, Qin W, Ma G, Fu J, Zhang Q. Effects of INSR genetic polymorphism on hippocampal volume and episodic memory in chinese type 2 diabetes. Acta Diabetol 2021; 58:1471-1480. [PMID: 34085146 DOI: 10.1007/s00592-021-01750-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Accepted: 05/26/2021] [Indexed: 11/28/2022]
Abstract
AIMS We aimed to investigate the effect of the type 2 diabetes-specific insulin/IGF signaling genetic variants on the hippocampal volume and their relationships with episodic memory in Chinese patients with type 2 diabetes. METHODS Analysis of variance was used to evaluate the genotype-by-diagnosis interaction effect on hippocampal volume in Chinese participants (109 patients with type 2 diabetes, 116 healthy controls). Mediation analysis was performed to test whether the hippocampal volume would mediate the association between genotype and episodic memory in patients with type 2 diabetes. RESULTS INSR (rs8101064) exhibited a significant genotype-by-diagnosis interaction effect on the bilateral hippocampal volumes (left, P = 0.020; right, P = 0.004, PFDR < 0.05). The T allele carriers exhibited smaller bilateral hippocampal volumes than the CC homozygotes in patients with type 2 diabetes (left, P = 0.004; right, P = 0.002). Mediation analysis revealed the significant mediation effect of the left hippocampal volume on the association between INSR (rs8101064) genetic polymorphism and the short- and long-term memory scores in patients with type 2 diabetes (short-term memory: 95% CI, -2.716, -0.266; long-term memory: 95% CI, -0.823, -0.103). CONCLUSIONS Hyperglycemia exposure and INSR (rs8101064) genetic polymorphism had an interaction effect on the hippocampal volume, and the T allele of the INSR (rs8101064) may serve as a risk factor for the decreased hippocampal volume in Chinese patients with type 2 diabetes. The left hippocampal volume mediated the effect of INSR (rs8101064) genetic polymorphism on episodic memory in Chinese patients with type 2 diabetes, which provided a biological pathway for understanding how the INSR (rs8101064) genetic polymorphism affects episodic memory in type 2 diabetes.
Collapse
Affiliation(s)
- Xin Du
- Department of Medical imaging and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, No. 154, Anshan Road, Heping District, Tianjin, 300052, China
| | - Yang Zhang
- Department of Medical imaging and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, No. 154, Anshan Road, Heping District, Tianjin, 300052, China
| | - Qiuyue Zhao
- Department of Medical imaging and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, No. 154, Anshan Road, Heping District, Tianjin, 300052, China
| | - Wen Qin
- Department of Medical imaging and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, No. 154, Anshan Road, Heping District, Tianjin, 300052, China
| | - Guangyang Ma
- Department of Radiology, Tianjin Medical University Metabolic Diseases Hospital, Tianjin, 300060, China
| | - Jilian Fu
- Department of Medical imaging and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, No. 154, Anshan Road, Heping District, Tianjin, 300052, China
| | - Quan Zhang
- Department of Medical imaging and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, No. 154, Anshan Road, Heping District, Tianjin, 300052, China.
| |
Collapse
|
44
|
Nitchingham A, Milne A, Toson B, Tuch B, Agar M, Close J, Caplan G. Intranasal insulin for treatment of delirium in older hospitalised patients: study protocol for a randomised controlled trial. BMJ Open 2021; 11:e050765. [PMID: 34667006 PMCID: PMC8527126 DOI: 10.1136/bmjopen-2021-050765] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
INTRODUCTION Delirium is one of the most common conditions diagnosed in hospitalised older people and is associated with numerous adverse outcomes, yet there are no proven pharmacological treatments. Recent research has identified cerebral glucose hypometabolism as a pathophysiological mechanism offering a therapeutic target in delirium. Insulin, delivered via the intranasal route, acts directly on the central nervous system and has been shown to enhance cerebral metabolism and improve cognition in patients with mild cognitive impairment and dementia. This trial will determine whether intranasal insulin can reduce the duration of delirium in older hospitalised patients. METHODS AND ANALYSIS This is a prospective randomised, placebo-controlled, double-blind study with 6 months follow-up. One hundred patients aged 65 years or older presenting to hospital with delirium admitted under geriatric medicine will be recruited. Participants will be randomised to intranasal insulin detemir or placebo administered twice daily until delirium resolves, defined as Confusion Assessment Method (CAM) negative for 2 days, or discharge from hospital. The primary outcome measure will be duration of delirium using the CAM. Secondary outcome measures will include length of hospital stay, severity of delirium, adherence to treatment, hospital complications, new admission to nursing home, mortality, use of antipsychotic medications during hospital stay and cognitive and physical function at 6 months postdischarge. ETHICS AND DISSEMINATION This trial has been approved by the South Eastern Sydney Human Research and Ethics Committee. Dissemination plans include submission to a peer-reviewed journal for publication and presentation at scientific conferences. TRIAL REGISTRATION NUMBER ACTRN12618000318280.
Collapse
Affiliation(s)
- Anita Nitchingham
- Department of Geriatric Medicine, Prince of Wales Hospital, Sydney, New South Wales, Australia
- Prince of Wales Clinical School, University of New South Wales, Sydney, New South Wales, Australia
| | - Andrew Milne
- Rural Clinical School, Coffs Harbour Health Campus, University of New South Wales, Coffs Harbour, New South Wales, Australia
| | - Barbara Toson
- Flinders Centre for Epidemiology and Biostatistics, Flinders University, Adelaide, South Australia, Australia
| | - Bernard Tuch
- Department of Molecular & Translational Science, Hudson Institute, Monash University, Melbourne, Victoria, Australia
| | - Meera Agar
- Faculty of Health, University of Technology Sydney, Sydney, New South Wales, Australia
| | - Jacqueline Close
- Department of Geriatric Medicine, Prince of Wales Hospital, Sydney, New South Wales, Australia
- Falls, Balance and Injury Research Centre, Neuroscience Research Australia, Randwick, New South Wales, Australia
| | - Gideon Caplan
- Department of Geriatric Medicine, Prince of Wales Hospital, Sydney, New South Wales, Australia
- Prince of Wales Clinical School, University of New South Wales, Sydney, New South Wales, Australia
| |
Collapse
|
45
|
Chen Y, Zhou Z, Liang Y, Tan X, Li Y, Qin C, Feng Y, Ma X, Mo Z, Xia J, Zhang H, Qiu S, Shen D. Classification of type 2 diabetes mellitus with or without cognitive impairment from healthy controls using high-order functional connectivity. Hum Brain Mapp 2021; 42:4671-4684. [PMID: 34213081 PMCID: PMC8410559 DOI: 10.1002/hbm.25575] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 06/03/2021] [Accepted: 06/08/2021] [Indexed: 12/12/2022] Open
Abstract
Type 2 diabetes mellitus (T2DM) is associated with cognitive impairment and may progress to dementia. However, the brain functional mechanism of T2DM-related dementia is still less understood. Recent resting-state functional magnetic resonance imaging functional connectivity (FC) studies have proved its potential value in the study of T2DM with cognitive impairment (T2DM-CI). However, they mainly used a mass-univariate statistical analysis that was not suitable to reveal the altered FC "pattern" in T2DM-CI, due to lower sensitivity. In this study, we proposed to use high-order FC to reveal the abnormal connectomics pattern in T2DM-CI with a multivariate, machine learning-based strategy. We also investigated whether such patterns were different between T2DM-CI and T2DM without cognitive impairment (T2DM-noCI) to better understand T2DM-induced cognitive impairment, on 23 T2DM-CI and 27 T2DM-noCI patients, as well as 50 healthy controls (HCs). We first built the large-scale high-order brain networks based on temporal synchronization of the dynamic FC time series among multiple brain region pairs and then used this information to classify the T2DM-CI (as well as T2DM-noCI) from the matched HC based on support vector machine. Our model achieved an accuracy of 79.17% in T2DM-CI versus HC differentiation, but only 59.62% in T2DM-noCI versus HC classification. We found abnormal high-order FC patterns in T2DM-CI compared to HC, which was different from that in T2DM-noCI. Our study indicates that there could be widespread connectivity alterations underlying the T2DM-induced cognitive impairment. The results help to better understand the changes in the central neural system due to T2DM.
Collapse
Affiliation(s)
- Yuna Chen
- The First School of Clinical MedicineGuangzhou University of Chinese MedicineGuangzhouGuangdongChina
- Department of Radiology and BRICUniversity of North Carolina at Chapel HillChapel HillNorth CarolinaUSA
| | - Zhen Zhou
- Department of Radiology and BRICUniversity of North Carolina at Chapel HillChapel HillNorth CarolinaUSA
| | - Yi Liang
- Department of RadiologyThe First Affiliated Hospital of Guangzhou University of Chinese MedicineGuangzhouGuangdongChina
| | - Xin Tan
- Department of RadiologyThe First Affiliated Hospital of Guangzhou University of Chinese MedicineGuangzhouGuangdongChina
| | - Yifan Li
- The First School of Clinical MedicineGuangzhou University of Chinese MedicineGuangzhouGuangdongChina
| | - Chunhong Qin
- Department of RadiologyThe First Affiliated Hospital of Guangzhou University of Chinese MedicineGuangzhouGuangdongChina
| | - Yue Feng
- The First School of Clinical MedicineGuangzhou University of Chinese MedicineGuangzhouGuangdongChina
| | - Xiaomeng Ma
- The First School of Clinical MedicineGuangzhou University of Chinese MedicineGuangzhouGuangdongChina
| | - Zhanhao Mo
- Department of Radiology and BRICUniversity of North Carolina at Chapel HillChapel HillNorth CarolinaUSA
- Department of RadiologyChina‐Japan Union Hospital of Jilin UniversityChangchunJilinChina
| | - Jing Xia
- Institute of Brain‐Intelligence Technology, Zhangjiang LabShanghaiChina
| | - Han Zhang
- Institute of Brain‐Intelligence Technology, Zhangjiang LabShanghaiChina
| | - Shijun Qiu
- Department of RadiologyThe First Affiliated Hospital of Guangzhou University of Chinese MedicineGuangzhouGuangdongChina
| | - Dinggang Shen
- School of Biomedical EngineeringShanghaiTech UniversityShanghaiChina
- Shanghai United Imaging Intelligence Co., Ltd.ShanghaiChina
- Department of Artificial IntelligenceKorea UniversitySeoulRepublic of Korea
| |
Collapse
|
46
|
Relation between Exogenous Insulin and Cognitive Function in Type 2 Diabetes Mellitus. MEDICINA-LITHUANIA 2021; 57:medicina57090943. [PMID: 34577866 PMCID: PMC8466467 DOI: 10.3390/medicina57090943] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 09/02/2021] [Accepted: 09/03/2021] [Indexed: 11/17/2022]
Abstract
Background and objectives: Although the role of insulin in the periphery is well understood, not as much is known about its multifactorial role in the brain. The aim of this study is to determine whether exogenous insulin, evaluated by daily insulin requirement, has an impact on mild cognitive impairment (MCI), and whether this relationship is mediated by insulin doses and other risk factors. Materials and methods: A sample of 100 participants with type 2 diabetes aged 40 and over was divided into case and control groups, according to their insulin requirement. Patients with an insulin requirement >1 IU/kg/day were assessed as the case group whereas those with an insulin dose <1 IU/kg were used as the control group. All participants underwent cognitive testing using MoCA questionnaire scoring and blood analysis to determine lipid and uric acid levels in plasma. Subjects were categorized as having normal cognitive function or MCI. Results: Results showed that the prevalence of MCI in Lithuanian elderly diabetic patients was high in the groups with a normal insulin requirement or high insulin requirement at 84.8% and 72.5%, respectively (p = 0.14). Age (p = 0.001) and insulin dose (p < 0.0001) were related to the MCI. Using ROC curve analysis, the highest rate risk of MCI occurred when the insulin dose was lower than 144 IU/d. Conclusions: In summary, the results of this study provided evidence that increased exogenous insulin supply improves cognitive function. Higher insulin dose (>144 IU/d) demonstrated a positive effect on cognitive function, especially in individuals with poorly controlled diabetes (HbA1c ≥ 9%). Finally, the prevalence of MCI in the T2DM population was found to be very high. Future research is needed to determine whether high exogenous insulin doses have a protective effect on MCI.
Collapse
|
47
|
Scherer T, Sakamoto K, Buettner C. Brain insulin signalling in metabolic homeostasis and disease. Nat Rev Endocrinol 2021; 17:468-483. [PMID: 34108679 DOI: 10.1038/s41574-021-00498-x] [Citation(s) in RCA: 80] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/22/2021] [Indexed: 02/06/2023]
Abstract
Insulin signalling in the central nervous system regulates energy homeostasis by controlling metabolism in several organs and by coordinating organ crosstalk. Studies performed in rodents, non-human primates and humans over more than five decades using intracerebroventricular, direct hypothalamic or intranasal application of insulin provide evidence that brain insulin action might reduce food intake and, more importantly, regulates energy homeostasis by orchestrating nutrient partitioning. This Review discusses the metabolic pathways that are under the control of brain insulin action and explains how brain insulin resistance contributes to metabolic disease in obesity, the metabolic syndrome and type 2 diabetes mellitus.
Collapse
Affiliation(s)
- Thomas Scherer
- Division of Endocrinology and Metabolism, Department of Medicine III, Medical University of Vienna, Vienna, Austria.
| | - Kenichi Sakamoto
- Division of Endocrinology, Metabolism & Nutrition, Department of Medicine, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| | - Christoph Buettner
- Division of Endocrinology, Metabolism & Nutrition, Department of Medicine, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA.
| |
Collapse
|
48
|
Gaddam M, Singh A, Jain N, Avanthika C, Jhaveri S, De la Hoz I, Sanka S, Goli SR. A Comprehensive Review of Intranasal Insulin and Its Effect on the Cognitive Function of Diabetics. Cureus 2021; 13:e17219. [PMID: 34540446 PMCID: PMC8442633 DOI: 10.7759/cureus.17219] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/16/2021] [Indexed: 12/01/2022] Open
Abstract
Diabetes mellitus continues to be a disease that affects a good percentage of our population. The majority affected need insulin on a day-to-day basis. Before the invention of the first manufactured insulin in 1978, dealing with diabetes took a significant toll on patient's lives. As technology and human innovation prevail, significant advancements have taken place in managing this chronic disease. Patients have an option to decide their mode of insulin delivery. Intranasal insulin, one such form, has a rapid mode of action while effectively controlling postprandial hyperglycemia. It has also been proven to reduce hypoglycemia and insulin resistance problems, which seem to be the main adverse effects of using conventional insulin regularly. However, due to the large dosages needed and high incurring costs, Intranasal Insulin is currently being used as adjunctive therapy along with conventional insulin. We conducted a literature search in PubMed indexed journals using the medical terms "Intranasal insulin," "diabetes," and "cognitive impairment" to provide an overview of the mechanism of action of Intranasal Insulin, its distinctive cognitive benefits, and how it can be compared to the standard parenteral insulin therapy. One unique feature of intranasal insulin is its ability to directly affect the central nervous system, bypassing the blood-brain barrier. Not only does this help in reducing the peripheral side effects of insulin, but it has also proven to play a role in improving the cognitive function of diabetics, especially those who have Alzheimer's or mild cognitive impairment, as decreased levels of insulin in the brain has been shown to impact cognitive function negatively. However, it does come with its limitations of poor absorption through the nasal mucosa due to mucociliary clearance and proteolytic enzymes, our body's natural defence mechanisms. This review focuses on the efficacy of intranasal insulin, its potential benefits, limitations, and role in cognitive improvement in people with diabetes with pre-existing cognitive impairment.
Collapse
Affiliation(s)
| | | | - Nidhi Jain
- Internal Medicine, Sir Ganga Ram Hospital, New Delhi, IND
| | | | - Sharan Jhaveri
- Internal Medicine, Smt. Nathiba Hargovandas Lakhmichand Municipal Medical College, Ahmedabad, IND
| | | | - Sujana Sanka
- Internal Medicine, JC Medical Center, Orlando, USA
| | - Sri Rupa Goli
- Internal Medicine, Shri Sathya Sai Medical College and Research Institute, Chennai, IND
| |
Collapse
|
49
|
Intranasal insulin rescues repeated anesthesia-induced deficits in synaptic plasticity and memory and prevents apoptosis in neonatal mice via mTORC1. Sci Rep 2021; 11:15490. [PMID: 34326413 PMCID: PMC8322102 DOI: 10.1038/s41598-021-94849-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Accepted: 07/16/2021] [Indexed: 02/06/2023] Open
Abstract
Long-lasting cognitive impairment in juveniles undergoing repeated general anesthesia has been observed in numerous preclinical and clinical studies, yet, the underlying mechanisms remain unknown and no preventive treatment is available. We found that daily intranasal insulin administration to juvenile mice for 7 days prior to repeated isoflurane anesthesia rescues deficits in hippocampus-dependent memory and synaptic plasticity in adulthood. Moreover, intranasal insulin prevented anesthesia-induced apoptosis of hippocampal cells, which is thought to underlie cognitive impairment. Inhibition of the mechanistic target of rapamycin complex 1 (mTORC1), a major intracellular effector of insulin receptor, blocked the beneficial effects of intranasal insulin on anesthesia-induced apoptosis. Consistent with this finding, mice lacking mTORC1 downstream translational repressor 4E-BP2 showed no induction of repeated anesthesia-induced apoptosis. Our study demonstrates that intranasal insulin prevents general anesthesia-induced apoptosis of hippocampal cells, and deficits in synaptic plasticity and memory, and suggests that the rescue effect is mediated via mTORC1/4E-BP2 signaling.
Collapse
|
50
|
Yao L, Yang C, Zhang W, Li S, Li Q, Chen L, Lui S, Kemp GJ, Biswal BB, Shah NJ, Li F, Gong Q. A multimodal meta-analysis of regional structural and functional brain alterations in type 2 diabetes. Front Neuroendocrinol 2021; 62:100915. [PMID: 33862036 DOI: 10.1016/j.yfrne.2021.100915] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 03/30/2021] [Accepted: 04/11/2021] [Indexed: 02/04/2023]
Abstract
Neuroimaging studies have identified brain structural and functional alterations of type 2 diabetes mellitus (T2DM) patients; however, there is no systematic information on the relations between abnormalities in these two domains. We conducted a multimodal meta-analysis of voxel-based morphometry and regional resting-state functional MRI studies in T2DM, including fifteen structural datasets (693 patients and 684 controls) and sixteen functional datasets (378 patients and 358 controls). We found, in patients with T2DM compared to controls, conjoint decreased regional gray matter volume (GMV) and altered intrinsic activity mainly in the default mode network including bilateral superior temporal gyrus/Rolandic operculum, left middle and inferior temporal gyrus, and left supramarginal gyrus; decreased GMV alone in the limbic system; and functional abnormalities alone in the cerebellum, insula, and visual cortex. This meta-analysis identified complicated patterns of conjoint and dissociated brain alterations in T2DM patients, which may help provide new insight into the neuropathology of T2DM.
Collapse
Affiliation(s)
- Li Yao
- Huaxi MR Research Center (HMRRC), Department of Radiology, West China Hospital of Sichuan University, NO. 37 Guoxue Xiang, Chengdu 610041, China; Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, NO. 37 Guoxue Xiang, Chengdu 610041, China; Functional and Molecular Imaging Key Laboratory of Sichuan University, NO. 37 Guoxue Xiang, Chengdu 610041, China
| | - Chengmin Yang
- Huaxi MR Research Center (HMRRC), Department of Radiology, West China Hospital of Sichuan University, NO. 37 Guoxue Xiang, Chengdu 610041, China; Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, NO. 37 Guoxue Xiang, Chengdu 610041, China; Functional and Molecular Imaging Key Laboratory of Sichuan University, NO. 37 Guoxue Xiang, Chengdu 610041, China
| | - Wenjing Zhang
- Huaxi MR Research Center (HMRRC), Department of Radiology, West China Hospital of Sichuan University, NO. 37 Guoxue Xiang, Chengdu 610041, China; Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, NO. 37 Guoxue Xiang, Chengdu 610041, China; Functional and Molecular Imaging Key Laboratory of Sichuan University, NO. 37 Guoxue Xiang, Chengdu 610041, China
| | - Siyi Li
- Huaxi MR Research Center (HMRRC), Department of Radiology, West China Hospital of Sichuan University, NO. 37 Guoxue Xiang, Chengdu 610041, China; Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, NO. 37 Guoxue Xiang, Chengdu 610041, China; Functional and Molecular Imaging Key Laboratory of Sichuan University, NO. 37 Guoxue Xiang, Chengdu 610041, China
| | - Qian Li
- Huaxi MR Research Center (HMRRC), Department of Radiology, West China Hospital of Sichuan University, NO. 37 Guoxue Xiang, Chengdu 610041, China; Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, NO. 37 Guoxue Xiang, Chengdu 610041, China; Functional and Molecular Imaging Key Laboratory of Sichuan University, NO. 37 Guoxue Xiang, Chengdu 610041, China
| | - Lizhou Chen
- Huaxi MR Research Center (HMRRC), Department of Radiology, West China Hospital of Sichuan University, NO. 37 Guoxue Xiang, Chengdu 610041, China; Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, NO. 37 Guoxue Xiang, Chengdu 610041, China; Functional and Molecular Imaging Key Laboratory of Sichuan University, NO. 37 Guoxue Xiang, Chengdu 610041, China
| | - Su Lui
- Huaxi MR Research Center (HMRRC), Department of Radiology, West China Hospital of Sichuan University, NO. 37 Guoxue Xiang, Chengdu 610041, China; Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, NO. 37 Guoxue Xiang, Chengdu 610041, China; Functional and Molecular Imaging Key Laboratory of Sichuan University, NO. 37 Guoxue Xiang, Chengdu 610041, China
| | - Graham J Kemp
- Liverpool Magnetic Resonance Imaging Centre (LiMRIC) and Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool L69 3BX, L3 5TR, United Kingdom
| | - Bharat B Biswal
- Department of Biomedical Engineering, New Jersey Institute of Technology, 323 Dr Martin Luther King Jr Blvd, Newark, NJ 07102, USA; The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Laboratory for Neuroinformation, University of Electronic Science and Technology of China, No.4, Section 2, North Jianshe Road, Chengdu 610054, China
| | - Nadim J Shah
- Institute of Neuroscience and Medicine (INM-4), Medical Imaging Physics, Forschungszentrum Jülich GmbH, 52425 Jülich, Germany
| | - Fei Li
- Huaxi MR Research Center (HMRRC), Department of Radiology, West China Hospital of Sichuan University, NO. 37 Guoxue Xiang, Chengdu 610041, China; Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, NO. 37 Guoxue Xiang, Chengdu 610041, China; Functional and Molecular Imaging Key Laboratory of Sichuan University, NO. 37 Guoxue Xiang, Chengdu 610041, China; Institute of Neuroscience and Medicine (INM-4), Medical Imaging Physics, Forschungszentrum Jülich GmbH, 52425 Jülich, Germany.
| | - Qiyong Gong
- Huaxi MR Research Center (HMRRC), Department of Radiology, West China Hospital of Sichuan University, NO. 37 Guoxue Xiang, Chengdu 610041, China; Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, NO. 37 Guoxue Xiang, Chengdu 610041, China; Functional and Molecular Imaging Key Laboratory of Sichuan University, NO. 37 Guoxue Xiang, Chengdu 610041, China.
| |
Collapse
|