1
|
de la Monte SM, Tong M. Dysregulated mTOR networks in experimental sporadic Alzheimer's disease. Front Cell Neurosci 2024; 18:1432359. [PMID: 39386180 PMCID: PMC11461251 DOI: 10.3389/fncel.2024.1432359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Accepted: 08/29/2024] [Indexed: 10/12/2024] Open
Abstract
Background Beyond the signature amyloid-beta plaques and neurofibrillary tangles, Alzheimer's disease (AD) has been shown to exhibit dysregulated metabolic signaling through insulin and insulin-like growth factor (IGF) networks that crosstalk with the mechanistic target of rapamycin (mTOR). Its broad impact on brain structure and function suggests that mTOR is likely an important therapeutic target for AD. Objective This study characterizes temporal lobe (TL) mTOR signaling abnormalities in a rat model of sporadic AD neurodegeneration. Methods Long Evans rats were given intracerebroventricular injections of streptozotocin (ic-STZ) or saline (control), and 4 weeks later, they were administered neurobehavioral tests followed by terminal harvesting of the TLs for histopathological study and measurement of AD biomarkers, neuroinflammatory/oxidative stress markers, and total and phosphorylated insulin/IGF-1-Akt-mTOR pathway signaling molecules. Results Rats treated with ic-STZ exhibited significantly impaired performance on Rotarod (RR) and Morris Water Maze (MWM) tests, brain atrophy, TL and hippocampal neuronal and white matter degeneration, and elevated TL pTau, AβPP, Aβ, AChE, 4-HNE, and GAPDH and reduced ubiquitin, IL-2, IL-6, and IFN-γ immunoreactivities. In addition, ic-STZ reduced TL pY1135/1136-IGF-1R, Akt, PTEN, pS380-PTEN, pS2448-mTOR, p70S6K, pT412-p70S6K, p/T-pT412-p70S6K, p/T-Rictor, and p/T-Raptor. Conclusion Experimental ic-STZ-induced sporadic AD-type neurodegeneration with neurobehavioral dysfunctions associated with inhibition of mTOR signaling networks linked to energy metabolism, plasticity, and white matter integrity.
Collapse
Affiliation(s)
- Suzanne M. de la Monte
- Departments of Medicine, Pathology and Laboratory Medicine, Neurology, and Neurosurgery, Rhode Island Hospital, Women and Infants Hospital, The Alpert Medical School at Brown University, Providence, RI, United States
| | - Ming Tong
- Department of Medicine, Rhode Island Hospital, The Alpert Medical School at Brown University, Providence, RI, United States
| |
Collapse
|
2
|
Barry CV, Chrysanthopoulou SA, Tallo V, Jarilla B, Vargas Z, McDonald E, Gundogan F, Friedman JF. The Impact of Prenatal Alcohol Exposure on Longitudinal Growth, Nutritional Status, and Insulin-Like Growth Factor 1 in Early Childhood in Leyte, the Philippines. J Pediatr 2024; 269:113977. [PMID: 38401788 PMCID: PMC11096041 DOI: 10.1016/j.jpeds.2024.113977] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 01/24/2024] [Accepted: 02/16/2024] [Indexed: 02/26/2024]
Abstract
OBJECTIVE To assess the impact and potential mechanistic pathways of prenatal alcohol exposure (PAE) on longitudinal growth and nutritional status in early childhood. STUDY DESIGN A cohort of 296 mother-infant dyads (32% with PAE vs 68% unexposed) were recruited in Leyte, the Philippines, and followed from early gestation through 24 months of age. PAE was assessed using serum phosphatidylethanol (PEth) captured twice prenatally and in cord blood and supplemented with self-reported alcohol consumption. Linear mixed models were used to examine longitudinal effects of PAE on growth from birth through 2 years including key potential mediating factors (placental histopathology, and infant serum leptin and Insulin-like Growth Factor 1 [IGF-1]). RESULTS After adjusting for potential confounders, we found that PAE was significantly associated with a delayed blunting of linear growth trajectories (height-for-age z-score, body length) and weight (weight-for-age z-score, body weight) that manifested between 4 and 6 months and continued through 12-24 months. PAE was also associated with a decreased rate of mid-upper-arm circumference growth from birth to 12 months, and a lower mean IGF-1 levels at birth and 6 months. CONCLUSION This study demonstrates a delayed impact of PAE on growth that manifested around 6 months of age, underscoring the importance of routine clinical monitoring in early childhood. Furthermore, the findings supported prior animal model findings that suggest a mechanistic role for IGF-1 in PAE-induced growth delay.
Collapse
Affiliation(s)
- Christopher V Barry
- Warren Alpert Medical School of Brown University, Providence, RI; Department of Epidemiology, Brown University School of Public Health, Providence, RI
| | | | - Veronica Tallo
- Research Institute for Tropical Medicine, Manila, Philippines
| | - Blanca Jarilla
- Research Institute for Tropical Medicine, Manila, Philippines
| | - Zorimel Vargas
- Center for International Health Research, Rhode Island Hospital, Warren Alpert Medical School of Brown University, Providence, RI
| | - Emily McDonald
- Center for International Health Research, Rhode Island Hospital, Warren Alpert Medical School of Brown University, Providence, RI; Department of Pediatrics, Warren Alpert Medical School of Brown University, Providence, RI
| | - Fusun Gundogan
- Department of Pathology, Warren Alpert Medical School of Brown University, Providence, RI; Women and Infants' Hospital, Care New England, Providence, RI
| | - Jennifer F Friedman
- Warren Alpert Medical School of Brown University, Providence, RI; Department of Epidemiology, Brown University School of Public Health, Providence, RI; Center for International Health Research, Rhode Island Hospital, Warren Alpert Medical School of Brown University, Providence, RI; Department of Pediatrics, Warren Alpert Medical School of Brown University, Providence, RI
| |
Collapse
|
3
|
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
|
4
|
BHUSARE NILAM, KUMAR MAUSHMI. A review on potential heterocycles for the treatment of glioblastoma targeting receptor tyrosine kinases. Oncol Res 2024; 32:849-875. [PMID: 38686058 PMCID: PMC11055995 DOI: 10.32604/or.2024.047042] [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: 10/23/2023] [Accepted: 01/10/2024] [Indexed: 05/02/2024] Open
Abstract
Glioblastoma, the most aggressive form of brain tumor, poses significant challenges in terms of treatment success and patient survival. Current treatment modalities for glioblastoma include radiation therapy, surgical intervention, and chemotherapy. Unfortunately, the median survival rate remains dishearteningly low at 12-15 months. One of the major obstacles in treating glioblastoma is the recurrence of tumors, making chemotherapy the primary approach for secondary glioma patients. However, the efficacy of drugs is hampered by the presence of the blood-brain barrier and multidrug resistance mechanisms. Consequently, considerable research efforts have been directed toward understanding the underlying signaling pathways involved in glioma and developing targeted drugs. To tackle glioma, numerous studies have examined kinase-downstream signaling pathways such as RAS-RAF-MEK-ERK-MPAK. By targeting specific signaling pathways, heterocyclic compounds have demonstrated efficacy in glioma therapeutics. Additionally, key kinases including phosphatidylinositol 3-kinase (PI3K), serine/threonine kinase, cytoplasmic tyrosine kinase (CTK), receptor tyrosine kinase (RTK) and lipid kinase (LK) have been considered for investigation. These pathways play crucial roles in drug effectiveness in glioma treatment. Heterocyclic compounds, encompassing pyrimidine, thiazole, quinazoline, imidazole, indole, acridone, triazine, and other derivatives, have shown promising results in targeting these pathways. As part of this review, we propose exploring novel structures with low toxicity and high potency for glioma treatment. The development of these compounds should strive to overcome multidrug resistance mechanisms and efficiently penetrate the blood-brain barrier. By optimizing the chemical properties and designing compounds with enhanced drug-like characteristics, we can maximize their therapeutic value and minimize adverse effects. Considering the complex nature of glioblastoma, these novel structures should be rigorously tested and evaluated for their efficacy and safety profiles.
Collapse
Affiliation(s)
- NILAM BHUSARE
- Somaiya Institute for Research & Consultancy, Somaiya Vidyavihar University, Vidyavihar (East), Mumbai, 400077, India
| | - MAUSHMI KUMAR
- Somaiya Institute for Research & Consultancy, Somaiya Vidyavihar University, Vidyavihar (East), Mumbai, 400077, India
| |
Collapse
|
5
|
Pan Y, Li J, Lin P, Wan L, Qu Y, Cao L, Wang L. A review of the mechanisms of abnormal ceramide metabolism in type 2 diabetes mellitus, Alzheimer's disease, and their co-morbidities. Front Pharmacol 2024; 15:1348410. [PMID: 38379904 PMCID: PMC10877008 DOI: 10.3389/fphar.2024.1348410] [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: 12/02/2023] [Accepted: 01/18/2024] [Indexed: 02/22/2024] Open
Abstract
The global prevalence of type 2 diabetes mellitus (T2DM) and Alzheimer's disease (AD) is rapidly increasing, revealing a strong association between these two diseases. Currently, there are no curative medication available for the comorbidity of T2DM and AD. Ceramides are structural components of cell membrane lipids and act as signal molecules regulating cell homeostasis. Their synthesis and degradation play crucial roles in maintaining metabolic balance in vivo, serving as important mediators in the development of neurodegenerative and metabolic disorders. Abnormal ceramide metabolism disrupts intracellular signaling, induces oxidative stress, activates inflammatory factors, and impacts glucose and lipid homeostasis in metabolism-related tissues like the liver, skeletal muscle, and adipose tissue, driving the occurrence and progression of T2DM. The connection between changes in ceramide levels in the brain, amyloid β accumulation, and tau hyper-phosphorylation is evident. Additionally, ceramide regulates cell survival and apoptosis through related signaling pathways, actively participating in the occurrence and progression of AD. Regulatory enzymes, their metabolites, and signaling pathways impact core pathological molecular mechanisms shared by T2DM and AD, such as insulin resistance and inflammatory response. Consequently, regulating ceramide metabolism may become a potential therapeutic target and intervention for the comorbidity of T2DM and AD. The paper comprehensively summarizes and discusses the role of ceramide and its metabolites in the pathogenesis of T2DM and AD, as well as the latest progress in the treatment of T2DM with AD.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Lei Wang
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| |
Collapse
|
6
|
de la Monte SM, Tong M. Agent Orange Herbicidal Toxin-Initiation of Alzheimer-Type Neurodegeneration. J Alzheimers Dis 2024; 97:1703-1726. [PMID: 38306038 PMCID: PMC10979462 DOI: 10.3233/jad-230881] [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] [Indexed: 02/03/2024]
Abstract
Background Agent Orange (AO) is a Vietnam War-era herbicide that contains a 1 : 1 ratio of 2,4-dichlorophenoxyacetic acid (2,4-D) and 2,4,5-trichlorophenoxyacetic acid (2,4,5-T). Emerging evidence suggests that AO exposures cause toxic and degenerative pathologies that may increase the risk for Alzheimer's disease (AD). Objective This study investigates the effects of the two main AO constituents on key molecular and biochemical indices of AD-type neurodegeneration. Methods Long Evans rat frontal lobe slice cultures treated with 250μg/ml of 2,4-D, 2,4,5-T, or both (D + T) were evaluated for cytotoxicity, oxidative injury, mitochondrial function, and AD biomarker expression. Results Treatment with the AO constituents caused histopathological changes corresponding to neuronal, white matter, and endothelial cell degeneration, and molecular/biochemical abnormalities indicative of cytotoxic injury, lipid peroxidation, DNA damage, and increased immunoreactivity to activated Caspase 3, glial fibrillary acidic protein, ubiquitin, tau, paired-helical filament phosphorylated tau, AβPP, Aβ, and choline acetyltransferase. Nearly all indices of cellular injury and degeneration were more pronounced in the D + T compared with 2,4-D or 2,4,5-T treated cultures. Conclusions Exposures to AO herbicidal chemicals damage frontal lobe brain tissue with molecular and biochemical abnormalities that mimic pathologies associated with early-stage AD-type neurodegeneration. Additional research is needed to evaluate the long-term effects of AO exposures in relation to aging and progressive neurodegeneration in Vietnam War Veterans.
Collapse
Affiliation(s)
- Suzanne M. de la Monte
- Departments of Pathology and Laboratory Medicine, Neurology, and Neurosurgery, Rhode Island Hospital, Lifespan Academic Institutions, and the Warren Alpert Medical School of Brown University, Providence, RI, USA
- Department of Medicine, Rhode Island Hospital, Lifespan Academic Institutions, and the Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Ming Tong
- Department of Medicine, Rhode Island Hospital, Lifespan Academic Institutions, and the Warren Alpert Medical School of Brown University, Providence, RI, USA
| |
Collapse
|
7
|
Bima A, Eldakhakhny B, Alamoudi AA, Awan Z, Alnami A, Abo-Elkhair SM, Sakr H, Ghoneim FM, Elsamanoudy A. Molecular Study of the Protective Effect of a Low-Carbohydrate, High-Fat Diet against Brain Insulin Resistance in an Animal Model of Metabolic Syndrome. Brain Sci 2023; 13:1383. [PMID: 37891752 PMCID: PMC10605073 DOI: 10.3390/brainsci13101383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 09/25/2023] [Accepted: 09/27/2023] [Indexed: 10/29/2023] Open
Abstract
Brain insulin resistance is linked to metabolic syndrome (MetS). A low-carbohydrate, high-fat (LCHF) diet has been proposed to have a protective effect. Therefore, this study aimed to investigate the brain insulin resistance markers in a rat animal model of MetS and the protective effects of the LCHF diet. Four groups of male rats (10/group) were created. Group I (Control) was fed a regular diet. Groups II-IV were injected with dexamethasone (DEX) to induce MetS. Group II received DEX with a regular diet. Group III (DEX + LCHF) rates were fed a low-carbohydrate, high-fat diet, while Group IV (DEX + HCLF) rats were fed a high-carbohydrate, low-fat (HCLF) diet. At the end of the four-week experiment, HOMA-IR was calculated. Moreover, cerebral gene expression analysis of S-100B, BDNF, TNF-α, IGF-1, IGF-1 R, IGFBP-2, IGFBP-5, Bax, Bcl-2, and caspase-3 was carried out. In the DEX group, rats showed a significant increase in the HOMA-IR and a decrease in the gene expression of IGF-1, IGF-1 R, IGFBP-2, IGFBP-5, BDNF, and Bcl2, with a concomitant rise in S100B, TNF-α, Bax, and caspase-3. The LCHF diet group showed a significantly opposite effect on all parameters. In conclusion, MetS is associated with dysregulated cerebral gene expression of BDNF, S100B, and TNF-α and disturbed IGF-1 signaling, with increased apoptosis and neuroinflammation. Moreover, the LCHF diet showed a protective effect, as evidenced by preservation of the investigated biochemical and molecular parameters.
Collapse
Affiliation(s)
- Abdulhadi Bima
- Clinical Biochemistry Department, Faculty of Medicine, King Abdulaziz University, Jeddah 21465, Saudi Arabia; (A.B.); (B.E.); (A.A.A.); (Z.A.); (A.A.)
| | - Basmah Eldakhakhny
- Clinical Biochemistry Department, Faculty of Medicine, King Abdulaziz University, Jeddah 21465, Saudi Arabia; (A.B.); (B.E.); (A.A.A.); (Z.A.); (A.A.)
- Food, Nutrition, and Lifestyle Research Unit, King Fahd for Medical Research Centre, King Abdulaziz University, Jeddah 21465, Saudi Arabia
| | - Aliaa A. Alamoudi
- Clinical Biochemistry Department, Faculty of Medicine, King Abdulaziz University, Jeddah 21465, Saudi Arabia; (A.B.); (B.E.); (A.A.A.); (Z.A.); (A.A.)
| | - Zuhier Awan
- Clinical Biochemistry Department, Faculty of Medicine, King Abdulaziz University, Jeddah 21465, Saudi Arabia; (A.B.); (B.E.); (A.A.A.); (Z.A.); (A.A.)
| | - Abrar Alnami
- Clinical Biochemistry Department, Faculty of Medicine, King Abdulaziz University, Jeddah 21465, Saudi Arabia; (A.B.); (B.E.); (A.A.A.); (Z.A.); (A.A.)
| | - Salwa Mohamed Abo-Elkhair
- Medical Biochemistry and Molecular Biology Department, Faculty of Medicine, Mansoura University, Mansoura 35516, Egypt;
| | - Hussein Sakr
- Physiology Department, College of Medicine and Health Sciences, Sultan Qaboos University, Muscat 123, Oman;
- Medical Physiology Department, Faculty of Medicine, Mansoura University, Mansoura 35516, Egypt
| | - Fatma Mohamed Ghoneim
- Faculty Development Unit, Physiological Science and Medical Education Department, Fakeeh College for Medical Sciences, Jeddah 23323, Saudi Arabia;
| | - Ayman Elsamanoudy
- Clinical Biochemistry Department, Faculty of Medicine, King Abdulaziz University, Jeddah 21465, Saudi Arabia; (A.B.); (B.E.); (A.A.A.); (Z.A.); (A.A.)
- Medical Biochemistry and Molecular Biology Department, Faculty of Medicine, Mansoura University, Mansoura 35516, Egypt;
| |
Collapse
|
8
|
de la Monte SM, Goel A, Tong M, Delikkaya B. Agent Orange Causes Metabolic Dysfunction and Molecular Pathology Reminiscent of Alzheimer's Disease. J Alzheimers Dis Rep 2023; 7:751-766. [PMID: 37662613 PMCID: PMC10473158 DOI: 10.3233/adr-230046] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Accepted: 06/19/2023] [Indexed: 09/05/2023] Open
Abstract
Background Agent Orange, an herbicide used during the Vietnam War, contains 2,4-dichlorophenoxyacetic acid (2,4-D) and 2,4,5-trichlorophenoxyacetic acid (2,4,5-T). Agent Orange has teratogenic and carcinogenic effects, and population-based studies suggest Agent Orange exposures lead to higher rates of toxic and degenerative pathologies in the peripheral and central nervous system (CNS). Objective This study examines the potential contribution of Agent Orange exposures to neurodegeneration. Methods Human CNS-derived neuroepithelial cells (PNET2) treated with 2,4-D and 2,4,5-T were evaluated for viability, mitochondrial function, and Alzheimer's disease (AD)-related proteins. Results Treatment with 250μg/ml 2,4-D or 2,4,5-T significantly impaired mitochondrial function, caused degenerative morphological changes, and reduced viability in PNET2 cells. Correspondingly, glyceraldehyde-3-phosphate dehydrogenase expression which is insulin-regulated and marks the integrity of carbohydrate metabolism, was significantly inhibited while 4-hydroxy-2-nonenal, a marker of lipid peroxidation, was increased. Tau neuronal cytoskeletal protein was significantly reduced by 2,4,5-T, and relative tau phosphorylation was progressively elevated by 2,4,5-T followed by 2,4-D treatment relative to control. Amyloid-β protein precursor (AβPP) was increased by 2,4,5-T and 2,4-D, and 2,4,5-T caused a statistical trend (0.05 < p<0.10) increase in Aβ. Finally, altered cholinergic function due to 2,4,5-T and 2,4-D exposures was marked by significantly increased choline acetyltransferase and decreased acetylcholinesterase expression, corresponding with responses in early-stage AD. Conclusion Exposures to Agent Orange herbicidal chemicals rapidly damage CNS neurons, initiating a path toward AD-type neurodegeneration. Additional research is needed to understand the permanency of these neuropathologic processes and the added risks of developing AD in Agent Orange-exposed aging Vietnam Veterans.
Collapse
Affiliation(s)
- Suzanne M. de la Monte
- Department of Pathology and Laboratory Medicine, Rhode Island Hospital, Lifespan Academic Institutions, and the Warren Alpert Medical School of Brown University, Providence, RI, USA
- Department of Medicine, Rhode Island Hospital, Lifespan Academic Institutions, and The Warren Alpert Medical School of Brown University, Providence, RI, USA
- Department of Neurology and Neurosurgery, Rhode Island Hospital, Lifespan Academic Institutions, and The Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Anuva Goel
- Department of Neuroscience, Brown University, Providence, RI, USA
| | - Ming Tong
- Department of Medicine, Rhode Island Hospital, Lifespan Academic Institutions, and The Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Busra Delikkaya
- Department of Pathology and Laboratory Medicine, Rhode Island Hospital, Lifespan Academic Institutions, and the Warren Alpert Medical School of Brown University, Providence, RI, USA
| |
Collapse
|
9
|
de la Monte SM, Tong M, Delikkaya B. Differential Early Mechanistic Frontal Lobe Responses to Choline Chloride and Soy Isoflavones in an Experimental Model of Fetal Alcohol Spectrum Disorder. Int J Mol Sci 2023; 24:7595. [PMID: 37108779 PMCID: PMC10145811 DOI: 10.3390/ijms24087595] [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: 02/26/2023] [Revised: 04/07/2023] [Accepted: 04/18/2023] [Indexed: 04/29/2023] Open
Abstract
Fetal alcohol spectrum disorder (FASD) is the most common preventable cause of neurodevelopmental defects, and white matter is a major target of ethanol neurotoxicity. Therapeutic interventions with choline or dietary soy could potentially supplement public health preventive measures. However, since soy contains abundant choline, it would be important to know if its benefits are mediated by choline or isoflavones. We compared early mechanistic responses to choline and the Daidzein+Genistein (D+G) soy isoflavones in an FASD model using frontal lobe tissue to assess oligodendrocyte function and Akt-mTOR signaling. Long Evans rat pups were binge administered 2 g/Kg of ethanol or saline (control) on postnatal days P3 and P5. P7 frontal lobe slice cultures were treated with vehicle (Veh), Choline chloride (Chol; 75 µM), or D+G (1 µM each) for 72 h without further ethanol exposures. The expression levels of myelin oligodendrocyte proteins and stress-related molecules were measured by duplex enzyme-linked immunosorbent assays (ELISAs), and mTOR signaling proteins and phosphoproteins were assessed using 11-plex magnetic bead-based ELISAs. Ethanol's main short-term effects in Veh-treated cultures were to increase GFAP and relative PTEN phosphorylation and reduce Akt phosphorylation. Chol and D+G significantly modulated the expression of oligodendrocyte myelin proteins and mediators of insulin/IGF-1-Akt-mTOR signaling in both control and ethanol-exposed cultures. In general, the responses were more robust with D+G; the main exception was that RPS6 phosphorylation was significantly increased by Chol and not D+G. The findings suggest that dietary soy, with the benefits of providing complete nutrition together with Choline, could be used to help optimize neurodevelopment in humans at risk for FASD.
Collapse
Affiliation(s)
- Suzanne M. de la Monte
- Departments of Pathology and Laboratory Medicine, Medicine, Neurology and Neurosurgery, Rhode Island Hospital, Lifespan Academic Institutions, The Warren Alpert Medical School of Brown University, Providence, RI 02903, USA
| | | | | |
Collapse
|
10
|
Michailidis M, Moraitou D, Tata DA, Kalinderi K, Papamitsou T, Papaliagkas V. Alzheimer's Disease as Type 3 Diabetes: Common Pathophysiological Mechanisms between Alzheimer's Disease and Type 2 Diabetes. Int J Mol Sci 2022; 23:2687. [PMID: 35269827 PMCID: PMC8910482 DOI: 10.3390/ijms23052687] [Citation(s) in RCA: 93] [Impact Index Per Article: 46.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 02/25/2022] [Accepted: 02/26/2022] [Indexed: 12/27/2022] Open
Abstract
Globally, the incidence of type 2 diabetes mellitus (T2DM) and Alzheimer's disease (AD) epidemics is increasing rapidly and has huge financial and emotional costs. The purpose of the current review article is to discuss the shared pathophysiological connections between AD and T2DM. Research findings are presented to underline the vital role that insulin plays in the brain's neurotransmitters, homeostasis of energy, as well as memory capacity. The findings of this review indicate the existence of a mechanistic interplay between AD pathogenesis with T2DM and, especially, disrupted insulin signaling. AD and T2DM are interlinked with insulin resistance, neuroinflammation, oxidative stress, advanced glycosylation end products (AGEs), mitochondrial dysfunction and metabolic syndrome. Beta-amyloid, tau protein and amylin can accumulate in T2DM and AD brains. Given that the T2DM patients are not routinely evaluated in terms of their cognitive status, they are rarely treated for cognitive impairment. Similarly, AD patients are not routinely evaluated for high levels of insulin or for T2DM. Studies suggesting AD as a metabolic disease caused by insulin resistance in the brain also offer strong support for the hypothesis that AD is a type 3 diabetes.
Collapse
Affiliation(s)
- Michalis Michailidis
- Laboratory of Psychology, School of Psychology, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (M.M.); (D.M.); (D.A.T.)
| | - Despina Moraitou
- Laboratory of Psychology, School of Psychology, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (M.M.); (D.M.); (D.A.T.)
| | - Despina A. Tata
- Laboratory of Psychology, School of Psychology, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (M.M.); (D.M.); (D.A.T.)
| | - Kallirhoe Kalinderi
- Laboratory of Medical Biology-Genetics, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece;
| | - Theodora Papamitsou
- Histology and Embryology Department, Faculty of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece;
| | - Vasileios Papaliagkas
- Department of Biomedical Sciences, School of Health Sciences, International Hellenic University, 57400 Thessaloniki, Greece
| |
Collapse
|
11
|
Insulin and Insulin Resistance in Alzheimer's Disease. Int J Mol Sci 2021; 22:ijms22189987. [PMID: 34576151 PMCID: PMC8472298 DOI: 10.3390/ijms22189987] [Citation(s) in RCA: 117] [Impact Index Per Article: 39.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 09/09/2021] [Accepted: 09/14/2021] [Indexed: 02/08/2023] Open
Abstract
Insulin plays a range of roles as an anabolic hormone in peripheral tissues. It regulates glucose metabolism, stimulates glucose transport into cells and suppresses hepatic glucose production. Insulin influences cell growth, differentiation and protein synthesis, and inhibits catabolic processes such as glycolysis, lipolysis and proteolysis. Insulin and insulin-like growth factor-1 receptors are expressed on all cell types in the central nervous system. Widespread distribution in the brain confirms that insulin signaling plays important and diverse roles in this organ. Insulin is known to regulate glucose metabolism, support cognition, enhance the outgrowth of neurons, modulate the release and uptake of catecholamine, and regulate the expression and localization of gamma-aminobutyric acid (GABA). Insulin is also able to freely cross the blood–brain barrier from the circulation. In addition, changes in insulin signaling, caused inter alia insulin resistance, may accelerate brain aging, and affect plasticity and possibly neurodegeneration. There are two significant insulin signal transduction pathways: the PBK/AKT pathway which is responsible for metabolic effects, and the MAPK pathway which influences cell growth, survival and gene expression. The aim of this study is to describe the role played by insulin in the CNS, in both healthy people and those with pathologies such as insulin resistance and Alzheimer’s disease.
Collapse
|
12
|
Sarahian N, Sarvazad H, Sajadi E, Rahnejat N, Eskandari Roozbahani N. Investigation of common risk factors between polycystic ovary syndrome and Alzheimer's disease: a narrative review. Reprod Health 2021; 18:156. [PMID: 34311759 PMCID: PMC8314638 DOI: 10.1186/s12978-021-01203-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 07/13/2021] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND The most common endocrine and metabolic disorders in premenopausal women is polycystic ovary syndrome (PCOS), characterized by hyperandrogenism, chronic anovulation, and/or ultrasound evidence of small ovarian cysts. Obesity and insulin resistance are also the main factors influencing the clinical manifestations of this syndrome. Alzheimer's disease (AD) is the most typical progressive neurodegenerative disorder of the brain, and recent studies suggest a relationship between endocrinal dysregulation and neuronal loss during AD pathology. AIM This study aimed to evaluate the common risk factors for Alzheimer's and PCOS based on previous studies. Knowing the common risk factors and eliminating them may prevent neurodegenerative Alzheimer's disease in the future. METHOD In this narrative review, international databases, including Google Scholar, Scopus, PubMed, and the Web of Science, were searched to retrieve the relevant studies. The relevant studies' summaries were categorized to discuss the possible pathways that may explain the association between Alzheimer's and PCOS signs/symptoms and complications. RESULTS According to our research, the factors involved in Alzheimer's and PCOS disorders may share some common risk factors. In patients with PCOS, increased LH to FSH ratio, decreased vitamin D, insulin resistance, and obesity are some of the most important factors that may increase the risk of Alzheimer's disease.
Collapse
Affiliation(s)
- Nahid Sarahian
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hosna Sarvazad
- Clinical Research Development Center, Imam Reza Hospital, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Elham Sajadi
- Department of Basic Science, School of Veterinary Medicine, Shiraz University, Shiraz, Iran
| | - Nasrin Rahnejat
- Faculty of Nursing and Midwifery, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Narges Eskandari Roozbahani
- Clinical Research Development Center, Imam Reza Hospital, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| |
Collapse
|
13
|
Zhao ZH, Du KJ, Wang T, Wang JY, Cao ZP, Chen XM, Song H, Zheng G, Shen XF. Maternal Lead Exposure Impairs Offspring Learning and Memory via Decreased GLUT4 Membrane Translocation. Front Cell Dev Biol 2021; 9:648261. [PMID: 33718391 PMCID: PMC7947239 DOI: 10.3389/fcell.2021.648261] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Accepted: 02/08/2021] [Indexed: 11/13/2022] Open
Abstract
Lead (Pb) can cause a significant neurotoxicity in both adults and children, leading to the impairment to brain function. Pb exposure plays a key role in the impairment of learning and memory through synaptic neurotoxicity, resulting in the cognitive function. Researches have demonstrated that Pb exposure plays an important role in the etiology and pathogenesis of neurodegenerative diseases, such as Alzheimer's disease. However, the underlying mechanisms remain unclear. In the current study, a gestational Pb exposure (GLE) rat model was established to investigate the underlying mechanisms of Pb-induced cognitive impairment. We demonstrated that low-level gestational Pb exposure impaired spatial learning and memory as well as hippocampal synaptic plasticity at postnatal day 30 (PND 30) when the blood concentration of Pb had already recovered to normal levels. Pb exposure induced a decrease in hippocampal glucose metabolism by reducing glucose transporter 4 (GLUT4) levels in the cell membrane through the phosphatidylinositol 3 kinase-protein kinase B (PI3K-Akt) pathway. In vivo and in vitro GLUT4 over-expression increased the membrane translocation of GLUT4 and glucose uptake, and reversed the Pb-induced impairment to synaptic plasticity and cognition. These findings indicate that Pb exposure impairs synaptic plasticity by reducing the level of GLUT4 in the cell membrane as well as glucose uptake via the PI3K-Akt signaling pathway, demonstrating a novel mechanism for Pb exposure-induced neurotoxicity.
Collapse
Affiliation(s)
- Zai-Hua Zhao
- Department of Occupational and Environmental Health and the Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, Fourth Military Medical University, Xi'an, China
| | - Ke-Jun Du
- Department of Occupational and Environmental Health and the Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, Fourth Military Medical University, Xi'an, China
| | - Tao Wang
- Department of Occupational and Environmental Health and the Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, Fourth Military Medical University, Xi'an, China
| | - Ji-Ye Wang
- Department of Occupational and Environmental Health and the Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, Fourth Military Medical University, Xi'an, China
| | - Zi-Peng Cao
- Department of Occupational and Environmental Health and the Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, Fourth Military Medical University, Xi'an, China
| | - Xiao-Ming Chen
- Department of Occupational and Environmental Health and the Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, Fourth Military Medical University, Xi'an, China
| | - Han Song
- Department of Occupational and Environmental Health and the Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, Fourth Military Medical University, Xi'an, China.,Department of Health Service, Chinese PLA General Hospital, Beijing, China
| | - Gang Zheng
- Department of Occupational and Environmental Health and the Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, Fourth Military Medical University, Xi'an, China
| | - Xue-Feng Shen
- Department of Occupational and Environmental Health and the Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, Fourth Military Medical University, Xi'an, China
| |
Collapse
|
14
|
A Neonatal Mild Defect in Brain Insulin Signaling Predisposes a Subclinical Model of Sporadic Alzheimer's to Develop the Disease. J Mol Neurosci 2021; 71:1473-1484. [PMID: 33492616 DOI: 10.1007/s12031-021-01797-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 01/11/2021] [Indexed: 10/22/2022]
Abstract
Brain insulin system dysfunction has been proposed as a key player in the pathogenesis of sporadic Alzheimer's disease (sAD). Given this fact, an adult rat model for sAD has been developed by intracerebroventricular injection of a subdiabetogenic streptozotocin dosage (icv-STZ). A low dose of icv-STZ in adult rats leads to a subclinical model of Alzheimer's disease. According to the brain developmental origin for sAD occurrence, the present study evaluated the effect of neonatal injection of icv-STZ on the development and progression of Alzheimer's disease later in the adult animals treated with a low dose of icv-STZ. Although no alteration was observed in the rats receiving an adult low dose of icv-STZ, these animals displayed cognitive deficits if they were also treated neonatally with icv-STZ. These impairments were associated with altered gene expression of insulin receptor, tau and choline acetyltransferase, along with increased astrocyte and dark neuron densities in the hippocampus. This study highlights neonatal brain insulin system dysfunction in the programming of brain insulin signaling sensitivity and provides more evidence for the developmental origin of sAD.
Collapse
|
15
|
Kshirsagar V, Thingore C, Juvekar A. Insulin resistance: a connecting link between Alzheimer's disease and metabolic disorder. Metab Brain Dis 2021; 36:67-83. [PMID: 32986168 DOI: 10.1007/s11011-020-00622-2] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Accepted: 09/22/2020] [Indexed: 12/11/2022]
Abstract
Recent evidence suggests that Alzheimer's disease (AD) is closely linked with insulin resistance, as seen in type 2 diabetes mellitus (T2DM). Insulin signaling is impaired in AD brains due to insulin resistance, ultimately resulting in the formation of neurofibrillary tangles (NFTs). AD and T2DM are connected at molecular, clinical, and epidemiological levels making it imperative to understand the contribution of T2DM, and other metabolic disorders, to AD pathogenesis. In this review, we have discussed various modalities involved in the pathogenesis of these two diseases and explained the contributing parameters. Insulin is vital for maintaining glucose homeostasis and it plays an important role in regulating inflammation. Here, we have discussed the roles of various contributing factors like miRNA, leptin hormone, neuroinflammation, metabolic dysfunction, and gangliosides in insulin impairment both in AD and T2DM. Understanding these mechanisms will be a big step forward for making molecular therapies that may help maintain or prevent both AD and T2DM, thus reducing the burden of both these diseases.
Collapse
Affiliation(s)
- Viplav Kshirsagar
- Department of Pharmaceutical Sciences and Technology, Institute of Chemical Technology, Nathalal Parekh Marg, Near Khalsa college, Matunga, Mumbai, Maharashtra, 400019, India
| | - Chetan Thingore
- Department of Pharmaceutical Sciences and Technology, Institute of Chemical Technology, Nathalal Parekh Marg, Near Khalsa college, Matunga, Mumbai, Maharashtra, 400019, India
| | - Archana Juvekar
- Department of Pharmaceutical Sciences and Technology, Institute of Chemical Technology, Nathalal Parekh Marg, Near Khalsa college, Matunga, Mumbai, Maharashtra, 400019, India.
| |
Collapse
|
16
|
Robbins J, Busquets O, Tong M, de la Monte SM. Dysregulation of Insulin-Linked Metabolic Pathways in Alzheimer's Disease: Co-Factor Role of Apolipoprotein E ɛ4. J Alzheimers Dis Rep 2020; 4:479-493. [PMID: 33344887 PMCID: PMC7739986 DOI: 10.3233/adr-200238] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
BACKGROUND Brain insulin resistance and deficiency are well-recognized abnormalities in Alzheimer's disease (AD) and likely mediators of impaired energy metabolism. Since apolipoprotein E (APOE) is a major risk factor for late-onset AD, it was of interest to examine its potential contribution to altered insulin-linked signaling networks in the brain. OBJECTIVE The main goal was to evaluate the independent and interactive contributions of AD severity and APOE ɛ4 dose on brain expression of insulin-related polypeptides and inflammatory mediators of metabolic dysfunction. METHODS Postmortem fresh frozen frontal lobe tissue from banked cases with known APOE genotypes and different AD Braak stages were used to measure insulin network polypeptide immunoreactivity with a commercial multiplex enzyme-linked immunosorbent assay (ELISA). RESULTS Significant AD Braak stage and APOE genotype-related abnormalities in insulin, C-peptide, gastric inhibitory polypeptide (GIP), glucaton-like peptide-1 (GLP-1), leptin, ghrelin, glucagon, resistin, and plasminogen activator inhibitor-1 (PAI-1) were detected. The main factors inhibiting polypeptide expression and promoting neuro-inflammatory responses included AD Braak stage and APOE ɛ4/ɛ4 rather than ɛ3/ɛ4. CONCLUSION This study demonstrates an expanded role for impaired expression of insulin-related network polypeptides as well as neuroinflammatory mediators of brain insulin resistance in AD pathogenesis and progression. In addition, the findings show that APOE has independent and additive effects on these aberrations in brain polypeptide expression, but the impact is decidedly greater for APOE ɛ4/ɛ4 than ɛ3/ɛ4.
Collapse
Affiliation(s)
- James Robbins
- Alpert Medical School of Brown University, Providence, RI, USA
| | - Oriol Busquets
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Institute of Neuroscience, University of Barcelona, Barcelona, Spain
| | - Ming Tong
- Alpert Medical School of Brown University, Providence, RI, USA,Department of Medicine, Rhode Island Hospital, Providence, RI, USA
| | - Suzanne M. de la Monte
- Alpert Medical School of Brown University, Providence, RI, USA,Department of Medicine, Rhode Island Hospital, Providence, RI, USA,Departments of Pathology and Laboratory Medicine Providence VA Medical Center, Rhode Island Hospital, and the Women and Infants Hospital of Rhode Island, Providence, RI, USA,Correspondence to: Dr. Suzanne M. de la Monte, MD, MPH, Rhode Island Hospital, 55 Claverick Street, Room 419, Providence, RI 02903, USA. Tel.: +1 401 444 7364; Fax: +1 401 444 2939; E-mail:
| |
Collapse
|
17
|
Cheng H, Gang X, Liu Y, Wang G, Zhao X, Wang G. Mitochondrial dysfunction plays a key role in the development of neurodegenerative diseases in diabetes. Am J Physiol Endocrinol Metab 2020; 318:E750-E764. [PMID: 31714795 DOI: 10.1152/ajpendo.00179.2019] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Mitochondria have an essential function in cell survival due to their role in bioenergetics, reactive oxygen species generation, calcium buffering, and other metabolic activities. Mitochondrial dysfunctions are commonly found in neurodegenerative diseases (NDs), and diabetes is a risk factor for NDs. However, the role of mitochondria in diabetic neurodegeneration is still unclear. In the present study, we review the latest evidence on the role of mitochondrial dysfunctions in the development of diabetes-related NDs and the underlying molecular mechanisms. Hypoglycemic agents, especially metformin, have been proven to have neuroprotective effects in the treatment of diabetes, in which mitochondria could act as one of the underlying mechanisms. Other hypoglycemic agents, including thiazolidinediones (TZDs), dipeptidyl peptidase 4 (DPP-4) inhibitors, and glucagon-like peptide 1 (GLP-1) receptor agonists, have gained more attention because of their beneficial effects on NDs, presumably by improving mitochondrial function. Our review highlights the notion that mitochondria could be a promising therapeutic target in the treatment of NDs in patients with diabetes.
Collapse
Affiliation(s)
- Han Cheng
- Department of Endocrinology and Metabolism, First Hospital of Jilin University, Changchun, Jilin, People's Republic of China
| | - Xiaokun Gang
- Department of Endocrinology and Metabolism, First Hospital of Jilin University, Changchun, Jilin, People's Republic of China
| | - Yujia Liu
- Department of Endocrinology and Metabolism, First Hospital of Jilin University, Changchun, Jilin, People's Republic of China
| | - Gang Wang
- Department of Endocrinology and Metabolism, First Hospital of Jilin University, Changchun, Jilin, People's Republic of China
| | - Xue Zhao
- Department of Endocrinology and Metabolism, First Hospital of Jilin University, Changchun, Jilin, People's Republic of China
| | - Guixia Wang
- Department of Endocrinology and Metabolism, First Hospital of Jilin University, Changchun, Jilin, People's Republic of China
| |
Collapse
|
18
|
de la Monte SM, Tong M, Daiello LA, Ott BR. Early-Stage Alzheimer's Disease Is Associated with Simultaneous Systemic and Central Nervous System Dysregulation of Insulin-Linked Metabolic Pathways. J Alzheimers Dis 2019; 68:657-668. [PMID: 30775986 PMCID: PMC10084886 DOI: 10.3233/jad-180906] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
BACKGROUND Brain insulin resistance is a well-recognized abnormality in Alzheimer's disease (AD) and the likely mediator of impaired glucose utilization that emerges early and progresses with disease severity. Moreover, the rates of mild cognitive impairment (MCI) or AD are significantly greater in people with diabetes mellitus or obesity. OBJECTIVE This study was designed to determine whether systemic and central nervous system (CNS) insulin resistant disease states emerge together and thus may be integrally related. METHODS Insulin-related molecules were measured in paired human serum and cerebrospinal fluid (CSF) samples from 19 with MCI or early AD, and 21 controls using a multiplex ELISA platform. RESULTS In MCI/AD, both the CSF and serum samples had significantly elevated mean levels of C-peptide and an incretin, and reduced expression of Visfatin, whereas only CSF showed significant reductions in insulin and leptin and only serum had increased glucagon, PAI-1, and ghrelin. Although the overall CSF and serum responses reflected insulin resistance together with insulin deficiency, the specific alterations measured in CSF and serum were different. CONCLUSION In MCI and early-stage AD, CNS and systemic insulin-related metabolic dysfunctions, including insulin resistance, occur simultaneously, suggesting that they are integrally related and possibly mediated similar pathogenic factors.
Collapse
Affiliation(s)
- Suzanne M de la Monte
- Department of Pathology and Laboratory Medicine (Neuropathology), Rhode Island Hospital, the Providence VA Medical Center, and the Alpert Medical School of Brown University, Providence, RI, USA.,Department of Neurology, Rhode Island Hospital and the Alpert Medical School of Brown University, Providence, RI, USA.,Department of Medicine, Rhode Island Hospital and the Alpert Medical School of Brown University, Providence, RI, USA
| | - Ming Tong
- Department of Medicine, Rhode Island Hospital and the Alpert Medical School of Brown University, Providence, RI, USA
| | - Lori A Daiello
- Department of Neurology, Rhode Island Hospital and the Alpert Medical School of Brown University, Providence, RI, USA.,The Alzheimer's Disease and Memory Disorders Center, Rhode Island Hospital and the Alpert Medical School of Brown University, Providence, RI, USA
| | - Brian R Ott
- Department of Neurology, Rhode Island Hospital and the Alpert Medical School of Brown University, Providence, RI, USA.,The Alzheimer's Disease and Memory Disorders Center, Rhode Island Hospital and the Alpert Medical School of Brown University, Providence, RI, USA
| |
Collapse
|
19
|
Loehfelm A, Boucsein A, Pretz D, Tups A. Timing Matters: Circadian Effects on Energy Homeostasis and Alzheimer's Disease. Trends Endocrinol Metab 2019; 30:132-143. [PMID: 30594436 DOI: 10.1016/j.tem.2018.12.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Revised: 11/09/2018] [Accepted: 12/03/2018] [Indexed: 10/27/2022]
Abstract
Metabolic syndrome and Alzheimer's disease (AD) are two major health issues in modern society causing an extraordinary financial burden for the global healthcare systems. A tight link between the pathologies of obesity and type 2 diabetes (T2D), and more recently between T2D and AD, has been discovered. Furthermore, in recent years it has become apparent that the circadian clock has an important function in controlling metabolism. This review integrates the role of the circadian clock in the development of these metabolic derangements and vice versa. Common features such as central insulin resistance, altered glycogen synthase kinase 3β (GSK3β) signalling, and central inflammation are discussed, and therapeutic interventions targeting those mechanisms are mentioned briefly.
Collapse
Affiliation(s)
- Aline Loehfelm
- Centre for Neuroendocrinology and Brain Health Research Centre, Department of Physiology, School of Medical Sciences, University of Otago, Dunedin 9054, New Zealand; These authors contributed equally to this work
| | - Alisa Boucsein
- Centre for Neuroendocrinology and Brain Health Research Centre, Department of Physiology, School of Medical Sciences, University of Otago, Dunedin 9054, New Zealand; These authors contributed equally to this work
| | - Dominik Pretz
- Centre for Neuroendocrinology and Brain Health Research Centre, Department of Physiology, School of Medical Sciences, University of Otago, Dunedin 9054, New Zealand; These authors contributed equally to this work
| | - Alexander Tups
- Centre for Neuroendocrinology and Brain Health Research Centre, Department of Physiology, School of Medical Sciences, University of Otago, Dunedin 9054, New Zealand.
| |
Collapse
|
20
|
de la Monte SM. The Full Spectrum of Alzheimer's Disease Is Rooted in Metabolic Derangements That Drive Type 3 Diabetes. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1128:45-83. [PMID: 31062325 PMCID: PMC9996398 DOI: 10.1007/978-981-13-3540-2_4] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The standard practice in neuropathology is to diagnose Alzheimer's disease (AD) based on the distribution and abundance of neurofibrillary tangles and Aβ deposits. However, other significant abnormalities including neuroinflammation, gliosis, white matter degeneration, non-Aβ microvascular disease, and insulin-related metabolic dysfunction require further study to understand how they could be targeted to more effectively remediate AD. This review addresses non-Aβ and non-pTau AD-associated pathologies, highlighting their major features, roles in neurodegeneration, and etiopathic links to deficits in brain insulin and insulin-like growth factor signaling and cognitive impairment. The discussion delineates why AD with its most characteristic clinical and pathological phenotypic profiles should be regarded as a brain form of diabetes, i.e., type 3 diabetes, and entertains the hypothesis that type 3 diabetes is just one of the categories of insulin resistance diseases that can occur independently or overlap with one or more of the others, including type 2 diabetes, metabolic syndrome, and nonalcoholic fatty liver disease.
Collapse
Affiliation(s)
- Suzanne M de la Monte
- Departments of Neurology, Neuropathology, and Neurosurgery, Rhode Island Hospital, and the Alpert Medical School of Brown University, Providence, RI, USA.
- Department of Pathology and Laboratory Medicine, Providence VA Medical Center, Providence, RI, USA.
| |
Collapse
|
21
|
Chronic ethanol forced administration from adolescence to adulthood reduces cell density in the rat spinal cord. Tissue Cell 2018; 55:77-82. [PMID: 30503063 DOI: 10.1016/j.tice.2018.10.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Revised: 10/15/2018] [Accepted: 10/20/2018] [Indexed: 11/21/2022]
Abstract
Ethanol (EtOH) consumption is a risk factor for central nervous system damage, especially during adolescence. This study aimed to investigate the possible effects of chronic EtOH forced administration on gray and white matter of the spinal cord, from adolescence to adulthood. For this, male Wistar rats were administered EtOH by gavage (6.5 g/kg/day; 22.5% w/v) from the 35th to the 90th day of life, while control animals received only distilled water. After exposure, animals were euthanized and their spinal cords processed to obtain cervical and thoracic segments for histological analyses. Quantitative analyses of total cell density and motor neurons of white and gray matter from the ventral horns were evaluated. Forced EtOH administration model showed a decrease in the motoneuron density in the spinal cord in both segments evaluated. Analyses of total cell density showed that the cervical segment was more susceptible to damages promoted by EtOH, with a significant decrease in cell density. Our results showed that chronic EtOH exposure during adolescence could promote injuries to the spinal cord, with neurodegeneration of motoneurons and other cell types present in neural parenchyma.
Collapse
|
22
|
Hussain R, Zubair H, Pursell S, Shahab M. Neurodegenerative Diseases: Regenerative Mechanisms and Novel Therapeutic Approaches. Brain Sci 2018; 8:E177. [PMID: 30223579 PMCID: PMC6162719 DOI: 10.3390/brainsci8090177] [Citation(s) in RCA: 119] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Revised: 09/03/2018] [Accepted: 09/12/2018] [Indexed: 12/12/2022] Open
Abstract
Regeneration refers to regrowth of tissue in the central nervous system. It includes generation of new neurons, glia, myelin, and synapses, as well as the regaining of essential functions: sensory, motor, emotional and cognitive abilities. Unfortunately, regeneration within the nervous system is very slow compared to other body systems. This relative slowness is attributed to increased vulnerability to irreversible cellular insults and the loss of function due to the very long lifespan of neurons, the stretch of cells and cytoplasm over several dozens of inches throughout the body, insufficiency of the tissue-level waste removal system, and minimal neural cell proliferation/self-renewal capacity. In this context, the current review summarized the most common features of major neurodegenerative disorders; their causes and consequences and proposed novel therapeutic approaches.
Collapse
Affiliation(s)
- Rashad Hussain
- Center for Translational Neuromedicine, University of Rochester, NY 14642, USA.
| | - Hira Zubair
- Department of Animal Sciences, Quaid-i-Azam University, Islamabad 45320, Pakistan.
| | - Sarah Pursell
- Center for Translational Neuromedicine, University of Rochester, NY 14642, USA.
| | - Muhammad Shahab
- Department of Animal Sciences, Quaid-i-Azam University, Islamabad 45320, Pakistan.
| |
Collapse
|
23
|
A transient insulin system dysfunction in newborn rat brain followed by neonatal intracerebroventricular administration of streptozotocin could be accompanied by a labile cognitive impairment. Neurosci Res 2017; 132:17-25. [PMID: 29055675 DOI: 10.1016/j.neures.2017.10.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2017] [Revised: 09/16/2017] [Accepted: 10/06/2017] [Indexed: 01/22/2023]
Abstract
The early postnatal period is a critical period of hippocampus development, which is highly dependent on insulin receptor (IR) signaling and very important in cognitive function. The present study was conducted in order to present a model of neonatal transient brain insulin system dysfunction through finding an appropriate dose of injection of streptozotocin (STZ) during the neonatal period. Sixty male Wistar rat pups were divided into 4 groups of 15 and received intracerebroventricular saline or STZ (icv-STZ) (15, 20 and 25μg/kg) on postnatal day 7. Gene expression of IR and target genes for IR signaling (choline acetyltransferase (ChAT) and Tau) were measured at the ages of 2 and 7 weeks. Behavioral tests were performed at the ages of 3 and 6 weeks to assess short- and long-term cognitive function. 20μg/kg dose of icv-STZ was estimated as the optimal dose causing transient alteration in gene expression of IR, ChAT and Tau. Additionally, cognitive function of the animals restored to normal level at the age of 6 weeks. Therefore, 20μg/kg dose of icv-STZ is proposed as a new approach to generating transient brain insulin system dysfunction associated with transient cognitive impairments at a critical postnatal period of brain development.
Collapse
|
24
|
de la Monte SM. Insulin Resistance and Neurodegeneration: Progress Towards the Development of New Therapeutics for Alzheimer's Disease. Drugs 2017; 77:47-65. [PMID: 27988872 PMCID: PMC5575843 DOI: 10.1007/s40265-016-0674-0] [Citation(s) in RCA: 203] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Alzheimer's disease (AD) should be regarded as a degenerative metabolic disease caused by brain insulin resistance and deficiency, and overlapping with the molecular, biochemical, pathophysiological, and metabolic dysfunctions in diabetes mellitus, non-alcoholic fatty liver disease, and metabolic syndrome. Although most of the diagnostic and therapeutic approaches over the past several decades have focused on amyloid-beta (Aβ42) and aberrantly phosphorylated tau, which could be caused by consequences of brain insulin resistance, the broader array of pathologies including white matter atrophy with loss of myelinated fibrils and leukoaraiosis, non-Aβ42 microvascular disease, dysregulated lipid metabolism, mitochondrial dysfunction, astrocytic gliosis, neuro-inflammation, and loss of synapses vis-à-vis growth of dystrophic neurites, is not readily accounted for by Aβ42 accumulations, but could be explained by dysregulated insulin/IGF-1 signaling with attendant impairments in signal transduction and gene expression. This review covers the diverse range of brain abnormalities in AD and discusses how insulins, incretins, and insulin sensitizers could be utilized to treat at different stages of neurodegeneration.
Collapse
Affiliation(s)
- Suzanne M de la Monte
- Department of Neurology, Rhode Island Hospital, and the Alpert Medical School of Brown University, Pierre Galletti Research Building, 55 Claverick Street, Room 419, Providence, RI, 02903, USA.
- Department of Neurosurgery, Rhode Island Hospital, and the Alpert Medical School of Brown University, Providence, RI, USA.
- Department of Neuropathology, Rhode Island Hospital, and the Alpert Medical School of Brown University, Providence, RI, USA.
- Department of Pathology, Rhode Island Hospital, and the Alpert Medical School of Brown University, Providence, RI, USA.
| |
Collapse
|
25
|
Tong M, Dominguez C, Didsbury J, de la Monte SM. Targeting Alzheimer's Disease Neuro-Metabolic Dysfunction with a Small Molecule Nuclear Receptor Agonist (T3D-959) Reverses Disease Pathologies. ACTA ACUST UNITED AC 2016; 6. [PMID: 27525190 PMCID: PMC4979550 DOI: 10.4172/2161-0460.1000238] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Background Alzheimer’s disease (AD) could be regarded as a brain form of diabetes since insulin resistance and deficiency develop early and progress with severity of neurodegeneration. Preserving insulin’s actions in the brain restores function and reduces neurodegeneration. T3D-959 is a dual nuclear receptor agonist currently in a Phase 2a trial in mild-to-moderate AD patients (ClinicalTrials.gov identifier NCT02560753). Herein, we show that T3D-959 improves motor function and reverses neurodegeneration in a sporadic model of AD. Methods Long Evans rats were administered intracerebral (i.c.) streptozotocin (STZ) or normal saline (control) and dosed orally with T3D-959 (1.0 mg/kg/day) or saline for 21 or 28 days. Rotarod tests evaluated motor function. Histopathology with image analysis was used to assess neurodegeneration. Results T3D-959 significantly improved motor performance, and preserved both cortical and normalized white matter structure in i.c STZ-treated rats. T3D-959 treatments were effective when dosed therapeutically, whether initiated 1 day or 7 days after i.c. STZ. Conclusion T3D-959’s targeting neuro-metabolic dysfunctions via agonism of PPAR delta and PPAR gamma nuclear receptors provides potential disease modification in AD.
Collapse
Affiliation(s)
- Ming Tong
- Liver Research Center, Warren Alpert Medical School of Brown University, Providence, RI, USA; Divisions of Gastroenterology, Warren Alpert Medical School of Brown University, Providence, RI, USA; Department of Medicine, Warren Alpert Medical School of Brown University, Providence, RI, USA
| | | | - John Didsbury
- T3D Pharmaceuticals, Raleigh Durham, NC, USA5Australian School of Advanced Medicine, Macquarie University, Sydney, Australia
| | - Suzanne M de la Monte
- Liver Research Center, Warren Alpert Medical School of Brown University, Providence, RI, USA; Divisions of Gastroenterology, Warren Alpert Medical School of Brown University, Providence, RI, USA; Department of Neuropathology, Warren Alpert Medical School of Brown University, Providence, RI, USA; Department of Medicine, Warren Alpert Medical School of Brown University, Providence, RI, USA; Department of Pathology, Warren Alpert Medical School of Brown University, Providence, RI, USA; Department of Neurology, Warren Alpert Medical School of Brown University, Providence, RI, USA; Department of Neurosurgery, Rhode Island Hospital and the Warren Alpert Medical School of Brown University, Providence, RI
| |
Collapse
|
26
|
Abstract
Several studies have indicated that Diabetes Mellitus (DM) can increase the risk
of developing Alzheimer's disease (AD). This review briefly describes current
concepts in mechanisms linking DM and insulin resistance/deficiency to AD.
Insulin/insulin-like growth factor (IGF) resistance can contribute to
neurodegeneration by several mechanisms which involve: energy and metabolism
deficits, impairment of Glucose transporter-4 function, oxidative and
endoplasmic reticulum stress, mitochondrial dysfunction, accumulation of AGEs,
ROS and RNS with increased production of neuro-inflammation and activation of
pro-apoptosis cascade. Impairment in insulin receptor function and increased
expression and activation of insulin-degrading enzyme (IDE) have also been
described. These processes compromise neuronal and glial function, with a
reduction in neurotransmitter homeostasis. Insulin/IGF resistance causes the
accumulation of AβPP-Aβ oligomeric fibrils or insoluble larger
aggregated fibrils in the form of plaques that are neurotoxic. Additionally,
there is production and accumulation of hyper-phosphorylated insoluble fibrillar
tau which can exacerbate cytoskeletal collapse and synaptic disconnection.
Collapse
Affiliation(s)
- Maria Niures P S Matioli
- Pós-graduanda, nível de Doutorado, Departamento de Neurologia da Faculdade de Medicina da Universidade de São Paulo
| | - Ricardo Nitrini
- Professor Titular da Disciplina de Neurologia da Faculdade de Medicina da Universidade de São Paulo. Orientador e Professor Responsável pela Pós-graduação do Departamento de Neurologia da Faculdade de Medicina da Universidade de São Paulo
| |
Collapse
|
27
|
Lourenco MV, Ferreira ST, De Felice FG. Neuronal stress signaling and eIF2α phosphorylation as molecular links between Alzheimer's disease and diabetes. Prog Neurobiol 2015; 129:37-57. [PMID: 25857551 DOI: 10.1016/j.pneurobio.2015.03.003] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2015] [Revised: 03/10/2015] [Accepted: 03/29/2015] [Indexed: 12/22/2022]
Abstract
Mounting evidence from clinical, epidemiological, neuropathology and preclinical studies indicates that mechanisms similar to those leading to peripheral metabolic deregulation in metabolic disorders, such as diabetes and obesity, take place in the brains of Alzheimer's disease (AD) patients. These include pro-inflammatory mechanisms, brain metabolic stress and neuronal insulin resistance. From a molecular and cellular perspective, recent progress has been made in unveiling novel pathways that act in an orchestrated way to cause neuronal damage and cognitive decline in AD. These pathways converge to the activation of neuronal stress-related protein kinases and excessive phosphorylation of eukaryotic translation initiation factor 2α (eIF2α-P), which plays a key role in control of protein translation, culminating in synapse dysfunction and memory loss. eIF2α-P signaling thus links multiple neuronal stress pathways to impaired neuronal function and neurodegeneration. Here, we present a critical analysis of recently discovered molecular mechanisms underlying impaired brain insulin signaling and metabolic stress, with emphasis on the role of stress kinase/eIF2α-P signaling as a hub that promotes brain and behavioral impairments in AD. Because very similar mechanisms appear to operate in peripheral metabolic deregulation in T2D and in brain defects in AD, we discuss the concept that targeting defective brain insulin signaling and neuronal stress mechanisms with anti-diabetes agents may be an attractive approach to fight memory decline in AD. We conclude by raising core questions that remain to be addressed toward the development of much needed therapeutic approaches for AD.
Collapse
Affiliation(s)
- Mychael V Lourenco
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, RJ 21941-902, Brazil.
| | - Sergio T Ferreira
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, RJ 21941-902, Brazil; Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, RJ 21941-902, Brazil
| | - Fernanda G De Felice
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, RJ 21941-902, Brazil.
| |
Collapse
|
28
|
de la Monte SM. Type 3 diabetes is sporadic Alzheimer׳s disease: mini-review. Eur Neuropsychopharmacol 2014; 24:1954-60. [PMID: 25088942 PMCID: PMC4444430 DOI: 10.1016/j.euroneuro.2014.06.008] [Citation(s) in RCA: 222] [Impact Index Per Article: 22.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2014] [Accepted: 06/20/2014] [Indexed: 01/16/2023]
Abstract
Alzheimer׳s disease (AD) is the most common cause of dementia in North America. Growing evidence supports the concept that AD is a metabolic disease mediated by impairments in brain insulin responsiveness, glucose utilization, and energy metabolism, which lead to increased oxidative stress, inflammation, and worsening of insulin resistance. In addition, metabolic derangements directly contribute to the structural, functional, molecular, and biochemical abnormalities that characterize AD, including neuronal loss, synaptic disconnection, tau hyperphosphorylation, and amyloid-beta accumulation. Because the fundamental abnormalities in AD represent effects of brain insulin resistance and deficiency, and the molecular and biochemical consequences overlap with Type 1 and Type 2 diabetes, we suggest the term "Type 3 diabetes" to account for the underlying abnormalities associated with AD-type neurodegeneration. In light of the rapid increases in sporadic AD prevalence rates and vastly expanded use of nitrites and nitrates in foods and agricultural products over the past 30-40 years, the potential role of nitrosamine exposures as mediators of Type 3 diabetes is discussed.
Collapse
Affiliation(s)
- Suzanne M de la Monte
- Departments of Medicine, Pathology, Neurology, and Neurosurgery, Rhode Island Hospital and the Warren Alpert Medical School of Brown University, 55 Claverick Street, Room 419, Providence, RI 02903, USA.
| |
Collapse
|
29
|
Dobson CC, Thevasundaram K, Mongillo DL, Winterborn A, Holloway AC, Brien JF, Reynolds JN. Chronic prenatal ethanol exposure alters expression of central and peripheral insulin signaling molecules in adult guinea pig offspring. Alcohol 2014; 48:687-93. [PMID: 25278255 DOI: 10.1016/j.alcohol.2014.09.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Maternal ethanol consumption during pregnancy can produce a range of teratogenic outcomes in offspring. The mechanism of ethanol teratogenicity is multi-faceted, but may involve alterations in insulin and insulin-like growth factor (IGF) signaling pathways. These pathways are not only important for metabolism, but are also critically involved in neuronal survival and plasticity, and they can be altered by chronic prenatal ethanol exposure (CPEE). The objective of this study was to test the hypothesis that CPEE alters expression of insulin and IGF signaling molecules in the prefrontal cortex and liver of adult guinea pig offspring. Pregnant Dunkin-Hartley-strain guinea pigs received ethanol (4 g/kg maternal body weight/day) or isocaloric-sucrose/pair-feeding (nutritional control) throughout gestation. Fasting blood glucose concentration was measured in male and female offspring at postnatal day 150-200, followed by euthanasia, collection of prefrontal cortex and liver, and RNA extraction. IGF-1, IGF-1 receptor (IGF-1R), IGF-2, IGF-2 receptor (IGF-2R), insulin receptor substrate (IRS)-1, IRS-2, and insulin receptor (INSR) mRNA expression levels were measured in tissues using quantitative real-time PCR. The mean maternal blood ethanol concentration was 281 ± 15 mg/dL at 1 h after the second divided dose of ethanol on GD 57. CPEE resulted in increased liver weight in adult offspring, but produced no difference in fasting blood glucose concentration compared with nutritional control. In the liver, CPEE decreased mRNA expression of IGF-1, IGF-1R, and IGF-2, and increased IRS-2 mRNA expression in male offspring only compared with nutritional control. Female CPEE offspring had decreased INSR hepatic mRNA expression compared with male CPEE offspring. In the prefrontal cortex, IRS-2 mRNA expression was increased in CPEE offspring compared with nutritional control. The data demonstrate that CPEE alters both central and peripheral expression of insulin and IGF signaling molecules at the mRNA level, which may be related to metabolic dysregulation in adult offspring. Furthermore, altered insulin and IGF signaling may be a mechanism of ethanol neurobehavioral teratogenicity.
Collapse
|
30
|
Shu X, Zhang Y, Xu H, Kang K, Cai D. Brain-derived neurotrophic factor inhibits glucose intolerance after cerebral ischemia. Neural Regen Res 2014; 8:2370-8. [PMID: 25206547 PMCID: PMC4146044 DOI: 10.3969/j.issn.1673-5374.2013.25.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2013] [Accepted: 07/20/2013] [Indexed: 11/18/2022] Open
Abstract
Brain-derived neurotrophic factor is associated with the insulin signaling pathway and glucose tabolism. We hypothesized that expression of brain-derived neurotrophic factor and its receptor may be involved in glucose intolerance following ischemic stress. To verify this hypothesis, this study aimed to observe the changes in brain-derived neurotrophic factor and tyrosine kinase B receptor expression in glucose metabolism-associated regions following cerebral ischemic stress in mice. At day 1 after middle cerebral artery occlusion, the expression levels of brain-derived neurotrophic factor were significantly decreased in the ischemic cortex, hypothalamus, liver, skeletal muscle, and pancreas. The expression levels of tyrosine kinase B receptor were decreased in the hypothalamus and liver, and increased in the skeletal muscle and pancreas, but remained unchanged in the cortex. Intrahypothalamic administration of brain-derived neurotrophic factor (40 ng) suppressed the decrease in insulin receptor and tyrosine-phosphorylated insulin receptor expression in the liver and skeletal muscle, and inhibited the overexpression of gluconeogenesis-associated phosphoenolpyruvate carboxykinase and glucose-6-phosphatase in the liver of cerebral ischemic mice. However, serum insulin levels remained unchanged. Our experimental findings indicate that brain-derived neurotrophic factor can promote glucose metabolism, reduce gluconeogenesis, and decrease blood glucose levels after cerebral ischemic stress. The low expression of brain-derived neurotrophic factor following cerebral ischemia may be involved in the development of glucose intolerance.
Collapse
Affiliation(s)
- Xiaoliang Shu
- Department of Nutrition, Affiliated Dongfang Hospital of Tongji University, Shanghai 200120, China
| | - Yongsheng Zhang
- Department of Nutrition, First Affiliated Hospital of Guangxi Medical University, Nanning 530027, Guangxi Zhuang Autonomous Region, China
| | - Han Xu
- Department of Nutrition, Affiliated Dongfang Hospital of Tongji University, Shanghai 200120, China
| | - Kai Kang
- Department of Nutrition, Affiliated Dongfang Hospital of Tongji University, Shanghai 200120, China
| | - Donglian Cai
- Department of Nutrition, Affiliated Changhai Hospital of the Second Military Medical University of Chinese PLA, Shanghai 200433, China
| |
Collapse
|
31
|
Role of receptor tyrosine kinases and their ligands in glioblastoma. Cells 2014; 3:199-235. [PMID: 24709958 PMCID: PMC4092852 DOI: 10.3390/cells3020199] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2013] [Revised: 03/12/2014] [Accepted: 03/21/2014] [Indexed: 01/04/2023] Open
Abstract
Glioblastoma multiforme is the most frequent, aggressive and fatal type of brain tumor. Glioblastomas are characterized by their infiltrating nature, high proliferation rate and resistance to chemotherapy and radiation. Recently, oncologic therapy experienced a rapid evolution towards “targeted therapy,” which is the employment of drugs directed against particular targets that play essential roles in proliferation, survival and invasiveness of cancer cells. A number of molecules involved in signal transduction pathways are used as molecular targets for the treatment of various tumors. In fact, inhibitors of these molecules have already entered the clinic or are undergoing clinical trials. Cellular receptors are clear examples of such targets and in the case of glioblastoma multiforme, some of these receptors and their ligands have become relevant. In this review, the importance of glioblastoma multiforme in signaling pathways initiated by extracellular tyrosine kinase receptors such as EGFR, PDGFR and IGF-1R will be discussed. We will describe their ligands, family members, structure, activation mechanism, downstream molecules, as well as the interaction among these pathways. Lastly, we will provide an up-to-date review of the current targeted therapies in cancer, in particular glioblastoma that employ inhibitors of these pathways and their benefits.
Collapse
|
32
|
Abstract
Epidemics of obesity, diabetes, nonalcoholic fatty liver disease, and cognitive impairment/Alzheimer disease have emerged over the past 3 to 4 decades. These diseases share in common target-organ insulin resistance with a constellation of molecular and biochemical abnormalities that lead to organ/tissue degeneration over time. This article discusses the fundamental links among these diseases and how peripheral organ insulin resistance diseases contribute to cognitive impairment and neurodegeneration. A future role of endocrinologists and diabetologists could be to provide integrative diagnostic and treatment approaches for this collection of diseases that seem to share pathophysiological and pathogenetic bases.
Collapse
Affiliation(s)
- Suzanne M de la Monte
- Department of Pathology (Neuropathology), Rhode Island Hospital, Warren Alpert Medical School of Brown University, Providence, RI, USA; Department of Neurology, Rhode Island Hospital, Warren Alpert Medical School of Brown University, Providence, RI, USA; Department of Neurosurgery, Rhode Island Hospital, Warren Alpert Medical School of Brown University, Providence, RI, USA; Department of Medicine, Rhode Island Hospital, Warren Alpert Medical School of Brown University, Providence, RI, USA.
| |
Collapse
|
33
|
de la Monte SM, Tong M. Brain metabolic dysfunction at the core of Alzheimer's disease. Biochem Pharmacol 2013; 88:548-59. [PMID: 24380887 DOI: 10.1016/j.bcp.2013.12.012] [Citation(s) in RCA: 320] [Impact Index Per Article: 29.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2013] [Revised: 12/16/2013] [Accepted: 12/16/2013] [Indexed: 02/06/2023]
Abstract
Growing evidence supports the concept that Alzheimer's disease (AD) is fundamentally a metabolic disease with molecular and biochemical features that correspond with diabetes mellitus and other peripheral insulin resistance disorders. Brain insulin/IGF resistance and its consequences can readily account for most of the structural and functional abnormalities in AD. However, disease pathogenesis is complicated by the fact that AD can occur as a separate disease process, or arise in association with systemic insulin resistance diseases, including diabetes, obesity, and non-alcoholic fatty liver disease. Whether primary or secondary in origin, brain insulin/IGF resistance initiates a cascade of neurodegeneration that is propagated by metabolic dysfunction, increased oxidative and ER stress, neuro-inflammation, impaired cell survival, and dysregulated lipid metabolism. These injurious processes compromise neuronal and glial functions, reduce neurotransmitter homeostasis, and cause toxic oligomeric pTau and (amyloid beta peptide of amyloid beta precursor protein) AβPP-Aβ fibrils and insoluble aggregates (neurofibrillary tangles and plaques) to accumulate in brain. AD progresses due to: (1) activation of a harmful positive feedback loop that progressively worsens the effects of insulin resistance; and (2) the formation of ROS- and RNS-related lipid, protein, and DNA adducts that permanently damage basic cellular and molecular functions. Epidemiologic data suggest that insulin resistance diseases, including AD, are exposure-related in etiology. Furthermore, experimental and lifestyle trend data suggest chronic low-level nitrosamine exposures are responsible. These concepts offer opportunities to discover and implement new treatments and devise preventive measures to conquer the AD and other insulin resistance disease epidemics.
Collapse
Affiliation(s)
- Suzanne M de la Monte
- Departments of Pathology (Neuropathology), Rhode Island Hospital and the Warren Alpert Medical School of Brown University, Providence, RI, USA; Departments of Neurology, Rhode Island Hospital and the Warren Alpert Medical School of Brown University, Providence, RI, USA; Departments of Neurosurgery, Rhode Island Hospital and the Warren Alpert Medical School of Brown University, Providence, RI, USA; Departments of Medicine, Rhode Island Hospital and the Warren Alpert Medical School of Brown University, Providence, RI, USA.
| | - Ming Tong
- Departments of Medicine, Rhode Island Hospital and the Warren Alpert Medical School of Brown University, Providence, RI, USA
| |
Collapse
|
34
|
Diaz MR, Vollmer CC, Zamudio-Bulcock PA, Vollmer W, Blomquist SL, Morton RA, Everett JC, Zurek AA, Yu J, Orser BA, Valenzuela CF. Repeated intermittent alcohol exposure during the third trimester-equivalent increases expression of the GABA(A) receptor δ subunit in cerebellar granule neurons and delays motor development in rats. Neuropharmacology 2013; 79:262-74. [PMID: 24316160 DOI: 10.1016/j.neuropharm.2013.11.020] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2012] [Revised: 10/17/2013] [Accepted: 11/25/2013] [Indexed: 02/01/2023]
Abstract
Exposure to ethanol (EtOH) during fetal development can lead to long-lasting alterations, including deficits in fine motor skills and motor learning. Studies suggest that these are, in part, a consequence of cerebellar damage. Cerebellar granule neurons (CGNs) are the gateway of information into the cerebellar cortex. Functionally, CGNs are heavily regulated by phasic and tonic GABAergic inhibition from Golgi cell interneurons; however, the effect of EtOH exposure on the development of GABAergic transmission in immature CGNs has not been investigated. To model EtOH exposure during the 3rd trimester-equivalent of human pregnancy, neonatal pups were exposed intermittently to high levels of vaporized EtOH from postnatal day (P) 2 to P12. This exposure gradually increased pup serum EtOH concentrations (SECs) to ∼60 mM (∼0.28 g/dl) during the 4 h of exposure. EtOH levels gradually decreased to baseline 8 h after the end of exposure. Surprisingly, basal tonic and phasic GABAergic currents in CGNs were not significantly affected by postnatal alcohol exposure (PAE). However, PAE increased δ subunit expression at P28 as detected by immunohistochemical and western blot analyses. Also, electrophysiological studies with an agonist that is highly selective for δ-containing GABA(A) receptors, 4,5,6,7-tetrahydroisoxazolo[4,5-c]pyridine-3-ol (THIP), showed an increase in THIP-induced tonic current. Behavioral studies of PAE rats did not reveal any deficits in motor coordination, except for a delay in the acquisition of the mid-air righting reflex that was apparent at P15 to P18. These findings demonstrate that repeated intermittent exposure to high levels of EtOH during the equivalent of the last trimester of human pregnancy has significant but relatively subtle effects on motor coordination and GABAergic transmission in CGNs in rats.
Collapse
Affiliation(s)
- Marvin R Diaz
- Department of Neurosciences, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
| | - Cyndel C Vollmer
- Department of Neurosciences, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
| | - Paula A Zamudio-Bulcock
- Department of Neurosciences, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
| | - William Vollmer
- Department of Neurosciences, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
| | - Samantha L Blomquist
- Department of Neurosciences, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
| | - Russell A Morton
- Department of Neurosciences, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
| | - Julie C Everett
- Department of Neurosciences, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
| | - Agnieszka A Zurek
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Jieying Yu
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Beverley A Orser
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada; Department of Anesthesia, University of Toronto, Toronto, Ontario, Canada; Department of Anesthesia, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada
| | - C Fernando Valenzuela
- Department of Neurosciences, University of New Mexico Health Sciences Center, Albuquerque, NM, USA.
| |
Collapse
|
35
|
de la Monte SM. Intranasal insulin therapy for cognitive impairment and neurodegeneration: current state of the art. Expert Opin Drug Deliv 2013; 10:1699-709. [PMID: 24215447 PMCID: PMC4551402 DOI: 10.1517/17425247.2013.856877] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
INTRODUCTION Growing evidence supports the concept that insulin resistance plays an important role in the pathogenesis of cognitive impairment and neurodegeneration, including in Alzheimer's disease (AD). The metabolic hypothesis has led to the development and utilization of insulin- and insulin agonist-based treatments. Therapeutic challenges faced include the ability to provide effective treatments that do not require repeated injections and also the ability to minimize the potentially hazardous off-target effects. AREAS COVERED This review covers the role of intranasal insulin therapy for cognitive impairment and neurodegeneration, particularly AD. The literature reviewed focuses on data published within the past 5 years as this field is evolving rapidly. The review provides evidence that brain insulin resistance is an important and early abnormality in AD, and that increasing brain supply and utilization of insulin improves cognition and memory. Emphasis was placed on discussing outcomes of clinical trials and interpreting discordant results to clarify the benefits and limitations of intranasal insulin therapy. EXPERT OPINION Intranasal insulin therapy can efficiently and directly target the brain to support energy metabolism, myelin maintenance, cell survival and neuronal plasticity, which begin to fail in the early stages of neurodegeneration. Efforts must continue toward increasing the safety, efficacy and specificity of intranasal insulin therapy.
Collapse
Affiliation(s)
- Suzanne M de la Monte
- Rhode Island Hospital and the Warren Alpert Medical School of Brown University, Departments of Pathology (Neuropathology), Neurology, and Neurosurgery , Pierre Galletti Research Building, Claverick Street, Room 419, Providence, RI 02903 , USA +1 401 444 7364 ; +1 401 444 2939 ;
| |
Collapse
|
36
|
McCarthy N, Wetherill L, Lovely CB, Swartz ME, Foroud TM, Eberhart JK. Pdgfra protects against ethanol-induced craniofacial defects in a zebrafish model of FASD. Development 2013; 140:3254-65. [PMID: 23861062 PMCID: PMC3931738 DOI: 10.1242/dev.094938] [Citation(s) in RCA: 92] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Human birth defects are highly variable and this phenotypic variability can be influenced by both the environment and genetics. However, the synergistic interactions between these two variables are not well understood. Fetal alcohol spectrum disorders (FASD) is the umbrella term used to describe the wide range of deleterious outcomes following prenatal alcohol exposure. Although FASD are caused by prenatal ethanol exposure, FASD are thought to be genetically modulated, although the genes regulating sensitivity to ethanol teratogenesis are largely unknown. To identify potential ethanol-sensitive genes, we tested five known craniofacial mutants for ethanol sensitivity: cyp26b1, gata3, pdgfra, smad5 and smoothened. We found that only platelet-derived growth factor receptor alpha (pdgfra) interacted with ethanol during zebrafish craniofacial development. Analysis of the PDGF family in a human FASD genome-wide dataset links PDGFRA to craniofacial phenotypes in FASD, prompting a mechanistic understanding of this interaction. In zebrafish, untreated pdgfra mutants have cleft palate due to defective neural crest cell migration, whereas pdgfra heterozygotes develop normally. Ethanol-exposed pdgfra mutants have profound craniofacial defects that include the loss of the palatal skeleton and hypoplasia of the pharyngeal skeleton. Furthermore, ethanol treatment revealed latent haploinsufficiency, causing palatal defects in ∼62% of pdgfra heterozygotes. Neural crest apoptosis partially underlies these ethanol-induced defects in pdgfra mutants, demonstrating a protective role for Pdgfra. This protective role is mediated by the PI3K/mTOR pathway. Collectively, our results suggest a model where combined genetic and environmental inhibition of PI3K/mTOR signaling leads to variability within FASD.
Collapse
Affiliation(s)
- Neil McCarthy
- Department of Molecular and Cell and Developmental Biology, Institute for Cellular and Molecular Biology and Institute for Neuroscience, University of Texas, Austin, TX 78712, USA
| | | | | | | | | | | |
Collapse
|
37
|
Lee S, Tong M, Hang S, Deochand C, de la Monte S. CSF and Brain Indices of Insulin Resistance, Oxidative Stress and Neuro-Inflammation in Early versus Late Alzheimer's Disease. ACTA ACUST UNITED AC 2013; 3:128. [PMID: 25035815 PMCID: PMC4096626 DOI: 10.4172/2161-0460.1000128] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Alzheimer’s disease (AD) is characterized by progressive impairments in cognitive and behavioral functions with deficits in learning, memory and executive reasoning. Growing evidence points toward brain insulin and insulin-like growth factor (IGF) resistance-mediated metabolic derangements as critical etiologic factors in AD. This suggests that indices of insulin/IGF resistance and their consequences, i.e. oxidative stress, neuro-inflammation, and reduced neuronal plasticity, should be included in biomarker panels for AD. Herein, we examine a range of metabolic, inflammatory, stress, and neuronal plasticity related proteins in early AD, late AD, and aged control postmortem brain, postmortem ventricular fluid (VF), and clinical cerebrospinal fluid (CSF) samples. In AD brain, VF, and CSF samples the trends with respect to alterations in metabolic, neurotrophin, and stress indices were similar, but for pro-inflammatory cytokines, the patterns were discordant. With the greater severities of dementia and neurodegeneration, the differences from control were more pronounced for late AD (VF and brain) than early or moderate AD (brain, VF and CSF). The findings suggest that the inclusion of metabolic, neurotrophin, stress biomarkers in AβPP-Aβ+pTau CSF-based panels could provide more information about the status and progression of neurodegeneration, as well as aid in predicting progression from early- to late-stage AD. Furthermore, standardized multi-targeted molecular assays of neurodegeneration could help streamline postmortem diagnoses, including assessments of AD severity and pathology.
Collapse
Affiliation(s)
- Sarah Lee
- Department of Medicine, Rhode Island Hospital, Warren Alpert Medical School, Providence, RI, USA
| | - Ming Tong
- Department of Medicine, Rhode Island Hospital, Warren Alpert Medical School, Providence, RI, USA
| | - Steven Hang
- Department of Medicine, Warren Alpert Medical School, Providence, RI, USA
| | - Chetram Deochand
- Departments of Medicine, Rhode Island Hospital, Brown University, Providence, RI, USA
| | - Suzanne de la Monte
- Department of Medicine, Pathology (Neuropathology), Neurology and Neurosurgery, Rhode Island Hospital, Warren Alpert Medical School, Brown University, Providence, RI, USA
| |
Collapse
|
38
|
Motor Function Deficits Following Chronic Prenatal Ethanol Exposure are Linked to Impairments in Insulin/IGF, Notch and Wnt Signaling in the Cerebellum. JOURNAL OF DIABETES & METABOLISM 2013; 4:238. [PMID: 25035811 PMCID: PMC4096945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Fetal alcohol spectrum disorder (FASD) is associated with deficits in cerebellar function that can persist through adolescence. Previous studies demonstrated striking inhibition of insulin and insulin-like growth factor (IGF) signaling in ethanol-exposed cerebella. OBJECTIVES We sought to determine if FASD-induced impairments in motor function were associated with deficits in insulin/IGF signaling in juvenile cerebella. Given the growing evidence that insulin/IGF pathways cross-talk with Notch and Wnt to promote brain development and maturation; we also examined the integrity of canonical Wnt and Notch signaling networks in the brain following chronic prenatal ethanol exposure. METHODS Pregnant Long Evans rats were fed isocaloric liquid diets containing 0% or 24% ethanol by caloric content from gestation day 6 through delivery. Pups were subjected to rotarod testing on postnatal days (P) 15-16 and sacrificed on P30. Cerebella were used for molecular and biochemical analysis of insulin/IGF-1, canonical Wnt, and Notch signaling mechanisms. RESULTS Prenatal ethanol exposures impaired rotarod performance, inhibited signaling through insulin and IGF-1 receptors, IRS-1, and Akt, increased activation of GSK-3β, and broadly suppressed genes mediating the canonical Wnt and Notch networks. CONCLUSIONS Abnormalities in cerebellar function following chronic prenatal ethanol exposure are associated with inhibition of insulin/IGF, canonical Wnt, and Notch networks that cross-talk via GSK-3β. Effective therapeutic measures for FASD may require multi-pronged support of interrelated signaling networks that regulate brain development.
Collapse
|
39
|
Clark I, Atwood C, Bowen R, Paz-Filho G, Vissel B. Tumor necrosis factor-induced cerebral insulin resistance in Alzheimer's disease links numerous treatment rationales. Pharmacol Rev 2012; 64:1004-26. [PMID: 22966039 DOI: 10.1124/pr.112.005850] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
The evident limitations of the amyloid theory of the pathogenesis of Alzheimer's disease are increasingly putting alternatives in the spotlight. We argue here that a number of independently developing approaches to therapy-including specific and nonspecific anti-tumor necrosis factor (TNF) agents, apolipoprotein E mimetics, leptin, intranasal insulin, the glucagon-like peptide-1 mimetics and glycogen synthase kinase-3 (GSK-3) antagonists-are all part of an interlocking chain of events. All these approaches inform us that inflammation and thence cerebral insulin resistance constitute the pathway on which to focus for a successful clinical outcome in treating this disease. The key link in this chain presently absent is a recognition by Alzheimer's research community of the long-neglected history of TNF induction of insulin resistance. When this is incorporated into the bigger picture, it becomes evident that the interventions we discuss are not competing alternatives but equally valid approaches to correcting different parts of the same pathway to Alzheimer's disease. These treatments can be expected to be at least additive, and conceivably synergistic, in effect. Thus the inflammation, insulin resistance, GSK-3, and mitochondrial dysfunction hypotheses are not opposing ideas but stages of the same fundamental, overarching, pathway of Alzheimer's disease pathogenesis. The insight this provides into progenitor cells, including those involved in adult neurogenesis, is a key part of this approach. This pathway also has therapeutic implications for other circumstances in which brain TNF is pathologically increased, such as stroke, traumatic brain injury, and the infectious disease encephalopathies.
Collapse
Affiliation(s)
- Ian Clark
- Division of Medical Science and Biochemistry, Research School of Biology, Australian National University, Canberra ACT, Australia.
| | | | | | | | | |
Collapse
|
40
|
de la Monte SM. Therapeutic targets of brain insulin resistance in sporadic Alzheimer's disease. Front Biosci (Elite Ed) 2012. [PMID: 22201977 DOI: 10.2741/482] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Growing evidence supports roles for brain insulin and insulin-like growth factor (IGF) resistance and metabolic dysfunction in the pathogenesis of Alzheimer's disease (AD). Whether the underlying problem stems from a primary disorder of central nervous system (CNS) neurons and glia, or secondary effects of systemic diseases such as obesity, Type 2 diabetes, or metabolic syndrome, the end-results include impaired glucose utilization, mitochondrial dysfunction, increased oxidative stress, neuroinflammation, and the propagation of cascades that result in the accumulation of neurotoxic misfolded, aggregated, and ubiquitinated fibrillar proteins. This article reviews the roles of impaired insulin and IGF signaling to AD-associated neuronal loss, synaptic disconnection, tau hyperphosphorylation, amyloid-beta accumulation, and impaired energy metabolism, and discusses therapeutic strategies and lifestyle approaches that could be used to prevent, delay the onset, or reduce the severity of AD. Finally, it is critical to recognize that AD is heterogeneous and has a clinical course that fully develops over a period of several decades. Therefore, early and multi-modal preventive and treatment approaches should be regarded as essential.
Collapse
Affiliation(s)
- Suzanne M de la Monte
- Department of Neurology, Rhode Island Hospital and Alpert Medical School of Brown University, Providence, RI, USA.
| |
Collapse
|
41
|
de la Monte SM. Therapeutic targets of brain insulin resistance in sporadic Alzheimer's disease. Front Biosci (Elite Ed) 2012; 4:1582-1605. [PMID: 22201977 PMCID: PMC4550311 DOI: 10.2741/e482] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/31/2023]
Abstract
Growing evidence supports roles for brain insulin and insulin-like growth factor (IGF) resistance and metabolic dysfunction in the pathogenesis of Alzheimer's disease (AD). Whether the underlying problem stems from a primary disorder of central nervous system (CNS) neurons and glia, or secondary effects of systemic diseases such as obesity, Type 2 diabetes, or metabolic syndrome, the end-results include impaired glucose utilization, mitochondrial dysfunction, increased oxidative stress, neuroinflammation, and the propagation of cascades that result in the accumulation of neurotoxic misfolded, aggregated, and ubiquitinated fibrillar proteins. This article reviews the roles of impaired insulin and IGF signaling to AD-associated neuronal loss, synaptic disconnection, tau hyperphosphorylation, amyloid-beta accumulation, and impaired energy metabolism, and discusses therapeutic strategies and lifestyle approaches that could be used to prevent, delay the onset, or reduce the severity of AD. Finally, it is critical to recognize that AD is heterogeneous and has a clinical course that fully develops over a period of several decades. Therefore, early and multi-modal preventive and treatment approaches should be regarded as essential.
Collapse
Affiliation(s)
- Suzanne M de la Monte
- Department of Neurology, Rhode Island Hospital and Alpert Medical School of Brown University, Providence, RI, USA.
| |
Collapse
|
42
|
Vangipuram SD, Lyman WD. Ethanol affects differentiation-related pathways and suppresses Wnt signaling protein expression in human neural stem cells. Alcohol Clin Exp Res 2011; 36:788-97. [PMID: 22150777 DOI: 10.1111/j.1530-0277.2011.01682.x] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
BACKGROUND Prenatal exposure of the fetus to ethanol (EtOH) can be teratogenic. We previously showed that EtOH alters the cell fate of human neural stem cells (NSC). As Wnt signaling plays an important role in fetal brain development, we hypothesized that EtOH suppresses Wnt signaling protein expression in differentiating NSC and thereby contributes to fetal alcohol spectrum disorder. METHODS NSC isolated from fetal human brains were cultured in mitogenic media to induce neurospheres, which were dissociated into single-cell suspensions and used for all experiments. Equal numbers of NSC were cultured on lysine/laminin-coated plates for 96 hours in differentiating media containing 0, 20, or 100 mM EtOH. Total mRNA was isolated from samples containing 0 or 100 mM EtOH and changes in expression of 263 genes associated with neurogenesis and NSC differentiation were determined by Oligo GEArray technology. The biological impact of gene changes was estimated using a systems biology approach with pathway express software and KEGG database. Based on the pathways identified, expression of Wnt proteins (Wnt3a and Wnt5a), Wnt-receptor complex proteins (p-LRP6, LRP6, DVL2, and DVL3), Wnt antagonist Naked-2 (NKD-2), and downstream Wnt proteins (β-catenin, Tyr-p-GSK3β, Ser-p-GSK3β) were analyzed by Western blot. RESULTS Of the 263 genes examined, the expressions of 22 genes in differentiating NSC were either upwardly or downwardly affected by EtOH. These genes are associated with 5 pathways/cellular processes: axon guidance; hedgehog signaling; TGF-β signaling; cell adhesion molecules; and Wnt signaling. When compared to controls, EtOH, at both 20 and 100 mM concentrations, suppressed the expression of Wnt3a and Wnt5a, receptor complex proteins p-LRP6, LRP6 and DVL2, and cytoplasmic proteins Ser-p-GSK3β and β-catenin. Expression of NKD-2 and DVL3 remained unchanged and the expression of active Tyr-p-GSK3β increased significantly. CONCLUSIONS EtOH can significantly alter neural differentiation pathway-related gene expression and suppress Wnt signaling proteins in differentiating human NSC.
Collapse
Affiliation(s)
- Sharada D Vangipuram
- Children's Research Center of Michigan, The Carman and Ann Adams Department of Pediatrics, Wayne State University School of Medicine, Detroit, USA.
| | | |
Collapse
|