1
|
Peng Y, Yao SY, Chen Q, Jin H, Du MQ, Xue YH, Liu S. True or false? Alzheimer's disease is type 3 diabetes: Evidences from bench to bedside. Ageing Res Rev 2024; 99:102383. [PMID: 38955264 DOI: 10.1016/j.arr.2024.102383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 06/12/2024] [Accepted: 06/17/2024] [Indexed: 07/04/2024]
Abstract
Globally, Alzheimer's disease (AD) is the most widespread chronic neurodegenerative disorder, leading to cognitive impairment, such as aphasia and agnosia, as well as mental symptoms, like behavioral abnormalities, that place a heavy psychological and financial burden on the families of the afflicted. Unfortunately, no particular medications exist to treat AD, as the current treatments only impede its progression.The link between AD and type 2 diabetes (T2D) has been increasingly revealed by research; the danger of developing both AD and T2D rises exponentially with age, with T2D being especially prone to AD. This has propelled researchers to investigate the mechanism(s) underlying this connection. A critical review of the relationship between insulin resistance, Aβ, oxidative stress, mitochondrial hypothesis, abnormal phosphorylation of Tau protein, inflammatory response, high blood glucose levels, neurotransmitters and signaling pathways, vascular issues in AD and diabetes, and the similarities between the two diseases, is presented in this review. Grasping the essential mechanisms behind this detrimental interaction may offer chances to devise successful therapeutic strategies.
Collapse
Affiliation(s)
- Yong Peng
- Department of Neurology, Affiliated First Hospital of Hunan Traditional Chinese Medical College, Zhuzhou, Hunan, China; Department of Neurology, Affiliated Provincial Traditional Chinese Medical Hospital of Hunan University of Chinese Medicine, Zhuzhou, Hunan, China.
| | - Shun-Yu Yao
- Department of Neurology, Affiliated First Hospital of Hunan Traditional Chinese Medical College, Zhuzhou, Hunan, China; Department of Neurology, Affiliated Provincial Traditional Chinese Medical Hospital of Hunan University of Chinese Medicine, Zhuzhou, Hunan, China
| | - Quan Chen
- Department of Neurology, Affiliated First Hospital of Hunan Traditional Chinese Medical College, Zhuzhou, Hunan, China; Department of Neurology, Affiliated Provincial Traditional Chinese Medical Hospital of Hunan University of Chinese Medicine, Zhuzhou, Hunan, China
| | - Hong Jin
- Department of Neurology, Affiliated First Hospital of Hunan Traditional Chinese Medical College, Zhuzhou, Hunan, China; Department of Neurology, Affiliated Provincial Traditional Chinese Medical Hospital of Hunan University of Chinese Medicine, Zhuzhou, Hunan, China
| | - Miao-Qiao Du
- Department of Neurology, Affiliated First Hospital of Hunan Traditional Chinese Medical College, Zhuzhou, Hunan, China; Department of Neurology, Affiliated Provincial Traditional Chinese Medical Hospital of Hunan University of Chinese Medicine, Zhuzhou, Hunan, China
| | - Ya-Hui Xue
- Department of Neurology, Affiliated First Hospital of Hunan Traditional Chinese Medical College, Zhuzhou, Hunan, China; Department of Neurology, Affiliated Provincial Traditional Chinese Medical Hospital of Hunan University of Chinese Medicine, Zhuzhou, Hunan, China
| | - Shu Liu
- Department of Neurology, Affiliated First Hospital of Hunan Traditional Chinese Medical College, Zhuzhou, Hunan, China; Department of Neurology, Affiliated Provincial Traditional Chinese Medical Hospital of Hunan University of Chinese Medicine, Zhuzhou, Hunan, China
| |
Collapse
|
2
|
Hernandez AR, Parker E, Babar M, Banerjee A, Ding S, Simley A, Buford TW. Microbiome-driven alterations in metabolic pathways and impaired cognition in aged female TgF344-AD rats. AGING BRAIN 2024; 5:100119. [PMID: 38881651 PMCID: PMC11179252 DOI: 10.1016/j.nbas.2024.100119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 05/09/2024] [Accepted: 05/28/2024] [Indexed: 06/18/2024] Open
Abstract
Alzheimer's disease (AD) not only affects cognition and neuropathology, but several other facets capable of negatively impacting quality of life and potentially driving impairments, including altered gut microbiome (GMB) composition and metabolism. Aged (20 + mo) female TgF344-AD and wildtype rats were cognitively characterized on several tasks incorporating several cognitive domains, including task acquisition, object recognition memory, anxiety-like behaviors, and spatial navigation. Additionally, metabolic phenotyping, GMB sequencing throughout the intestinal tract (duodenum, jejunum, ileum, colon, and feces), neuropathological burden assessment and marker gene functional abundance predictions (PICRUSt2) were conducted. TgF344-AD rats demonstrated significant cognitive impairment in multiple domains, as well as regionally specific GMB dysbiosis. Relationships between peripheral factors were investigated using Canonical Correspondence Analysis (CCA), revealing correlations between GMB changes and both cognitive and metabolic factors. Moreover, communities of gut microbes contributing to essential metabolic pathways were significantly altered in TgF344-AD rats. These data indicate dysbiosis may affect cognitive outcomes in AD through alterations in metabolism-related enzymatic pathways that are necessary for proper brain function. Moreover, these changes were mostly observed in intestinal segments required for carbohydrate digestion, not fecal samples. These data support the targeting of intestinal and microbiome health for the treatment of AD.
Collapse
Affiliation(s)
- Abbi R Hernandez
- Department of Medicine, Division of Geriatrics, Gerontology & Palliative Care, University of Alabama at Birmingham, Birmingham, AL 35205, USA
| | - Erik Parker
- Department of Epidemiology and Biostatistics, School of Public Health, Indiana University-Bloomington, Bloomington, IN 47405, USA
| | - Maham Babar
- Department of Medicine, Division of Geriatrics, Gerontology & Palliative Care, University of Alabama at Birmingham, Birmingham, AL 35205, USA
| | - Anisha Banerjee
- Department of Medicine, Division of Geriatrics, Gerontology & Palliative Care, University of Alabama at Birmingham, Birmingham, AL 35205, USA
| | - Sarah Ding
- Department of Medicine, Division of Geriatrics, Gerontology & Palliative Care, University of Alabama at Birmingham, Birmingham, AL 35205, USA
| | - Alexis Simley
- Department of Medicine, Division of Geriatrics, Gerontology & Palliative Care, University of Alabama at Birmingham, Birmingham, AL 35205, USA
| | - Thomas W Buford
- Department of Medicine, Division of Geriatrics, Gerontology & Palliative Care, University of Alabama at Birmingham, Birmingham, AL 35205, USA
- Birmingham/Atlanta VA GRECC, Birmingham VA Medical Center, Birmingham, AL 35244, USA
| |
Collapse
|
3
|
de la Monte SM. Conquering Insulin Network Dysfunctions in Alzheimer's Disease: Where Are We Today? J Alzheimers Dis 2024; 101:S317-S343. [PMID: 39422949 DOI: 10.3233/jad-240069] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
Functional impairments in the brain's insulin and insulin-like growth factor (IGF) signal transduction networks are recognized mediators of dysregulated energy metabolism, a major driver of the Alzheimer's disease (AD) neurodegeneration cascade. AD-associated insulin-deficient and insulin-resistant states mimic those of diabetes mellitus and affect all cell types in the brain. Besides accounting for abundant amyloid-β and hyperphosphorylated tau lesions in AD, insulin/IGF pathway dysfunctions cause cortical atrophy, loss of synaptic plasticity, white matter myelin/oligodendrocyte degeneration, astrocyte and microglial neuroinflammation and oxidative stress, deficits in energy metabolism, mitochondrial dysfunction, and microvascular disease. These same neuropathological processes have been linked to cognitive impairment in type 2 diabetes mellitus, Parkinson's disease, and vascular dementia. Strategies to address metabolic mediators of cognitive impairment have been borrowed from diabetes and other insulin-resistant diseases and leveraged on preclinical AD model data. The repurposing of diabetes drugs led to clinical trials with intranasal insulin, followed by insulin sensitizers including metformin and peroxisome-proliferator-activated receptor agonists, and then incretin mimetics primarily targeting GLP-1 receptors. In addition, other glucose-lowering agents have been tested for their efficacy in preventing cognitive declines. The strengths and limitations of these approaches are discussed. The main conclusion of this review is that we have now arrived at a stage in which it is time to address long-term deficits in trophic factor availability and receptor responsiveness, signaling abnormalities that extend beyond insulin and include IGFs and interconnected pathways, and the need for multi-pronged rather than single-pronged therapeutic targeting to remediate AD and other forms of neurodegeneration.
Collapse
Affiliation(s)
- Suzanne M de la Monte
- Departments of Pathology and Laboratory Medicine, Medicine, Neurology and Neurosurgery, Rhode Island Hospital, Lifespan Academic Institutions, and the Warren Alpert Medical School of Brown University, Providence, RI, USA
| |
Collapse
|
4
|
Sharma P, Aggarwal K, Awasthi R, Kulkarni GT, Sharma B. Behavioral and biochemical investigations to explore the efficacy of quercetin and folacin in experimental diabetes induced vascular endothelium dysfunction and associated dementia in rats. J Basic Clin Physiol Pharmacol 2023; 34:603-615. [PMID: 34161695 DOI: 10.1515/jbcpp-2020-0159] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2020] [Accepted: 04/16/2021] [Indexed: 12/19/2022]
Abstract
OBJECTIVES Vascular dementia (VaD), being strongly associated with metabolic conditions is a major health concern around the world. Diabetes is a major risk factor for the development of VaD. This study investigates the efficacy of quercetin and folacin in diabetes induced vascular endothelium dysfunction and related dementia. METHODS Single dose streptozotocin (STZ) (50 mg/kg i.p) was administered to albino Wistar rats (male, 200-250 g) by dissolving in citrate buffer. Morris water maze (MWM) and attentional set shifting tests were used to assess the spatial learning, memory, reversal learning, and executive functioning in animals. Body weight, serum glucose, serum nitrite/nitrate, vascular endothelial function, aortic superoxide anion, brains' oxidative markers (thiobarbituric acid reactive species-TBARS, reduced glutathione-GSH, superoxide dismutase-SOD, and catalase-CAT), mitochondrial enzyme complex (I, II, and IV), inflammatory markers (interleukin-IL-6, IL-10, tumor necrosis factor-TNF-α, and myeloperoxidase-MPO), and acetylcholinesterase activity-AChE were also assessed. Quercetin (30 mg kg-1/60 mg kg-1) and folacin (30 mg kg-1/60 mg kg-1) were used as the treatment drugs. Donepezil (0.5 mg kg-1) was used as a positive control. RESULTS STZ administered rats showed reduction in learning, memory, reversal learning, executive functioning, impairment in endothelial function, increase in brains' oxidative stress; inflammation; AChE activity, and decrease in mitochondrial complex (I, II, and IV) activity. Administration of quercetin and folacin in two different doses, significantly attenuated the STZ induced diabetes induced impairments in the behavioral, endothelial, and biochemical parameters. CONCLUSIONS STZ administration caused diabetes and VaD which was attenuated by the administration of quercetin and folacin. Therefore, these agents may be studied further for the assessment of their full potential in diabetes induced VaD conditions.
Collapse
Affiliation(s)
- Poonam Sharma
- Department of Pharmacology, Amity Institute of Pharmacy, Amity University, Noida, Uttar Pradesh, India
| | - Khushboo Aggarwal
- Department of Pharmacology, School of Pharmacy, Bharat Institute of Technology, Meerut, India
| | - Rajendra Awasthi
- Amity Institute of Pharmacy, Amity University, Noida, Uttar Pradesh, India
| | - Giriraj T Kulkarni
- Amity Institute of Pharmacy, Amity University, Noida, Uttar Pradesh, India
| | - Bhupesh Sharma
- Department of Pharmacology, Amity Institute of Pharmacy, Amity University, Noida, Uttar Pradesh, India
- CNS Pharmacology, Conscience Research, Delhi, India
| |
Collapse
|
5
|
Richter E, Geetha T, Burnett D, Broderick TL, Babu JR. The Effects of Momordica charantia on Type 2 Diabetes Mellitus and Alzheimer's Disease. Int J Mol Sci 2023; 24:ijms24054643. [PMID: 36902074 PMCID: PMC10002567 DOI: 10.3390/ijms24054643] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 02/21/2023] [Accepted: 02/23/2023] [Indexed: 03/06/2023] Open
Abstract
T2DM is a complex metabolic disorder characterized by hyperglycemia and glucose intolerance. It is recognized as one of the most common metabolic disorders and its prevalence continues to raise major concerns in healthcare globally. Alzheimer's disease (AD) is a gradual neurodegenerative brain disorder characterized by the chronic loss of cognitive and behavioral function. Recent research suggests a link between the two diseases. Considering the shared characteristics of both diseases, common therapeutic and preventive agents are effective. Certain bioactive compounds such as polyphenols, vitamins, and minerals found in vegetables and fruits can have antioxidant and anti-inflammatory effects that allow for preventative or potential treatment options for T2DM and AD. Recently, it has been estimated that up to one-third of patients with diabetes use some form of complementary and alternative medicine. Increasing evidence from cell or animal models suggests that bioactive compounds may have a direct effect on reducing hyperglycemia, amplifying insulin secretion, and blocking the formation of amyloid plaques. One plant that has received substantial recognition for its numerous bioactive properties is Momordica charantia (M. charantia), otherwise known as bitter melon, bitter gourd, karela, and balsam pear. M. charantia is utilized for its glucose-lowering effects and is often used as a treatment for diabetes and related metabolic conditions amongst the indigenous populations of Asia, South America, India, and East Africa. Several pre-clinical studies have documented the beneficial effects of M. charantia through various postulated mechanisms. Throughout this review, the underlying molecular mechanisms of the bioactive components of M. charantia will be highlighted. More studies will be necessary to establish the clinical efficacy of the bioactive compounds within M. charantia to effectively determine its pertinence in the treatment of metabolic disorders and neurodegenerative diseases, such as T2DM and AD.
Collapse
Affiliation(s)
- Erika Richter
- Department of Nutritional Sciences, Auburn University, Auburn, AL 36849, USA
| | - Thangiah Geetha
- Department of Nutritional Sciences, Auburn University, Auburn, AL 36849, USA
- Boshell Metabolic Diseases and Diabetes Program, Auburn University, Auburn, AL 36849, USA
| | - Donna Burnett
- Department of Nutritional Sciences, Auburn University, Auburn, AL 36849, USA
- Boshell Metabolic Diseases and Diabetes Program, Auburn University, Auburn, AL 36849, USA
| | - Tom L. Broderick
- Department of Physiology, Laboratory of Diabetes and Exercise Metabolism, College of Graduate Studies, Midwestern University, Glendale, AZ 85308, USA
| | - Jeganathan Ramesh Babu
- Department of Nutritional Sciences, Auburn University, Auburn, AL 36849, USA
- Boshell Metabolic Diseases and Diabetes Program, Auburn University, Auburn, AL 36849, USA
- Correspondence: ; Tel.: +1-223-844-3840
| |
Collapse
|
6
|
Bakrim S, Aboulaghras S, El Menyiy N, El Omari N, Assaggaf H, Lee LH, Montesano D, Gallo M, Zengin G, AlDhaheri Y, Bouyahya A. Phytochemical Compounds and Nanoparticles as Phytochemical Delivery Systems for Alzheimer's Disease Management. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27249043. [PMID: 36558176 PMCID: PMC9781052 DOI: 10.3390/molecules27249043] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Revised: 12/04/2022] [Accepted: 12/09/2022] [Indexed: 12/23/2022]
Abstract
Alzheimer's disease remains one of the most widespread neurodegenerative reasons for dementia worldwide and is associated with considerable mortality and morbidity. Therefore, it has been considered a priority for research. Indeed, several risk factors are involved in the complexity of the therapeutic ways of this pathology, including age, traumatic brain injury, genetics, exposure to aluminum, infections, diabetes, vascular diseases, hypertension, dyslipidemia, and obesity. The pathophysiology of Alzheimer's disease is mostly associated with hyperphosphorylated protein in the neuronal cytoplasm and extracellular plaques of the insoluble β-amyloid peptide. Therefore, the management of this pathology needs the screening of drugs targeting different pathological levels, such as acetylcholinesterase (AchE), amyloid β formation, and lipoxygenase inhibitors. Among the pharmacological strategies used for the management of Alzheimer's disease, natural drugs are considered a promising therapeutic strategy. Indeed, bioactive compounds isolated from different natural sources exhibit important anti-Alzheimer effects by their effectiveness in promoting neuroplasticity and protecting against neurodegeneration as well as neuroinflammation and oxidative stress in the brain. These effects involve different sub-cellular, cellular, and/or molecular mechanisms, such as the inhibition of acetylcholinesterase (AchE), the modulation of signaling pathways, and the inhibition of oxidative stress. Moreover, some nanoparticles were recently used as phytochemical delivery systems to improve the effects of phytochemical compounds against Alzheimer's disease. Therefore, the present work aims to provide a comprehensive overview of the key advances concerning nano-drug delivery applications of phytochemicals for Alzheimer's disease management.
Collapse
Affiliation(s)
- Saad Bakrim
- Geo-Bio-Environment Engineering and Innovation Laboratory, Molecular Engineering, Biotechnology and Innovation Team, Polydisciplinary Faculty of Taroudant, Ibn Zohr University, Agadir 80000, Morocco
| | - Sara Aboulaghras
- Physiology and Physiopathology Team, Faculty of Sciences, Genomic of Human Pathologies Research, Mohammed V University in Rabat, Rabat 10100, Morocco
| | - Naoual El Menyiy
- Laboratory of Pharmacology, National Agency of Medicinal and Aromatic Plants, Taounate 34025, Morocco
| | - Nasreddine El Omari
- Laboratory of Histology, Embryology and Cytogenetic, Faculty of Medicine and Pharmacy, Mohammed V University in Rabat, Rabat 10100, Morocco
| | - Hamza Assaggaf
- Department of Laboratory Medicine, Faculty of Applied Medical Sciences, Umm Al-Qura University, Makkah 21955, Saudi Arabia
| | - Learn-Han Lee
- Novel Bacteria and Drug Discovery Research Group (NBDD), Microbiome and Bioresource Research Strength (MBRS), Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Subang Jaya 47500, Malaysia
| | - Domenico Montesano
- Department of Pharmacy, University of Naples Federico II, Via D. Montesano 49, 80131 Naples, Italy
| | - Monica Gallo
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Via Pansini 5, 80131 Naples, Italy
| | - Gokhan Zengin
- Department of Biology, Science Faculty, Selcuk University, 42130 Konya, Turkey
- Correspondence: (G.Z.); (Y.A.); (A.B.)
| | - Yusra AlDhaheri
- Department of Biology, College of Science, United Arab Emirates University, Al Ain 15551, United Arab Emirates
- Correspondence: (G.Z.); (Y.A.); (A.B.)
| | - Abdelhakim Bouyahya
- Laboratory of Human Pathologies Biology, Department of Biology, Faculty of Sciences, Mohammed V University in Rabat, Rabat 10106, Morocco
- Correspondence: (G.Z.); (Y.A.); (A.B.)
| |
Collapse
|
7
|
Yang J, Wang Z, Fu Y, Xu J, Zhang Y, Qin W, Zhang Q. Prediction value of the genetic risk of type 2 diabetes on the amnestic mild cognitive impairment conversion to Alzheimer’s disease. Front Aging Neurosci 2022; 14:964463. [PMID: 36185474 PMCID: PMC9521369 DOI: 10.3389/fnagi.2022.964463] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Accepted: 08/23/2022] [Indexed: 11/23/2022] Open
Abstract
Amnestic mild cognitive impairment (aMCI) and Type 2 diabetes mellitus (T2DM) are both important risk factors for Alzheimer’s disease (AD). We aimed to investigate whether a T2DM-specific polygenic risk score (PRSsT2DM) can predict the conversion of aMCI to AD and further explore the underlying neurological mechanism. All aMCI patients were from the Alzheimer’s disease Neuroimaging Initiative (ADNI) database and were divided into conversion (aMCI-C, n = 164) and stable (aMCI-S, n = 222) groups. PRSsT2DM was calculated by PRSice-2 software to explore the predictive efficacy of the aMCI conversion to AD. We found that PRSsT2DM could independently predict the aMCI conversion to AD after removing the common variants of these two diseases. PRSsT2DM was significantly negatively correlated with gray matter volume (GMV) of the right superior frontal gyrus in the aMCI-C group. In all aMCI patients, PRSsT2DM was significantly negatively correlated with the cortical volume of the right superior occipital gyrus. The cortical volume of the right superior occipital gyrus could significantly mediate the association between PRSsT2DM and aMCI conversion. Gene-based analysis showed that T2DM-specific genes are highly expressed in cortical neurons and involved in ion and protein binding, neural development and generation, cell junction and projection, and PI3K-Akt and MAPK signaling pathway, which might increase the aMCI conversion by affecting the Tau phosphorylation and amyloid-beta (Aβ) accumulation. Therefore, the PRSsT2DM could be used as a measure to predict the conversion of aMCI to AD.
Collapse
|
8
|
Querfurth H, Marshall J, Parang K, Rioult-Pedotti MS, Tiwari R, Kwon B, Reisinger S, Lee HK. A PDK-1 allosteric agonist neutralizes insulin signaling derangements and beta-amyloid toxicity in neuronal cells and in vitro. PLoS One 2022; 17:e0261696. [PMID: 35061720 PMCID: PMC8782417 DOI: 10.1371/journal.pone.0261696] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Accepted: 12/08/2021] [Indexed: 01/09/2023] Open
Abstract
The Alzheimer's brain is affected by multiple pathophysiological processes, which include a unique, organ-specific form of insulin resistance that begins early in its course. An additional complexity arises from the four-fold risk of Alzheimer's Disease (AD) in type 2 diabetics, however there is no definitive proof of causation. Several strategies to improve brain insulin signaling have been proposed and some have been clinically tested. We report findings on a small allosteric molecule that reverses several indices of insulin insensitivity in both cell culture and in vitro models of AD that emphasize the intracellular accumulation of β-amyloid (Aβi). PS48, a chlorophenyl pentenoic acid, is an allosteric activator of PDK-1, which is an Akt-kinase in the insulin/PI3K pathway. PS48 was active at 10 nM to 1 μM in restoring normal insulin-dependent Akt activation and in mitigating Aβi peptide toxicity. Synaptic plasticity (LTP) in prefrontal cortical slices from normal rat exposed to Aβ oligomers also benefited from PS48. During these experiments, neither overstimulation of PI3K/Akt signaling nor toxic effects on cells was observed. Another neurotoxicity model producing insulin insensitivity, utilizing palmitic acid, also responded to PS48 treatment, thus validating the target and indicating that its therapeutic potential may extend outside of β-amyloid reliance. The described in vitro and cell based-in vitro coupled enzymatic assay systems proved suitable platforms to screen a preliminary library of new analogs.
Collapse
Affiliation(s)
- Henry Querfurth
- Department of Neurology, Tufts Medical Center, Boston, MA, United States of America
| | - John Marshall
- Department of Molecular Pharmacology, Physiology, and Biotechnology, Brown University, Providence, RI, United States of America
| | - Keykavous Parang
- Center for Targeted Drug Delivery, Chapman University, School of Pharmacology, Irvine, CA United States of America
| | - Mengia S. Rioult-Pedotti
- Department of Molecular Pharmacology, Physiology, and Biotechnology, Brown University, Providence, RI, United States of America
- Department of Neurology, Clinical Neurorehabilitation, University of Zurich, Zurich, Switzerland
| | - Rakesh Tiwari
- Center for Targeted Drug Delivery, Chapman University, School of Pharmacology, Irvine, CA United States of America
| | - Bumsup Kwon
- Department of Neurology, Rhode Island Hospital, Providence, RI, United States of America
| | | | - Han-Kyu Lee
- Department of Neurology, Tufts Medical Center, Boston, MA, United States of America
| |
Collapse
|
9
|
Role of Phytoconstituents as PPAR Agonists: Implications for Neurodegenerative Disorders. Biomedicines 2021; 9:biomedicines9121914. [PMID: 34944727 PMCID: PMC8698906 DOI: 10.3390/biomedicines9121914] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 12/09/2021] [Accepted: 12/11/2021] [Indexed: 12/16/2022] Open
Abstract
Peroxisome proliferator-activated receptors (PPAR-γ, PPAR-α, and PPAR-β/δ) are ligand-dependent nuclear receptors that play a critical role in the regulation of hundreds of genes through their activation. Their expression and targeted activation play an important role in the treatment of a variety of diseases, including neurodegenerative, cardiovascular, diabetes, and cancer. In recent years, several reviews have been published describing the therapeutic potential of PPAR agonists (natural or synthetic) in the disorders listed above; however, no comprehensive report defining the role of naturally derived phytoconstituents as PPAR agonists targeting neurodegenerative diseases has been published. This review will focus on the role of phytoconstituents as PPAR agonists and the relevant preclinical studies and mechanistic insights into their neuroprotective effects. Exemplary research includes flavonoids, fatty acids, cannabinoids, curcumin, genistein, capsaicin, and piperine, all of which have been shown to be PPAR agonists either directly or indirectly. Additionally, a few studies have demonstrated the use of clinical samples in in vitro investigations. The role of the fruit fly Drosophila melanogaster as a potential model for studying neurodegenerative diseases has also been highlighted.
Collapse
|
10
|
Behl T, Arora A, Sehgal A, Singh S, Sharma N, Bhatia S, Al-Harrasi A, Bungau S, Mostafavi E. Molecular and Biochemical Pathways Encompassing Diabetes Mellitus and Dementia. CNS & NEUROLOGICAL DISORDERS-DRUG TARGETS 2021; 21:542-556. [PMID: 34758720 DOI: 10.2174/1871527320666211110115257] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 08/25/2021] [Accepted: 09/15/2021] [Indexed: 11/22/2022]
Abstract
Diabetes mellitus is a major metabolic disorder that has now emerged as an epidemic, and it affects the brain through an array of pathways. Diabetes mellitus patients can develop pathological changes in the brain, which eventually take the shape of mild cognitive impairment progressing to Alzheimer's Disease. A number of preclinical and clinical studies demonstrate this fact, and it comes out to be those molecular pathways such as amyloidogenesis, oxidative stress, inflammation, and impaired insulin signaling are identical in diabetes mellitus and dementia. However, the critical player involved in the vicious cycle of diabetes mellitus and dementia is insulin, whose signaling, when impaired in diabetes mellitus (both type 1 and 2), leads to a decline in cognition, although other pathways are also essential contributors. Moreover, it is not only that diabetes mellitus patients indicate cognitive decline at a later stage; many Alzheimer's Disease patients also reflect symptoms of diabetes mellitus, thus creating a vicious cycle inculcating a web of complex molecular mechanisms and hence categorizing Alzheimer's Disease as 'brain diabetes'. Thus, it is practical to suggest that anti-diabetic drugs are beneficial in Alzheimer's Disease; but only smaller trials, not the larger ones, have showcased positive outcomes mainly because of the late onset of therapy. Therefore, it is extremely important to develop more of such molecules that target insulin in dementia patients along with such methods that diagnose impaired insulin signaling and the associated cognitive decline so that early therapy may be initiated and the progression of the disease be prevented.
Collapse
Affiliation(s)
- Tapan Behl
- Chitkara College of Pharmacy, Chitkara University, Punjab. India
| | - Arpita Arora
- Chitkara College of Pharmacy, Chitkara University, Punjab. India
| | - Aayush Sehgal
- Chitkara College of Pharmacy, Chitkara University, Punjab. India
| | - Sukhbir Singh
- Chitkara College of Pharmacy, Chitkara University, Punjab. India
| | - Neelam Sharma
- Chitkara College of Pharmacy, Chitkara University, Punjab. India
| | - Saurabh Bhatia
- Amity Institute of Pharmacy, Amity University, Haryana. India
| | - Ahmed Al-Harrasi
- Natural & Medical Sciences Research Centre, University of Nizwa, Nizwa. Oman
| | - Simona Bungau
- Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, Oradea. Romania
| | - Ebrahim Mostafavi
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA. United States
| |
Collapse
|
11
|
Ahmad W, Ebert PR. Suppression of a core metabolic enzyme dihydrolipoamide dehydrogenase ( dld) protects against amyloid beta toxicity in C. elegans model of Alzheimer's disease. Genes Dis 2021; 8:849-866. [PMID: 34522713 PMCID: PMC8427249 DOI: 10.1016/j.gendis.2020.08.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 07/24/2020] [Accepted: 08/14/2020] [Indexed: 01/24/2023] Open
Abstract
A decrease in energy metabolism is associated with Alzheimer's disease (AD), but it is not known whether the observed decrease exacerbates or protects against the disease. The importance of energy metabolism in AD is reinforced by the observation that variants of dihydrolipoamide dehydrogenase (DLD), is genetically linked to late-onset AD. To determine whether DLD is a suitable therapeutic target, we suppressed the dld-1 gene in Caenorhabditis elegans that express human Aβ peptide in either muscles or neurons. Suppression of the dld-1 gene resulted in significant restoration of vitality and function that had been degraded by Aβ pathology. This included protection of neurons and muscles cells. The observed decrease in proteotoxicity was associated with a decrease in the formation of toxic oligomers rather than a decrease in the abundance of the Aβ peptide. The mitochondrial uncoupler, carbonyl cyanide 4-(trifluoromethoxy) phenylhydrazone (FCCP), which like dld-1 gene expression inhibits ATP synthesis, had no significant effect on Aβ toxicity. Proteomics data analysis revealed that beneficial effects after dld-1 suppression could be due to change in energy metabolism and activation of the pathways associated with proteasomal degradation, improved cell signaling and longevity. Thus, some features unique to dld-1 gene suppression are responsible for the therapeutic benefit. By direct genetic intervention, we have shown that acute inhibition of dld-1 gene function may be therapeutically beneficial. This result supports the hypothesis that lowering energy metabolism protects against Aβ pathogenicity and that DLD warrants further investigation as a therapeutic target.
Collapse
Affiliation(s)
- Waqar Ahmad
- School of Biological Sciences, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Paul R. Ebert
- School of Biological Sciences, The University of Queensland, Brisbane, QLD 4072, Australia
| |
Collapse
|
12
|
Nutraceutical and Probiotic Approaches to Examine Molecular Interactions of the Amyloid Precursor Protein APP in Drosophila Models of Alzheimer's Disease. Int J Mol Sci 2021; 22:ijms22137022. [PMID: 34209883 PMCID: PMC8269328 DOI: 10.3390/ijms22137022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 06/24/2021] [Accepted: 06/24/2021] [Indexed: 12/12/2022] Open
Abstract
Studies using animal models have shed light into the molecular and cellular basis for the neuropathology observed in patients with Alzheimer’s disease (AD). In particular, the role of the amyloid precursor protein (APP) plays a crucial role in the formation of senile plaques and aging-dependent degeneration. Here, we focus our review on recent findings using the Drosophila AD model to expand our understanding of APP molecular function and interactions, including insights gained from the fly homolog APP-like (APPL). Finally, as there is still no cure for AD, we review some approaches that have shown promising results in ameliorating AD-associated phenotypes, with special attention on the use of nutraceuticals and their molecular effects, as well as interactions with the gut microbiome. Overall, the phenomena described here are of fundamental significance for understanding network development and degeneration. Given the highly conserved nature of fundamental signaling pathways, the insight gained from animal models such as Drosophila melanogaster will likely advance the understanding of the mammalian brain, and thus be relevant to human health.
Collapse
|
13
|
Nanjundaiah S, Chidambaram H, Chandrashekar M, Chinnathambi S. Role of Microglia in Regulating Cholesterol and Tau Pathology in Alzheimer's Disease. Cell Mol Neurobiol 2021; 41:651-668. [PMID: 32468440 DOI: 10.1007/s10571-020-00883-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Accepted: 05/19/2020] [Indexed: 01/21/2023]
Abstract
Cholesterol, a principal constituent of the cell membrane, plays a crucial role in the brain by regulating the synaptic transmission, neuronal signaling, as well as neurodegenerative diseases. Defects in the cholesterol trafficking are associated with enhanced generation of hyperphosphorylated Tau and Amyloid-β protein. Tau, a major microtubule-associated protein in the brain, is the key regulator of the mature neuron. Abnormally hyperphosphorylated Tau hampers the major functions related to microtubule assembly by promoting neurofibrillary tangles of paired helical filaments, twisted ribbons, and straight filaments. The observed pathological changes due to impaired cholesterol and Tau protein accumulation cause Alzheimer's disease. Thus, in order to regulate the pathogenesis of Alzheimer's disease, regulation of cholesterol metabolism, as well as Tau phosphorylation, is essential. The current review provides an overview of (1) cholesterol synthesis in the brain, neurons, astrocytes, and microglia; (2) the mechanism involved in modulating cholesterol concentration between the astrocytes and brain; (3) major mechanisms involved in the hyperphosphorylation of Tau and amyloid-β protein; and (4) microglial involvement in its regulation. Thus, the answering key questions will provide an in-depth information on microglia involvement in managing the pathogenesis of cholesterol-modulated hyperphosphorylated Tau protein.
Collapse
Affiliation(s)
- Shwetha Nanjundaiah
- Neurobiology Group, Division of Biochemical Sciences, CSIR-National Chemical Laboratory (CSIR-NCL), Dr. Homi Bhabha Road, Pune, 411008, India
| | - Hariharakrishnan Chidambaram
- Neurobiology Group, Division of Biochemical Sciences, CSIR-National Chemical Laboratory (CSIR-NCL), Dr. Homi Bhabha Road, Pune, 411008, India
- Academy of Scientific and Innovative Research (AcSIR), New Delhi, 110025, India
| | - Madhura Chandrashekar
- School of Biomedical Engineering and Sciences, MIT University, Loni Kalbhor, Pune, 412201, India
| | - Subashchandrabose Chinnathambi
- Neurobiology Group, Division of Biochemical Sciences, CSIR-National Chemical Laboratory (CSIR-NCL), Dr. Homi Bhabha Road, Pune, 411008, India.
- Academy of Scientific and Innovative Research (AcSIR), New Delhi, 110025, India.
| |
Collapse
|
14
|
Ettcheto M, Busquets O, Espinosa-Jiménez T, Verdaguer E, Auladell C, Camins A. A Chronological Review of Potential Disease-Modifying Therapeutic Strategies for Alzheimer's Disease. Curr Pharm Des 2020; 26:1286-1299. [PMID: 32066356 DOI: 10.2174/1381612826666200211121416] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2019] [Accepted: 12/18/2019] [Indexed: 01/28/2023]
Abstract
Late-onset Alzheimer's disease (LOAD) is a neurodegenerative disorder that has become a worldwide health problem. This pathology has been classically characterized for its affectation on cognitive function and the presence of depositions of extracellular amyloid β-protein (Aβ) and intracellular neurofibrillary tangles (NFT) composed of hyperphosphorylated Tau protein. To this day, no effective treatment has been developed. Multiple strategies have been proposed over the years with the aim of finding new therapeutic approaches, such as the sequestration of Aβ in plasma or the administration of anti-inflammatory drugs. Also, given the significant role of the insulin receptor in the brain in the proper maintenance of cognitive function, drugs focused on the amelioration of insulin resistance have been proposed as potentially useful and effective in the treatment of AD. In the present review, taking into account the molecular complexity of the disease, it has been proposed that the most appropriate therapeutic strategy is a combinatory treatment of several drugs that will regulate a wide spectrum of the described altered pathological pathways.
Collapse
Affiliation(s)
- Miren Ettcheto
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Science, University of Barcelona, Barcelona, Spain.,Department of Biochemistry and Biotechnology, Faculty of Medicine and Life Sciences, University Rovira i Virgili, Reus, Spain.,Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Carlos III Health Institute, Madrid, Spain.,Institute of Neurosciences, University of Barcelona, Barcelona, Spain
| | - Oriol Busquets
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Science, University of Barcelona, Barcelona, Spain.,Department of Biochemistry and Biotechnology, Faculty of Medicine and Life Sciences, University Rovira i Virgili, Reus, Spain.,Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Carlos III Health Institute, Madrid, Spain.,Institute of Neurosciences, University of Barcelona, Barcelona, Spain
| | - Triana Espinosa-Jiménez
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Science, University of Barcelona, Barcelona, Spain.,Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Carlos III Health Institute, Madrid, Spain.,Institute of Neurosciences, University of Barcelona, Barcelona, Spain
| | - Ester Verdaguer
- Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Carlos III Health Institute, Madrid, Spain.,Institute of Neurosciences, University of Barcelona, Barcelona, Spain.,Department of Cellular Biology, Physiology and Immunology, Faculty of Biology, University of Barcelona, Barcelona, Spain
| | - Carme Auladell
- Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Carlos III Health Institute, Madrid, Spain.,Institute of Neurosciences, University of Barcelona, Barcelona, Spain.,Department of Cellular Biology, Physiology and Immunology, Faculty of Biology, University of Barcelona, Barcelona, Spain
| | - Antoni Camins
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Science, University of Barcelona, Barcelona, Spain.,Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Carlos III Health Institute, Madrid, Spain.,Institute of Neurosciences, University of Barcelona, Barcelona, Spain
| |
Collapse
|
15
|
Bomba M, Granzotto A, Castelli V, Onofrj M, Lattanzio R, Cimini A, Sensi SL. Exenatide Reverts the High-Fat-Diet-Induced Impairment of BDNF Signaling and Inflammatory Response in an Animal Model of Alzheimer's Disease. J Alzheimers Dis 2020; 70:793-810. [PMID: 31256135 DOI: 10.3233/jad-190237] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Alzheimer's disease (AD) is a multifactorial condition in which, along with amyloid-β (Aβ) and tau-related pathology, the synergistic activity of co-morbidity factors promote the onset and progression of the disease. Epidemiological evidence indicates that glucose intolerance, deficits in insulin secretion, or type-2 diabetes mellitus (T2DM) participate in increasing cognitive impairment or dementia risk. Insulin plays a pivotal role in the process as the hormone critically regulates brain functioning. GLP-1, the glucagon-like peptide 1, facilitates insulin signaling, regulates glucose homeostasis, and modulates synaptic plasticity. Exenatide is a synthetic GLP-1 analog employed in T2DM. However, exenatide has also been shown to affect the signaling of the brain-derived neurotrophic factor (BDNF), synaptic plasticity, and cognitive performances in animal models. In this study, we tested whether exenatide exerts neuroprotection in a preclinical AD model set to mimic the clinical complexity of the human disease. We investigated the effects of exenatide treatment in 3xTg-AD mice challenged with a high-fat diet (HFD). Endpoints of the study were variations in systemic metabolism, insulin and neurotrophic signaling, neuroinflammation, Aβ and tau pathology, and cognitive performances. Results of the study indicate that exenatide reverts the adverse changes of BDNF signaling and the neuroinflammation status of 3xTg-AD mice undergoing HFD without affecting systemic metabolism or promoting changes in cognitive performances.
Collapse
Affiliation(s)
- Manuela Bomba
- Center of Excellence on Aging and Translational Medicine - CeSI-MeT, University G. d'Annunzio of Chieti-Pescara, Italy.,Department of Neuroscience, Imaging, and Clinical Sciences, University G. d'Annunzio of Chieti-Pescara, Italy
| | - Alberto Granzotto
- Center of Excellence on Aging and Translational Medicine - CeSI-MeT, University G. d'Annunzio of Chieti-Pescara, Italy.,Department of Neuroscience, Imaging, and Clinical Sciences, University G. d'Annunzio of Chieti-Pescara, Italy
| | - Vanessa Castelli
- Department of Life, Health and Environmental Sciences, University of L'Aquila, Italy
| | - Marco Onofrj
- Center of Excellence on Aging and Translational Medicine - CeSI-MeT, University G. d'Annunzio of Chieti-Pescara, Italy.,Department of Neuroscience, Imaging, and Clinical Sciences, University G. d'Annunzio of Chieti-Pescara, Italy
| | - Rossano Lattanzio
- Center of Excellence on Aging and Translational Medicine - CeSI-MeT, University G. d'Annunzio of Chieti-Pescara, Italy.,Department of Medical, Oral, and Biotechnological Sciences, University G. d'Annunzio of Chieti-Pescara, Italy
| | - Annamaria Cimini
- Department of Life, Health and Environmental Sciences, University of L'Aquila, Italy.,Sbarro Institute for Cancer Research and Molecular Medicine and Center for Biotechnology, Temple University, Philadelphia, PA, USA.,National Institute for Nuclear Physics (INFN), Gran Sasso National Laboratory (LNGS), Assergi, Italy
| | - Stefano L Sensi
- Center of Excellence on Aging and Translational Medicine - CeSI-MeT, University G. d'Annunzio of Chieti-Pescara, Italy.,Department of Neuroscience, Imaging, and Clinical Sciences, University G. d'Annunzio of Chieti-Pescara, Italy.,Departments of Neurology and Pharmacology, Institute for Mind Impairments and Neurological Disorders - iMIND, University of California - Irvine, Irvine, CA, USA
| |
Collapse
|
16
|
Yuan X, Wang L, Tandon N, Sun H, Tian J, Du H, Pascual JM, Guo L. Triheptanoin Mitigates Brain ATP Depletion and Mitochondrial Dysfunction in a Mouse Model of Alzheimer's Disease. J Alzheimers Dis 2020; 78:425-437. [PMID: 33016909 PMCID: PMC8502101 DOI: 10.3233/jad-200594] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
BACKGROUND Brain energy failure is an early pathological event associated with synaptic dysfunction in Alzheimer's disease (AD). Thus, mitigation or enhancement of brain energy metabolism may offer a therapeutic avenue. However, there is uncertainty as to what metabolic process(es) may be more appropriate to support or augment since metabolism is a multiform process such that each of the various metabolic precursors available is utilized via a specific metabolic pathway. In the brain, these pathways sustain not only a robust rate of energy production but also of carbon replenishment. OBJECTIVE Triheptanoin, an edible odd-chain fatty acid triglyceride, is uncommon in that it replenishes metabolites in the tricarboxylic acid cycle (TCA) cycle via anaplerosis in addition to fueling the cycle via oxidation, thus potentially leading to both carbon replenishment and enhanced mitochondrial ATP production. METHODS To test the hypothesis that triheptanoin is protective in AD, we supplied mice with severe brain amyloidosis (5×FAD mice) with dietary triheptanoin for four and a half months, followed by biological and biochemical experiments to examine mice metabolic as well as synaptic function. RESULTS Triheptanoin treatment had minimal impact on systemic metabolism and brain amyloidosis as well as tauopathy while attenuating brain ATP deficiency and mitochondrial dysfunction including respiration and redox balance in 5×FAD mice. Synaptic density, a disease hallmark, was also preserved in hippocampus and neocortex despite profound amyloid deposition. None of these effects took place in treated control mice. CONCLUSION These findings support the energy failure hypothesis of AD and justify investigating the mechanisms in greater depth with ultimate therapeutic intent.
Collapse
Affiliation(s)
- Xiaodong Yuan
- Department of Biological Sciences, The University of Texas at Dallas, Richardson, TX, USA.,Health Management Center, Jinan Central Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Lu Wang
- Department of Biological Sciences, The University of Texas at Dallas, Richardson, TX, USA
| | - Neha Tandon
- Department of Biological Sciences, The University of Texas at Dallas, Richardson, TX, USA
| | - Huili Sun
- Department of Biological Sciences, The University of Texas at Dallas, Richardson, TX, USA
| | - Jing Tian
- Department of Biological Sciences, The University of Texas at Dallas, Richardson, TX, USA
| | - Heng Du
- Department of Biological Sciences, The University of Texas at Dallas, Richardson, TX, USA.,Department of Pharmacology & Toxicology, The University of Kansas, Lawrence, KS, USA.,Higuchi Biosciences Center, The University of Kansas, Lawrence, KS, USA
| | - Juan M Pascual
- Department of Neurology and Neurotherapeutics, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Lan Guo
- Department of Biological Sciences, The University of Texas at Dallas, Richardson, TX, USA.,Department of Pharmacology & Toxicology, The University of Kansas, Lawrence, KS, USA.,Higuchi Biosciences Center, The University of Kansas, Lawrence, KS, USA
| |
Collapse
|
17
|
Jiang Y, Li L, Dai CL, Zhou R, Gong CX, Iqbal K, Gu JH, Liu F. Effect of Peripheral Insulin Administration on Phosphorylation of Tau in the Brain. J Alzheimers Dis 2020; 75:1377-1390. [PMID: 32417781 DOI: 10.3233/jad-200147] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Abnormally hyperphosphorylated tau is the major protein of neurofibrillary tangles in Alzheimer's disease. Insulin activates PI3K-AKT signaling and regulates tau phosphorylation. Impaired brain insulin signaling is involved in Alzheimer's disease pathogenesis. However, the effect of peripheral insulin on tau phosphorylation is controversial. OBJECTIVE In the present study, we determined the effect of peripheral insulin administration on tau phosphorylation in brain. METHODS We intraperitoneally injected a super physiological dose of insulin to mice and analyzed PI3K-AKT signaling and tau phosphorylation in brains by western blots. RESULTS We found that peripherally administered insulin activated the PI3K-AKT signaling pathway immediately in the liver, but not in the brain. Tau phosphorylation in the mouse brain was found to be first decreased (15 min) and then increased (30 min and 60 min) after peripheral insulin administration and these changes correlated inversely with body temperature and the level of brain protein O-GlcNAcylation. Maintaining body temperature of mice post peripheral insulin administration prevented the insulin/hypoglycemia-induced tau hyperphosphorylation after peripheral insulin administration. CONCLUSION These findings suggest that peripheral insulin can induce tau hyperphosphorylation through both hypothermia and downregulation of brain protein O-GlcNAcylation during hypoglycemia.
Collapse
Affiliation(s)
- Yanli Jiang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education of China, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu, China.,Department of Neurochemistry, Inge Grundke-Iqbal Research Floor, New York State Institute for Basic Research in Developmental Disabilities, Staten Island, NY, USA
| | - Longfei Li
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education of China, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu, China.,Department of Neurochemistry, Inge Grundke-Iqbal Research Floor, New York State Institute for Basic Research in Developmental Disabilities, Staten Island, NY, USA
| | - Chun-Ling Dai
- Department of Neurochemistry, Inge Grundke-Iqbal Research Floor, New York State Institute for Basic Research in Developmental Disabilities, Staten Island, NY, USA
| | - Ranran Zhou
- Department of Endocrinology, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
| | - Cheng-Xin Gong
- Department of Neurochemistry, Inge Grundke-Iqbal Research Floor, New York State Institute for Basic Research in Developmental Disabilities, Staten Island, NY, USA
| | - Khalid Iqbal
- Department of Neurochemistry, Inge Grundke-Iqbal Research Floor, New York State Institute for Basic Research in Developmental Disabilities, Staten Island, NY, USA
| | - Jin-Hua Gu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education of China, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu, China.,Department of Neurochemistry, Inge Grundke-Iqbal Research Floor, New York State Institute for Basic Research in Developmental Disabilities, Staten Island, NY, USA.,Department of Clinical Pharmacy, Affiliated Maternity & Child Health Care Hospital of Nantong University, Nantong, Jiangsu, China
| | - Fei Liu
- Department of Neurochemistry, Inge Grundke-Iqbal Research Floor, New York State Institute for Basic Research in Developmental Disabilities, Staten Island, NY, USA
| |
Collapse
|
18
|
Kadoyama K, Matsuura K, Takano M, Otani M, Tomiyama T, Mori H, Matsuyama S. Proteomic analysis involved with synaptic plasticity improvement by GABA A receptor blockade in hippocampus of a mouse model of Alzheimer's disease. Neurosci Res 2020; 165:61-68. [PMID: 32348793 DOI: 10.1016/j.neures.2020.04.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2019] [Revised: 04/03/2020] [Accepted: 04/15/2020] [Indexed: 12/31/2022]
Abstract
GABAergic system plays a part in synaptic plasticity in the hippocampus. We had reported a long-term potentiation (LTP)-like facilitation in vivo, known as synaptic plasticity, through GABAA receptor blockade by bicuculline and the expression of proteins involved with this synaptic plasticity in mouse hippocampus. In the present study, we aimed to show improvement of impaired synaptic plasticity through GABAA receptor blockade and to clarify the molecular mechanisms involved with this improvement in the hippocampus of mice overexpressing human amyloid precursor protein with the E693Δ mutation (APPOSK-Tg) as an Alzheimer's disease model showing impaired synaptic plasticity. Electrophysiological study showed that the LTP-like facilitation expressed with application of bicuculline in vivo was significantly greater than impaired tetanic LTP in APPOSK-Tg mice, which was improved by bicuculline. Proteomic analysis showed that the expression of 11 proteins in the hippocampus was significantly changed 8 h after bicuculline application to APPOSK-Tg mice. The identified proteins could be functionally classified as chaperone, cytoskeletal protein, energy metabolism, metabolism, neuronal development, and synaptic component. Additionally, western blotting validated the changes in four proteins. We therefore propose that the improvement of impaired synaptic plasticity through GABAA receptor blockade could be mediated by the changed expression of these proteins.
Collapse
Affiliation(s)
- Keiichi Kadoyama
- Department of Pharmaceutical Health Care, Faculty of Pharmaceutical Sciences, Himeji Dokkyo University, Himeji 670-8524, Japan
| | - Kenji Matsuura
- Faculty of Pharmacy, Osaka-Ohtani University, Tondabayashi 584-8540, Japan
| | - Masaoki Takano
- Department of Life Sciences Pharmacy, School of Pharmaceutical Sciences, Kobe Gakuin University, Kobe 650-8586, Japan
| | - Mieko Otani
- Department of Life Sciences Pharmacy, School of Pharmaceutical Sciences, Kobe Gakuin University, Kobe 650-8586, Japan
| | - Takami Tomiyama
- Department of Translational Neuroscience, Osaka City University Graduate School of Medicine, Osaka 545-8585, Japan
| | - Hiroshi Mori
- Department of Clinical Neuroscience, Osaka City University Medical School, Osaka 545-8585, Japan
| | - Shogo Matsuyama
- Biosignal Research Center, Kobe University, Kobe 657-8501, Japan.
| |
Collapse
|
19
|
Alhowail AH, Alfawzan GS, Aldubayan MA, S. Alsalam L. Effect of Acute Chemotherapy on Glucose Levels in Rats. INT J PHARMACOL 2020. [DOI: 10.3923/ijp.2020.276.281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
20
|
Tabassum R, Jeong NY, Jung J. Protective effect of hydrogen sulfide on oxidative stress-induced neurodegenerative diseases. Neural Regen Res 2020; 15:232-241. [PMID: 31552888 PMCID: PMC6905340 DOI: 10.4103/1673-5374.265543] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Accepted: 06/06/2019] [Indexed: 12/11/2022] Open
Abstract
Hydrogen sulfide is an antioxidant molecule that has a wide range of biological effects against oxidative stress. Balanced oxidative stress is also vital for maintaining cellular function in biological system, where reactive oxygen species are the main source of oxidative stress. When the normal redox balance is disturbed, deoxyribonucleic acid, lipid, and protein molecules are oxidized under pathological conditions, like diabetes mellitus that leads to diabetic peripheral neuropathy. In diabetes mellitus-induced diabetic peripheral neuropathy, due to hyperglycemia, pancreatic beta cell (β cell) shows resistance to insulin secretion. As a consequence, glucose metabolism is disturbed in neuronal cells which are distracted from providing proper cell signaling pathway. Not only diabetic peripheral neuropathy but also other central damages occur in brain neuropathy. Neurological studies regarding type 1 diabetes mellitus patients with Alzheimer's disease, Parkinson's disease, and amyotrophic lateral sclerosis have shown changes in the central nervous system because high blood glucose levels (HbA1c) appeared with poor cognitive function. Oxidative stress plays a role in inhibiting insulin signaling that is necessary for brain function. Hydrogen sulfide exhibits antioxidant effects against oxidative stress, where cystathionine β synthase, cystathionine γ lyase, and 3-mercaptopyruvate sulfurtransferase are the endogenous sources of hydrogen sulfide. This review is to explore the pathogenesis of diabetes mellitus-induced diabetic peripheral neuropathy and other neurological comorbid disorders under the oxidative stress condition and the anti-oxidative effects of hydrogen sulfide.
Collapse
Affiliation(s)
- Rubaiya Tabassum
- Department of Anatomy and Cell Biology, College of Medicine, Dong-A University, Seo-gu, Busan, Korea
- Department of Medicine, Graduate School, Dong-A University, Seo-gu, Busan, Korea
| | - Na Young Jeong
- Department of Anatomy and Cell Biology, College of Medicine, Dong-A University, Seo-gu, Busan, Korea
- Department of Medicine, Graduate School, Dong-A University, Seo-gu, Busan, Korea
| | - Junyang Jung
- Department of Anatomy and Neurobiology, College of Medicine, Kyung Hee University, Dongdaemun-gu, Seoul, Korea
| |
Collapse
|
21
|
Gerzson MFB, Bona NP, Soares MSP, Teixeira FC, Rahmeier FL, Carvalho FB, da Cruz Fernandes M, Onzi G, Lenz G, Gonçales RA, Spanevello RM, Stefanello FM. Tannic Acid Ameliorates STZ-Induced Alzheimer's Disease-Like Impairment of Memory, Neuroinflammation, Neuronal Death and Modulates Akt Expression. Neurotox Res 2020; 37:1009-1017. [PMID: 31997154 DOI: 10.1007/s12640-020-00167-3] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Revised: 12/18/2019] [Accepted: 01/22/2020] [Indexed: 12/28/2022]
Abstract
Tannic acid (TA) is a hydrolysable glycosidic polyphenol polymer of gallic acid, which possesses neuroprotective properties. The aim of this study was to evaluate the effect of TA treatment on cognitive performance and neurochemical changes in an experimental model of sporadic dementia of Alzheimer's type (SDAT) induced by intracerebroventricular (ICV) injection of streptozotocin (STZ) and to explore the potential cellular and molecular mechanisms underlying these effects. Adult male rats were divided into four groups: control, TA, STZ, and TA + STZ. Animals from TA and TA + STZ groups were treated with TA (30 mg/kg) daily, by gavage, for 21 days; others groups received water (1 mL/kg). Subsequently, an ICV injection of STZ (3 mg/kg) was administered into the lateral ventricles of animals from STZ and TA + STZ groups, while other groups received citrate buffer. Cognitive deficits (short-term memory), neuronal survival, neuroinflammation as well as expression of SNAP-25, Akt, and pAkt were evaluated in the cerebral cortex. TA treatment protected against the impairment of memory in STZ-induced SDAT. STZ promoted an increase in neuronal death and the levels of proinflammatory cytokines (IL-6 and TNF-α) and a decrease in Akt and pAkt expression; TA was able to restore these changes. Neither STZ nor TA altered SNAP-25 expression or the levels of IL-12 and IL-4 in the cerebral cortex. Our study highlights that treatment with TA prevents memory deficits and reestablishes Akt and pAkt expression, protecting against neuronal death and neuroinflammation in STZ-induced SDAT in rats.
Collapse
Affiliation(s)
- Mariana F B Gerzson
- Laboratório de Biomarcadores, Centro de Ciências Químicas, Farmacêuticas e de Alimentos, Universidade Federal de Pelotas, Campus Universitário s/n, Pelotas, RS, Brazil
| | - Natália P Bona
- Laboratório de Biomarcadores, Centro de Ciências Químicas, Farmacêuticas e de Alimentos, Universidade Federal de Pelotas, Campus Universitário s/n, Pelotas, RS, Brazil
| | - Mayara S P Soares
- Laboratório de Neuroquímica, Inflamação e Câncer, Centro de Ciências Químicas, Farmacêuticas e de Alimentos, Universidade Federal de Pelotas, Campus Universitário s/n, Pelotas, RS, Brazil
| | - Fernanda C Teixeira
- Laboratório de Neuroquímica, Inflamação e Câncer, Centro de Ciências Químicas, Farmacêuticas e de Alimentos, Universidade Federal de Pelotas, Campus Universitário s/n, Pelotas, RS, Brazil
| | - Francine L Rahmeier
- Laboratório de Pesquisa em Patologia, Universidade Federal de Ciências da Saúde de Porto Alegre, Rua Sarmento Leite, 245, Porto Alegre, RS, Brazil
| | - Fabiano B Carvalho
- Laboratório de Pesquisa em Patologia, Universidade Federal de Ciências da Saúde de Porto Alegre, Rua Sarmento Leite, 245, Porto Alegre, RS, Brazil
| | - Marilda da Cruz Fernandes
- Laboratório de Pesquisa em Patologia, Universidade Federal de Ciências da Saúde de Porto Alegre, Rua Sarmento Leite, 245, Porto Alegre, RS, Brazil
| | - Giovana Onzi
- Laboratório de Sinalização e Plasticidade Celular, Departamento de Biofísica, Universidade Federal do Rio Grande do Sul , Porto Alegre, RS, Brazil
| | - Guido Lenz
- Laboratório de Sinalização e Plasticidade Celular, Departamento de Biofísica, Universidade Federal do Rio Grande do Sul , Porto Alegre, RS, Brazil
| | - Relber A Gonçales
- Laboratório de Biologia Molecular, Universidade Federal de Goiás, Goiânia, GO, Brazil
| | - Roselia M Spanevello
- Laboratório de Neuroquímica, Inflamação e Câncer, Centro de Ciências Químicas, Farmacêuticas e de Alimentos, Universidade Federal de Pelotas, Campus Universitário s/n, Pelotas, RS, Brazil.
| | - Francieli M Stefanello
- Laboratório de Biomarcadores, Centro de Ciências Químicas, Farmacêuticas e de Alimentos, Universidade Federal de Pelotas, Campus Universitário s/n, Pelotas, RS, Brazil.
| |
Collapse
|
22
|
Sullivan CR, Koene RH, Hasselfeld K, O'Donovan S, Ramsey A, McCullumsmith RE. Neuron-specific deficits of bioenergetic processes in the dorsolateral prefrontal cortex in schizophrenia. Mol Psychiatry 2019; 24:1319-1328. [PMID: 29497148 PMCID: PMC6119539 DOI: 10.1038/s41380-018-0035-3] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Revised: 12/07/2017] [Accepted: 01/15/2018] [Indexed: 12/20/2022]
Abstract
Schizophrenia is a devastating illness that affects over 2 million people in the United States and costs society billions of dollars annually. New insights into the pathophysiology of schizophrenia are needed to provide the conceptual framework to facilitate development of new treatment strategies. We examined bioenergetic pathways in the dorsolateral prefrontal cortex (DLPFC) of subjects with schizophrenia and control subjects using western blot analysis, quantitative real-time polymerase chain reaction, and enzyme/substrate assays. Laser-capture microdissection-quantitative polymerase chain reaction was used to examine these pathways at the cellular level. We found decreases in hexokinase (HXK) and phosphofructokinase (PFK) activity in the DLPFC, as well as decreased PFK1 mRNA expression. In pyramidal neurons, we found an increase in monocarboxylate transporter 1 mRNA expression, and decreases in HXK1, PFK1, glucose transporter 1 (GLUT1), and GLUT3 mRNA expression. These results suggest abnormal bioenergetic function, as well as a neuron-specific defect in glucose utilization, in the DLPFC in schizophrenia.
Collapse
Affiliation(s)
- Courtney R. Sullivan
- Corresponding author: , Phone number: 513-558-4855, Mail address: 231 Albert Sabin Way, Care 5830, Cincinnati, Ohio, 45267-2827
| | - Rachael H. Koene
- Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati, Cincinnati, OH
| | - Kathryn Hasselfeld
- Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati, Cincinnati, OH
| | - Sinead O'Donovan
- Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati, Cincinnati, OH
| | - Amy Ramsey
- Department of Pharmacology and Toxicology, University of Toronto, ON, Canada
| | - Robert E. McCullumsmith
- Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati, Cincinnati, OH
| |
Collapse
|
23
|
Ansari SA, Emerald BS. The Role of Insulin Resistance and Protein O-GlcNAcylation in Neurodegeneration. Front Neurosci 2019; 13:473. [PMID: 31143098 PMCID: PMC6521730 DOI: 10.3389/fnins.2019.00473] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Accepted: 04/26/2019] [Indexed: 12/15/2022] Open
Abstract
Metabolic syndrome including obesity and type 2 diabetes is increasing at an alarming rate worldwide. Similarly, there has been an increase in the cases of neurodegenerative diseases such as Alzheimer’s disease (AD) possibility due to increase in elderly population in the past few decades. Both, metabolic diseases and AD have one common feature that is insulin resistance. Recent studies suggest a link between the regulatory functions of insulin in the brain and AD. Hypoglycemia, a characteristic feature of AD may be a result of impaired insulin signaling in the affected regions of the brain. O-GlcNAcylation is a post-translational protein modification, the levels of which are dependent on the availability of glucose inside the cells. Hyperphosphorylation of Tau is a major molecular feature, which leads to its aggregation and neurotoxicity in AD. In addition, impaired processing of Amyloid precursor protein (APP) leading to toxic amyloid β (Aβ) aggregation is also implicated in the pathogenesis of AD. Both APP and Tau are also found to be O-GlcNAcylated. Reduced O-GlcNAcylation of APP and Tau due to hypoglycemia is found to be associated with their pathological features in AD brain. Recent studies have also identified perturbed O-GlcNAcylation/phosphorylation of several other proteins important for normal neuronal function, which may be contributing to the neuropathological development in AD. Herein, we discuss about the uptake and distribution of insulin inside the brain, brain insulin signaling and insulin resistance as well as its relation to neurodegenerative diseases with a special focus on protein O-GlcNAcylation and its potential role in the treatment of AD.
Collapse
Affiliation(s)
- Suraiya A Ansari
- Department of Biochemistry, College of Medicine and Health Sciences, UAE University, Al Ain, United Arab Emirates
| | - Bright Starling Emerald
- Department of Anatomy, College of Medicine and Health Sciences, UAE University, Al Ain, United Arab Emirates
| |
Collapse
|
24
|
Das TK, Chakrabarti SK, Zulkipli IN, Abdul Hamid MR. Curcumin Ameliorates the Impaired Insulin Signaling Involved in the Pathogenesis of Alzheimer's Disease in Rats. J Alzheimers Dis Rep 2019; 3:59-70. [PMID: 31025030 PMCID: PMC6481473 DOI: 10.3233/adr-180091] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/23/2019] [Indexed: 12/15/2022] Open
Abstract
To date, dysregulation of the insulin signaling pathway in the brain has not been demonstrated unequivocally in Alzheimer's disease (AD). The purpose of the study was to examine the possible dysregulation of insulin signaling pathway in an AD rat model. Furthermore, the present study investigated the effect of Donepezil and Curcumin on insulin signaling, insulin, and glucose levels in AD rat brain. The rats were induced to develop AD by intraperitoneal administration of Scopolamine. We found that glucose levels in plasma and brain were decreased in AD rats, whereas the insulin levels was increased in plasma but decreased in brain in AD rats. In addition, insulin signaling proteins IR-β, IGF-1, IRS-1, IRS-2 p-Akt (Ser473), and Akt were markedly reduced in the AD rats. Furthermore, GLUT3 and GLUT4 levels in the brain were markedly reduced in AD rats. All these data were compared to Saline-treated control rats. Curcumin significantly increased glucose levels in plasma and in brain. However, insulin levels was decreased in plasma and was increased in AD rats' brain. Moreover, GLUT3 and GLUT4 levels were significantly increased in Curcumin-treated AD rats. All these data were compared to Scopolamine- induced AD rats. Thus amelioration of impaired insulin signaling and improved glucose regulation in AD rats by Curcumin may be beneficial in the management of AD.
Collapse
Affiliation(s)
- Tushar Kanti Das
- Universiti Brunei Darussalam, PAPRSB Institute of Health Sciences, Jalan Tungku Link, Gadong, Brunei Darussalam, Brunei
- Institute of Reproductive Medicine, Salt Lake, Kolkata, India
| | | | - Ihsan Nazurah Zulkipli
- Universiti Brunei Darussalam, PAPRSB Institute of Health Sciences, Jalan Tungku Link, Gadong, Brunei Darussalam, Brunei
| | - Mas R.W. Abdul Hamid
- Universiti Brunei Darussalam, PAPRSB Institute of Health Sciences, Jalan Tungku Link, Gadong, Brunei Darussalam, Brunei
| |
Collapse
|
25
|
Babic Perhoc A, Osmanovic Barilar J, Knezovic A, Farkas V, Bagaric R, Svarc A, Grünblatt E, Riederer P, Salkovic-Petrisic M. Cognitive, behavioral and metabolic effects of oral galactose treatment in the transgenic Tg2576 mice. Neuropharmacology 2018; 148:50-67. [PMID: 30571958 DOI: 10.1016/j.neuropharm.2018.12.018] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Revised: 12/13/2018] [Accepted: 12/14/2018] [Indexed: 12/23/2022]
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disorder associated with insulin resistance and glucose hypometabolism in the brain. Oral administration of galactose, a nutrient that provides an alternative source of energy, prevents and ameliorates early cognitive impairment in a streptozotocin-induced model (STZ-icv) of the sporadic AD (sAD). Here we explored the influence of 2-month oral galactose treatment (200 mg/kg/day) in the familial AD (fAD) by using 5- (5M) and 10- (10M) month-old transgenic Tg2576 mice mimicking the presymptomatic and the mild stage of fAD, and compared it to that observed in 7-month old STZ-icv rats mimicking mild-to-moderate sAD. Cognitive and behavioral performance was tested by Morris Water Maze, Open Field and Elevated Plus Maze tests, and metabolic status by intraperitoneal glucose tolerance test and fluorodeoxyglucose Positron-Emission Tomography scan. The level of insulin, glucagon-like peptide-1 (GLP-1) and soluble amyloid β1-42 (sAβ1-42) was measured by ELISA and the protein expression of insulin receptor (IR), glycogen synthase kinase-3β (GSK-3β), and pre-/post-synaptic markers by Western blot analysis. Although galactose normalized alterations in cerebral glucose metabolism in all Tg2576 mice (5M+2M; 10M+2M) and STZ-icv rats, it did not improve cognitive impairment in either model. Improvement of reduced grooming behavior and normalization in reduced plasma insulin levels were seen only in 5M+2M Tg2576 mice while in 10M+2M Tg2576 mice oral galactose induced metabolic exacerbation at the level of plasma insulin, GLP-1 homeostasis and glucose intolerance, and additionally increased hippocampal sAβ1-42 level, decreased IR expression and increased GSK-3β activity. The results indicate that therapeutic potential of oral galactose seems to depend on the stage and the type/model of AD and to differ in the absence and the presence of AD-like pathology.
Collapse
Affiliation(s)
- Ana Babic Perhoc
- Department of Pharmacology, University of Zagreb School of Medicine, Salata 11, HR-10 000, Zagreb, Croatia
| | - Jelena Osmanovic Barilar
- Department of Pharmacology, University of Zagreb School of Medicine, Salata 11, HR-10 000, Zagreb, Croatia
| | - Ana Knezovic
- Department of Pharmacology, University of Zagreb School of Medicine, Salata 11, HR-10 000, Zagreb, Croatia
| | - Vladimir Farkas
- Department of Experimental Physics, Rudjer Boskovic Institute, Bijenicka 54, HR-10 000, Zagreb, Croatia
| | - Robert Bagaric
- Department of Experimental Physics, Rudjer Boskovic Institute, Bijenicka 54, HR-10 000, Zagreb, Croatia
| | - Alfred Svarc
- Department of Experimental Physics, Rudjer Boskovic Institute, Bijenicka 54, HR-10 000, Zagreb, Croatia
| | - Edna Grünblatt
- Department of Child and Adolescent Psychiatry and Psychotherapy, University Hospital of Psychiatry Zurich, University of Zurich, Zurich, Switzerland; Neuroscience Center Zurich, University of Zurich and ETH Zurich, Zurich, Switzerland; Zurich Center for Integrative Human Physiology, University of Zurich, Switzerland
| | - Peter Riederer
- Center of Mental Health, Department of Psychiatry, Psychosomatics and Psychotherapy, University Hospital, Würzburg, Füchsleinstrasse 15, 97080, Würzburg, Germany; Department of Clinical Research and Psychiatry, University of Southern Denmark Odense, Odense, Denmark
| | - Melita Salkovic-Petrisic
- Department of Pharmacology, University of Zagreb School of Medicine, Salata 11, HR-10 000, Zagreb, Croatia; Research Centre of Excellence of Fundamental, Clinical and Translational Neuroscience, Croatian Institute for Brain Research, University of Zagreb School of Medicine, Salata 12, HR-10 000, Zagreb, Croatia.
| |
Collapse
|
26
|
The Role of Long Noncoding RNAs in Diabetic Alzheimer's Disease. J Clin Med 2018; 7:jcm7110461. [PMID: 30469430 PMCID: PMC6262561 DOI: 10.3390/jcm7110461] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Revised: 11/17/2018] [Accepted: 11/19/2018] [Indexed: 12/16/2022] Open
Abstract
Long noncoding RNAs (lncRNAs) are involved in diverse physiological and pathological processes by modulating gene expression. They have been found to be dysregulated in the brain and cerebrospinal fluid of patients with neurodegenerative diseases, and are considered promising therapeutic targets for treatment. Among the various neurodegenerative diseases, diabetic Alzheimer's disease (AD) has been recently emerging as an important issue due to several unexpected reports suggesting that metabolic issues in the brain, such as insulin resistance and glucose dysregulation, could be important risk factors for AD. To facilitate understanding of the role of lncRNAs in this field, here we review recent studies on lncRNAs in AD and diabetes, and summarize them with different categories associated with the pathogenesis of the diseases including neurogenesis, synaptic dysfunction, amyloid beta accumulation, neuroinflammation, insulin resistance, and glucose dysregulation. It is essential to understand the role of lncRNAs in the pathogenesis of diabetic AD from various perspectives for therapeutic utilization of lncRNAs in the near future.
Collapse
|
27
|
Gomez-Pinilla F, Yang X. System biology approach intersecting diet and cell metabolism with pathogenesis of brain disorders. Prog Neurobiol 2018; 169:76-90. [PMID: 30059718 PMCID: PMC6231047 DOI: 10.1016/j.pneurobio.2018.07.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Accepted: 07/15/2018] [Indexed: 12/21/2022]
Abstract
The surge in meals high in calories has prompted an epidemic of metabolic disorders around the world such that the elevated incidence of obese and diabetic individuals is alarming. New research indicates that metabolic disorders pose a risk for neurological and psychiatric conditions including stroke, Alzheimer's disease, Huntington's disease, and depression, all of which have a metabolic component. These relationships are rooted to a dysfunctional interaction between molecular processes that regulate energy metabolism and synaptic plasticity. The strong adaptive force of dietary factors on shaping the brain during evolution can be manipulated to transform the interaction between cell bioenergetics and epigenome with the aptitude to promote long-lasting brain healthiness. A thorough understanding of the association between the broad action of nutrients and brain fitness requires high level data processing empowered with the capacity to integrate information from a multitude of molecular entities and pathways. Nutritional systems biology is emerging as a viable approach to elucidate the multiple molecular layers involved in information processing in cells, tissues, and organ systems in response to diet. Information about the wide range of cellular and molecular interactions elicited by foods on the brain and cognitive plasticity is crucial for the design of public health initiatives for curtailing the epidemic of metabolic and brain disorders.
Collapse
Affiliation(s)
- Fernando Gomez-Pinilla
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA, 90095, USA; Department of Neurosurgery, UCLA Brain Injury Research Center, University of California, Los Angeles, Los Angeles, CA, 90095, USA.
| | - Xia Yang
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| |
Collapse
|
28
|
Ahmad W, Ebert PR. 5-Methoxyindole-2-carboxylic acid (MICA) suppresses Aβ-mediated pathology in C. elegans. Exp Gerontol 2018; 108:215-225. [DOI: 10.1016/j.exger.2018.04.021] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Revised: 04/02/2018] [Accepted: 04/26/2018] [Indexed: 12/22/2022]
|
29
|
Niu G, Guo J, Tian Y, Zhao K, Li J, Xiao Q. α‑lipoic acid can greatly alleviate the toxic effect of AGES on SH‑SY5Y cells. Int J Mol Med 2018; 41:2855-2864. [PMID: 29436603 DOI: 10.3892/ijmm.2018.3477] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Accepted: 01/22/2018] [Indexed: 11/05/2022] Open
Abstract
The aim of the study was to explore the influence of α‑lipoic acid (α‑LA) on the cytotoxicity of advanced glycation end‑products (AGEs) against SH‑SY5Y cells. AGE‑bovine serum albumin (BSA) was incubated in vitro using SH‑SY5Y cells as a target model, and the control group was set. Cells were exposed to AGE‑BSA, and α‑LA was selectively added to the cells. Cell growth and death was determined by the MTT assay, which measures cellular metabolic rate, lactate dehydrogenase (LDH) leakage rate and cellular axonal length. Immunocytochemistry was employed to detect the expression of β‑amyloid (Aβ) protein in cells, and mRNA expression of amyloid precursor protein (APP) and the receptor for AGE (RAGE) were assayed by PT‑PCR. The metabolism of MTT was clearly increased, the rate of LDH leakage was significantly decreased, and axonal length was significantly increased in cells treated with α‑LA (0.1 g/l) as compared to untreated cells. Furthermore, the expression levels of Aβ protein were also decreased. In addition, α‑LA (0.1 g/l) markedly inhibited the expression of RAGE mRNA, and did not influence APP mRNA expression as compared the control group. α‑LA (0.1 g/l) was effective at dampening the cytotoxicity of AGE‑BSA, a preliminary observation that confirms the ability of α‑LA to significantly alleviate the cytotoxicity of AGEs against SH‑SY5Y cells.
Collapse
Affiliation(s)
- Guifen Niu
- Department of Endocrinology, Liaocheng City People's Hospital, Liaocheng, Shandong 252000, P.R. China
| | - Jianfei Guo
- Department of Endocrinology, Liaocheng City People's Hospital, Liaocheng, Shandong 252000, P.R. China
| | - Yaqiang Tian
- Department of Endocrinology, Liaocheng Brain Hospital, Liaocheng, Shandong 252000, P.R. China
| | - Kexiang Zhao
- Department of the Elderly, Chongqing Medical University, Chongqing 400016, P.R. China
| | - Jian Li
- Department of Endocrinology, Liaocheng City People's Hospital, Liaocheng, Shandong 252000, P.R. China
| | - Qian Xiao
- Department of the Elderly, Chongqing Medical University, Chongqing 400016, P.R. China
| |
Collapse
|
30
|
Bazazzadegan N, Dehghan Shasaltaneh M, Saliminejad K, Kamali K, Banan M, Nazari R, Riazi GH, Khorram Khorshid HR. Effects of Ectoine on Behavior and Candidate Genes Expression in ICV-STZ Rat Model of Sporadic Alzheimer's Disease. Adv Pharm Bull 2018; 7:629-636. [PMID: 29399553 PMCID: PMC5788218 DOI: 10.15171/apb.2017.075] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2017] [Revised: 11/29/2017] [Accepted: 12/04/2017] [Indexed: 01/02/2023] Open
Abstract
Purpose: Alzheimer's disease (AD) is pathologically defined by the presence of amyloid plaques and tangles in the brain, therefore, any drug or compound with potential effect on lowering amyloid plaques, could be noticed for AD management especially in the primary phases of the disease. Ectoine constitutes a group of small molecule chaperones (SMCs). SMCs inhibit proteins and other changeable macromolecular structures misfolding from environmental stresses. Ectoine has been reported successfully prohibit insulin amyloid formation in vitro. Methods: We selected eight genes, DAXX, NFκβ, VEGF, PSEN1, MTAP2, SYP, MAPK3 and TNFα genes which had previously showed significant differential expression in Alzheimer human brain and STZ- rat model. We considered the neuroprotective efficacy by comparing the expression of candidate genes levels in the hippocampus of rat model of Sopradic Alzheimer's disease (SAD), using qPCR in compound-treated and control groups as well as therapeutic effects at learning and memory levels by using Morris Water Maze (MWM) test. Results: Our results showed significant down-regulation of Syp, Mapk3 and Tnfα and up-regulation of Vegf in rat's hippocampus after treatment with ectoine comparing to the STZ-induced group. In MWM, there was no significant change in swimming distance and time for finding the hidden platform in treated comparing to STZ-induced group. In addition, it wasn't seen significant change in compound-treated comparing to STZ-induced and control groups in memory level. Conclusion: It seems this compound may have significant effect on expression level of some AD- related genes but not on clinical levels.
Collapse
Affiliation(s)
- Niloofar Bazazzadegan
- Genetics Research Center, University of Social Welfare and Rehabilitation Sciences, Tehran, Iran
| | - Marzieh Dehghan Shasaltaneh
- Laboratory of Neuro-organic Chemistry, Institute of Biochemistry and Biophysics (IBB), University of Tehran, Tehran, Iran
| | - Kioomars Saliminejad
- Reproductive Biotechnology Research Center, Avicenna Research Institute, ACECR, Tehran, Iran
| | - Koorosh Kamali
- Reproductive Biotechnology Research Center, Avicenna Research Institute, ACECR, Tehran, Iran
| | - Mehdi Banan
- Genetics Research Center, University of Social Welfare and Rehabilitation Sciences, Tehran, Iran
| | - Reza Nazari
- Laboratory of Neuro-organic Chemistry, Institute of Biochemistry and Biophysics (IBB), University of Tehran, Tehran, Iran
| | - Gholam Hossein Riazi
- Laboratory of Neuro-organic Chemistry, Institute of Biochemistry and Biophysics (IBB), University of Tehran, Tehran, Iran
| | | |
Collapse
|
31
|
Ribarič S. Peptides as Potential Therapeutics for Alzheimer's Disease. Molecules 2018; 23:E283. [PMID: 29385735 PMCID: PMC6017258 DOI: 10.3390/molecules23020283] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Revised: 01/26/2018] [Accepted: 01/28/2018] [Indexed: 12/22/2022] Open
Abstract
Intracellular synthesis, folding, trafficking and degradation of proteins are controlled and integrated by proteostasis. The frequency of protein misfolding disorders in the human population, e.g., in Alzheimer's disease (AD), is increasing due to the aging population. AD treatment options are limited to symptomatic interventions that at best slow-down disease progression. The key biochemical change in AD is the excessive accumulation of per-se non-toxic and soluble amyloid peptides (Aβ(1-37/44), in the intracellular and extracellular space, that alters proteostasis and triggers Aβ modification (e.g., by reactive oxygen species (ROS)) into toxic intermediate, misfolded soluble Aβ peptides, Aβ dimers and Aβ oligomers. The toxic intermediate Aβ products aggregate into progressively less toxic and less soluble protofibrils, fibrils and senile plaques. This review focuses on peptides that inhibit toxic Aβ oligomerization, Aβ aggregation into fibrils, or stabilize Aβ peptides in non-toxic oligomers, and discusses their potential for AD treatment.
Collapse
Affiliation(s)
- Samo Ribarič
- Institute of Pathophysiology, Faculty of Medicine, Zaloška 4, SI-1000 Ljubljana, Slovenia.
| |
Collapse
|
32
|
Ahmad W, Ijaz B, Shabbiri K, Ahmed F, Rehman S. Oxidative toxicity in diabetes and Alzheimer's disease: mechanisms behind ROS/ RNS generation. J Biomed Sci 2017; 24:76. [PMID: 28927401 PMCID: PMC5606025 DOI: 10.1186/s12929-017-0379-z] [Citation(s) in RCA: 218] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Accepted: 09/05/2017] [Indexed: 02/07/2023] Open
Abstract
Reactive oxidative species (ROS) toxicity remains an undisputed cause and link between Alzheimer’s disease (AD) and Type-2 Diabetes Mellitus (T2DM). Patients with both AD and T2DM have damaged, oxidized DNA, RNA, protein and lipid products that can be used as possible disease progression markers. Although the oxidative stress has been anticipated as a main cause in promoting both AD and T2DM, multiple pathways could be involved in ROS production. The focus of this review is to summarize the mechanisms involved in ROS production and their possible association with AD and T2DM pathogenesis and progression. We have also highlighted the role of current treatments that can be linked with reduced oxidative stress and damage in AD and T2DM.
Collapse
Affiliation(s)
- Waqar Ahmad
- School of Biological Sciences, University of Queensland, Brisbane, 4072, Australia.
| | - Bushra Ijaz
- Centre of Excellence in Molecular Biology, University of the Punjab, Thokar Niaz Baig, Lahore, 54000, Pakistan
| | - Khadija Shabbiri
- School of Biological Sciences, University of Queensland, Brisbane, 4072, Australia
| | - Fayyaz Ahmed
- Centre of Excellence in Molecular Biology, University of the Punjab, Thokar Niaz Baig, Lahore, 54000, Pakistan
| | - Sidra Rehman
- COMSATS Institute of Information Technology Abbottabad, Abbottabad, 22010, Pakistan
| |
Collapse
|
33
|
Li L. The Molecular Mechanism of Glucagon-Like Peptide-1 Therapy in Alzheimer's Disease, Based on a Mechanistic Target of Rapamycin Pathway. CNS Drugs 2017; 31:535-549. [PMID: 28540646 DOI: 10.1007/s40263-017-0431-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The mechanistic target of rapamycin (mTOR) is an important molecule that connects aging, lifespan, energy balance, glucose and lipid metabolism, and neurodegeneration. Rapamycin exerts effects in numerous biological activities via its target protein, playing a key role in energy balance, regulation of autophagy, extension of lifespan, immunosuppression, and protection against neurodegeneration. There are many similar pathophysiological processes and molecular pathways between Alzheimer's disease (AD) and type 2 diabetes mellitus (T2DM), and pharmacologic agents used to treat T2DM, including glucagon-like peptide-1 (GLP-1) analogs, seem to be beneficial for AD. mTOR mediates the effects of GLP-1 analogs in the treatment of T2DM; hence, I hypothesize that mTOR is a key molecule for mediating the protective effects of GLP-1 for AD.
Collapse
Affiliation(s)
- Lin Li
- Key Laboratory of Cellular Physiology, Shanxi Medical University, Taiyuan, Shanxi, China.
| |
Collapse
|
34
|
Tundo GR, Sbardella D, Ciaccio C, Grasso G, Gioia M, Coletta A, Polticelli F, Di Pierro D, Milardi D, Van Endert P, Marini S, Coletta M. Multiple functions of insulin-degrading enzyme: a metabolic crosslight? Crit Rev Biochem Mol Biol 2017. [PMID: 28635330 DOI: 10.1080/10409238.2017.1337707] [Citation(s) in RCA: 74] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Insulin-degrading enzyme (IDE) is a ubiquitous zinc peptidase of the inverzincin family, which has been initially discovered as the enzyme responsible for insulin catabolism; therefore, its involvement in the onset of diabetes has been largely investigated. However, further studies on IDE unraveled its ability to degrade several other polypeptides, such as β-amyloid, amylin, and glucagon, envisaging the possible implication of IDE dys-regulation in the "aggregopathies" and, in particular, in neurodegenerative diseases. Over the last decade, a novel scenario on IDE biology has emerged, pointing out a multi-functional role of this enzyme in several basic cellular processes. In particular, latest advances indicate that IDE behaves as a heat shock protein and modulates the ubiquitin-proteasome system, suggesting a major implication in proteins turnover and cell homeostasis. In addition, recent observations have highlighted that the regulation of glucose metabolism by IDE is not merely based on its largely proposed role in the degradation of insulin in vivo. There is increasing evidence that improper IDE function, regulation, or trafficking might contribute to the etiology of metabolic diseases. In addition, the enzymatic activity of IDE is affected by metals levels, thus suggesting a role also in the metal homeostasis (metallostasis), which is thought to be tightly linked to the malfunction of the "quality control" machinery of the cell. Focusing on the physiological role of IDE, we will address a comprehensive vision of the very complex scenario in which IDE takes part, outlining its crucial role in interconnecting several relevant cellular processes.
Collapse
Affiliation(s)
- Grazia R Tundo
- a Department of Clinical Sciences and Translation Medicine , University of Roma Tor Vergata , Roma , Italy.,b CIRCMSB , Bari , Italy
| | - Diego Sbardella
- a Department of Clinical Sciences and Translation Medicine , University of Roma Tor Vergata , Roma , Italy.,b CIRCMSB , Bari , Italy.,c Center for TeleInfrastructures, University of Roma Tor Vergata , Roma , Italy
| | - Chiara Ciaccio
- a Department of Clinical Sciences and Translation Medicine , University of Roma Tor Vergata , Roma , Italy.,b CIRCMSB , Bari , Italy
| | - Giuseppe Grasso
- d Department of Chemistry , University of Catania , Catania , Italy.,e CNR IBB , Catania , Italy
| | - Magda Gioia
- a Department of Clinical Sciences and Translation Medicine , University of Roma Tor Vergata , Roma , Italy.,b CIRCMSB , Bari , Italy
| | - Andrea Coletta
- f Department of Chemistry , University of Aarhus , Aarhus , Denmark
| | | | - Donato Di Pierro
- a Department of Clinical Sciences and Translation Medicine , University of Roma Tor Vergata , Roma , Italy.,b CIRCMSB , Bari , Italy
| | | | - Peter Van Endert
- h Université Paris Descartes, INSERM, U1151, CNRS , Paris , France
| | - Stefano Marini
- a Department of Clinical Sciences and Translation Medicine , University of Roma Tor Vergata , Roma , Italy.,b CIRCMSB , Bari , Italy.,c Center for TeleInfrastructures, University of Roma Tor Vergata , Roma , Italy
| | - Massimo Coletta
- a Department of Clinical Sciences and Translation Medicine , University of Roma Tor Vergata , Roma , Italy.,b CIRCMSB , Bari , Italy.,c Center for TeleInfrastructures, University of Roma Tor Vergata , Roma , Italy
| |
Collapse
|
35
|
Grape powder consumption affects the expression of neurodegeneration-related brain proteins in rats chronically fed a high-fructose–high-fat diet. J Nutr Biochem 2017; 43:132-140. [DOI: 10.1016/j.jnutbio.2017.02.013] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Revised: 01/11/2017] [Accepted: 02/08/2017] [Indexed: 01/19/2023]
|
36
|
Enche Ady CNA, Lim SM, Teh LK, Salleh MZ, Chin AV, Tan MP, Poi PJH, Kamaruzzaman SB, Abdul Majeed AB, Ramasamy K. Metabolomic-guided discovery of Alzheimer's disease biomarkers from body fluid. J Neurosci Res 2017; 95:2005-2024. [PMID: 28301062 DOI: 10.1002/jnr.24048] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Revised: 01/31/2017] [Accepted: 02/15/2017] [Indexed: 12/11/2022]
Abstract
The rapid increase in the older population has made age-related diseases like Alzheimer's disease (AD) a global concern. Given that there is still no cure for this neurodegenerative disease, the drastic growth in the number of susceptible individuals represents a major emerging threat to public health. The poor understanding of the mechanisms underlying AD is deemed the greatest stumbling block against progress in definitive diagnosis and management of this disease. There is a dire need for biomarkers that can facilitate early diagnosis, classification, prognosis, and treatment response. Efforts have been directed toward discovery of reliable and distinctive AD biomarkers but with very little success. With the recent emergence of high-throughput technology that is able to collect and catalogue vast datasets of small metabolites, metabolomics offers hope for a better understanding of AD and subsequent identification of biomarkers. This review article highlights the potential of using multiple metabolomics platforms as useful means in uncovering AD biomarkers from body fluids. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Che Nor Adlia Enche Ady
- Faculty of Pharmacy, University Teknologi MARA (UiTM), 42300 Bandar Puncak Alam, Selangor Darul Ehsan, Malaysia.,Collaborative Drug Discovery Research (CDDR) Group, Pharmaceutical and Life Sciences Community of Research, Universiti Teknologi MARA (UiTM), 40450 Shah Alam, Selangor Darul Ehsan, Malaysia
| | - Siong Meng Lim
- Faculty of Pharmacy, University Teknologi MARA (UiTM), 42300 Bandar Puncak Alam, Selangor Darul Ehsan, Malaysia.,Collaborative Drug Discovery Research (CDDR) Group, Pharmaceutical and Life Sciences Community of Research, Universiti Teknologi MARA (UiTM), 40450 Shah Alam, Selangor Darul Ehsan, Malaysia
| | - Lay Kek Teh
- Integrative Pharmacogenomics Institute (iPROMISE), Universiti Teknologi MARA (UiTM), 42300 Bandar Puncak Alam, Selangor Darul Ehsan, Malaysia
| | - Mohd Zaki Salleh
- Integrative Pharmacogenomics Institute (iPROMISE), Universiti Teknologi MARA (UiTM), 42300 Bandar Puncak Alam, Selangor Darul Ehsan, Malaysia
| | - Ai-Vyrn Chin
- Division of Geriatric Medicine, Department of Medicine, Faculty of Medicine, University of Malaya, 50603 Kuala Lumpur, Malaysia
| | - Maw Pin Tan
- Division of Geriatric Medicine, Department of Medicine, Faculty of Medicine, University of Malaya, 50603 Kuala Lumpur, Malaysia
| | - Philip Jun Hua Poi
- Division of Geriatric Medicine, Department of Medicine, Faculty of Medicine, University of Malaya, 50603 Kuala Lumpur, Malaysia
| | - Shahrul Bahyah Kamaruzzaman
- Division of Geriatric Medicine, Department of Medicine, Faculty of Medicine, University of Malaya, 50603 Kuala Lumpur, Malaysia
| | - Abu Bakar Abdul Majeed
- Faculty of Pharmacy, University Teknologi MARA (UiTM), 42300 Bandar Puncak Alam, Selangor Darul Ehsan, Malaysia.,Brain Degeneration and Therapeutics Group, Pharmaceutical and Life Sciences Community of Research, Universiti Teknologi MARA (UiTM), 40450 Shah Alam, Selangor Darul Ehsan, Malaysia
| | - Kalavathy Ramasamy
- Faculty of Pharmacy, University Teknologi MARA (UiTM), 42300 Bandar Puncak Alam, Selangor Darul Ehsan, Malaysia.,Collaborative Drug Discovery Research (CDDR) Group, Pharmaceutical and Life Sciences Community of Research, Universiti Teknologi MARA (UiTM), 40450 Shah Alam, Selangor Darul Ehsan, Malaysia
| |
Collapse
|
37
|
Ashok BS, Ajith TA, Sivanesan S. Hypoxia-inducible factors as neuroprotective agent in Alzheimer's disease. Clin Exp Pharmacol Physiol 2017; 44:327-334. [PMID: 28004401 DOI: 10.1111/1440-1681.12717] [Citation(s) in RCA: 84] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Revised: 11/17/2016] [Accepted: 12/15/2016] [Indexed: 07/31/2024]
Abstract
Beta amyloid (Aβ)-42 peptide and phosphorylated tau protein have been demonstrated as the pathological hallmarks of Alzheimer's disease (AD). A gradual decline of oxygen and glucose supply to the brain during aging or hypoxia was manifested as a contributing factor to hypometabolism. The brain regions susceptible to hypometabolism are the hippocampus, entorhinal cortex and cognition-associated neocortical regions like parietal, temporal and frontal cortex. In AD patients, the brain regions with hypometabolism can trigger overexpression of amyloid precursor protein and decrease the clearance of Aβ. Aβ and hypoxia can evoke inflammation, oxidative stress and finally neuronal cell death. Among the transcription factors involved in the compensatory mechanism, hypoxia-inducible factor-1 alpha (HIF-1α) has a major role in the cellular adaptation by inducing the expression of several proteins, including vascular endothelial growth factor, erythropoietin and inducible nitric oxide synthase. Therefore, maintaining the HIF-1α level by inhibiting the prolyl 4-hydroxylase was effective to attenuate the nerve damage during hypoxia and postpone the incidence of AD. Agents such as iron chelators, and heavy metals like cobalt and nickel were demonstrated to be effective in maintaining the HIF-1α level in the nerve. This review article discusses the possible role of HIF-1α as a neuroprotector in AD and the future perspectives.
Collapse
Affiliation(s)
- Ben Sundra Ashok
- Department of Biochemistry, Sri Ramachandra Medical College and Research Institute, Sri Ramachandra University, Chennai, Tamil Nadu, India
| | | | | |
Collapse
|
38
|
Zhao HY, Wu HJ, He JL, Zhuang JH, Liu ZY, Huang LQ, Zhao ZX. Chronic Sleep Restriction Induces Cognitive Deficits and Cortical Beta-Amyloid Deposition in Mice via BACE1-Antisense Activation. CNS Neurosci Ther 2017; 23:233-240. [PMID: 28145081 DOI: 10.1111/cns.12667] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Revised: 11/27/2016] [Accepted: 11/29/2016] [Indexed: 12/11/2022] Open
Abstract
AIMS To clarify the correlation between chronic sleep restriction (CSR) and sporadic Alzheimer disease (AD), we determined in wild-type mice the impact of CSR, on cognitive performance, beta-amyloid (Aβ) peptides, and its feed-forward regulators regarding AD pathogenesis. METHODS Sixteen nine-month-old C57BL/6 male mice were equally divided into the CSR and control groups. CSR was achieved by application of a slowly rotating drum for 2 months. The Morris water maze test was used to assess cognitive impairment. The concentrations of Aβ peptides, amyloid precursor protein (APP) and β-secretase 1 (BACE1), and the mRNA levels of BACE1 and BACE1-antisense (BACE1-AS) were measured. RESULTS Following CSR, impairments of spatial learning and memory consolidation were observed in the mice, accompanied by Aβ plaque deposition and an increased Aβ concentration in the prefrontal and temporal lobe cortex. CSR also upregulated the β-secretase-induced cleavage of APP by increasing the protein and mRNA levels of BACE1, particularly the BACE1-AS. CONCLUSIONS This study shows that a CSR accelerates AD pathogenesis in wild-type mice. An upregulation of the BACE1 pathway appears to participate in both cortical Aβ plaque deposition and memory impairment caused by CSR. BACE1-AS is likely activated to initiate a cascade of events that lead to AD pathogenesis. Our study provides, therefore, a molecular mechanism that links CSR to sporadic AD.
Collapse
Affiliation(s)
- Hong-Yi Zhao
- Department of Neurology, Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Hui-Juan Wu
- Department of Neurology, Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Jia-Lin He
- Academy of Clinical Medicine, Second Military Medical University, Shanghai, China
| | - Jian-Hua Zhuang
- Department of Neurology, Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Zhen-Yu Liu
- Department of Neurology, Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Liu-Qing Huang
- Department of Neurology, Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Zhong-Xin Zhao
- Department of Neurology, Changzheng Hospital, Second Military Medical University, Shanghai, China
| |
Collapse
|
39
|
Cao J, Tang Y, Li Y, Gao K, Shi X, Li Z. Behavioral Changes and Hippocampus Glucose Metabolism in APP/PS1 Transgenic Mice via Electro-acupuncture at Governor Vessel Acupoints. Front Aging Neurosci 2017; 9:5. [PMID: 28174534 PMCID: PMC5259686 DOI: 10.3389/fnagi.2017.00005] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Accepted: 01/10/2017] [Indexed: 11/22/2022] Open
Abstract
Objective: Investigating the effects of electro-acupuncture (EA) treatment on mice with Alzheimer’s disease (AD), using Morris water maze (MWM) for spatial learning and memory behavior tests combined with micro-positron emission tomography (micro-PET) imaging for glucose metabolism in hippocampus. Methods: Thirty seven-month-old APP/PS1 mice were randomly divided into AD Model group (AD group), medicine group (M group) and EA group, C57BL/6 mice were used for Normal control group (N group), n = 10 in each group. Mice in M group received donepezil intervention by gavage with dose at 0.92 mg/kg. EA was applied at Baihui (GV20) and Yintang (GV29) acupoints for 20 min then pricked at Shuigou (GV26) acupoint, while mice in N, M and AD groups were received restriction for 20 min, with all treatment administrated once a day for 15 consecutive days. After the treatment, MWM was performed to observe behavioral changes in mice, then hippocampus glucose metabolism level was tested by micro-PET imaging. Results: Compared with that of AD group, the escape latency of M and EA groups declined significantly (P < 0.01), while the proportion of the platform quadrant swimming distance in total swimming distance showed an obvious increase (P < 0.01), and EA group occupied a higher percentage than that in M group. The micro-PET imaging showed that mice in AD group performed a lower glucose metabolic rate in hippocampus compared with N group (P < 0.01). Both M and EA groups presented a significant higher injected dose compared with AD group (P < 0.01), and the uptake rate of EA group was higher than M group. Conclusion: Both donepezil and EA have therapeutic effects on AD mice. To a certain extent, EA shows a better efficacy in treatment of AD by improving the spatial learning and memory ability, while also enhancing glucose metabolism in hippocampus.
Collapse
Affiliation(s)
- Jin Cao
- School of Acupuncture Moxibustion and Tuina, Beijing University of Chinese Medicine Beijing, China
| | - Yinshan Tang
- Department of Rehabilitation in Traditional Chinese Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine Hangzhou, China
| | - Yujie Li
- School of Acupuncture Moxibustion and Tuina, Beijing University of Chinese Medicine Beijing, China
| | - Kai Gao
- Institute of Medical Laboratory Animal Science, Chinese Academy of Medical Sciences Beijing, China
| | - Xudong Shi
- Institute of Medical Laboratory Animal Science, Chinese Academy of Medical Sciences Beijing, China
| | - Zhigang Li
- School of Acupuncture Moxibustion and Tuina, Beijing University of Chinese Medicine Beijing, China
| |
Collapse
|
40
|
Cardoso S, Seiça R, Moreira PI. Diabesity and Brain Energy Metabolism: The Case of Alzheimer's Disease. ADVANCES IN NEUROBIOLOGY 2017; 19:117-150. [PMID: 28933063 DOI: 10.1007/978-3-319-63260-5_5] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/21/2023]
Abstract
It is widely accepted that high calorie diets and a sedentary lifestyle sturdily influence the incidence and outcome of type 2 diabetes and obesity, which can occur simultaneously, a situation called diabesity. Tightly linked with metabolic and energy regulation, a close association between diabetes and Alzheimer's disease (AD) has been proposed. Among the common pathogenic mechanisms that underpin both conditions, insulin resistance, brain glucose hypometabolism, and metabolic dyshomeostasis appear to have a pivotal role. This century is an unprecedented diabetogenic period in human history, so therapeutic strategies and/or approaches to control and/or revert this evolving epidemic is of utmost importance. This chapter will make a brief contextualization about the impact that diabetes and obesity can exert in brain structure and function alongside with a brief survey about the role of insulin in normal brain function, exploring its roles in cognition and brain glucose metabolism. Later, attention will be given to the intricate relation of diabesity, insulin resistance, and AD. Finally, both pharmacological and lifestyle interventions will also be reviewed as strategies aimed at fighting diabesity and/or AD-related metabolic effects.
Collapse
Affiliation(s)
- Susana Cardoso
- Institute for Interdisciplinary Research, University of Coimbra, Coimbra, Portugal.
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal.
| | - Raquel Seiça
- Institute of Physiology, Institute for Biomedical Imaging and Life Sciences-IBILI, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Paula I Moreira
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
- Institute of Physiology, Institute for Biomedical Imaging and Life Sciences-IBILI, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| |
Collapse
|
41
|
Fong S, Teo E, Ng LF, Chen CB, Lakshmanan LN, Tsoi SY, Moore PK, Inoue T, Halliwell B, Gruber J. Energy crisis precedes global metabolic failure in a novel Caenorhabditis elegans Alzheimer Disease model. Sci Rep 2016; 6:33781. [PMID: 27653553 PMCID: PMC5031964 DOI: 10.1038/srep33781] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2016] [Accepted: 09/01/2016] [Indexed: 01/01/2023] Open
Abstract
Alzheimer Disease (AD) is a progressive neurological disorder characterized by the deposition of amyloid beta (Aβ), predominantly the Aβ1–42 form, in the brain. Mitochondrial dysfunction and impaired energy metabolism are important components of AD pathogenesis. However, the causal and temporal relationships between them and AD pathology remain unclear. Using a novel C. elegans AD strain with constitutive neuronal Aβ1–42 expression that displays neuromuscular defects and age-dependent behavioural dysfunction reminiscent of AD, we have shown that mitochondrial bioenergetic deficit is an early event in AD pathogenesis, preceding dysfunction of mitochondrial electron transfer chain (ETC) complexes and the onset of global metabolic failure. These results are consistent with an emerging view that AD may be a metabolic neurodegenerative disease, and also confirm that Aβ-driven metabolic and mitochondrial effects can be reproduced in organisms separated by large evolutionary distances.
Collapse
Affiliation(s)
- Sheng Fong
- Internal Medicine Residency Programme, SingHealth Group, Singapore
| | - Emelyne Teo
- NUS Graduate School for Integrative Sciences and Engineering, National University of Singapore, Singapore
| | - Li Fang Ng
- Science Division, Yale-NUS College, Singapore
| | - Ce-Belle Chen
- Department of Biochemistry, National University of Singapore, Singapore
| | | | | | - Philip Keith Moore
- NUS Graduate School for Integrative Sciences and Engineering, National University of Singapore, Singapore
| | - Takao Inoue
- Department of Biochemistry, National University of Singapore, Singapore
| | - Barry Halliwell
- Department of Biochemistry, National University of Singapore, Singapore
| | - Jan Gruber
- Science Division, Yale-NUS College, Singapore.,Department of Biochemistry, National University of Singapore, Singapore
| |
Collapse
|
42
|
Kandimalla R, Thirumala V, Reddy PH. Is Alzheimer's disease a Type 3 Diabetes? A critical appraisal. Biochim Biophys Acta Mol Basis Dis 2016; 1863:1078-1089. [PMID: 27567931 DOI: 10.1016/j.bbadis.2016.08.018] [Citation(s) in RCA: 384] [Impact Index Per Article: 42.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Revised: 08/07/2016] [Accepted: 08/17/2016] [Indexed: 12/20/2022]
Abstract
Recently researchers proposed the term 'Type-3-Diabetes' for Alzheimer's disease (ad) because of the shared molecular and cellular features among Type-1-Diabetes, Type-2-Diabetes and insulin resistance associated with memory deficits and cognitive decline in elderly individuals. Recent clinical and basic studies on patients with diabetes and AD revealed previously unreported cellular and pathological among diabetes, insulin resistance and AD. These studies are also strengthened by various basic biological studies that decipher the effects of insulin in the pathology of AD through cellular and molecular mechanisms. For instance, insulin is involved in the activation of glycogen synthase kinase 3β, which in turn causes phosphorylation of tau, which involved in the formation of neurofibrillary tangles. Interestingly, insulin also plays a crucial role in the formation amyloid plaques. In this review, we discussed significant shared mechanisms between AD and diabetes and we also provided therapeutic avenues for diabetes and AD. This article is part of a Special Issue entitled: Oxidative Stress and Mitochondrial Quality in Diabetes/Obesity and Critical Illness Spectrum of Diseases - edited by P. Hemachandra Reddy.
Collapse
Affiliation(s)
- Ramesh Kandimalla
- Garrison Institute on Aging, Texas Tech University Health Sciences Center, 3601 4th Street, MS 9424, Lubbock, TX 79430, United States.
| | - Vani Thirumala
- Garrison Institute on Aging, Texas Tech University Health Sciences Center, 3601 4th Street, MS 9424, Lubbock, TX 79430, United States; BSA Neuroscience, University of Texas at Austin, Austin, TX 78712, USA
| | - P Hemachandra Reddy
- Garrison Institute on Aging, Texas Tech University Health Sciences Center, 3601 4th Street, MS 9424, Lubbock, TX 79430, United States; Departments of Cell Biology & Biochemistry, Neuroscience & Pharmacology and Neurology, Texas Tech University Health Sciences Center, 3601 4th Street, MS 9424, Lubbock, TX 79430, United States
| |
Collapse
|
43
|
Adlimoghaddam A, Sabbir MG, Albensi BC. Ammonia as a Potential Neurotoxic Factor in Alzheimer's Disease. Front Mol Neurosci 2016; 9:57. [PMID: 27551259 PMCID: PMC4976099 DOI: 10.3389/fnmol.2016.00057] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2016] [Accepted: 07/06/2016] [Indexed: 11/13/2022] Open
Abstract
Ammonia is known to be a potent neurotoxin that causes severe negative effects on the central nervous system. Excessive ammonia levels have been detected in the brain of patients with neurological disorders such as Alzheimer disease (AD). Therefore, ammonia could be a factor contributing to the progression of AD. In this review, we provide an introduction to the toxicity of ammonia and putative ammonia transport proteins. We also hypothesize how ammonia may be linked to AD. Additionally, we discuss the evidence that support the hypothesis that ammonia is a key factor contributing to AD progression. Lastly, we summarize the old and new experimental evidence that focuses on energy metabolism, mitochondrial function, inflammatory responses, excitatory glutamatergic, and GABAergic neurotransmission, and memory in support of our ammonia-related hypotheses of AD.
Collapse
Affiliation(s)
- Aida Adlimoghaddam
- Division of Neurodegenerative Disorders, St. Boniface Hospital Research Winnipeg, MB, Canada
| | - Mohammad G Sabbir
- Division of Neurodegenerative Disorders, St. Boniface Hospital Research Winnipeg, MB, Canada
| | - Benedict C Albensi
- Division of Neurodegenerative Disorders, St. Boniface Hospital ResearchWinnipeg, MB, Canada; Department of Pharmacology & Therapeutics, University of ManitobaWinnipeg, MB, Canada
| |
Collapse
|
44
|
Ribarič S. The Rationale for Insulin Therapy in Alzheimer's Disease. Molecules 2016; 21:molecules21060689. [PMID: 27240327 PMCID: PMC6273626 DOI: 10.3390/molecules21060689] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Revised: 05/14/2016] [Accepted: 05/19/2016] [Indexed: 12/30/2022] Open
Abstract
Alzheimer's disease (AD) is the most common form of dementia, with a prevalence that increases with age. By 2050, the worldwide number of patients with AD is projected to reach more than 140 million. The prominent signs of AD are progressive memory loss, accompanied by a gradual decline in cognitive function and premature death. AD is the clinical manifestation of altered proteostasis. The initiating step of altered proteostasis in most AD patients is not known. The progression of AD is accelerated by several chronic disorders, among which the contribution of diabetes to AD is well understood at the cell biology level. The pathological mechanisms of AD and diabetes interact and tend to reinforce each other, thus accelerating cognitive impairment. At present, only symptomatic interventions are available for treating AD. To optimise symptomatic treatment, a personalised therapy approach has been suggested. Intranasal insulin administration seems to open the possibility for a safe, and at least in the short term, effective symptomatic intervention that delays loss of cognition in AD patients. This review summarizes the interactions of AD and diabetes from the cell biology to the patient level and the clinical results of intranasal insulin treatment of cognitive decline in AD.
Collapse
Affiliation(s)
- Samo Ribarič
- Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, Zaloška 4, SI-1000 Ljubljana, Slovenia.
| |
Collapse
|
45
|
Yang SH, Li W, Sumien N, Forster M, Simpkins JW, Liu R. Alternative mitochondrial electron transfer for the treatment of neurodegenerative diseases and cancers: Methylene blue connects the dots. Prog Neurobiol 2015; 157:273-291. [PMID: 26603930 DOI: 10.1016/j.pneurobio.2015.10.005] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2015] [Revised: 09/10/2015] [Accepted: 10/20/2015] [Indexed: 12/21/2022]
Abstract
Brain has exceptional high requirement for energy metabolism with glucose as the exclusive energy source. Decrease of brain energy metabolism and glucose uptake has been found in patients of Alzheimer's, Parkinson's and other neurodegenerative diseases, providing a clear link between neurodegenerative disorders and energy metabolism. On the other hand, cancers, including glioblastoma, have increased glucose uptake and rely on aerobic glycolysis for energy metabolism. The switch of high efficient oxidative phosphorylation to low efficient aerobic glycolysis pathway (Warburg effect) provides macromolecule for biosynthesis and proliferation. Current research indicates that methylene blue, a century old drug, can receive electron from NADH in the presence of complex I and donates it to cytochrome c, providing an alternative electron transfer pathway. Methylene blue increases oxygen consumption, decrease glycolysis, and increases glucose uptake in vitro. Methylene blue enhances glucose uptake and regional cerebral blood flow in rats upon acute treatment. In addition, methylene blue provides protective effect in neuron and astrocyte against various insults in vitro and in rodent models of Alzheimer's, Parkinson's, and Huntington's disease. In glioblastoma cells, methylene blue reverses Warburg effect by enhancing mitochondrial oxidative phosphorylation, arrests glioma cell cycle at s-phase, and inhibits glioma cell proliferation. Accordingly, methylene blue activates AMP-activated protein kinase, inhibits downstream acetyl-coA carboxylase and cyclin-dependent kinases. In summary, there is accumulating evidence providing a proof of concept that enhancement of mitochondrial oxidative phosphorylation via alternative mitochondrial electron transfer may offer protective action against neurodegenerative diseases and inhibit cancers proliferation.
Collapse
Affiliation(s)
- Shao-Hua Yang
- Center for Neuroscience Discovery, University of North Texas Health Science Center, Fort Worth, TX 76107, USA.
| | - Wenjun Li
- Center for Neuroscience Discovery, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| | - Nathalie Sumien
- Center for Neuroscience Discovery, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| | - Michael Forster
- Center for Neuroscience Discovery, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| | - James W Simpkins
- Department of Physiology and Pharmacology, Center for Neuroscience, Health Science Center, West Virginia University, Medical Center Drive, Morgantown, WV 26506, USA
| | - Ran Liu
- Center for Neuroscience Discovery, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| |
Collapse
|
46
|
Goldman JG, Aggarwal NT, Schroeder CD. Mild cognitive impairment: an update in Parkinson's disease and lessons learned from Alzheimer's disease. Neurodegener Dis Manag 2015; 5:425-43. [PMID: 26517759 DOI: 10.2217/nmt.15.34] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Cognitive dysfunction is an important focus of research in Parkinson's disease (PD) and Alzheimer's disease (AD). While the concept of amnestic mild cognitive impairment (MCI) as a prodrome to AD has been recognized for many years, the construct of MCI in PD is a relative newcomer with recent development of diagnostic criteria, biomarker research programs and treatment trials. Controversies and challenges, however, regarding PD-MCI's definition, application, heterogeneity and different trajectories have arisen. This review will highlight current research advances and challenges in PD-MCI. Furthermore, lessons from the AD field, which has witnessed an evolution in MCI/AD definitions, relevant advances in biomarker research and development of disease-modifying and targeted therapeutic trials will be discussed.
Collapse
Affiliation(s)
- Jennifer G Goldman
- Rush University Medical Center, Department of Neurological Sciences, Section of Parkinson Disease & Movement Disorders, 1725 W. Harrison Street, Suite 755, Chicago, IL 60612, USA
| | - Neelum T Aggarwal
- Rush University Medical Center, Department of Neurological Sciences & Rush Alzheimer's Disease Center, 600 South Paulina, Suite 1038, Chicago, IL 60612, USA
| | - Cynthia D Schroeder
- Rush University Medical Center, Department of Neurological Sciences, 1735 W. Harrison Street, Suite 306, Chicago, IL 60612, USA
| |
Collapse
|
47
|
Jha NK, Jha SK, Kumar D, Kejriwal N, Sharma R, Ambasta RK, Kumar P. Impact of Insulin Degrading Enzyme and Neprilysin in Alzheimer’s Disease Biology: Characterization of Putative Cognates for Therapeutic Applications. J Alzheimers Dis 2015; 48:891-917. [DOI: 10.3233/jad-150379] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Niraj Kumar Jha
- Molecular Neuroscience and Functional Genomics Laboratory, Department of Biotechnology, Delhi Technological University (Formerly DCE), Shahbad Daulatpur, Delhi, India
| | - Saurabh Kumar Jha
- Molecular Neuroscience and Functional Genomics Laboratory, Department of Biotechnology, Delhi Technological University (Formerly DCE), Shahbad Daulatpur, Delhi, India
| | - Dhiraj Kumar
- Molecular Neuroscience and Functional Genomics Laboratory, Department of Biotechnology, Delhi Technological University (Formerly DCE), Shahbad Daulatpur, Delhi, India
| | - Noopur Kejriwal
- Molecular Neuroscience and Functional Genomics Laboratory, Department of Biotechnology, Delhi Technological University (Formerly DCE), Shahbad Daulatpur, Delhi, India
| | - Renu Sharma
- Molecular Neuroscience and Functional Genomics Laboratory, Department of Biotechnology, Delhi Technological University (Formerly DCE), Shahbad Daulatpur, Delhi, India
| | - Rashmi K. Ambasta
- Molecular Neuroscience and Functional Genomics Laboratory, Department of Biotechnology, Delhi Technological University (Formerly DCE), Shahbad Daulatpur, Delhi, India
| | - Pravir Kumar
- Molecular Neuroscience and Functional Genomics Laboratory, Department of Biotechnology, Delhi Technological University (Formerly DCE), Shahbad Daulatpur, Delhi, India
- Department of Neurology, Tufts University School of Medicine, Boston, MA, USA
| |
Collapse
|
48
|
Oliveira LT, Leon GVO, Provance DW, de Mello FG, Sorenson MM, Salerno VP. Exogenous β-amyloid peptide interferes with GLUT4 localization in neurons. Brain Res 2015; 1615:42-50. [PMID: 25912430 DOI: 10.1016/j.brainres.2015.04.026] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2014] [Accepted: 04/15/2015] [Indexed: 01/14/2023]
Abstract
Aging represents a major risk factor for numerous illnesses that are of increasing importance to society, including two of the most prevalent: diabetes and Alzheimer's disease. Studies have shown that diabetes is a risk factor for spontaneous Alzheimer's disease. While these studies suggest that diabetes can contribute to Alzheimer's disease, the implications of AD on diabetes are practically unexplored. The major mediator of the pathophysiological effects, the Aβ42 peptide, has been shown to enter neurons and lead to an alteration of the intracellular distribution of the molecular motor myosin Vb. Myosin Vb functions in memory and learning by participating in the strengthening of the long-term potentiation (LTP) of synaptic transmissions. It has also been implicated in the translocation of the glucose transporter, GLUT4, to the plasma membrane in response to insulin, a process that is defective in diabetes. Here, the effect on GLUT4 upon entry of the Aβ42 peptide into cultured chick retinal neurons was explored. The results suggest an alteration in distribution and a reduced level at the cell surface, as well as an increased colocalization with myosin Vb, which can partially explain the changes in glucose metabolism associated with AD. It is also shown that the presence of the Aβ40 peptide inhibits the internalization of the Aβ42 peptide in cultured cells. Together, the results provide additional targets for the development of therapeutics against the progression and effects of Alzheimer's disease.
Collapse
Affiliation(s)
- Leandro T Oliveira
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, RJ 21944-590, Brazil; School of Physical Education and Sports, Federal University of Rio deJaneiro, RJ 21944-590, Brazil
| | - Gabbriela V O Leon
- School of Physical Education and Sports, Federal University of Rio deJaneiro, RJ 21944-590, Brazil
| | - D William Provance
- Center for the Development of Technology in Health (CDTS)/National Institute of Science and Technology for Innovation in Neglected Diseases (INCT-IDN), Oswaldo Cruz Foundation, RJ 21040-361, Brazil
| | - Fernando G de Mello
- Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, RJ 21944-590, Brazil
| | - Martha M Sorenson
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, RJ 21944-590, Brazil
| | - Verônica P Salerno
- School of Physical Education and Sports, Federal University of Rio deJaneiro, RJ 21944-590, Brazil.
| |
Collapse
|
49
|
Effects of ageing and experimental diabetes on insulin-degrading enzyme expression in male rat tissues. Biogerontology 2015; 16:473-84. [PMID: 25792373 DOI: 10.1007/s10522-015-9569-9] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2014] [Accepted: 03/11/2015] [Indexed: 10/23/2022]
Abstract
Due to an increasing life expectancy in developing countries, cases of type 2 diabetes and Alzheimer's disease (AD) in the elderly are growing exponentially. Despite a causative link between diabetes and AD, general molecular mechanisms underlying pathogenesis of these disorders are still far from being understood. One of the factors leading to cell death and cognitive impairment characteristic of AD is accumulation in the brain of toxic aggregates of amyloid-β peptide (Aβ). In the normally functioning brain Aβ catabolism is regulated by a cohort of proteolytic enzymes including insulin-degrading enzyme (IDE) and their deficit with ageing can result in Aβ accumulation and increased risk of AD. The aim of this study was a comparative analysis of IDE expression in the brain structures involved in AD, as well as in peripheral organs (the liver and kidney) of rats, during natural ageing and after experimentally-induced diabetes. It was found that ageing is accompanied by a significant decrease of IDE mRNA and protein content in the liver (by 32 and 81%) and brain structures (in the cortex by 58 and 47% and in the striatum by 53 and 68%, respectively). In diabetic animals, IDE protein level was increased in the liver (by 36%) and in the striatum (by 42%) while in the brain cortex and hippocampus it was 20-30% lower than in control animals. No significant IDE protein changes were observed in the kidney of diabetic rats. These data testify that ageing and diabetes are accompanied by a deficit of IDE in the brain structures where accumulation of Aβ was reported in AD patients, which might be one of the factors predisposing to development of the sporadic form of AD in the elderly, and especially in diabetics.
Collapse
|
50
|
Doert A, Pilatus U, Zanella F, Müller WE, Eckert GP. ¹H- and ¹³C-NMR spectroscopy of Thy-1-APPSL mice brain extracts indicates metabolic changes in Alzheimer's disease. J Neural Transm (Vienna) 2015; 122:541-50. [PMID: 25742870 DOI: 10.1007/s00702-015-1387-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2014] [Accepted: 02/22/2015] [Indexed: 01/17/2023]
Abstract
Biochemical alterations underlying the symptoms and pathomechanisms of Alzheimer's disease (AD) are not fully understood. However, alterations of glucose metabolism and mitochondrial dysfunction certainly play an important role. (1)H- and (13)C-NMR spectroscopy exhibits promising results in providing information about those alterations in vivo in patients and animals, especially regarding the mitochondrial tricarboxylic acid (TCA) cycle. Accordingly, transgenic mice expressing mutant human amyloid precursor protein (APP(SL))-serving as a model of neuropathological changes in AD-were examined with in vitro 1D (1)H- and 2D (1)H-(13)C-HSQC-NMR spectroscopy after oral administration of 1-(13)C-glucose and acquisition of brain material after 30 min. Perchloric acid extracts were measured using a 500 MHz spectrometer, providing more detailed information compared to in vivo spectra achievable nowadays. Area under curve (AUC) data of metabolite peaks were obtained and normalized in relation to the creatine signal, serving as internal reference. Besides confirming well-known metabolic alterations in AD like decreased N-acetylaspartate (NAA)/Creatine (Cr) ratio, new findings such as a decrease in phosphorylcholine (PC) are presented. Glutamate (Glu) and glutamine (Gln) concentrations were decreased while γ-aminobutyric acid (GABA) was elevated in Thy1-APP(SL) mice. (13)C-NMR spectroscopy revealed a shift in the Glx-2/Glx-4-ratio-where Glx represents a combined Glu/Gln-signal-towards Glx-2 in AD. These findings correlated well with the NAA/Cr-ratio. The Gln-4/Glu-4-ratio is altered in favor of Glu. Our findings suggest that glutamine synthetase (GS), which is predominantly present in glial cells may be impaired in the brain of Thy1-APP(SL) transgenic mice. Since GS is an ATP-dependent enzyme, mitochondrial dysfunction might contribute to reduced activity, which might also account for the increased metabolism of glutamate via the GABA shunt, a metabolic pathway to bypass intra-mitochondrial α-ketoglutarate-dehydrogenase, resulting in elevated GABA levels.
Collapse
Affiliation(s)
- A Doert
- Institute of Neuroradiology, Goethe-University Hospital, Frankfurt, Germany
| | | | | | | | | |
Collapse
|