51
|
Uddin MS, Kabir MT, Rahman MH, Alim MA, Rahman MM, Khatkar A, Al Mamun A, Rauf A, Mathew B, Ashraf GM. Exploring the Multifunctional Neuroprotective Promise of Rasagiline Derivatives for Multi-Dysfunctional Alzheimer's Disease. Curr Pharm Des 2021; 26:4690-4698. [PMID: 32250219 DOI: 10.2174/1381612826666200406075044] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Accepted: 02/19/2020] [Indexed: 01/07/2023]
Abstract
Alzheimer's disease (AD) is a chronic, age-related, and irreversible brain disorder that typically develops slowly and gets worse over time. The potent auspicious drug candidate for the treatment of AD is supposed to perform the simultaneous modulation of several targets linked to AD. The new therapeutic approach involves drug candidates that are designed to act on multiple targets and have various pharmacological properties. This trend has triggered the development of various multimodal drugs including TV-3326 (i.e. ladostigil) and M-30 (i.e. a new multitarget iron chelator). TV-3326 combines the neurorestorative/neuroprotective effects of the cholinesterase (ChE) inhibitory activity of rivastigmine with rasagiline (a selective monoamine oxidase-B inhibitor and novel antiparkinsonian agent) in a single molecule. M-30, the second derivative of rasagiline, was developed by combining the propargyl moiety of rasagiline into the skeleton of VK-28 (i.e. a novel brain permeable neuroprotective iron chelator). It has been revealed that both the compounds possess anti-AD effects and therefore, the clinical development is directed to the treatment of this type of neurodegenerative diseases (NDs). In this article, we have reviewed the neuroprotective molecular mechanisms and multimodal effects of TV-3326 and M-30.
Collapse
Affiliation(s)
- Md Sahab Uddin
- Department of Pharmacy, Southeast University, Dhaka, Bangladesh,Pharmakon Neuroscience Research Network, Dhaka, Bangladesh
| | | | - Md Habibur Rahman
- Department of Environmental Medical Biology, Wonju College of Medicine, Yonsei University, Wonju, Republic of Korea
| | - Md Abdul Alim
- Department of Chemistry, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, Bangladesh,Graduate School of Innovative Life Science, Faculty of Engineering, University of Toyama, Toyama, Japan
| | - Md Motiar Rahman
- Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Anurag Khatkar
- Laboratory for Preservation Technology and Enzyme Inhibition Studies, Faculty of Pharmaceutical
Sciences, Maharshi Dayanand University, Rohtak, India
| | - Abdullah Al Mamun
- Department of Pharmacy, Southeast University, Dhaka, Bangladesh,Pharmakon Neuroscience Research Network, Dhaka, Bangladesh
| | - Abdur Rauf
- Department of Chemistry, University of Swabi, Swabi, Anbar 23561, Khyber Pakhtunkhwa, Pakistan
| | - Bijo Mathew
- Division of Drug Design and Medicinal Chemistry Research Lab, Department of Pharmaceutical Chemistry, Ahalia School of Pharmacy, Palakkad, India
| | - Ghulam Md Ashraf
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia,Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University,
Jeddah, Saudi Arabia
| |
Collapse
|
52
|
Jang CH, Oh J, Lim JS, Kim HJ, Kim JS. Fermented Soy Products: Beneficial Potential in Neurodegenerative Diseases. Foods 2021; 10:foods10030636. [PMID: 33803607 PMCID: PMC8003083 DOI: 10.3390/foods10030636] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 03/11/2021] [Accepted: 03/15/2021] [Indexed: 12/12/2022] Open
Abstract
Fermented soybean products, such as cheonggukjang (Japanese natto), doenjang (soy paste), ganjang (soy sauce), and douchi, are widely consumed in East Asian countries and are major sources of bioactive compounds. The fermentation of cooked soybean with bacteria (Bacillus spp.) and fungi (Aspergillus spp. and Rhizopus spp.) produces a variety of novel compounds, most of which possess health benefits. This review is focused on the preventive and ameliorative potential of fermented soy foods and their components to manage neurodegenerative diseases, including Alzheimer's and Parkinson's diseases.
Collapse
Affiliation(s)
- Chan Ho Jang
- School of Food Science and Biotechnology, Kyungpook National University, Daegu 41566, Korea;
| | - Jisun Oh
- Institute of Agricultural Science and Technology, Kyungpook National University, Daegu 41566, Korea; (J.O.); (J.S.L.)
| | - Ji Sun Lim
- Institute of Agricultural Science and Technology, Kyungpook National University, Daegu 41566, Korea; (J.O.); (J.S.L.)
| | - Hyo Jung Kim
- Department of Korean Medicine Development, National Institute for Korean Medicine Development, Gyeongsan 38540, Korea;
| | - Jong-Sang Kim
- School of Food Science and Biotechnology, Kyungpook National University, Daegu 41566, Korea;
- Institute of Agricultural Science and Technology, Kyungpook National University, Daegu 41566, Korea; (J.O.); (J.S.L.)
- Department of Integrative Biotechnology, Kyungpook National University, Daegu 41566, Korea
- Correspondence: ; Tel.: +82-53-950-5752; Fax: +82-53-950-6750
| |
Collapse
|
53
|
Iqubal A, Iqubal MK, Fazal SA, Pottoo FH, Haque SE. Nutraceuticals and their Derived Nano-formulations for the Prevention and Treatment of Alzheimer's disease. Curr Mol Pharmacol 2021; 15:23-50. [PMID: 33687906 DOI: 10.2174/1874467214666210309115605] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 11/22/2020] [Accepted: 12/02/2020] [Indexed: 11/22/2022]
Abstract
Alzheimer's disease is one of the common chronic neurological disorders and associated with cognitive dysfunction, depression and progressive dementia. Presence of β-amyloid or senile plaques, hyper-phosphorylated tau proteins, neurofibrillary tangle, oxidative-nitrative stress, mitochondrial dysfunction, endoplasmic reticulum stress, neuroinflammation and derailed neurotransmitter status are the hallmark of AD. Currently, donepezil, memantine, rivastigmine and galantamine are approved by the FDA for symptomatic management. It is well-known that these approved drugs only exert symptomatic relief and possess poor patient-compliance. Additionally, various published evidence shows the neuroprotective potential of various nutraceuticals via their antioxidant, anti-inflammatory and anti-apoptotic effects in the preclinical and clinical studies. These nutraceuticals possess a significant neuroprotective potential and hence, can be a future pharmacotherapeutic for the management and treatment of AD. However, nutraceutical suffers from certain major limitations such as poor solubility, low bioavailability, low stability, fast hepatic-metabolism and larger particle size. These pharmacokinetic attributes restrict their entry into the brain via the blood-brain barrier. Therefore, to over such issues, various nanoformulation of nutraceuticals was developed, that allows their effective delivery into brain owning to reduced particle size, increased lipophilicity increased bioavailability and avoidance of fast hepatic metabolism. Thus, in this review, we have discussed the etiology of AD, focused on the pharmacotherapeutics of nutraceuticals with preclinical and clinical evidence, discussed pharmaceutical limitation and regulatory aspects of nutraceuticals to ensure safety and efficacy. We further explored the latitude of various nanoformulation of nutraceuticals as a novel approach to overcome the existing pharmaceutical limitation and for effective delivery into the brain.
Collapse
Affiliation(s)
- Ashif Iqubal
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi-110062. India
| | - Mohammad Kashif Iqubal
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi-110062. India
| | - Syed Abul Fazal
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi-110062. India
| | - Faheem Hyder Pottoo
- Department of Pharmacology, College of Clinical Pharmacy, Imam Abdulrahman Bin Faisal, University, P.O.BOX 1982, Damman, 31441. Saudi Arabia
| | - Syed Ehtaishamul Haque
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi-110062. India
| |
Collapse
|
54
|
Duro-Castano A, Borrás C, Herranz-Pérez V, Blanco-Gandía MC, Conejos-Sánchez I, Armiñán A, Mas-Bargues C, Inglés M, Miñarro J, Rodríguez-Arias M, García-Verdugo JM, Viña J, Vicent MJ. Targeting Alzheimer's disease with multimodal polypeptide-based nanoconjugates. SCIENCE ADVANCES 2021; 7:7/13/eabf9180. [PMID: 33771874 PMCID: PMC7997513 DOI: 10.1126/sciadv.abf9180] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Accepted: 02/08/2021] [Indexed: 05/04/2023]
Abstract
Alzheimer's disease (AD), the most prevalent form of dementia, remains incurable mainly due to our failings in the search for effective pharmacological strategies. Here, we describe the development of targeted multimodal polypeptide-based nanoconjugates as potential AD treatments. Treatment with polypeptide nanoconjugates bearing propargylamine moieties and bisdemethoxycurcumin or genistein afforded neuroprotection and displayed neurotrophic effects, as evidenced by an increase in dendritic density of pyramidal neurons in organotypic hippocampal culture. The additional conjugation of the Angiopep-2 targeting moiety enhanced nanoconjugate passage through the blood-brain barrier and modulated brain distribution with nanoconjugate accumulation in neurogenic areas, including the olfactory bulb. Nanoconjugate treatment effectively reduced neurotoxic β amyloid aggregate levels and rescued impairments to olfactory memory and object recognition in APP/PS1 transgenic AD model mice. Overall, this study provides a description of a targeted multimodal polyglutamate-based nanoconjugate with neuroprotective and neurotrophic potential for AD treatment.
Collapse
Affiliation(s)
- A Duro-Castano
- Polymer Therapeutics Lab., Centro de Investigación Príncipe Felipe (CIPF), Av. Eduardo Primo Yúfera 3, 46012 Valencia, Spain
| | - C Borrás
- Grupo de Investigación FRESHAGE, Departamento de Fisiología, Facultad de Medicina, Univ.. Valencia, CIBERFES-ISCIII, INCLIVA, Av. Blasco Ibáñez 15, 46010 Valencia, Spain
| | - V Herranz-Pérez
- Laboratory of Comparative Neurobiology, Cavanilles Institute of Biodiversity and Evolutionary Biology, Univ. València, CIBERNED, 46980 Valencia, Spain
- Predepartamental Unit of Medicine, Faculty of Health Sciences, Univ. Jaume I, 12071 Castelló de la Plana, Spain
| | - M C Blanco-Gandía
- Departamento de Psicología y Sociología, Facultad de Ciencias Sociales y Humanas, Univ. Zaragoza, Teruel, Spain
| | - I Conejos-Sánchez
- Polymer Therapeutics Lab., Centro de Investigación Príncipe Felipe (CIPF), Av. Eduardo Primo Yúfera 3, 46012 Valencia, Spain
| | - A Armiñán
- Polymer Therapeutics Lab., Centro de Investigación Príncipe Felipe (CIPF), Av. Eduardo Primo Yúfera 3, 46012 Valencia, Spain
| | - C Mas-Bargues
- Grupo de Investigación FRESHAGE, Departamento de Fisiología, Facultad de Medicina, Univ.. Valencia, CIBERFES-ISCIII, INCLIVA, Av. Blasco Ibáñez 15, 46010 Valencia, Spain
| | - M Inglés
- Unidad de Investigación Psicobiología de las Drogodependencias, Departamento de Psicobiología, Facultad de Psicología, Univ. Valencia, Valencia, Spain
| | - J Miñarro
- Unidad de Investigación Psicobiología de las Drogodependencias, Departamento de Psicobiología, Facultad de Psicología, Univ. Valencia, Valencia, Spain
| | - M Rodríguez-Arias
- Unidad de Investigación Psicobiología de las Drogodependencias, Departamento de Psicobiología, Facultad de Psicología, Univ. Valencia, Valencia, Spain
| | - J M García-Verdugo
- Laboratory of Comparative Neurobiology, Cavanilles Institute of Biodiversity and Evolutionary Biology, Univ. València, CIBERNED, 46980 Valencia, Spain
| | - J Viña
- Grupo de Investigación FRESHAGE, Departamento de Fisiología, Facultad de Medicina, Univ.. Valencia, CIBERFES-ISCIII, INCLIVA, Av. Blasco Ibáñez 15, 46010 Valencia, Spain
| | - M J Vicent
- Polymer Therapeutics Lab., Centro de Investigación Príncipe Felipe (CIPF), Av. Eduardo Primo Yúfera 3, 46012 Valencia, Spain.
| |
Collapse
|
55
|
Kabir MT, Uddin MS, Mathew B, Das PK, Perveen A, Ashraf GM. Emerging Promise of Immunotherapy for Alzheimer's Disease: A New Hope for the Development of Alzheimer's Vaccine. Curr Top Med Chem 2021; 20:1214-1234. [PMID: 32321405 DOI: 10.2174/1568026620666200422105156] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 03/13/2020] [Accepted: 03/13/2020] [Indexed: 12/21/2022]
Abstract
BACKGROUND Alzheimer's disease (AD) is a chronic neurodegenerative disorder and the characteristics of this devastating disorder include the progressive and disabling deficits in the cognitive functions including reasoning, attention, judgment, comprehension, memory, and language. OBJECTIVE In this article, we have focused on the recent progress that has been achieved in the development of an effective AD vaccine. SUMMARY Currently, available treatment options of AD are limited to deliver short-term symptomatic relief only. A number of strategies targeting amyloid-beta (Aβ) have been developed in order to treat or prevent AD. In order to exert an effective immune response, an AD vaccine should contain adjuvants that can induce an effective anti-inflammatory T helper 2 (Th2) immune response. AD vaccines should also possess the immunogens which have the capacity to stimulate a protective immune response against various cytotoxic Aβ conformers. The induction of an effective vaccine's immune response would necessitate the parallel delivery of immunogen to dendritic cells (DCs) and their priming to stimulate a Th2-polarized response. The aforesaid immune response is likely to mediate the generation of neutralizing antibodies against the neurotoxic Aβ oligomers (AβOs) and also anti-inflammatory cytokines, thus preventing the AD-related inflammation. CONCLUSION Since there is an age-related decline in the immune functions, therefore vaccines are more likely to prevent AD instead of providing treatment. AD vaccines might be an effective and convenient approach to avoid the treatment-related huge expense.
Collapse
Affiliation(s)
| | - Md Sahab Uddin
- Department of Pharmacy, Southeast University, Dhaka, Bangladesh.,Pharmakon Neuroscience Research Network, Dhaka, Bangladesh
| | - Bijo Mathew
- Division of Drug Design and Medicinal Chemistry Research Lab, Department of Pharmaceutical Chemistry, Ahalia School of Pharmacy, Palakkad, India
| | | | - Asma Perveen
- Glocal School of Life Sciences, Glocal University, Saharanpur, India
| | - Ghulam Md Ashraf
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia.,Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
56
|
Aziz MA, Sarwar MS, Akter T, Uddin MS, Xun S, Zhu Y, Islam MS, Hongjie Z. Polyphenolic molecules targeting STAT3 pathway for the treatment of cancer. Life Sci 2021; 268:118999. [PMID: 33421525 DOI: 10.1016/j.lfs.2020.118999] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Revised: 12/22/2020] [Accepted: 12/24/2020] [Indexed: 01/17/2023]
Abstract
Cancer is accounted as the second-highest cause of morbidity and mortality throughout the world. Numerous preclinical and clinical investigations have consistently highlighted the role of natural polyphenolic compounds against various cancers. A plethora of potential bioactive polyphenolic molecules, primarily flavonoids, phenolic acids, lignans and stilbenes, have been explored from the natural sources for their chemopreventive and chemoprotective activities. Moreover, combinations of these polyphenols with current chemotherapeutic agents have also demonstrated their strong role against both progression and resistance of malignancies. Signal transducer and activator of transcription 3 (STAT3) is a ubiquitously-expressed signaling molecule in almost all body cells. Thousands of literatures have revealed that STAT3 plays significant roles in promoting the cellular proliferation, differentiation, cell cycle progression, metastasis, angiogenesis and immunosuppression as well as chemoresistance through the regulation of its downstream target genes such as Bcl-2, Bcl-xL, cyclin D1, c-Myc and survivin. For its key role in cancer development, researchers considered STAT3 as a major target for cancer therapy that mainly focuses on abrogating the expression (activation or phosphorylation) of STAT3 in tumor cells both directly and indirectly. Polyphenolic molecules have explicated their protective actions in malignant cells via targeting STAT3 both in vitro and in vivo. In this article, we reviewed how polyphenolic compounds as well as their combinations with other chemotherapeutic drugs inhibit cancer cells by targeting STAT3 signaling pathway.
Collapse
Affiliation(s)
- Md Abdul Aziz
- Department of Pharmacy, Faculty of Science, Noakhali Science and Technology University, Noakhali 3814, Bangladesh
| | - Md Shahid Sarwar
- Department of Pharmacy, Faculty of Science, Noakhali Science and Technology University, Noakhali 3814, Bangladesh.
| | - Tahmina Akter
- Department of Pharmacy, Faculty of Science, Noakhali Science and Technology University, Noakhali 3814, Bangladesh
| | - Md Sahab Uddin
- Department of Pharmacy, Southeast University, Dhaka, Bangladesh; Pharmakon Neuroscience Research Network, Dhaka, Bangladesh
| | - Song Xun
- School of Pharmaceutical Science, Health Science Center, Shenzhen University, Shenzhen, China
| | - Yu Zhu
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, SAR, China
| | - Mohammad Safiqul Islam
- Department of Pharmacy, Faculty of Science, Noakhali Science and Technology University, Noakhali 3814, Bangladesh
| | - Zhang Hongjie
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, SAR, China.
| |
Collapse
|
57
|
Potential Therapeutic Role of Phytochemicals to Mitigate Mitochondrial Dysfunctions in Alzheimer's Disease. Antioxidants (Basel) 2020; 10:antiox10010023. [PMID: 33379372 PMCID: PMC7823298 DOI: 10.3390/antiox10010023] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 12/22/2020] [Accepted: 12/23/2020] [Indexed: 12/15/2022] Open
Abstract
Alzheimer’s disease (AD) is a progressive neurodegenerative disorder characterized by a decline in cognitive function and neuronal damage. Although the precise pathobiology of AD remains elusive, accumulating evidence suggests that mitochondrial dysfunction is one of the underlying causes of AD. Mutations in mitochondrial or nuclear DNA that encode mitochondrial components may cause mitochondrial dysfunction. In particular, the dysfunction of electron transport chain complexes, along with the interactions of mitochondrial pathological proteins are associated with mitochondrial dysfunction in AD. Mitochondrial dysfunction causes an imbalance in the production of reactive oxygen species, leading to oxidative stress (OS) and vice versa. Neuroinflammation is another potential contributory factor that induces mitochondrial dysfunction. Phytochemicals or other natural compounds have the potential to scavenge oxygen free radicals and enhance cellular antioxidant defense systems, thereby protecting against OS-mediated cellular damage. Phytochemicals can also modulate other cellular processes, including autophagy and mitochondrial biogenesis. Therefore, pharmacological intervention via neuroprotective phytochemicals can be a potential strategy to combat mitochondrial dysfunction as well as AD. This review focuses on the role of phytochemicals in mitigating mitochondrial dysfunction in the pathogenesis of AD.
Collapse
|
58
|
Viral Infection and Autophagy Dysregulation: The Case of HHV-6, EBV and KSHV. Cells 2020; 9:cells9122624. [PMID: 33297368 PMCID: PMC7762304 DOI: 10.3390/cells9122624] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 11/30/2020] [Accepted: 12/04/2020] [Indexed: 12/22/2022] Open
Abstract
Human Herpes Virus-6 (HHV-6), Epstein-Barr Virus (EBV) and Kaposi Sarcoma Herpes Virus (KSHV) are viruses that share with other member of the Herpesvirus family the capacity to interfere with the autophagic process. In this paper, mainly based on the findings of our laboratory, we describe how, through different mechanisms, these viruses converge in reducing autophagy to impair DC immune function and how, by infecting and dysregulating autophagy in different cell types, they promote the pathologies associated with their infection, from the neurodegenerative diseases such Alzheimer’s disease to cancer.
Collapse
|
59
|
Qu Z, Sun J, Zhang W, Yu J, Zhuang C. Transcription factor NRF2 as a promising therapeutic target for Alzheimer's disease. Free Radic Biol Med 2020; 159:87-102. [PMID: 32730855 DOI: 10.1016/j.freeradbiomed.2020.06.028] [Citation(s) in RCA: 78] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 06/15/2020] [Accepted: 06/17/2020] [Indexed: 12/12/2022]
Abstract
Oxidative stress is considered as one of the pathogenesis of Alzheimer's disease (AD) and plays an important role in the occurrence and development of AD. Nuclear factor erythroid 2 related factor 2 (NRF2) is a key regulatory of oxidative stress defence. There is growing evidence indicating the relationship between NRF2 and AD. NRF2 activation mitigates multiple pathogenic processes involved in AD by upregulating antioxidative defense, inhibiting neuroinflammation, improving mitochondrial function, maintaining proteostasis, and inhibiting ferroptosis. In addition, several NRF2 activators are currently being evaluated as AD therapeutic agents in clinical trials. Thus, targeting NRF2 has been the focus of a new strategy for prevention and treatment of AD. In this review, the role of NRF2 in AD and the NRF2 activators advanced into clinical and preclinical studies will be summarized.
Collapse
Affiliation(s)
- Zhuo Qu
- School of Pharmacy, Key Laboratory of Hui Ethnic Medicine Modernization, Ministry of Education, Ningxia Medical University, 1160 Shengli Street, Yinchuan, 750004, China
| | - Jiachen Sun
- School of Biotechnology and Food Science, Tianjin University of Commerce, 409 Guangrong Road, Tianjin, 300134, China
| | - Wannian Zhang
- School of Pharmacy, Key Laboratory of Hui Ethnic Medicine Modernization, Ministry of Education, Ningxia Medical University, 1160 Shengli Street, Yinchuan, 750004, China; School of Pharmacy, Second Military Medical University, 325 Guohe Road, Shanghai, 200433, China
| | - Jianqiang Yu
- School of Pharmacy, Key Laboratory of Hui Ethnic Medicine Modernization, Ministry of Education, Ningxia Medical University, 1160 Shengli Street, Yinchuan, 750004, China.
| | - Chunlin Zhuang
- School of Pharmacy, Key Laboratory of Hui Ethnic Medicine Modernization, Ministry of Education, Ningxia Medical University, 1160 Shengli Street, Yinchuan, 750004, China; School of Pharmacy, Second Military Medical University, 325 Guohe Road, Shanghai, 200433, China.
| |
Collapse
|
60
|
Sekikawa A, Higashiyama A, Lopresti BJ, Ihara M, Aizenstein H, Watanabe M, Chang Y, Kakuta C, Yu Z, Mathis C, Kokubo Y, Klunk W, Lopez OL, Kuller LH, Miyamoto Y, Cui C. Associations of equol-producing status with white matter lesion and amyloid-β deposition in cognitively normal elderly Japanese. ALZHEIMER'S & DEMENTIA (NEW YORK, N. Y.) 2020; 6:e12089. [PMID: 33117881 PMCID: PMC7580022 DOI: 10.1002/trc2.12089] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 08/10/2020] [Accepted: 08/17/2020] [Indexed: 11/09/2022]
Abstract
INTRODUCTION Equol, a metabolite of a soy isoflavone transformed by the gut microbiome, is anti-oxidant and anti-amyloidogenic. We assessed the associations of equol with white matter lesion normalized to total brain volume (WML%) and amyloid beta (Aβ) deposition. METHODS From 2016 to 2018, 91 cognitively normal elderly Japanese aged 75 to 89 underwent brain magnetic resonance imaging and positron emission tomography using 11C-Pittsburgh compound-B. Serum equol was measured using stored samples from 2008 to 2012. Equol producers were defined as individuals with serum levels >0. Producers were further divided into high (> the median) and low (≤ the median) producers. RESULTS The median (interquartile range) WML% was 1.10 (0.59 to 1.61); 24.2% were Aβ positive, and 51% were equol producers. Equol-producing status (non-producers, low and high) was significantly inversely associated with WML%: 1.19, 0.89, and 0.58, respectively (trend P < .01). Equol-producing status was not associated with Aβ status. DISCUSSION A randomized-controlled trial of equol targeting WML volume is warranted.
Collapse
Affiliation(s)
- Akira Sekikawa
- Department of EpidemiologyGraduate School of Public HealthUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Aya Higashiyama
- Department of Preventive CardiologyNational Cerebral and Cardiovascular CenterSuitaOsakaJapan
| | - Brian J Lopresti
- Department of RadiologyUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Masafumi Ihara
- Department of NeurologyNational Cerebral and Cardiovascular CenterSuitaOsakaJapan
| | - Howard Aizenstein
- Department of PsychiatryUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Makoto Watanabe
- Department of Preventive CardiologyNational Cerebral and Cardiovascular CenterSuitaOsakaJapan
| | - Yuefang Chang
- Department of Neurological SurgeryUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Chikage Kakuta
- Department of NeurologyNational Cerebral and Cardiovascular CenterSuitaOsakaJapan
| | - Zheming Yu
- Department of RadiologyUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Chester Mathis
- Department of RadiologyUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Yoshihioro Kokubo
- Department of Preventive CardiologyNational Cerebral and Cardiovascular CenterSuitaOsakaJapan
| | - William Klunk
- Department of PsychiatryUniversity of PittsburghPittsburghPennsylvaniaUSA
- Department of NeurologyUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Oscar L. Lopez
- Department of NeurologyUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Lewis H. Kuller
- Department of EpidemiologyGraduate School of Public HealthUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Yoshihiro Miyamoto
- Department of Preventive CardiologyNational Cerebral and Cardiovascular CenterSuitaOsakaJapan
- Open Innovation CenterNational Cerebral and Cardiovascular CenterSuitaOsakaJapan
| | - Chendi Cui
- Department of EpidemiologyGraduate School of Public HealthUniversity of PittsburghPittsburghPennsylvaniaUSA
| |
Collapse
|
61
|
Petry FDS, Hoppe JB, Klein CP, Dos Santos BG, Hözer RM, Bifi F, Matté C, Salbego CG, Trindade VMT. Genistein attenuates amyloid-beta-induced cognitive impairment in rats by modulation of hippocampal synaptotoxicity and hyperphosphorylation of Tau. J Nutr Biochem 2020; 87:108525. [PMID: 33065257 DOI: 10.1016/j.jnutbio.2020.108525] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Revised: 07/07/2020] [Accepted: 10/07/2020] [Indexed: 11/26/2022]
Abstract
Alzheimer's disease is a progressive neurodegenerative disorder characterized by extracellular accumulation of amyloid-beta (Aβ) peptide, which induces synaptic dysfunction, alteration of intracellular signaling pathways, hyperphosphorylation of the Tau protein, and cognitive impairment. Genistein, one of the major isoflavones present in soy and soy products, has been shown to modulate some of the pathogenic events associated with the neurodegeneration process. However, its underlying mechanisms remain to be clarified. Therefore, the objectives of the present study were to evaluate the ability of genistein to protect against Aβ1-42-induced cognitive impairment in rats and to elucidate some of the possible mechanisms involved in its neuroprotective effects in the hippocampus. Male Wistar rats received bilateral intracerebroventricular infusions of Aβ1-42 (2 nmol) and genistein 10 mg/kg orally for 10 days. The Aβ-infused animals showed significant impairment of memory, which was accompanied by the following neurochemical alterations in the hippocampus: decreased levels of the synaptic proteins synaptophysin and postsynaptic density protein 95 (PSD-95), hyperphosphorylation of Tau with increased activation of glycogen synthase kinase-3β and c-Jun N-terminal kinase, and inactivation of ERK. Treatment with genistein improved Aβ-induced cognitive impairment by attenuation of synaptotoxicity, hyperphosphorylation of Tau, and inactivation of ERK. Furthermore, treatment with this soy isoflavone did not cause systemic toxicity. These findings provide further evidence of the neuroprotective effect of genistein in an in vivo model of Aβ toxicity and, importantly, extend the current knowledge concerning the mechanisms associated with the neuroprotective effects of this compound in the hippocampus.
Collapse
Affiliation(s)
- Fernanda Dos Santos Petry
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil.
| | - Juliana Bender Hoppe
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Caroline Peres Klein
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Bernardo Gindri Dos Santos
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Régis Mateus Hözer
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Felippo Bifi
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Cristiane Matté
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil; Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Christianne Gazzana Salbego
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil; Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Vera Maria Treis Trindade
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil; Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| |
Collapse
|
62
|
Khadka B, Lee JY, Park DH, Kim KT, Bae JS. The Role of Natural Compounds and their Nanocarriers in the Treatment of CNS Inflammation. Biomolecules 2020; 10:E1401. [PMID: 33019651 PMCID: PMC7601486 DOI: 10.3390/biom10101401] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2020] [Revised: 09/25/2020] [Accepted: 09/26/2020] [Indexed: 12/16/2022] Open
Abstract
Neuroinflammation, which is involved in various inflammatory cascades in nervous tissues, can result in persistent and chronic apoptotic neuronal cell death and programmed cell death, triggering various degenerative disorders of the central nervous system (CNS). The neuroprotective effects of natural compounds against neuroinflammation are mainly mediated by their antioxidant, anti-inflammatory, and antiapoptotic properties that specifically promote or inhibit various molecular signal transduction pathways. However, natural compounds have several limitations, such as their pharmacokinetic properties and stability, which hinder their clinical development and use as medicines. This review discusses the molecular mechanisms of neuroinflammation and degenerative diseases of CNS. In addition, it emphasizes potential natural compounds and their promising nanocarriers for overcoming their limitations in the treatment of neuroinflammation. Moreover, recent promising CNS inflammation-targeted nanocarrier systems implementing lesion site-specific active targeting strategies for CNS inflammation are also discussed.
Collapse
Affiliation(s)
- Bikram Khadka
- Department of Biomedicine, Health & Life Convergence Sciences, BK21 Four, Mokpo National University, Muan-gun, Jeonnam 58554, Korea;
| | - Jae-Young Lee
- College of Pharmacy, Chungnam National University, Daejeon 34134, Korea;
| | - Dong Ho Park
- Department of Ophthalmology, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu 41944, Korea;
| | - Ki-Taek Kim
- Department of Biomedicine, Health & Life Convergence Sciences, BK21 Four, Mokpo National University, Muan-gun, Jeonnam 58554, Korea;
- College of Pharmacy and Natural Medicine Research Institute, Mokpo National University, Muan-gun, Jeonnam 58554, Korea
| | - Jong-Sup Bae
- College of Pharmacy, CMR1, Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu 41566, Korea
| |
Collapse
|
63
|
Emerging Therapeutic Promise of Ketogenic Diet to Attenuate Neuropathological Alterations in Alzheimer's Disease. Mol Neurobiol 2020; 57:4961-4977. [PMID: 32820459 DOI: 10.1007/s12035-020-02065-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Accepted: 08/07/2020] [Indexed: 12/18/2022]
Abstract
Alzheimer's disease (AD) is a multifactorial and chronic neurodegenerative disorder that interferes with memory, thinking, and behavior. The consumption of dietary fat has been considered a vital factor for AD as this disease is related to blood-brain barrier function and cholesterol signaling. The ε4 allele of apolipoprotein E (APOE4) is a primary genetic risk factor that encodes one of many proteins accountable for the transport of cholesterol and it is deemed as the leading cholesterol transport proteins in the brain. In case of AD development, the causative factor is the high level of serum/plasma cholesterol. However, this statement is arguable and, in the meantime, the levels of brain cholesterol in individuals with AD are extremely inconstant and levels of cholesterol in the brain and serum/plasma of AD individuals do not reflect cholesterol as a risk factor. In fact, APOE4 is neither fundamental nor sufficient for the advancement of AD; it just acts as a synergistic and increases the danger of AD. Another noticeable characteristic of AD is area-specific decreases in the metabolism of brain glucose. It has been found that the brain cells cannot efficiently metabolize fats; hence, they totally rely upon glucose as a vitality substrate. Thus, suppression of glucose metabolism can possess an intense effect on brain actions. Hypometabolism is frequently found in AD and has quite recently achieved impressive consideration as a plausible target for interfering in the progression of the disease. One promising approach is to keep up the normal supply of glucose to the brain with ketone bodies from the ketogenic diet signifies a potential therapeutic agent for AD. Therefore, this review represents the role of ketogenic diets to combat AD pathogenesis by considering the influence of APOE.
Collapse
|
64
|
Uddin MS, Hossain MF, Mamun AA, Shah MA, Hasana S, Bulbul IJ, Sarwar MS, Mansouri RA, Ashraf GM, Rauf A, Abdel-Daim MM, Bin-Jumah MN. Exploring the multimodal role of phytochemicals in the modulation of cellular signaling pathways to combat age-related neurodegeneration. THE SCIENCE OF THE TOTAL ENVIRONMENT 2020; 725:138313. [PMID: 32464743 DOI: 10.1016/j.scitotenv.2020.138313] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/02/2020] [Revised: 03/27/2020] [Accepted: 03/28/2020] [Indexed: 06/11/2023]
Abstract
Neurodegeneration is the progressive loss of neuronal structures and functions that lead to copious disorders like Alzheimer's (AD), Parkinson's (PD), Huntington's (HD), amyotrophic lateral sclerosis (ALS), and other less recurring diseases. Aging is the prime culprit for most neurodegenerative events. Moreover, the shared pathogenic factors of many neurodegenerative processes are inflammatory responses and oxidative stress (OS). Unfortunately, it is very complicated to treat neurodegeneration and there is no effective remedy. The rapid progression of the neurodegenerative diseases that exacerbate the burden and the concurrent absence of effective treatment strategies force the researchers to investigate more therapeutic approaches that ultimately target the causative factors of the neurodegeneration. Phytochemicals have great potential to exert their neuroprotective effects by targeting various mechanisms, such as OS, neuroinflammation, abnormal protein aggregation, neurotrophic factor deficiency, disruption in mitochondrial function, and apoptosis. Therefore, this review represents the molecular mechanisms of neuroprotection by multifunctional phytochemicals to combat age-linked neurodegenerative disorders.
Collapse
Affiliation(s)
- Md Sahab Uddin
- Department of Pharmacy, Southeast University, Dhaka, Bangladesh; Pharmakon Neuroscience Research Network, Dhaka, Bangladesh.
| | - Md Farhad Hossain
- Pharmakon Neuroscience Research Network, Dhaka, Bangladesh; Department of Physical Therapy, Graduate School of Inje University, Gimhae, South Korea
| | - Abdullah Al Mamun
- Department of Pharmacy, Southeast University, Dhaka, Bangladesh; Pharmakon Neuroscience Research Network, Dhaka, Bangladesh
| | - Muhammad Ajmal Shah
- Department of Pharmacognosy, Faculty of Pharmaceutical Sciences, Government College University, Faisalabad, Pakistan
| | - Sharifa Hasana
- Department of Pharmacy, Southeast University, Dhaka, Bangladesh
| | | | - Md Shahid Sarwar
- Department of Pharmacy, Noakhali Science and Technology University, Noakhali, Bangladesh
| | - Rasha A Mansouri
- Department of Biochemistry, Faculty of Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Ghulam Md Ashraf
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia; Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Abdur Rauf
- Department of Chemistry, University of Swabi, Swabi, Anbar, Khyber Pakhtunkhwa, Pakistan
| | - Mohamed M Abdel-Daim
- Department of Zoology, College of Science, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia; Pharmacology Department, Faculty of Veterinary Medicine, Suez Canal University, Ismailia 41522, Egypt
| | - May N Bin-Jumah
- Department of Biology, College of Science, Princess Nourah Bint Abdulrahman University, Riyadh 11474, Saudi Arabia
| |
Collapse
|
65
|
Uddin MS, Tewari D, Mamun AA, Kabir MT, Niaz K, Wahed MII, Barreto GE, Ashraf GM. Circadian and sleep dysfunction in Alzheimer's disease. Ageing Res Rev 2020; 60:101046. [PMID: 32171783 DOI: 10.1016/j.arr.2020.101046] [Citation(s) in RCA: 99] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Revised: 02/05/2020] [Accepted: 03/10/2020] [Indexed: 12/12/2022]
Abstract
Alzheimer's disease (AD) is a devastating and irreversible cognitive impairment and the most common type of dementia. Along with progressive cognitive impairment, dysfunction of the circadian rhythms also plays a pivotal role in the progression of AD. A mutual relationship among circadian rhythms, sleep, and AD has been well-recommended. The etiopathogenesis of the disturbances of the circadian system and AD share some general features that also unlock the outlook of observing them as a mutually dependent pathway. Indeed, the burden of amyloid β (Aβ), neurofibrillary tangles (NFTs), neuroinflammation, oxidative stress, and dysfunction of circadian rhythms may lead to AD. Aging can alter both sleep timings and quality that can be strongly disrupted in AD. Increased production of Aβ and reduced Aβ clearance are caused by a close interplay of Aβ, sleep disturbance and raised wakefulness. Besides Aβ, the impact of tau pathology is possibly noteworthy to the sleep deprivation found in AD. Hence, this review is focused on the primary mechanistic complexities linked to disruption of circadian rhythms, sleep deprivation, and AD. Furthermore, this review also highlights the potential therapeutic strategies to abate AD pathogenesis.
Collapse
|
66
|
Kabir MT, Uddin MS, Setu JR, Ashraf GM, Bin-Jumah MN, Abdel-Daim MM. Exploring the Role of PSEN Mutations in the Pathogenesis of Alzheimer's Disease. Neurotox Res 2020; 38:833-849. [PMID: 32556937 DOI: 10.1007/s12640-020-00232-x] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Revised: 05/24/2020] [Accepted: 05/28/2020] [Indexed: 12/25/2022]
Abstract
Alzheimer's disease (AD) is the most common cause of dementia. Mutations of presenilin (PSEN) genes that encode presenilin proteins have been found as the vital causal factors for early-onset familial AD (FAD). AD pathological features such as memory loss, synaptic dysfunction, and formation of plaques have been successfully mimicked in the transgenic mouse models that coexpress FAD-related presenilin and amyloid precursor protein (APP) variants. γ-Secretase (GS) is an enzyme that plays roles in catalyzing intramembranous APP proteolysis to release pathogenic amyloid beta (Aβ). It has been found that presenilins can play a role as the GS's catalytic subunit. FAD-related mutations in presenilins can modify the site of GS cleavage in a way that can elevate the production of longer and highly fibrillogenic Aβ. Presenilins can interact with β-catenin to generate presenilin complexes. Aforesaid interactions have also been studied to observe the mutational and physiological activities in the catenin signal transduction pathway. Along with APP, GS can catalyze intramembrane proteolysis of various substrates that play a vital role in synaptic function. PSEN mutations can cause FAD with autosomal dominant inheritance and early onset of the disease. In this article, we have reviewed the current progress in the analysis of PSENs and the correlation of PSEN mutations and AD pathogenesis.
Collapse
Affiliation(s)
| | - Md Sahab Uddin
- Department of Pharmacy, Southeast University, Dhaka, Bangladesh. .,Pharmakon Neuroscience Research Network, Dhaka, Bangladesh.
| | | | - Ghulam Md Ashraf
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia.,Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - May N Bin-Jumah
- Department of Biology, College of Science, Princess Nourah bint Abdulrahman University, Riyadh, 11474, Saudi Arabia
| | - Mohamed M Abdel-Daim
- Department of Zoology, College of Science, King Saud University, P.O. Box 2455, Riyadh, 11451, Saudi Arabia.,Pharmacology Department, Faculty of Veterinary Medicine, Suez Canal University, Ismailia, 41522, Egypt
| |
Collapse
|
67
|
Kabir MT, Sufian MA, Uddin MS, Begum MM, Akhter S, Islam A, Mathew B, Islam MS, Amran MS, Md Ashraf G. NMDA Receptor Antagonists: Repositioning of Memantine as a Multitargeting Agent for Alzheimer's Therapy. Curr Pharm Des 2020; 25:3506-3518. [PMID: 31604413 DOI: 10.2174/1381612825666191011102444] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Accepted: 10/08/2019] [Indexed: 12/28/2022]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disease that causes problems with memory, thinking, and behavior. Currently, there is no drug that can reduce the pathological events of this degenerative disease but symptomatic relief is possible that can abate the disease condition. N-methyl-D-aspartate (NMDA) receptors exert a critical role for synaptic plasticity as well as transmission. Overstimulation of glutamate receptors, predominantly NMDA type, may cause excitotoxic effects on neurons and is recommended as a mechanism for neurodegeneration. Atypical activation of the NMDA receptor has been suggested for AD by synaptic dysfunction. NMDA receptor antagonists especially memantine block the NMDA receptor and can reduce the influx of calcium (Ca2+) ions into neuron, thus, toxic intracellular events are not activated. This review represents the role of NMDA receptors antagonists as potential therapeutic agents to reduce AD. Moreover, this review highlights the repositioning of memantine as a potential novel therapeutic multitargeting agent for AD.
Collapse
Affiliation(s)
| | | | - Md Sahab Uddin
- Department of Pharmacy, Southeast University, Dhaka, Bangladesh.,Pharmakon Neuroscience Research Network, Dhaka, Bangladesh
| | | | - Shammi Akhter
- Department of Pharmacy, Southeast University, Dhaka, Bangladesh
| | - Ariful Islam
- Department of Cell Biology and Neuroscience, Rowan University School of Osteopathic Medicine, Stratford, United States
| | - Bijo Mathew
- Division of Drug Design and Medicinal Chemistry Research Lab, Department of Pharmaceutical Chemistry, Ahalia School of Pharmacy, Palakkad, India
| | | | - Md Shah Amran
- Department of Pharmaceutical Chemistry, University of Dhaka, Dhaka, Bangladesh
| | - Ghulam Md Ashraf
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia.,Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
68
|
Uddin MS, Mamun AA, Rahman MA, Kabir MT, Alkahtani S, Alanazi IS, Perveen A, Ashraf GM, Bin-Jumah MN, Abdel-Daim MM. Exploring the Promise of Flavonoids to Combat Neuropathic Pain: From Molecular Mechanisms to Therapeutic Implications. Front Neurosci 2020; 14:478. [PMID: 32587501 PMCID: PMC7299068 DOI: 10.3389/fnins.2020.00478] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Accepted: 04/17/2020] [Indexed: 01/10/2023] Open
Abstract
Neuropathic pain (NP) is the result of irregular processing in the central or peripheral nervous system, which is generally caused by neuronal injury. The management of NP represents a great challenge owing to its heterogeneous profile and the significant undesirable side effects of the frequently prescribed psychoactive agents, including benzodiazepines (BDZ). Currently, several established drugs including antidepressants, anticonvulsants, topical lidocaine, and opioids are used to treat NP, but they exert a wide range of adverse effects. To reduce the burden of adverse effects, we need to investigate alternative therapeutics for the management of NP. Flavonoids are the most common secondary metabolites of plants used in folkloric medicine as tranquilizers, and have been claimed to have a selective affinity to the BDZ binding site. Several studies in animal models have reported that flavonoids can reduce NP. In this paper, we emphasize the potentiality of flavonoids for the management of NP.
Collapse
Affiliation(s)
- Md Sahab Uddin
- Department of Pharmacy, Southeast University, Dhaka, Bangladesh.,Pharmakon Neuroscience Research Network, Dhaka, Bangladesh
| | - Abdullah Al Mamun
- Department of Pharmacy, Southeast University, Dhaka, Bangladesh.,Pharmakon Neuroscience Research Network, Dhaka, Bangladesh
| | - Md Ataur Rahman
- Center for Neuroscience, Brain Science Institute, Korea Institute of Science and Technology, Seoul, South Korea
| | | | - Saad Alkahtani
- Department of Zoology, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Ibtesam S Alanazi
- Department of Biology, Faculty of Sciences, Univesity of Hafr Al Batin, Hafr Al Batin, Saudi Arabia
| | - Asma Perveen
- Glocal School of Life Sciences, Glocal University, Saharanpur, India
| | - Ghulam Md Ashraf
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia.,Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - May N Bin-Jumah
- Department of Biology, College of Science, Princess Nourah bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Mohamed M Abdel-Daim
- Department of Zoology, College of Science, King Saud University, Riyadh, Saudi Arabia.,Pharmacology Department, Faculty of Veterinary Medicine, Suez Canal University, Ismailia, Egypt
| |
Collapse
|
69
|
Uddin MS, Kabir MT, Tewari D, Mamun AA, Mathew B, Aleya L, Barreto GE, Bin-Jumah MN, Abdel-Daim MM, Ashraf GM. Revisiting the role of brain and peripheral Aβ in the pathogenesis of Alzheimer's disease. J Neurol Sci 2020; 416:116974. [PMID: 32559516 DOI: 10.1016/j.jns.2020.116974] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 05/15/2020] [Accepted: 06/01/2020] [Indexed: 12/12/2022]
Abstract
Amyloid beta (Aβ) is an intricate molecule that interacts with several biomolecules and/or produces insoluble assemblies and eventually the nonphysiological depositions of its alternate with normal neuronal conditions leading to Alzheimer's disease (AD). Aβ is formed through the proteolytic cleavage of the amyloid precursor protein (APP). Significant efforts are being made to explore the exact role of Aβ in AD pathogenesis. It is believed that the deposition of Aβ in the brain takes place from Aβ components which are derived from the brain itself. However, recent evidence suggests that Aβ derived also from the periphery and hence the Aβ circulating in the blood is capable of penetrating the blood-brain barrier (BBB) and the role of Aβ derived from the periphery is largely unknown so far. Therefore, Aβ origin determination and the underlying mechanisms of its pathological effects are of considerable interest in exploring effective therapeutic strategies. The purpose of this review is to provide a novel insight into AD pathogenesis based on Aβ in both the brain and periphery and highlight new therapeutic avenues to combat AD pathogenesis.
Collapse
Affiliation(s)
- Md Sahab Uddin
- Department of Pharmacy, Southeast University, Dhaka, Bangladesh; Pharmakon Neuroscience Research Network, Dhaka, Bangladesh.
| | | | - Devesh Tewari
- Department of Pharmacognosy, School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, India
| | - Abdullah Al Mamun
- Department of Pharmacy, Southeast University, Dhaka, Bangladesh; Pharmakon Neuroscience Research Network, Dhaka, Bangladesh
| | - Bijo Mathew
- Department of Pharmaceutical Chemistry, Ahalia School of Pharmacy, Palakkad, India
| | - Lotfi Aleya
- Chrono-Environnement Laboratory, CNRS 6249, Bourgogne Franche-Comté University, Besançon, France
| | - George E Barreto
- Department of Biological Sciences, University of Limerick, Limerick, Ireland; Health Research Institute, University of Limerick, Limerick, Ireland
| | - May N Bin-Jumah
- Department of Biology, College of Science, Princess Nourah bint Abdulrahman University, Riyadh 11474, Saudi Arabia
| | - Mohamed M Abdel-Daim
- Department of Zoology, College of Science, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia; Pharmacology Department, Faculty of Veterinary Medicine, Suez Canal University, Ismailia 41522, Egypt
| | - Ghulam Md Ashraf
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia; Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia.
| |
Collapse
|
70
|
Uddin MS, Rahman MA, Kabir MT, Behl T, Mathew B, Perveen A, Barreto GE, Bin-Jumah MN, Abdel-Daim MM, Ashraf GM. Multifarious roles of mTOR signaling in cognitive aging and cerebrovascular dysfunction of Alzheimer's disease. IUBMB Life 2020; 72:1843-1855. [PMID: 32472959 DOI: 10.1002/iub.2324] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Revised: 05/10/2020] [Accepted: 05/13/2020] [Indexed: 12/19/2022]
Abstract
Age-related cognitive failure is a main devastating incident affecting even healthy people. Alzheimer's disease (AD) is the utmost common form of dementia among the geriatric community. In the pathogenesis of AD, cerebrovascular dysfunction is revealed before the beginning of the cognitive decline. Mounting proof shows a precarious impact of cerebrovascular dysregulation in the development of AD pathology. Recent studies document that the mammalian target of rapamycin (mTOR) acts as a crucial effector of cerebrovascular dysregulation in AD. The mTOR contributes to brain vascular dysfunction and subsequence cerebral blood flow deficits as well as cognitive impairment. Furthermore, mTOR causes the blood-brain barrier (BBB) breakdown in AD models. Inhibition of mTOR hyperactivity protects the BBB integrity in AD. Furthermore, mTOR drives cognitive defect and cerebrovascular dysfunction, which are greatly prevalent in AD, but the central molecular mechanisms underlying these alterations are obscure. This review represents the crucial and current research findings regarding the role of mTOR signaling in cognitive aging and cerebrovascular dysfunction in the pathogenesis of AD.
Collapse
Affiliation(s)
- Md Sahab Uddin
- Department of Pharmacy, Southeast University, Dhaka, Bangladesh.,Pharmakon Neuroscience Research Network, Dhaka, Bangladesh
| | - Md Ataur Rahman
- Center for Neuroscience, Brain Science Institute, Korea Institute of Science and Technology, Seoul, Republic of Korea
| | | | - Tapan Behl
- Chitkara College of Pharmacy, Chitkara University, Patiala, India
| | - Bijo Mathew
- Division of Drug Design and Medicinal Chemistry Research Lab, Department of Pharmaceutical Chemistry, Ahalia School of Pharmacy, Palakkad, India
| | - Asma Perveen
- Glocal School of Life Sciences, Glocal University, Saharanpur, India
| | - George E Barreto
- Department of Biological Sciences, University of Limerick, Limerick, Ireland.,Instituto de Ciencias Biomédicas, Universidad Autónoma de Chile, Santiago, Chile
| | - May N Bin-Jumah
- Department of Biology, College of Science, Princess Nourah bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Mohamed M Abdel-Daim
- Department of Zoology, College of Science, King Saud University, Riyadh, Saudi Arabia.,Pharmacology Department, Faculty of Veterinary Medicine, Suez Canal University, Ismailia, Egypt
| | - Ghulam Md Ashraf
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia.,Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
71
|
Vaiserman A, Koliada A, Lushchak O. Neuroinflammation in pathogenesis of Alzheimer's disease: Phytochemicals as potential therapeutics. Mech Ageing Dev 2020; 189:111259. [PMID: 32450086 DOI: 10.1016/j.mad.2020.111259] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2020] [Revised: 05/02/2020] [Accepted: 05/02/2020] [Indexed: 02/07/2023]
Abstract
Accumulation of neurotoxic forms of amyloid-β proteins in senile plaques and hyperphosphorylated tau proteins in neurofibrillary tangles is a well-known pathophysiological hallmark of Alzheimer's disease (AD). However, clinical trials with drugs targeting amyloid-β and tau have failed to demonstrate efficacy in treating AD. All currently FDA-approved anti-AD drugs have symptomatic effects only and are not able to cure this disease. This makes necessary to search for alternative therapeutic targets. Accumulating evidence suggests that systemic inflammation and related vascular dysfunction play important etiological roles in AD and precede its clinical manifestation. Therefore, novel therapeutic modalities targeted at these pathophysiological components of AD are intensively developed now. Phytochemicals such as resveratrol, curcumin, quercetin, genistein and catechins are promising anti-AD therapeutics due to their ability to affect major pathogenetic mechanisms of AD, including oxidative stress, neuroinflammation and mitochondrial dysfunction. The implementation of innovative approaches for phytochemical delivery, including the nanotechnology-based ones which enable to significantly enhance their oral bioavailability, would likely provide an opportunity to address many challenges of conventional anti-AD therapies. In this review, roles of inflammation and vascular dysregulation in AD are described and phytobioactive compound-based treatment strategies for AD are discussed.
Collapse
Affiliation(s)
- Alexander Vaiserman
- Laboratory of Epigenetics, D.F. Chebotarev Institute of Gerontology, NAMS, 67 Vyshgorodska str., Kyiv, 04114, Ukraine.
| | - Alexander Koliada
- Laboratory of Epigenetics, D.F. Chebotarev Institute of Gerontology, NAMS, 67 Vyshgorodska str., Kyiv, 04114, Ukraine
| | - Oleh Lushchak
- Department of Biochemistry and Biotechnology, Vasyl Stefanyk Precarpathian National University, 57 Shevchenka str., Ivano-Frankivsk, 76018, Ukraine
| |
Collapse
|
72
|
Uddin MS, Tewari D, Sharma G, Kabir MT, Barreto GE, Bin-Jumah MN, Perveen A, Abdel-Daim MM, Ashraf GM. Molecular Mechanisms of ER Stress and UPR in the Pathogenesis of Alzheimer's Disease. Mol Neurobiol 2020; 57:2902-2919. [PMID: 32430843 DOI: 10.1007/s12035-020-01929-y] [Citation(s) in RCA: 98] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2020] [Accepted: 05/01/2020] [Indexed: 01/01/2023]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disease involving aggregation of misfolded proteins inside the neuron causing prolonged cellular stress. The neuropathological hallmarks of AD include the formation of senile plaques and neurofibrillary tangles in specific brain regions that lead to synaptic loss and neuronal death. The exact mechanism of neuron dysfunction in AD remains obscure. In recent years, endoplasmic reticulum (ER) dysfunction has been implicated in neuronal degeneration seen in AD. Apart from AD, many other diseases also involve misfolded proteins aggregations in the ER, a condition referred to as ER stress. The response of the cell to ER stress is to activate a group of signaling pathways called unfolded protein response (UPR) that stimulates a particular transcriptional program to restore ER function and ensure cell survival. ER stress also involves the generation of reactive oxygen species (ROS) that, together with mitochondrial ROS and decreased effectiveness of antioxidant mechanisms, producing a condition of chronic oxidative stress. The unfolded proteins may not always produce a response that leads to the restoration of cellular functions, but they may also lead to inflammation by a set of different pathways with deleterious consequences. In this review, we extensively discuss the role of ER stress and how to target it using different pharmacological approaches in AD development and onset.
Collapse
Affiliation(s)
- Md Sahab Uddin
- Department of Pharmacy, Southeast University, Dhaka, Bangladesh.
- Pharmakon Neuroscience Research Network, Dhaka, Bangladesh.
| | - Devesh Tewari
- Department of Pharmacognosy, School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, India
| | - Gaurav Sharma
- Department of Physiology, AIIMS Jodhpur, Jodhpur, India
| | | | - George E Barreto
- Department of Biological Sciences, University of Limerick, Limerick, Ireland.
- Instituto de Ciencias Biomédicas, Universidad Autónoma de Chile, Santiago, Chile.
| | - May N Bin-Jumah
- Department of Biology, College of Science, Princess Nourah bint Abdulrahman University, Riyadh 11474, Saudi Arabia
| | - Asma Perveen
- Glocal School of Life Sciences, Glocal University, Saharanpur, India
| | - Mohamed M Abdel-Daim
- Department of Zoology, College of Science, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia
- Pharmacology Department, Faculty of Veterinary Medicine, Suez Canal University, Ismailia 41522, Egypt
| | - Ghulam Md Ashraf
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia.
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia.
| |
Collapse
|
73
|
Mamun AA, Uddin MS, Bin Bashar MF, Zaman S, Begum Y, Bulbul IJ, Islam MS, Sarwar MS, Mathew B, Amran MS, Md Ashraf G, Bin-Jumah MN, Mousa SA, Abdel-Daim MM. Molecular Insight into the Therapeutic Promise of Targeting APOE4 for Alzheimer's Disease. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:5086250. [PMID: 32509144 PMCID: PMC7245681 DOI: 10.1155/2020/5086250] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Accepted: 04/17/2020] [Indexed: 11/17/2022]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disease that causes chronic cognitive dysfunction. Most of the AD cases are late onset, and the apolipoprotein E (APOE) isoform is a key genetic risk factor. The APOE gene has 3 key alleles in humans including APOE2, APOE3, and APOE4. Among them, APOE4 is the most potent genetic risk factor for late-onset AD (LOAD), while APOE2 has a defensive effect. Research data suggest that APOE4 leads to the pathogenesis of AD through various processes such as accelerated beta-amyloid aggregations that raised neurofibrillary tangle formation, cerebrovascular diseases, aggravated neuroinflammation, and synaptic loss. However, the precise mode of actions regarding in what way APOE4 leads to AD pathology remains unclear. Since APOE contributes to several pathological pathways of AD, targeting APOE4 might serve as a promising strategy for the development of novel drugs to combat AD. In this review, we focus on the recent studies about APOE4-targeted therapeutic strategies that have been advanced in animal models and are being prepared for use in humans for the management of AD.
Collapse
Affiliation(s)
- Abdullah Al Mamun
- Department of Pharmacy, Southeast University, Dhaka, Bangladesh
- Pharmakon Neuroscience Research Network, Dhaka, Bangladesh
| | - Md. Sahab Uddin
- Department of Pharmacy, Southeast University, Dhaka, Bangladesh
- Pharmakon Neuroscience Research Network, Dhaka, Bangladesh
| | - Md. Fahim Bin Bashar
- Department of Pharmacy, University of Development Alternative, Dhaka, Bangladesh
| | - Sonia Zaman
- Department of Pharmacy, Southeast University, Dhaka, Bangladesh
| | - Yesmin Begum
- Department of Pharmacy, Southeast University, Dhaka, Bangladesh
| | | | | | - Md. Shahid Sarwar
- Department of Pharmacy, Noakhali Science and Technology University, Noakhali, Bangladesh
| | - Bijo Mathew
- Division of Drug Design and Medicinal Chemistry Research Lab, Department of Pharmaceutical Chemistry, Ahalia School of Pharmacy, Palakkad, India
| | - Md. Shah Amran
- Department of Pharmaceutical Chemistry, University of Dhaka, Dhaka, Bangladesh
| | - Ghulam Md Ashraf
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - May N. Bin-Jumah
- Department of Biology, College of Science, Princess Nourah bint Abdulrahman University, Riyadh 11474, Saudi Arabia
| | - Shaker A. Mousa
- Pharmaceutical Research Institute, Albany College of Pharmacy and Health Sciences, New York, NY 12144, USA
| | - Mohamed M. Abdel-Daim
- Department of Zoology, College of Science, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia
- Pharmacology Department, Faculty of Veterinary Medicine, Suez Canal University, Ismailia 41522, Egypt
| |
Collapse
|
74
|
Kabir MT, Uddin MS, Mamun AA, Jeandet P, Aleya L, Mansouri RA, Ashraf GM, Mathew B, Bin-Jumah MN, Abdel-Daim MM. Combination Drug Therapy for the Management of Alzheimer's Disease. Int J Mol Sci 2020; 21:E3272. [PMID: 32380758 PMCID: PMC7246721 DOI: 10.3390/ijms21093272] [Citation(s) in RCA: 78] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 03/19/2020] [Accepted: 03/19/2020] [Indexed: 02/02/2023] Open
Abstract
Alzheimer's disease (AD) is the leading cause of dementia worldwide. Even though the number of AD patients is rapidly growing, there is no effective treatment for this neurodegenerative disorder. At present, implementation of effective treatment approaches for AD is vital to meet clinical needs. In AD research, priorities concern the development of disease-modifying therapeutic agents to be used in the early phases of AD and the optimization of the symptomatic treatments predominantly dedicated to the more advanced AD stages. Until now, available therapeutic agents for AD treatment only provide symptomatic treatment. Since AD pathogenesis is multifactorial, use of a multimodal therapeutic intervention addressing several molecular targets of AD-related pathological processes seems to be the most practical approach to modify the course of AD progression. It has been demonstrated through numerous studies, that the clinical efficacy of combination therapy (CT) is higher than that of monotherapy. In case of AD, CT is more effective, mostly when started early, at slowing the rate of cognitive impairment. In this review, we have covered the major studies regarding CT to combat AD pathogenesis. Moreover, we have also highlighted the safety, tolerability, and efficacy of CT in the treatment of AD.
Collapse
Affiliation(s)
- Md. Tanvir Kabir
- Department of Pharmacy, BRAC University, Dhaka 1212, Bangladesh;
| | - Md. Sahab Uddin
- Department of Pharmacy, Southeast University, Dhaka 1213, Bangladesh;
- Pharmakon Neuroscience Research Network, Dhaka 1207, Bangladesh
| | - Abdullah Al Mamun
- Department of Pharmacy, Southeast University, Dhaka 1213, Bangladesh;
- Pharmakon Neuroscience Research Network, Dhaka 1207, Bangladesh
| | - Philippe Jeandet
- Research Unit, Induced Resistance and Plant Bioprotection, EA 4707, SFR Condorcet FR CNRS 3417, Faculty of Sciences, University of Reims Champagne-Ardenne, P.O. Box 1039, 51687 Reims CEDEX 2, France;
| | - Lotfi Aleya
- Chrono-Environnement Laboratory, UMR CNRS 6249, Bourgogne Franche-Comté University, F-25030 Besançon, France;
| | - Rasha A. Mansouri
- Department of Biochemistry, Faculty of Sciences, King Abdulaziz University, Jeddah 21589, Saudi Arabia;
| | - Ghulam Md Ashraf
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21589, Saudi Arabia;
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Bijo Mathew
- Division of Drug Design and Medicinal Chemistry Research Lab, Department of Pharmaceutical Chemistry, Ahalia School of Pharmacy, Palakkad 678557, India;
| | - May N. Bin-Jumah
- Department of Biology, College of Science, Princess Nourah bint Abdulrahman University, Riyadh 11474, Saudi Arabia;
| | - Mohamed M. Abdel-Daim
- Department of Zoology, College of Science, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia;
- Pharmacology Department, Faculty of Veterinary Medicine, Suez Canal University, Ismailia 41522, Egypt
| |
Collapse
|
75
|
Uddin MS, Kabir MT, Jeandet P, Mathew B, Ashraf GM, Perveen A, Bin-Jumah MN, Mousa SA, Abdel-Daim MM. Novel Anti-Alzheimer's Therapeutic Molecules Targeting Amyloid Precursor Protein Processing. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:7039138. [PMID: 32411333 PMCID: PMC7206886 DOI: 10.1155/2020/7039138] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/15/2020] [Revised: 03/27/2020] [Accepted: 04/01/2020] [Indexed: 02/04/2023]
Abstract
Alzheimer's disease (AD) is the most common cause of dementia among older people, and the prevalence of this disease is estimated to rise quickly in the upcoming years. Unfortunately, almost all of the drug candidates tested for AD until now have failed to exhibit any efficacy. Henceforth, there is an increased necessity to avert and/or slow down the advancement of AD. It is known that one of the major pathological characteristics of AD is the presence of senile plaques (SPs) in the brain. These SPs are composed of aggregated amyloid beta (Aβ), derived from the amyloid precursor protein (APP). Pharmaceutical companies have conducted a number of studies in order to identify safe and effective anti-Aβ drugs to combat AD. It is known that α-, β-, and γ-secretases are the three proteases that are involved in APP processing. Furthermore, there is a growing interest in these proteases, as they have a contribution to the modulation and production of Aβ. It has been observed that small compounds can be used to target these important proteases. Indeed, these compounds must satisfy the common strict requirements of a drug candidate targeted for brain penetration and selectivity toward different proteases. In this article, we have focused on the auspicious molecules which are under development for targeting APP-processing enzymes. We have also presented several anti-AD molecules targeting Aβ accumulation and phosphorylation signaling in APP processing. This review highlights the structure-activity relationship and other physicochemical features of several pharmacological candidates in order to successfully develop new anti-AD drugs.
Collapse
Affiliation(s)
- Md. Sahab Uddin
- Department of Pharmacy, Southeast University, Dhaka, Bangladesh
- Pharmakon Neuroscience Research Network, Dhaka, Bangladesh
| | | | - Philippe Jeandet
- Research Unit, Induced Resistance and Plant Bioprotection, EA 4707, SFR Condorcet FR CNRS 3417, Faculty of Sciences, University of Reims Champagne-Ardenne, PO Box 1039, 51687 Reims Cedex 2, France
| | - Bijo Mathew
- Division of Drug Design and Medicinal Chemistry Research Lab, Department of Pharmaceutical Chemistry, Ahalia School of Pharmacy, Palakkad, India
| | - Ghulam Md Ashraf
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Asma Perveen
- Glocal School of Life Sciences, Glocal University, Saharanpur, India
| | - May N. Bin-Jumah
- Department of Biology, College of Science, Princess Nourah bint Abdulrahman University, Riyadh 11474, Saudi Arabia
| | - Shaker A. Mousa
- Pharmaceutical Research Institute, Albany College of Pharmacy and Health Sciences, New York, NY 12144, USA
| | - Mohamed M. Abdel-Daim
- Department of Zoology, College of Science, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia
- Pharmacology Department, Faculty of Veterinary Medicine, Suez Canal University, Ismailia 41522, Egypt
| |
Collapse
|
76
|
Uddin MS, Kabir MT, Mamun AA, Barreto GE, Rashid M, Perveen A, Ashraf GM. Pharmacological approaches to mitigate neuroinflammation in Alzheimer's disease. Int Immunopharmacol 2020; 84:106479. [PMID: 32353686 DOI: 10.1016/j.intimp.2020.106479] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2019] [Revised: 03/13/2020] [Accepted: 04/02/2020] [Indexed: 12/12/2022]
Abstract
Alzheimer's disease (AD) is one of the most prevalent neurodegenerative diseases characterized by the formation of extracellular amyloid beta (Aβ) plaques and intracellular neurofibrillary tangles (NFTs). Growing evidence suggested that there is an association between neuronal dysfunction and neuroinflammation (NI) in AD, coordinated by the chronic activation of astrocytes and microglial cells along with the subsequent excessive generation of the proinflammatory molecule. Therefore, a better understanding of the relationship between the nervous and immune systems is important in order to delay or avert the neurodegenerative events of AD. The inflammatory/immune pathways and the mechanisms to control these pathways may provide a novel arena to develop new drugs in order to target NI in AD. In this review, we represent the influence of cellular mediators which are involved in the NI process, with regards to the progression of AD. We also discuss the processes and the current status of multiple anti-inflammatory agents which are used in AD and have gone through or going through clinical trials. Moreover, new prospects for targeting NI in the development of AD drugs have also been highlighted.
Collapse
Affiliation(s)
- Md Sahab Uddin
- Department of Pharmacy, Southeast University, Dhaka, Bangladesh; Pharmakon Neuroscience Research Network, Dhaka, Bangladesh.
| | | | - Abdullah Al Mamun
- Department of Pharmacy, Southeast University, Dhaka, Bangladesh; Pharmakon Neuroscience Research Network, Dhaka, Bangladesh
| | - George E Barreto
- Department of Biological Sciences, University of Limerick, Limerick, Ireland; Instituto de Ciencias Biomédicas, Universidad Autónoma de Chile, Santiago, Chile
| | - Mamunur Rashid
- Department of Pharmacy, University of Rajshahi, Rajshahi, Bangladesh
| | - Asma Perveen
- School of Life Sciences, The Glocal University, Saharanpur, Uttar Pradesh 247121, India
| | - Ghulam Md Ashraf
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia; Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia.
| |
Collapse
|
77
|
Integrated metabolomics and network pharmacology strategy for ascertaining the quality marker of flavonoids for Sophora flavescens. J Pharm Biomed Anal 2020; 186:113297. [PMID: 32325403 DOI: 10.1016/j.jpba.2020.113297] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Revised: 03/22/2020] [Accepted: 04/01/2020] [Indexed: 12/20/2022]
Abstract
Traditional Chinese medicines (TCMs) have been widely used in Asian countries for thousands of years due to their supreme quality and good clinical efficacy. However, the increasing demand for TCMs in recent decades warrants effective quality control methodology to avoid clinical problems. Therefore, comprehensive quality evaluation systems should be established for ensuring TCM's quality, in terms of chemical components, as well as bioactivity for identifying quality markers in TCM and developing suitable analytical methods for quality control. In this study, we selected Sophora flavescens (S. flavescens) as the research object and developed a novel integrated strategy combining metabolomics and network pharmacology to explore the quality markers. Firstly, we determined the targeted metabolomic profiles of seventy-four batches of S. flavescens (aged from 1 to 6 years) by UHPLC/QE-MS. Six potential markers were successfully screened, quantified and reverse-verified as the most influential effective compounds by UHPLC/QE-MS and multivariate statistical analysis. Secondly, the network of "components-targets-pathways" was constructed, and the pharmacological activities of six potential markers were predicted. Finally, we determined the anti-tumor activity of six flavonoids (kurarinone, norkurarinone, kuraridin, kushenol N, trifolirhizin, and genistein) as the quality markers for Sophora flavescens, evaluated their pharmacokinetic profiles and reviewed their existing pharmacological researches. Thus, integrated metabolomics and network pharmacology technology were applied for the effective discovery of quality markers of Chinese material medica.
Collapse
|
78
|
Wei C, Fan J, Sun X, Yao J, Guo Y, Zhou B, Shang Y. Acetyl-11-keto-β-boswellic acid ameliorates cognitive deficits and reduces amyloid-β levels in APPswe/PS1dE9 mice through antioxidant and anti-inflammatory pathways. Free Radic Biol Med 2020; 150:96-108. [PMID: 32109514 DOI: 10.1016/j.freeradbiomed.2020.02.022] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Revised: 02/16/2020] [Accepted: 02/20/2020] [Indexed: 02/07/2023]
Abstract
Alzheimer's disease (AD) is a complex disease involved oxidative stress and inflammation in its pathogenesis. Acetyl-11-keto-β-boswellic acid (AKBA) is an active triterpenoid compound from extracts of Boswellia serrata, which has been widely used as an antioxidant and anti-inflammatory agent. The present study was to determine whether AKBA, a novel candidate, could protect against cognitive and neuropathological impairments in AD. We found that AKBA treatment resulted in a significant improvement of learning and memory deficits, a dramatic decrease in cerebral amyloid-β (Aβ) levels and plaque burden, a profound alleviation in oxidative stress and inflammation, and a marked reduction in activated glial cells and synaptic defects in the APPswe/PS1dE9 mice. Furthermore, amyloid precursor protein (APP) processing was remarkably suppressed with AKBA treatment by inhibiting beta-site APP cleaving enzyme 1 (BACE1) protein expression to produce Aβ in the APPswe/PS1dE9 mice brains. Mechanistically, AKBA modulated antioxidant and anti-inflammatory pathways via increasing nuclear erythroid 2-related factor 2 (Nrf2) and heme oxygenase-1 (HO-1) expression, and via declining phosphorylation of inhibitor of nuclear factor-kappa B alpha (IκBα) and p65. Collectively, our findings provide evidence that AKBA protects neurons against oxidative stress and inflammation in AD, and this neuroprotective effect involves the Nrf2/HO-1 and nuclear factor-kappa B (NF-κB) signaling pathways.
Collapse
Affiliation(s)
- Chao Wei
- Department of Neurology, The Second Medical Center, Chinese PLA General Hospital, Beijing, 100853, China.
| | - Jiao Fan
- Institute of Geriatrics, National Clinical Research Center of Geriatrics Disease, Chinese PLA General Hospital, Beijing, 100853, China
| | - Xuan Sun
- Department of Neurology, The Second Medical Center, Chinese PLA General Hospital, Beijing, 100853, China
| | - Jiarui Yao
- Department of Neurology, The Second Medical Center, Chinese PLA General Hospital, Beijing, 100853, China
| | - Yane Guo
- Department of Neurology, The Second Medical Center, Chinese PLA General Hospital, Beijing, 100853, China
| | - Bo Zhou
- Department of Neurology, The Second Medical Center, Chinese PLA General Hospital, Beijing, 100853, China
| | - Yanchang Shang
- Department of Neurology, The Second Medical Center, Chinese PLA General Hospital, Beijing, 100853, China
| |
Collapse
|
79
|
Uddin MS, Kabir MT, Niaz K, Jeandet P, Clément C, Mathew B, Rauf A, Rengasamy KR, Sobarzo-Sánchez E, Ashraf GM, Aleya L. Molecular Insight into the Therapeutic Promise of Flavonoids against Alzheimer's Disease. Molecules 2020; 25:molecules25061267. [PMID: 32168835 PMCID: PMC7143946 DOI: 10.3390/molecules25061267] [Citation(s) in RCA: 83] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 03/05/2020] [Accepted: 03/10/2020] [Indexed: 12/12/2022] Open
Abstract
Alzheimer’s disease (AD) is one of the utmost chronic neurodegenerative disorders, which is characterized from a neuropathological point of view by the aggregates of amyloid beta (Aβ) peptides that are deposited as senile plaques and tau proteins which form neurofibrillary tangles (NFTs). Even though advancement has been observed in order to understand AD pathogenesis, currently available therapeutic methods can only deliver modest symptomatic relief. Interestingly, naturally occurring dietary flavonoids have gained substantial attention due to their antioxidative, anti-inflammatory, and anti-amyloidogenic properties as alternative candidates for AD therapy. Experimental proof provides support to the idea that some flavonoids might protect AD by interfering with the production and aggregation of Aβ peptides and/or decreasing the aggregation of tau. Flavonoids have the ability to promote clearance of Aβ peptides and inhibit tau phosphorylation by the mTOR/autophagy signaling pathway. Moreover, due to their cholinesterase inhibitory potential, flavonoids can represent promising symptomatic anti-Alzheimer agents. Several processes have been suggested for the aptitude of flavonoids to slow down the advancement or to avert the onset of Alzheimer’s pathogenesis. To enhance cognitive performance and to prevent the onset and progress of AD, the interaction of flavonoids with various signaling pathways is proposed to exert their therapeutic potential. Therefore, this review elaborates on the probable therapeutic approaches of flavonoids aimed at averting or slowing the progression of the AD pathogenesis.
Collapse
Affiliation(s)
- Md. Sahab Uddin
- Department of Pharmacy, Southeast University, Dhaka 1213, Bangladesh
- Pharmakon Neuroscience Research Network, Dhaka 1207, Bangladesh
- Correspondence: or (M.S.U.); (P.J.); Tel.: +880-1710220110 (M.S.U.); +33-3-26913-341 (P.J.)
| | | | - Kamal Niaz
- Department of Pharmacology and Toxicology, Faculty of Bio-Sciences, Cholistan University of Veterinary and Animal Sciences (CUVAS), Bahawalpur 63100, Pakistan
| | - Philippe Jeandet
- Research Unit, Induced Resistance and Plant Bioprotection, EA 4707, SFR Condorcet FR CNRS 3417, Faculty of Sciences, University of Reims Champagne-Ardenne, PO Box 1039, 51687 Reims CEDEX 2, France
- Correspondence: or (M.S.U.); (P.J.); Tel.: +880-1710220110 (M.S.U.); +33-3-26913-341 (P.J.)
| | - Christophe Clément
- Research Unit, Induced Resistance and Plant Bioprotection, EA 4707, SFR Condorcet FR CNRS 3417, Faculty of Sciences, University of Reims Champagne-Ardenne, PO Box 1039, 51687 Reims CEDEX 2, France
| | - Bijo Mathew
- Division of Drug Design and Medicinal Chemistry Research Lab, Department of Pharmaceutical Chemistry, Ahalia School of Pharmacy, Palakkad, Kerala 678557, India
| | - Abdur Rauf
- Department of Chemistry, University of Swabi, Anbar 23561, Khyber Pakhtunkhwa, Pakistan
| | | | - Eduardo Sobarzo-Sánchez
- Instituto de Investigación e Innovación en Salud, Facultad de Ciencias de la Salud, Universidad Central de Chile, Santiago 8330507, Chile
- Department of Organic Chemistry, Faculty of Pharmacy, University of Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - Ghulam Md Ashraf
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Lotfi Aleya
- Chrono-Environnement Laboratory, UMR CNRS 6249, Bourgogne Franche-Comté University, F-25030 Besançon, France
| |
Collapse
|
80
|
Uddin MS, Mamun AA, Jakaria M, Thangapandiyan S, Ahmad J, Rahman MA, Mathew B, Abdel-Daim MM, Aleya L. Emerging promise of sulforaphane-mediated Nrf2 signaling cascade against neurological disorders. THE SCIENCE OF THE TOTAL ENVIRONMENT 2020; 707:135624. [PMID: 31784171 DOI: 10.1016/j.scitotenv.2019.135624] [Citation(s) in RCA: 107] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Revised: 11/15/2019] [Accepted: 11/17/2019] [Indexed: 06/10/2023]
Abstract
Neurological disorders represent a great challenge and are the leading cause of death and disability globally. Although numerous complicated mechanisms are involved in the progressions of chronic and acute neurodegenerative disorders, most of the diseases share mutual pathogenic features such as oxidative stress, mitochondrial dysfunction, neuroinflammation, protein misfolding, excitotoxicity, and neuronal damage, all of these are the common targets of nuclear factor erythroid 2 related factor 2 (Nrf2) signaling cascade. No cure has yet been discovered to tackle these disorders, so, intervention approaches targeting phytochemicals have been recommended as an alternative form of treatment. Sulforaphane is a sulfur-rich dietary phytochemical which has several activities such as antioxidant, anti-inflammatory, and anti-tumor via multiple targets and various mechanisms. Given its numerous actions, sulforaphane has drawn considerable attention for neurological disorders in recent years. Nrf2 is one of the most crucial targets of sulforaphane which has potential in regulating the series of cytoprotective enzyme expressions that have neuroprotective, antioxidative, and detoxification actions. Neurological disorders are auspicious candidates for Nrf2-targeted treatment strategy. Sulforaphane protects various neurological disorders by regulating the Nrf2 pathway. In this article, we recapitulate current studies of sulforaphane-mediated Nrf2 activation in the treatment of various neurological disorders.
Collapse
Affiliation(s)
- Md Sahab Uddin
- Department of Pharmacy, Southeast University, Dhaka, Bangladesh; Pharmakon Neuroscience Research Network, Dhaka, Bangladesh.
| | - Abdullah Al Mamun
- Department of Pharmacy, Southeast University, Dhaka, Bangladesh; Pharmakon Neuroscience Research Network, Dhaka, Bangladesh
| | - Md Jakaria
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC, Australia
| | | | - Jamil Ahmad
- Department of Human Nutrition, The University of Agriculture Peshawar, Khyber Pakhtunkhwa, Pakistan
| | - Md Ataur Rahman
- Center for Neuroscience, Brain Science Institute, Korea Institute of Science and Technology, Seoul, Republic of Korea
| | - Bijo Mathew
- Division of Drug Design and Medicinal Chemistry Research Lab, Department of Pharmaceutical Chemistry, Ahalia School of Pharmacy, Palakkad, India
| | - Mohamed M Abdel-Daim
- Department of Zoology, Science College, King Saud University, Riyadh 11451, Saudi Arabia; Department of Pharmacology, Faculty of Veterinary Medicine, Suez Canal University, Ismailia 41522, Egypt
| | - Lotfi Aleya
- Chrono-Environnement Laboratory, UMR CNRS 6249, Bourgogne Franche-Comté University, Besançon, France.
| |
Collapse
|
81
|
Al Mamun A, Uddin MS, Kabir MT, Khanum S, Sarwar MS, Mathew B, Rauf A, Ahmed M, Ashraf GM. Exploring the Promise of Targeting Ubiquitin-Proteasome System to Combat Alzheimer’s Disease. Neurotox Res 2020; 38:8-17. [DOI: 10.1007/s12640-020-00185-1] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 02/15/2020] [Accepted: 02/18/2020] [Indexed: 12/13/2022]
|
82
|
Uddin MS, Kabir MT, Rahman MM, Mathew B, Shah MA, Ashraf GM. TV 3326 for Alzheimer's dementia: a novel multimodal ChE and MAO inhibitors to mitigate Alzheimer's-like neuropathology. ACTA ACUST UNITED AC 2020; 72:1001-1012. [PMID: 32149402 DOI: 10.1111/jphp.13244] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Revised: 01/30/2020] [Accepted: 02/09/2020] [Indexed: 12/24/2022]
Abstract
OBJECTIVES Alzheimer's disease (AD) is one of the most prevalent neurodegenerative disorders and a well-recognized cause of dementia with ageing. In this review, we have represented the ChE and MAO inhibitory potential of TV 3326 against AD based on current scientific evidence. KEY FINDINGS The aetiology of AD is quite complex and not completely understood. However, it has been observed that AD involves the deposition of abnormal amyloid beta (Aβ), along with hyperphosphorylation of tau, oxidative stress, low acetylcholine (ACh) level and biometal dyshomeostasis. Due to the complex nature of AD aetiology, active research is required in the areas of development of multitarget drugs with 2 or more complementary biological functions, as they might represent significant progress in the AD treatment. Interestingly, it has been found that TV 3326 (i.e. ladostigil) is regarded as a novel therapeutic agent since it has the potential to cause inhibition of monoamine oxidase (MAO) A and B, and acetylcholinesterase (AChE) and butyrylcholinesterase (BuChE) in the brain. Furthermore, it has the capacity to reverse memory impairments, which further suggests the ability of this drug to elevate cholinergic activity in the brain. SUMMARY TV 3326 can avert oxidative-nitrative stress and gliosis. It has also been confirmed that TV 3326 contains neuroprotective and anti-apoptotic properties. Therefore, this distinctive combined inhibition of ChE and MAO along with its neuroprotective property makes TV 3326 a useful drug in the treatment of AD.
Collapse
Affiliation(s)
- Md Sahab Uddin
- Department of Pharmacy, Southeast University, Dhaka, Bangladesh.,Pharmakon Neuroscience Research Network, Dhaka, Bangladesh
| | | | - Md Motiar Rahman
- Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Bijo Mathew
- Division of Drug Design and Medicinal Chemistry Research Lab, Department of Pharmaceutical Chemistry, Ahalia School of Pharmacy, Palakkad, India
| | - Muhammad Ajmal Shah
- Department of Pharmacognosy, Faculty of Pharmaceutical Sciences, Government College University, Faisalabad, Pakistan
| | - Ghulam Md Ashraf
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia.,Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
83
|
Wang Y, Wang Z. Identification of dysregulated genes and pathways of different brain regions in Alzheimer's disease. Int J Neurosci 2020; 130:1082-1094. [PMID: 32019384 DOI: 10.1080/00207454.2020.1720677] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Background: Alzheimer's disease (AD) is a degenerative neurologic disease. The study aimed to identify the key differentially expressed genes (DEGs) and pathways in AD pathogenesis and obtain potential biomarkers in AD diagnosis.Methods: An integrated analysis of publicly available Gene Expression Omnibus datasets of AD was performed. DEGs in hippocampus tissue (HIP), temporal gyrus tissue (TG), frontal gyrus tissue (FG) and whole blood (WB) were identified. Bioinformatics analyses were used to insight into the functions of DEGs. The expression levels of candidate DEGs were preliminarily validated in GSE1297. The discriminatory ability of candidate DEGs in WB samples of AD patients and healthy individuals was evaluated in GSE63060 and GSE63061 datasets through receiver operating characteristic (ROC) analysis.Results: The DEGs in HIP, TG and FG tissues of AD were identified. Functions involved in regulation of apoptotic process, apoptotic process and cell death were significantly enriched from DEGs in AD. MAPK signaling pathway and Wnt signaling pathway were significantly enriched. YAP1, MAPK9 and GJA1 were the hub proteins in protein-protein interaction network in HIP, TG and FG. The expression levels of 14 DEGs in GSE1297 dataset were consistent with our integrated analysis. Moreover, 7 out of 14 DEGs had the diagnostic value in distinguishing AD patients from healthy controls in both GSE630060 and GSE630061 datasets.Conclusion: The DEGs including YAP1, MAPK1, GJA1 and pathways including MAPK signaling pathway and Wnt signaling pathway may be related to AD progression. RAD51C, SAFB2, SSH3 and TXNDC9 might be potential biomarkers in AD diagnosis.
Collapse
Affiliation(s)
- Yaping Wang
- Department of Neurology, Tianjin First Central Hospital, Nankai District, Tianjin, China
| | - Zhiyun Wang
- Department of Neurology, Tianjin First Central Hospital, Nankai District, Tianjin, China
| |
Collapse
|
84
|
Mamun AA, Uddin MS, Mathew B, Ashraf GM. Toxic tau: structural origins of tau aggregation in Alzheimer's disease. Neural Regen Res 2020; 15:1417-1420. [PMID: 31997800 PMCID: PMC7059584 DOI: 10.4103/1673-5374.274329] [Citation(s) in RCA: 97] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Alzheimer’s disease is characterized by the extracellular accumulation of the amyloid β in the form of amyloid plaques and the intracellular deposition of the microtubule-associated protein tau in the form of neurofibrillary tangles. Most of the Alzheimer’s drugs targeting amyloid β have been failed in clinical trials. Particularly, tau pathology connects greatly in the pathogenesis of Alzheimer’s disease. Tau protein enhances the stabilization of microtubules that leads to the appropriate function of the neuron. Changes in the quantity or the conformation of tau protein could affect its function as a microtubules stabilizer and some of the processes wherein it is involved. The molecular mechanisms leading to the accumulation of tau are principally signified by numerous posttranslational modifications that change its conformation and structural state. Therefore, aberrant phosphorylation, as well as truncation of tau protein, has come into focus as significant mechanisms that make tau protein in a pathological entity. Furthermore, the shape-shifting nature of tau advocates to comprehend the progression of Alzheimer’s disease precisely. In this review, we emphasize the recent studies about the toxic and shape-shifting nature of tau in the pathogenesis of Alzheimer’s disease.
Collapse
Affiliation(s)
| | - Md Sahab Uddin
- Department of Pharmacy, Southeast University; Pharmakon Neuroscience Research Network, Dhaka, Bangladesh
| | - Bijo Mathew
- Division of Drug Design and Medicinal Chemistry Research Lab, Department of Pharmaceutical Chemistry, Ahalia School of Pharmacy, Palakkad, India
| | - Ghulam Md Ashraf
- King Fahd Medical Research Center; Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|