1
|
Xiong G, Sachdeva M, Yosipovitch G, Ziv M, Dodiuk-Gad RP. Pruritus and Neuropsychiatric Symptoms Among Patients with Darier Disease-An Overlooked and Interconnected Challenge. J Clin Med 2025; 14:1818. [PMID: 40142630 PMCID: PMC11942685 DOI: 10.3390/jcm14061818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Revised: 02/24/2025] [Accepted: 03/06/2025] [Indexed: 03/28/2025] Open
Abstract
(1) Background: Darier disease (DD) is a rare autosomal dominant disorder caused by mutations in ATP2A2, a gene that encodes the sarco(endo)plasmic reticulum calcium-ATPase 2 enzyme, which disrupts calcium homeostasis in keratinocytes. Pruritus, a frequently overlooked symptom in DD, can lead to physical and emotional complications, especially in patients with DD who are genetically predisposed to psychiatric comorbidities. (2) Methods: This study aimed to analyze pruritus and other related symptoms in patients with DD and explore their correlation with neuropsychiatric conditions, psychological challenges, disease severity, and body surface area (BSA) involvement through a retrospective review of a tertiary center. (3) Results: Data from 76 patients (equal gender distribution, mean age 44 years) revealed a prevalence of pruritus of 90.8%, surpassing symptoms such as pain (34.3%) and malodor (43.4%). Burning sensations due to DD lesions were significantly correlated with the diagnosis of comorbid neuropsychiatric conditions (p = 0.047) and psychiatric medication use (p = 0.019). While pruritus correlated with disease severity and %BSA involvement, the findings were not statistically significant. Patients reporting pruritus had a significantly higher Dermatology Life Quality Index symptom score (2.4 ± 1.0), which is defined as the presence of itch, soreness, pain, or stinging, than those who did not (1.5 ± 0.6), indicating accurate symptom reporting. (4) Conclusions: In conclusion, a striking majority of patients with DD experience pruritus, with higher prevalence among those with neuropsychiatric challenges, severe Darier disease, and greater %BSA skin involvement. Clinicians should recognize pruritus as a key therapeutic target and adopt comprehensive treatment approaches that both address the neuropsychiatric comorbidities and the added psychological burden of pruritus in patients with DD.
Collapse
Affiliation(s)
- Grace Xiong
- Michael G. DeGroote School of Medicine, McMaster University, Hamilton, ON L8S 4L8, Canada;
| | - Muskaan Sachdeva
- Division of Dermatology, Department of Medicine, University of Toronto, Toronto, ON M5S 1A1, Canada
| | - Gil Yosipovitch
- Miami Itch Centre, Dr. Phillip Frost Department of Dermatology and Cutaneous Surgery, University of Miami, Leonard M. Miller School of Medicine, Coral Gables, FL 33136, USA;
| | - Michael Ziv
- Department of Dermatology, Emek Medical Centre, Rappaport Faculty of Medicine, Technion—Institute of Technology, Haifa 1834111, Israel
| | - Roni P. Dodiuk-Gad
- Division of Dermatology, Department of Medicine, University of Toronto, Toronto, ON M5S 1A1, Canada
- Department of Dermatology, Emek Medical Centre, Rappaport Faculty of Medicine, Technion—Institute of Technology, Haifa 1834111, Israel
| |
Collapse
|
2
|
Goetzfried SK, Hakkennes MLA, Busemann A, Bonnet S. Toward the Treatment of Glioblastoma Tumors Using Photoactivated Chemotherapy: In Vitro Evaluation of Efficacy and Safety. ACS Pharmacol Transl Sci 2025; 8:484-498. [PMID: 39974641 PMCID: PMC11833736 DOI: 10.1021/acsptsci.4c00600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 01/09/2025] [Accepted: 01/10/2025] [Indexed: 02/21/2025]
Abstract
Glioblastoma multiforme (GBM) is highly aggressive, necessitating new therapies. Photoactivated chemotherapy (PACT) offers a promising approach by activating prodrugs with visible light at the tumor site. This study evaluated the anticancer activity of ruthenium-based PACT compounds in U-87MG glioblastoma cells and their safety in SH-SY5Y neuron-like cells. The compound [3](PF6)2 showed promising light-activated anticancer effects in U-87MG cells, while [1](PF6)2 was inactive, and [2](PF6)2 was nonactivated. Interestingly, in SH-SY5Y cells, light-activated [3](PF6)2 increased cell proliferation, similar to donepezil, without causing cell death. Increased Ca2+ uptake was observed, possibly via interaction with the AMPA receptor, as suggested by docking studies. These findings suggest ruthenium-based PACT compounds may serve as potential treatments for GBM, effectively attacking cancer cells while preserving healthy neuronal cells.
Collapse
Affiliation(s)
| | - Matthijs L. A. Hakkennes
- Leiden Institute of Chemistry, Leiden University, Einsteinweg 55, 2333CC Leiden, The Netherlands
| | - Anja Busemann
- Leiden Institute of Chemistry, Leiden University, Einsteinweg 55, 2333CC Leiden, The Netherlands
| | - Sylvestre Bonnet
- Leiden Institute of Chemistry, Leiden University, Einsteinweg 55, 2333CC Leiden, The Netherlands
| |
Collapse
|
3
|
Allami P, Yazdanpanah N, Rezaei N. The role of neuroinflammation in PV interneuron impairments in brain networks; implications for cognitive disorders. Rev Neurosci 2025:revneuro-2024-0153. [PMID: 39842401 DOI: 10.1515/revneuro-2024-0153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Accepted: 12/30/2024] [Indexed: 01/24/2025]
Abstract
Fast spiking parvalbumin (PV) interneuron is an inhibitory gamma-aminobutyric acid (GABA)ergic interneuron diffused in different brain networks, including the cortex and hippocampus. As a key component of brain networks, PV interneurons collaborate in fundamental brain functions such as learning and memory by regulating excitation and inhibition (E/I) balance and generating gamma oscillations. The unique characteristics of PV interneurons, like their high metabolic demands and long branching axons, make them too vulnerable to stressors. Neuroinflammation is one of the most significant stressors that have an adverse, long-lasting impact on PV interneurons. Neuroinflammation affects PV interneurons through specialized inflammatory pathways triggered by cytokines such as tumor necrosis factor (TNF) and interleukin 6 (IL-6). The crucial cells in neuroinflammation, microglia, also play a significant role. The destructive effect of inflammation on PV interneurons can have comprehensive effects and cause neurological disorders such as schizophrenia, Alzheimer's disease (AD), autism spectrum disorder (ASD), and bipolar disorder. In this article, we provide a comprehensive review of mechanisms in which neuroinflammation leads to PV interneuron hypofunction in these diseases. The integrated knowledge about the role of PV interneurons in cognitive networks of the brain and mechanisms involved in PV interneuron impairment in the pathology of these diseases can help us with better therapeutic interventions.
Collapse
Affiliation(s)
- Pantea Allami
- Student's Scientific Research Center, School of Medicine, 48439 Tehran University of Medical Sciences , Pour Sina St, Tehran 1416634793, Iran
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Children's Medical Center Hospital, Dr. Qarib St, Keshavarz Blvd, Tehran 14194, Iran
| | - Niloufar Yazdanpanah
- Student's Scientific Research Center, School of Medicine, 48439 Tehran University of Medical Sciences , Pour Sina St, Tehran 1416634793, Iran
- Research Center for Immunodeficiencies, Children's Medical Center, 48439 Tehran University of Medical Sciences, Children's Medical Center Hospital , Dr. Qarib St, Keshavarz Blvd, Tehran 14194, Iran
- Department of Immunology, School of Medicine, 48439 Tehran University of Medical Sciences , Pour Sina St, Tehran 1416634793, Tehran, Iran
| | - Nima Rezaei
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Children's Medical Center Hospital, Dr. Qarib St, Keshavarz Blvd, Tehran 14194, Iran
- Research Center for Immunodeficiencies, Children's Medical Center, 48439 Tehran University of Medical Sciences, Children's Medical Center Hospital , Dr. Qarib St, Keshavarz Blvd, Tehran 14194, Iran
- Department of Immunology, School of Medicine, 48439 Tehran University of Medical Sciences , Pour Sina St, Tehran 1416634793, Tehran, Iran
| |
Collapse
|
4
|
Zhang L, Huang T, He H, Xu F, Yang C, Lu L, Tian G, Wang L, Mi J. Unraveling the molecular mechanisms of Ace2-mediated post-COVID-19 cognitive dysfunction through systems genetics approach. Exp Neurol 2024; 381:114921. [PMID: 39142369 DOI: 10.1016/j.expneurol.2024.114921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 08/03/2024] [Accepted: 08/10/2024] [Indexed: 08/16/2024]
Abstract
The dysregulation of Angiotensin-converting enzyme 2 (ACE2) in central nervous system is believed associates with COVID-19 induced cognitive dysfunction. However, the detailed mechanism remains largely unknown. In this study, we performed a comprehensive system genetics analysis on hippocampal ACE2 based on BXD mice panel. Expression quantitative trait loci (eQTLs) mapping showed that Ace2 was strongly trans-regulated, and the elevation of Ace2 expression level was significantly correlated with impaired cognitive functions. Further Gene co-expression analysis showed that Ace2 may be correlated with the membrane proteins in Calcium signaling pathway. Further, qRT-PCR confirmed that SARS-CoV-2 spike S1 protein upregulated ACE2 expression together with eight membrane proteins in Calcium Signaling pathway. Moreover, such elevation can be attenuated by recombinant ACE2. Collectively, our findings revealed a potential mechanism of Ace2 in cognitive dysfunction, which could be beneficial for COVID-19-induced cognitive dysfunction prevention and potential treatment.
Collapse
Affiliation(s)
- Liyuan Zhang
- Shandong Technology Innovation Center of Molecular Targeting and Intelligent Diagnosis and Treatment, Binzhou Medical University, Shandong, Yantai 264003, China
| | - Tingting Huang
- Shandong Technology Innovation Center of Molecular Targeting and Intelligent Diagnosis and Treatment, Binzhou Medical University, Shandong, Yantai 264003, China
| | - Hongjie He
- Shandong Technology Innovation Center of Molecular Targeting and Intelligent Diagnosis and Treatment, Binzhou Medical University, Shandong, Yantai 264003, China
| | - Fuyi Xu
- Shandong Technology Innovation Center of Molecular Targeting and Intelligent Diagnosis and Treatment, Binzhou Medical University, Shandong, Yantai 264003, China
| | - Chunhua Yang
- Shandong Technology Innovation Center of Molecular Targeting and Intelligent Diagnosis and Treatment, Binzhou Medical University, Shandong, Yantai 264003, China
| | - Lu Lu
- University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Geng Tian
- Shandong Technology Innovation Center of Molecular Targeting and Intelligent Diagnosis and Treatment, Binzhou Medical University, Shandong, Yantai 264003, China
| | - Lei Wang
- Harbin Medical University, Harbin 150086, Heilongjiang Province, China.
| | - Jia Mi
- Shandong Technology Innovation Center of Molecular Targeting and Intelligent Diagnosis and Treatment, Binzhou Medical University, Shandong, Yantai 264003, China.
| |
Collapse
|
5
|
Gao J, Li H, Lv H, Cheng X. Mutation of TRPML1 Channel and Pathogenesis of Neurodegeneration in Haimeria. Mol Neurobiol 2024; 61:4992-5001. [PMID: 38157120 DOI: 10.1007/s12035-023-03874-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Accepted: 12/11/2023] [Indexed: 01/03/2024]
Abstract
Neurodegenerative diseases, a group of debilitating disorders, have garnered increasing attention due to their escalating prevalence, particularly among aging populations. Alzheimer's disease (AD) reigns as a prominent exemplar within this category, distinguished by its relentless progression of cognitive impairment and the accumulation of aberrant protein aggregates within the intricate landscape of the brain. While the intricate pathogenesis of neurodegenerative diseases has been the subject of extensive investigation, recent scientific inquiry has unveiled a novel player in this complex scenario-transient receptor potential mucolipin 1 (TRPML1) channels. This comprehensive review embarks on an exploration of the intricate interplay between TRPML1 channels and neurodegenerative diseases, with an explicit spotlight on Alzheimer's disease. It immerses itself in the intricate molecular mechanisms governing TRPML1 channel functionality and elucidates their profound implications for the well-being of neurons. Furthermore, the review ventures into the realm of therapeutic potential, pondering the possibilities and challenges associated with targeting TRPML1 channels as a promising avenue for the amelioration of neurodegenerative disorders. As we traverse this multifaceted terrain of neurodegeneration and the enigmatic role of TRPML1 channels, we embark on a journey that not only broadens our understanding of the intricate machinery governing neuronal health but also holds promise for the development of innovative therapeutic interventions in the relentless battle against neurodegenerative diseases.
Collapse
Affiliation(s)
- Junqing Gao
- Department of Neurology, Shaanxi Provincial People's Hospital, Shaanxi, Xi'an, 710068, China
| | - Huanhuan Li
- Department of Neurology, Tangdu Hospital, The Fourth Military Medical University, Shaanxi, Xi'an, 710038, China
| | - Hua Lv
- Department of Neurology, Shaanxi Provincial People's Hospital, Shaanxi, Xi'an, 710068, China
| | - Xiansong Cheng
- Department of Neurology, Shaanxi Provincial People's Hospital, Shaanxi, Xi'an, 710068, China.
| |
Collapse
|
6
|
Ramakrishna S, Radhakrishna BK, Kaladiyil AP, Shah NM, Basavaraju N, Freude KK, Kommaddi RP, Muddashetty RS. Distinct calcium sources regulate temporal profiles of NMDAR and mGluR-mediated protein synthesis. Life Sci Alliance 2024; 7:e202402594. [PMID: 38749544 PMCID: PMC11096670 DOI: 10.26508/lsa.202402594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Revised: 05/02/2024] [Accepted: 05/02/2024] [Indexed: 05/18/2024] Open
Abstract
Calcium signaling is integral for neuronal activity and synaptic plasticity. We demonstrate that the calcium response generated by different sources modulates neuronal activity-mediated protein synthesis, another process essential for synaptic plasticity. Stimulation of NMDARs generates a protein synthesis response involving three phases-increased translation inhibition, followed by a decrease in translation inhibition, and increased translation activation. We show that these phases are linked to NMDAR-mediated calcium response. Calcium influx through NMDARs elicits increased translation inhibition, which is necessary for the successive phases. Calcium through L-VGCCs acts as a switch from translation inhibition to the activation phase. NMDAR-mediated translation activation requires the contribution of L-VGCCs, RyRs, and SOCE. Furthermore, we show that IP3-mediated calcium release and SOCE are essential for mGluR-mediated translation up-regulation. Finally, we signify the relevance of our findings in the context of Alzheimer's disease. Using neurons derived from human fAD iPSCs and transgenic AD mice, we demonstrate the dysregulation of NMDAR-mediated calcium and translation response. Our study highlights the complex interplay between calcium signaling and protein synthesis, and its implications in neurodegeneration.
Collapse
Affiliation(s)
- Sarayu Ramakrishna
- Centre for Brain Research, Indian Institute of Science, Bangalore, India
| | - Bindushree K Radhakrishna
- Centre for Brain Research, Indian Institute of Science, Bangalore, India
- Manipal Academy of Higher Education, Manipal, India
| | - Ahamed P Kaladiyil
- Centre for Brain Research, Indian Institute of Science, Bangalore, India
| | - Nisa Manzoor Shah
- Centre for Brain Research, Indian Institute of Science, Bangalore, India
- Manipal Academy of Higher Education, Manipal, India
| | - Nimisha Basavaraju
- Centre for Brain Research, Indian Institute of Science, Bangalore, India
- Manipal Academy of Higher Education, Manipal, India
| | - Kristine K Freude
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg C, Denmark
| | | | - Ravi S Muddashetty
- Centre for Brain Research, Indian Institute of Science, Bangalore, India
| |
Collapse
|
7
|
Cruchaga C, Bradley J, Western D, Wang C, Lucio Da Fonseca E, Neupane A, Kurup J, Ray NI, Jean-Francois M, Gorijala P, Bergmann K, Budde J, Martin E, Pericak-Vance M, Cuccaro M, Kunkle B, Morris J, Holtzman D, Perrin R, Naj A, Haines J, Schellenberg G, Fernandez V, Reitz C, Beecham G. Novel early-onset Alzheimer-associated genes influence risk through dysregulation of glutamate, immune activation, and intracell signaling pathways. RESEARCH SQUARE 2024:rs.3.rs-4480585. [PMID: 38883718 PMCID: PMC11177996 DOI: 10.21203/rs.3.rs-4480585/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2024]
Abstract
Alzheimer Disease (AD) is a highly polygenic disease that presents with relatively earlier onset (≤70yo; EOAD) in about 5% of cases. Around 90% of these EOAD cases remain unexplained by pathogenic mutations. Using data from EOAD cases and controls, we performed a genome-wide association study (GWAS) and trans-ancestry meta-analysis on non-Hispanic Whites (NHW, NCase=6,282, NControl=13,386), African Americans (AA NCase=782, NControl=3,663) and East Asians (NCase=375, NControl=838 CO). We identified eight novel significant loci: six in the ancestry-specific analyses and two in the trans-ancestry analysis. By integrating gene-based analysis, eQTL, pQTL and functional annotations, we nominate four novel genes that are involved in microglia activation, glutamate production, and signaling pathways. These results indicate that EOAD, although sharing many genes with LOAD, harbors unique genes and pathways that could be used to create better prediction models or target identification for this type of AD.
Collapse
Affiliation(s)
| | | | - Daniel Western
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
| | | | | | | | | | | | | | | | | | | | | | | | - Michael Cuccaro
- The John P. Hussman Institute for Human Genomics, University of Miami, Miami, Florida
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
8
|
Andrade VHBD, M Rodrigues EY, Dias NAF, Ferreira GFC, Carvalho DBD, das Neves AR, Coronel PMV, Yonekawa MKA, Parisotto EB, Santos EAD, Souza AS, Paredes-Gamero EJ, de Sousa KS, Souza LLD, Resstel LBM, Baroni ACM, Lagatta DC. Neuroprotective Profile of Triazole Grandisin Analogue against Amyloid-Beta Oligomer-Induced Cognitive Impairment. ACS Chem Neurosci 2023; 14:4298-4310. [PMID: 38048522 DOI: 10.1021/acschemneuro.3c00443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/06/2023] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder caused by accumulation of amyloid-β oligomers (AβO) in the brain, neuroinflammation, oxidative stress, and cognitive decline. Grandisin, a tetrahydrofuran neolignan, exhibits relevant anti-inflammatory and antioxidant properties. Interestingly, grandisin-based compounds were shown to prevent AβO-induced neuronal death in vitro. However, no study has assessed the effect of these compounds on the AD animal model. This study focuses on a triazole grandisin analogue (TGA) synthesized using simplification and bioisosteric drug design, which resulted in improved potency and solubility compared with the parent compound. This study aimed to investigate the possible in vivo effects of TGA against AβO-induced AD. Male C57/Bl6 mice underwent stereotaxic intracerebroventricular AβO (90 μM) or vehicle injections. 24 h after surgery, animals received intraperitoneal treatment with TGA (1 mg/kg) or vehicle, administered on a 14 day schedule. One day after treatment completion, a novel object recognition task (NORT) was performed. Memantine (10 mg/kg) was administered as a positive control. NORT retention sessions were performed on days 8 and 16 after AβO injection. Immediately after retention sessions, animals were euthanized for cortex and hippocampus collection. Specimens were subjected to oxidative stress and cytokine analyses. TGA reduced the level of cortex/hippocampus lipoperoxidation and prevented cognitive impairment in AβO-injected mice. Additionally, TGA reduced tumor necrosis factor (TNF) and interferon-γ (IFN-γ) levels in the hippocampus. By contrast, memantine failed to prevent cortex/hippocampus lipid peroxidation, recognition memory decline, and AβO-induced increases in TNF and IFN-γ levels in the hippocampus. Thus, memantine was unable to avoid the AβO-induced persistent cognitive impairment. The results showed that TGA may prevent memory impairment by exerting antioxidant and anti-inflammatory effects in AβO-injected mice. Moreover, TGA exhibited a persistent neuroprotective effect compared to memantine, reflecting an innovative profile of this promising agent against neurodegenerative diseases, such as AD.
Collapse
Affiliation(s)
- Victor H B de Andrade
- Pharmaceutical Sciences, Food and Nutrition School, Federal University of Mato Grosso do Sul (UFMS), Campo Grande 79051-470, Brazil
| | - Erick Y M Rodrigues
- Pharmaceutical Sciences, Food and Nutrition School, Federal University of Mato Grosso do Sul (UFMS), Campo Grande 79051-470, Brazil
| | - Nayara A F Dias
- School of Medicine, Federal University of Mato Grosso do Sul (UFMS), Campo Grande 79051-470, Brazil
| | - Gabriela F C Ferreira
- School of Medicine, Federal University of Mato Grosso do Sul (UFMS), Campo Grande 79051-470, Brazil
| | - Diego B de Carvalho
- Pharmaceutical Sciences, Food and Nutrition School, Federal University of Mato Grosso do Sul (UFMS), Campo Grande 79051-470, Brazil
| | - Amarith R das Neves
- Pharmaceutical Sciences, Food and Nutrition School, Federal University of Mato Grosso do Sul (UFMS), Campo Grande 79051-470, Brazil
| | - Paola M V Coronel
- Pharmaceutical Sciences, Food and Nutrition School, Federal University of Mato Grosso do Sul (UFMS), Campo Grande 79051-470, Brazil
| | - Murilo K A Yonekawa
- Institute of Biosciences, Federal University of Mato Grosso do Sul (INBIO-UFMS), Campo Grande 79051-470, Brazil
| | - Eduardo B Parisotto
- Pharmaceutical Sciences, Food and Nutrition School, Federal University of Mato Grosso do Sul (UFMS), Campo Grande 79051-470, Brazil
| | - Edson A Dos Santos
- Institute of Biosciences, Federal University of Mato Grosso do Sul (INBIO-UFMS), Campo Grande 79051-470, Brazil
| | - Albert S Souza
- Institute of Biosciences, Federal University of Mato Grosso do Sul (INBIO-UFMS), Campo Grande 79051-470, Brazil
| | - Edgar J Paredes-Gamero
- Pharmaceutical Sciences, Food and Nutrition School, Federal University of Mato Grosso do Sul (UFMS), Campo Grande 79051-470, Brazil
| | - Kamylla S de Sousa
- Pharmaceutical Sciences, Food and Nutrition School, Federal University of Mato Grosso do Sul (UFMS), Campo Grande 79051-470, Brazil
| | - Letícia L de Souza
- Pharmaceutical Sciences, Food and Nutrition School, Federal University of Mato Grosso do Sul (UFMS), Campo Grande 79051-470, Brazil
| | - Leonardo B M Resstel
- School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto 14049-900, Brazil
| | - Adriano C M Baroni
- Pharmaceutical Sciences, Food and Nutrition School, Federal University of Mato Grosso do Sul (UFMS), Campo Grande 79051-470, Brazil
| | - Davi C Lagatta
- Pharmaceutical Sciences, Food and Nutrition School, Federal University of Mato Grosso do Sul (UFMS), Campo Grande 79051-470, Brazil
| |
Collapse
|
9
|
Puentes-Díaz N, Chaparro D, Morales-Morales D, Alí-Torres J, Flores-Gaspar A. Pincer Ligands as Multifunctional Agents for Alzheimer's Copper Dysregulation and Oxidative Stress: A Computational Evaluation. Chempluschem 2023; 88:e202300405. [PMID: 37756039 DOI: 10.1002/cplu.202300405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2023] [Revised: 09/22/2023] [Accepted: 09/26/2023] [Indexed: 09/28/2023]
Abstract
Alzheimer's disease (AD) is the most common form of dementia worldwide, affecting millions of people around the globe. AD is characterized by different pathologies being beta-amyloid (Aβ) plaque formation, metal ion dysregulation, and oxidative stress (OS) central topics under investigation. Copper-Aβ complexes have been shown to induce catalytic hydrogen peroxide formation and increase OS in the brain leading to neuronal death. Pincer-type compounds are tridentate ligands that coordinate metals in a planar fashion whose properties can be tuned via group substitutions, giving rise to many possibilities in catalysis and drug discovery. In this work we evaluated the potential pharmaceutical activity of 26 pincer compounds in AD's copper ion-related oxidative stress framework. In this sense, four key aspects were considered: 1) Lipinski's rule of five, 2) blood-brain barrier permeation, 3) standard reduction potential (SRP) of the formed copper complexes, and 4) the ligand's affinity towards copper cations. The evaluation of these criteria was performed by means of bioinformatic tools and electronic structure calculations at the DFT level of theory. Our results suggest that two compounds from this set are potential antioxidant agents, whereas five of them are promissory distributor-like compounds in the context of AD.
Collapse
Affiliation(s)
- Nicolás Puentes-Díaz
- Department of Chemistry, Universidad Nacional de Colombia, Av. Cra. 30 #45-03, Bogotá
| | - Diego Chaparro
- Department of Chemistry, Universidad Nacional de Colombia, Av. Cra. 30 #45-03, Bogotá
- Department of Chemistry, Universidad Militar Nueva Granada, Kilómetro 2, vía Cajicá-Zipaquira, costado oriental, Cajicá, Colombia
| | - David Morales-Morales
- Institute of Chemistry, Universidad Nacional Autónoma de México, Ciudad de México, 04510, Mexico
| | - Jorge Alí-Torres
- Department of Chemistry, Universidad Nacional de Colombia, Av. Cra. 30 #45-03, Bogotá
| | - Areli Flores-Gaspar
- Department of Chemistry, Universidad Militar Nueva Granada, Kilómetro 2, vía Cajicá-Zipaquira, costado oriental, Cajicá, Colombia
| |
Collapse
|
10
|
Rothschild SC, Lai G, Tombes RM, Clements WK. Constitutively active CaMKII Drives B lineage acute lymphoblastic leukemia/lymphoma in tp53 mutant zebrafish. PLoS Genet 2023; 19:e1011102. [PMID: 38117861 PMCID: PMC10766190 DOI: 10.1371/journal.pgen.1011102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 01/04/2024] [Accepted: 12/07/2023] [Indexed: 12/22/2023] Open
Abstract
Acute lymphoblastic leukemia/lymphoma (ALL) is the most common pediatric cancer and is a malignancy of T or B lineage lymphoblasts. Dysregulation of intracellular Ca2+ levels has been observed in patients with ALL, leading to improper activation of downstream signaling. Here we describe a new zebrafish model of B ALL, generated by expressing human constitutively active CaMKII (CA-CaMKII) in tp53 mutant lymphocytes. In this model, B cell hyperplasia in the kidney marrow and spleen progresses to overt leukemia/lymphoma, with only 29% of zebrafish surviving the first year of life. Leukemic fish have reduced productive genomic VDJ recombination in addition to reduced expression and improper splicing of ikaros1, a gene often deleted or mutated in patients with B ALL. Inhibiting CaMKII in human pre-B ALL cells induced cell death, further supporting a role for CaMKII in leukemogenesis. This research provides novel insight into the role of Ca2+-directed signaling in lymphoid malignancy and will be useful in understanding disease development and progression.
Collapse
Affiliation(s)
- Sarah C. Rothschild
- Life Sciences, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Guanhua Lai
- Pathology, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Robert M. Tombes
- Life Sciences, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Wilson K. Clements
- Experimental Hematology, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States of America
| |
Collapse
|
11
|
Liu B, Zhu X, Zhou Q, Su Y, Qian Y, Ma Z, Gu X, Xia T. Activating ryanodine receptor improves isoflurane-induced cognitive dysfunction. Brain Res Bull 2023; 204:110790. [PMID: 37852420 DOI: 10.1016/j.brainresbull.2023.110790] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2023] [Revised: 09/30/2023] [Accepted: 10/16/2023] [Indexed: 10/20/2023]
Abstract
BACKGROUND Postoperative cognitive dysfunction (POCD) is characterized by impaired learning and memory. 6 h duration isoflurane anesthesia is an important factor to induce POCD, and the dysfunction of ryanodine receptor (RyR) in the hippocampus may be involved in this process. We investigated the expression of RyR3 in the hippocampus of mice after 6-h duration isoflurane anesthesia, as well as the improvement of RyR receptor agonist caffeine on POCD mice, while attempting to identify the underlying molecular mechanism. MATERIALS We constructed a POCD model using 8-week-old male C57BL/6J mice that were exposed to 6-h duration isoflurane. Prior to the three-day cognitive behavioral experiment, RyR agonist caffeine were injected. Fear conditioning and location memory tests were used in behavioral studies. We also exposed the mouse neuroblastoma cell line Neuro-2a (N2A) to 6-h duration isoflurane exposure to simulate the conditions of in vivo cognitive dysfunction. We administered ryanodine receptor agonist (caffeine) and inhibitor (ryanodine) to N2a cells. Following that, we performed a series of bioinformatics analysis to discover proteins that are involved in the development of cognitive dysfunction. Rt-PCR and Western blot were used to assess mRNA level and protein expression. RESULTS 6-h duration isoflurane anesthesia induced cognitive dysfunction and increased RyR3 mRNA levels in hippocampus. The mRNA levels of RyR3 in cultured N2a cells after anesthesia were comparable to those in vivo, and the RyR agonist caffeine corrected the expression of some cognitive-related phenotypic proteins that were disturbed after anesthesia. Intraperitoneal injection of RyR agonist caffeine can improve cognitive function after isoflurane anesthesia in mice, and bioinformatics analyses suggest that CaMKⅣ may be involved in the molecular mechanism. CONCLUSION Ryanodine receptor agonist caffeine may improve cognitive dysfunction in mice after isoflurane anesthesia.
Collapse
Affiliation(s)
- Binwen Liu
- Department of Anesthesiology, Affiliated Drum Tower Hospital of Medical School of Nanjing University, Nanjing 210008, China; Medical School, Nanjing University, Nanjing 210093, China; Jiangsu Key Laboratory of Molecular Medicine, Nanjing University, Nanjing, China; State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, China.
| | - Xurui Zhu
- Department of Anesthesiology, Affiliated Drum Tower Hospital of Medical School of Nanjing University, Nanjing 210008, China.
| | - Qingyun Zhou
- Department of Anesthesiology, Affiliated Drum Tower Hospital of Medical School of Nanjing University, Nanjing 210008, China; Medical School, Nanjing University, Nanjing 210093, China; Jiangsu Key Laboratory of Molecular Medicine, Nanjing University, Nanjing, China; State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, China.
| | - Yan Su
- Department of Anesthesiology, Affiliated Drum Tower Hospital of Medical School of Nanjing University, Nanjing 210008, China; Medical School, Nanjing University, Nanjing 210093, China; Jiangsu Key Laboratory of Molecular Medicine, Nanjing University, Nanjing, China; State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, China.
| | - Yue Qian
- Department of Anesthesiology, Affiliated Drum Tower Hospital of Medical School of Nanjing University, Nanjing 210008, China.
| | - Zhengliang Ma
- Department of Anesthesiology, Affiliated Drum Tower Hospital of Medical School of Nanjing University, Nanjing 210008, China.
| | - Xiaoping Gu
- Department of Anesthesiology, Affiliated Drum Tower Hospital of Medical School of Nanjing University, Nanjing 210008, China.
| | - Tianjiao Xia
- Medical School, Nanjing University, Nanjing 210093, China; Jiangsu Key Laboratory of Molecular Medicine, Nanjing University, Nanjing, China; State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, China.
| |
Collapse
|
12
|
Kim J, Seo S, Park JHY, Lee KW, Kim J, Kim JC. Ca 2+-Permeable TRPV1 Receptor Mediates Neuroprotective Effects in a Mouse Model of Alzheimer's Disease via BDNF/CREB Signaling Pathway. Mol Cells 2023; 46:319-328. [PMID: 37070458 PMCID: PMC10183797 DOI: 10.14348/molcells.2023.2156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 01/04/2023] [Accepted: 01/06/2023] [Indexed: 04/19/2023] Open
Abstract
Transient receptor potential vanilloid 1 (TRPV1) protein is a Ca2+-permeable non-selective cation channel known for its pain modulation pathway. In a previous study, it was discovered that a triple-transgenic Alzheimer's disease (AD) mouse model (3xTg-AD+/+) has anti-AD effects. The expression of proteins in the brain-derived neurotrophic factor (BDNF)/cAMP response element binding protein (CREB) pathway in a 3xTg-AD/TRPV1 transgenic mice model was investigated to better understand the AD regulatory effect of TRPV1 deficiency. The results show that TRPV1 deficiency leads to CREB activation by increasing BDNF levels and promoting phosphorylation of tyrosine receptor kinase B (TrkB), extracellular signal-regulated kinase (ERK), protein kinase B (Akt), and CREB in the hippocampus. Additionally, TRPV1 deficiency-induced CREB activation increases the antiapoptotic factor B-cell lymphoma 2 (Bcl-2) gene, which consequently downregulates Bcl-2-associated X (Bax) expression and decreases cleaved caspase-3 and cleaved poly (ADP-ribose) polymerase (PARP), which leads to the prevention of hippocampal apoptosis. In conclusion, TRPV1 deficiency exhibits neuroprotective effects by preventing apoptosis through the BDNF/CREB signal transduction pathway in the hippocampus of 3xTg-AD mice.
Collapse
Affiliation(s)
- Juyong Kim
- Department of Agricultural Biotechnology, Seoul National University, Seoul 08826, Korea
- Natural Product Research Center, Korea Institute of Science and Technology (KIST), Gangneung 25451, Korea
| | - Sangwoo Seo
- Department of Agricultural Biotechnology, Seoul National University, Seoul 08826, Korea
| | | | - Ki Won Lee
- Department of Agricultural Biotechnology, Seoul National University, Seoul 08826, Korea
- Bio-MAX Institute, Seoul National University, Seoul 08826, Korea
- Advanced Institutes of Convergence Technology, Seoul National University, Suwon 16229, Korea
- Center for Food and Bioconvergence, College of Agriculture and Life Sciences, Seoul National University, Seoul 08826, Korea
| | - Jiyoung Kim
- Center for Food and Bioconvergence, College of Agriculture and Life Sciences, Seoul National University, Seoul 08826, Korea
| | - Jin-Chul Kim
- Natural Product Research Center, Korea Institute of Science and Technology (KIST), Gangneung 25451, Korea
- Division of Bio-Medical Science & Technology, University of Science and Technology, Daejeon 34113, Korea
| |
Collapse
|
13
|
Vera R, Hong N, Jiang B, Liang G, Eckenhoff MF, Kincaid HJ, Browne V, Chellaraj V, Gisewhite D, Greenberg M, Ranjan S, Zhu G, Wei H. Effects of intranasal dantrolene nanoparticles on brain concentration and behavior in PS19 tau transgenic mice. RESEARCH SQUARE 2023:rs.3.rs-2802620. [PMID: 37214948 PMCID: PMC10197765 DOI: 10.21203/rs.3.rs-2802620/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
Background Repurposing dantrolene as a potential disease-modifying treatment for Alzheimer's disease has been shown to be effective in amyloid transgenic mouse models but has not been examined in a model of tauopathy. Objective The effects of a nanoparticle intranasal formulation, the Eagle Research Formulation of Ryanodex (ERFR), in young adult and aged wild type and PS19 tau transgenic mice was investigated. Methods The bioavailability of intranasal ERFR was measured in 2 months and 9-12 month old C57BL/6J male mice. Mice received a single intranasal dose of ERFR and, after 20 min, blood and brain samples were collected. Dantrolene concentrations in the plasma and brain were analyzed by High Performance Liquid Chromatography. Animal behavior was examined in PS19 tau transgenic mice, with/without acrolein treatment to exacerbate cognitive deficits. Behavioral tests included cognition (cued and contextual fear conditioning, y-maze), motor function (rotarod), and olfaction (buried food test). Results Dantrolene concentration in the blood and brain decreased with age, though the decrease was greater in the blood resulting in a higher brain to blood concentration ratio. The behavioral assays showed no significant changes in cognition, olfaction or motor function in the PS19 mice compared to controls after chronic ERFR treatment even with acrolein treatment. Conclusion Our studies suggest that while we did not find PS19 mice to be a reliable Alzheimer animal model to test the therapeutic efficacy of dantrolene, the results suggest a potential for ERFR to be an effective chronic therapy for Alzheimer's disease and that further studies are indicated.
Collapse
Affiliation(s)
- Robert Vera
- University of Pennsylvania, Perelman School of Medicine
| | - Nicholas Hong
- University of Pennsylvania, Perelman School of Medicine
| | | | - Grace Liang
- University of Pennsylvania, Perelman School of Medicine Maryellen
| | | | | | | | | | | | | | | | | | - Huafeng Wei
- University of Pennsylvania, Perelman School of Medicine
| |
Collapse
|
14
|
Ren X, Zhang S, Yang Y, Song A, Liang F, Zhang Y, Dong Y, Wu X, Xie Z. Ketamine Induces Delirium-Like Behavior and Interferes With Endosomal Tau Trafficking. Anesth Analg 2023; 136:779-788. [PMID: 35880774 PMCID: PMC9873837 DOI: 10.1213/ane.0000000000006146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
BACKGROUND Ketamine is an intravenous anesthetic. However, whether ketamine can induce neurotoxicity and neurobehavioral deficits remains largely unknown. Delirium is a syndrome of acute brain dysfunction associated with anesthesia and surgery in patients, and tau protein may contribute to postoperative delirium. Finally, ketamine may affect the function of the endosome, the key organelle for tau release from neurons. Therefore, we set out to determine the effects of ketamine on delirium-like behavior in mice and on tau trafficking in cultured cells. METHODS We used the buried-food test, open-field test, and Y-maze test in adult mice to assess the presence of delirium-like behavior in mice. We quantified tau amounts in the serum of mice. We used cell fraction methods to determine the effects of ketamine on tau intracellular trafficking, extracellular release, and endosome trafficking in cultured cells. RESULTS Ketamine induced delirium-like behavior in mice and increased tau amounts in serum of mice. The ketamine treatments also led to increased accumulation of endosomes, as evidenced by increased endosomal markers Rab5 and Rab7. Moreover, ketamine inhibited endosome maturation, demonstrated by decreased membrane-bound but increased cytoplasm amounts of Rab5 and Rab7. Consequently, ketamine increased tau in the endosomes of cultured cells and the cell culture medium. CONCLUSIONS These data suggest that ketamine may interfere with intracellular tau trafficking and induce delirium-like behavior, promoting future research regarding the potential neurotoxicity of anesthetics.
Collapse
Affiliation(s)
- Xinghua Ren
- School of Forensic Medicine, China Medical University, No. 77, Puhe Road, Shenyang North New Area, Shenyang, Liaoning 110122, China
- Geriatric Anesthesia Research Unit, Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, 149 13 Street, Charlestown, MA 02129, USA
| | - Siyi Zhang
- School of Forensic Medicine, China Medical University, No. 77, Puhe Road, Shenyang North New Area, Shenyang, Liaoning 110122, China
- Genetics and Aging Research Unit, Department of Neurology, Massachusetts General Hospital, 114 16 Street, Charlestown, MA 02129, USA
| | - Yongyan Yang
- Geriatric Anesthesia Research Unit, Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, 149 13 Street, Charlestown, MA 02129, USA
- Department of Anesthesiology, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin 300052, China
| | - Annie Song
- Geriatric Anesthesia Research Unit, Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, 149 13 Street, Charlestown, MA 02129, USA
| | - Feng Liang
- Geriatric Anesthesia Research Unit, Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, 149 13 Street, Charlestown, MA 02129, USA
| | - Yiying Zhang
- Geriatric Anesthesia Research Unit, Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, 149 13 Street, Charlestown, MA 02129, USA
| | - Yuanlin Dong
- Geriatric Anesthesia Research Unit, Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, 149 13 Street, Charlestown, MA 02129, USA
| | - Xu Wu
- School of Forensic Medicine, China Medical University, No. 77, Puhe Road, Shenyang North New Area, Shenyang, Liaoning 110122, China
| | - Zhongcong Xie
- Geriatric Anesthesia Research Unit, Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, 149 13 Street, Charlestown, MA 02129, USA
| |
Collapse
|
15
|
Tiwari V, Singh J, Tiwari P, Chaturvedi S, Gupta S, Mishra A, Singh S, Wahajuddin M, Hanif K, Shukla S. ACE2/ANG-(1-7)/Mas receptor axis activation prevents inflammation and improves cognitive functions in streptozotocin induced rat model of Alzheimer's disease-like phenotypes. Eur J Pharmacol 2023; 946:175623. [PMID: 36871666 DOI: 10.1016/j.ejphar.2023.175623] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 01/25/2023] [Accepted: 02/23/2023] [Indexed: 03/06/2023]
Abstract
Activation of the renin-angiotensin system (RAS), by Angiotensin converting enzyme/Angiotensin II/Angiotensin receptor-1 (ACE/Ang II/AT1 R) axis elicits amyloid deposition and cognitive impairment. Furthermore, ACE2 induced release of Ang-(1-7) binds with the Mas receptor and autoinhibits ACE/Ang II/AT1 axis activation. Inhibition of ACE by perindopril has been reported to improve memory in preclinical settings. However, the functional significance and mechanism by which ACE2/Mas receptor regulate cognitive functions and amyloid pathology is not known. The present study is aimed to determine the role of ACE2/Ang-(1-7)/Mas receptor axis in STZ induced rat model of Alzheimer's disease (AD). We have used pharmacological, biochemical and behavioural approaches to identify the role of ACE2/Ang-(1-7)/Mas receptor axis activation on AD-like pathology in both in vitro and invivo models. STZ treatment enhances ROS formation, inflammation markers and NFκB/p65 levels which are associated with reduced ACE2/Mas receptor levels, acetylcholine activity and mitochondrial membrane potential in N2A cells. DIZE mediated ACE2/Ang-(1-7)/Mas receptor axis activation resulted in reduced ROS generation, astrogliosis, NFκB level and inflammatory molecules and improved mitochondrial functions along with Ca2+ influx in STZ treated N2A cells. Interestingly, DIZE induced activation of ACE2/Mas receptor significantly restored acetylcholine levels and reduced amyloid-beta and phospho-tau deposition in cortex and hippocampus that resulted in improved cognitive function in STZ induced rat model of AD-like phenotypes. Our data indicate that ACE2/Mas receptor activation is sufficient to prevented cognitive impairment and progression of amyloid pathology in STZ induced rat model of AD-like phenotypes. These findings suggest the potential role of ACE2/Ang-(1-7)/Mas axis in AD pathophysiology by regulating inflammation cognitive functions.
Collapse
Affiliation(s)
- Virendra Tiwari
- Division of Neuroscience and Ageing Biology, CSIR- Central Drug Research Institute, Lucknow, 226031, (U.P), India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Jitendra Singh
- Division of Neuroscience and Ageing Biology, CSIR- Central Drug Research Institute, Lucknow, 226031, (U.P), India
| | - Priya Tiwari
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India; Division of Pharmacology, CSIR- Central Drug Research Institute, Lucknow, 226031, (U.P), India
| | - Swati Chaturvedi
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India; Division of Pharmaceutics and Pharmacokinetics, CSIR - Central Drug Research Institute, Lucknow, 226031, (U.P), India
| | - Shivangi Gupta
- Division of Neuroscience and Ageing Biology, CSIR- Central Drug Research Institute, Lucknow, 226031, (U.P), India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Akanksha Mishra
- Division of Neuroscience and Ageing Biology, CSIR- Central Drug Research Institute, Lucknow, 226031, (U.P), India; Department of Cell Biology and Anatomy, New York Medical College, Valhalla, NY, 01595, USA
| | - Sonu Singh
- Division of Neuroscience and Ageing Biology, CSIR- Central Drug Research Institute, Lucknow, 226031, (U.P), India; Department of Neuroscience, School of Medicine, University of Connecticut (Uconn) Health Center, 263 Farmington Avenue, L-4078, Farmington, CT, 06030, USA
| | - Muhammad Wahajuddin
- Division of Pharmaceutics and Pharmacokinetics, CSIR - Central Drug Research Institute, Lucknow, 226031, (U.P), India; Institute of Cancer Therapeutics, University of Bradford, Bradford, BD7 1DP, United Kingdom
| | - Kashif Hanif
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India; Division of Pharmacology, CSIR- Central Drug Research Institute, Lucknow, 226031, (U.P), India
| | - Shubha Shukla
- Division of Neuroscience and Ageing Biology, CSIR- Central Drug Research Institute, Lucknow, 226031, (U.P), India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India.
| |
Collapse
|
16
|
Gunawan R, Yang M, Lau C. X-RAY MEASUREMENT OF INTRACELLULAR CHLORIDE AND OTHER IONS IN MAMMALIAN CELLS. TALANTA OPEN 2023. [DOI: 10.1016/j.talo.2023.100189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
|
17
|
Sharma N, Banerjee R, Davis RL. Early Mitochondrial Defects in the 5xFAD Mouse Model of Alzheimer's Disease. J Alzheimers Dis 2023; 91:1323-1338. [PMID: 36617782 DOI: 10.3233/jad-220884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
BACKGROUND Mitochondrial (MT) dysfunction is a hallmark of Alzheimer's disease (AD). Amyloid-β protein precursor and amyloid-β peptides localize to MT and lead to MT dysfunction in familial forms of AD. This dysfunction may trigger subsequent types of pathology. OBJECTIVE To identify the MT phenotypes that occur early in order to help understand the cascade of AD pathophysiology. METHODS The 5xFAD mouse model was used to explore the time course of MT pathologies in both sexes. Protein biomarkers for MT dynamics were measured biochemically and MT function was measured using oxygen consumption and ATP assays. RESULTS We discovered progressive alterations in mitochondrial dynamics (biogenesis, fission, fusion, and mitophagy) and function (O2 consumption, ATP generation, and Ca2+ import) in the hippocampus of 5xFAD mice in both sexes as early as 2 months of age. Thus, mitochondrial dynamics and function become altered at young ages, consistent with an early role for mitochondria in the AD pathological cascade. CONCLUSION Our study offers the baseline information required to understand the hierarchical relationship between the multiple pathologies that develop in this mouse model and provides early biomarkers for MT dysfunction. This will aid in dissecting the temporal cascade of pathologies, understanding sex-specific differences, and in testing the efficacy of putative mitochondrial therapeutics.
Collapse
Affiliation(s)
- Neelam Sharma
- Department of Neuroscience, University of Florida Scripps Biomedical Research Institute, Jupiter, FL, USA
| | - Rupkatha Banerjee
- Department of Neuroscience, University of Florida Scripps Biomedical Research Institute, Jupiter, FL, USA
| | - Ronald L Davis
- Department of Neuroscience, University of Florida Scripps Biomedical Research Institute, Jupiter, FL, USA
| |
Collapse
|
18
|
Role of Microglia and Astrocytes in Alzheimer’s Disease: From Neuroinflammation to Ca2+ Homeostasis Dysregulation. Cells 2022; 11:cells11172728. [PMID: 36078138 PMCID: PMC9454513 DOI: 10.3390/cells11172728] [Citation(s) in RCA: 75] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 08/26/2022] [Accepted: 08/30/2022] [Indexed: 12/12/2022] Open
Abstract
Alzheimer’s disease (AD) is the most common form of dementia worldwide, with a complex, poorly understood pathogenesis. Cerebral atrophy, amyloid-β (Aβ) plaques, and neurofibrillary tangles represent the main pathological hallmarks of the AD brain. Recently, neuroinflammation has been recognized as a prominent feature of the AD brain and substantial evidence suggests that the inflammatory response modulates disease progression. Additionally, dysregulation of calcium (Ca2+) homeostasis represents another early factor involved in the AD pathogenesis, as intracellular Ca2+ concentration is essential to ensure proper cellular and neuronal functions. Although growing evidence supports the involvement of Ca2+ in the mechanisms of neurodegeneration-related inflammatory processes, scant data are available on its contribution in microglia and astrocytes functioning, both in health and throughout the AD continuum. Nevertheless, AD-related aberrant Ca2+ signalling in astrocytes and microglia is crucially involved in the mechanisms underpinning neuroinflammatory processes that, in turn, impact neuronal Ca2+ homeostasis and brain function. In this light, we attempted to provide an overview of the current understanding of the interactions between the glia cells-mediated inflammatory responses and the molecular mechanisms involved in Ca2+ homeostasis dysregulation in AD.
Collapse
|
19
|
Lin YK, Liang CS, Tsai CK, Tsai CL, Lee JT, Sung YF, Chou CH, Shang HS, Yang BH, Lin GY, Su MW, Yang FC. A Metallomic Approach to Assess Associations of Plasma Metal Levels with Amnestic Mild Cognitive Impairment and Alzheimer's Disease: An Exploratory Study. J Clin Med 2022; 11:jcm11133655. [PMID: 35806940 PMCID: PMC9267221 DOI: 10.3390/jcm11133655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 06/10/2022] [Accepted: 06/22/2022] [Indexed: 02/01/2023] Open
Abstract
Alzheimer’s disease (AD) involves the abnormal activity of transition metals and metal ion dyshomeostasis; however, the potential of trace metal biomarkers in predicting cognitive decline has not been evaluated. This study aimed to assess the potential of 36 trace elements in predicting cognitive decline in patients with amnestic mild cognitive impairment (aMCI) or AD. Participants (9 controls, 23 aMCI due to AD, and 8 AD dementia) underwent comprehensive cognitive tests, including the Mini-Mental State Examination (MMSE) and trace metal analysis. The correlations between the plasma trace element levels and annual MMSE changes during follow-up were analyzed. We found that an increase in disease severity was linked to lower plasma levels of boron (B), bismuth (Bi), thorium (Th), and uranium (U) (adjusted p < 0.05). Higher baseline calcium levels (r = 0.50, p = 0.026) were associated with less annual cognitive decline; those of B (r = −0.70, p = 0.001), zirconium (r = −0.58, p = 0.007), and Th (r = −0.52, p = 0.020) with rapid annual cognitive decline in the aMCI group; and those of manganese (r = −0.91, p = 0.035) with rapid annual cognitive decline in the AD group. Overall, our exploratory study suggests that plasma metal levels have great potential as in vivo biomarkers for aMCI and AD. Larger sample studies are necessary to confirm these results.
Collapse
Affiliation(s)
- Yu-Kai Lin
- Department of Neurology, Tri-Service General Hospital, National Defense Medical Center, Taipei 114, Taiwan; (Y.-K.L.); (C.-K.T.); (C.-L.T.); (J.-T.L.); (Y.-F.S.); (C.-H.C.); (G.-Y.L.)
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei 114, Taiwan;
| | - Chih-Sung Liang
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei 114, Taiwan;
- Department of Psychiatry, Beitou Branch, Tri-Service General Hospital, National Defense Medical Center, Taipei 112, Taiwan
| | - Chia-Kuang Tsai
- Department of Neurology, Tri-Service General Hospital, National Defense Medical Center, Taipei 114, Taiwan; (Y.-K.L.); (C.-K.T.); (C.-L.T.); (J.-T.L.); (Y.-F.S.); (C.-H.C.); (G.-Y.L.)
| | - Chia-Lin Tsai
- Department of Neurology, Tri-Service General Hospital, National Defense Medical Center, Taipei 114, Taiwan; (Y.-K.L.); (C.-K.T.); (C.-L.T.); (J.-T.L.); (Y.-F.S.); (C.-H.C.); (G.-Y.L.)
| | - Jiunn-Tay Lee
- Department of Neurology, Tri-Service General Hospital, National Defense Medical Center, Taipei 114, Taiwan; (Y.-K.L.); (C.-K.T.); (C.-L.T.); (J.-T.L.); (Y.-F.S.); (C.-H.C.); (G.-Y.L.)
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei 114, Taiwan;
| | - Yueh-Feng Sung
- Department of Neurology, Tri-Service General Hospital, National Defense Medical Center, Taipei 114, Taiwan; (Y.-K.L.); (C.-K.T.); (C.-L.T.); (J.-T.L.); (Y.-F.S.); (C.-H.C.); (G.-Y.L.)
| | - Chung-Hsing Chou
- Department of Neurology, Tri-Service General Hospital, National Defense Medical Center, Taipei 114, Taiwan; (Y.-K.L.); (C.-K.T.); (C.-L.T.); (J.-T.L.); (Y.-F.S.); (C.-H.C.); (G.-Y.L.)
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei 114, Taiwan;
| | - Hung-Sheng Shang
- Division of Clinical Pathology, Department of Pathology, Tri-Service General Hospital, National Defense Medical Center, Taipei 114, Taiwan; (H.-S.S.); (B.-H.Y.)
| | - Bing-Heng Yang
- Division of Clinical Pathology, Department of Pathology, Tri-Service General Hospital, National Defense Medical Center, Taipei 114, Taiwan; (H.-S.S.); (B.-H.Y.)
| | - Guan-Yu Lin
- Department of Neurology, Tri-Service General Hospital, National Defense Medical Center, Taipei 114, Taiwan; (Y.-K.L.); (C.-K.T.); (C.-L.T.); (J.-T.L.); (Y.-F.S.); (C.-H.C.); (G.-Y.L.)
- Department of Neurology, Songshan Branch, Tri-Service General Hospital, National Defense Medical Center, Taipei 105, Taiwan
| | - Ming-Wei Su
- Institute of Biomedical Sciences, Academia Sinica, Taipei 115, Taiwan;
| | - Fu-Chi Yang
- Department of Neurology, Tri-Service General Hospital, National Defense Medical Center, Taipei 114, Taiwan; (Y.-K.L.); (C.-K.T.); (C.-L.T.); (J.-T.L.); (Y.-F.S.); (C.-H.C.); (G.-Y.L.)
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei 114, Taiwan;
- Correspondence: ; Tel.: +886-2-87923311; Fax: +886-87927174
| |
Collapse
|
20
|
Jyoti Dutta B, Singh S, Seksaria S, Das Gupta G, Bodakhe SH, Singh A. Potential role of IP3/Ca 2+ signaling and phosphodiesterases: Relevance to neurodegeneration in Alzheimer's disease and possible therapeutic strategies. Biochem Pharmacol 2022; 201:115071. [PMID: 35525328 DOI: 10.1016/j.bcp.2022.115071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 04/27/2022] [Accepted: 04/28/2022] [Indexed: 11/02/2022]
Abstract
Despite large investments by industry and governments, no disease-modifying medications for the treatment of patients with Alzheimer's disease (AD) have been found. The failures of various clinical trials indicate the need for a more in-depth understanding of the pathophysiology of AD and for innovative therapeutic strategies for its treatment. Here, we review the rational for targeting IP3 signaling, cytosolic calcium dysregulation, phosphodiesterases (PDEs), and secondary messengers like cGMP and cAMP, as well as their correlations with the pathophysiology of AD. Various drugs targeting these signaling cascades are still in pre-clinical and clinical trials which support the ideas presented in this article. Further, we describe different molecular mechanisms and medications currently being used in various pre-clinical and clinical trials involving IP3/Ca+2 signaling. We also highlight various isoforms, as well as the functions and pharmacology of the PDEs broadly expressed in different parts of the brain and attempt to unravel the potential benefits of PDE inhibitors for use as novel medications to alleviate the pathogenesis of AD.
Collapse
Affiliation(s)
- Bhaskar Jyoti Dutta
- Department of Pharmacology, ISF College of Pharmacy, GT Road, Ghal Kalan, Moga-142001, Punjab, India
| | - Shamsher Singh
- Department of Pharmacology, ISF College of Pharmacy, GT Road, Ghal Kalan, Moga-142001, Punjab, India
| | - Sanket Seksaria
- Department of Pharmacology, ISF College of Pharmacy, GT Road, Ghal Kalan, Moga-142001, Punjab, India
| | - Ghanshyam Das Gupta
- Department of Pharmacology, ISF College of Pharmacy, GT Road, Ghal Kalan, Moga-142001, Punjab, India
| | - Surendra H Bodakhe
- Department of Pharmacy, Guru Ghasidas Vishwavidyalaya (A Central University), Bilaspur - 495009, Chhattisgarh, India
| | - Amrita Singh
- Department of Pharmacology, ISF College of Pharmacy, GT Road, Ghal Kalan, Moga-142001, Punjab, India.
| |
Collapse
|
21
|
Thapak P, Khare P, Bishnoi M, Sharma SS. Neuroprotective Effect of 2-Aminoethoxydiphenyl Borate (2-APB) in Amyloid β-Induced Memory Dysfunction: A Mechanistic Study. Cell Mol Neurobiol 2022; 42:1211-1223. [PMID: 33219878 PMCID: PMC11441215 DOI: 10.1007/s10571-020-01012-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Accepted: 11/13/2020] [Indexed: 02/08/2023]
Abstract
β-Amyloid (Aβ) peptide is a characteristic feature of Alzheimer's disease (AD) and accumulation of Aβ is associated with loss of synaptic plasticity and neuronal cell death. Aggregation of Aβ initiates numerous molecular signalling pathways leading to oxidative stress, mitochondrial dysfunction as well as an imbalance of calcium ion influx homeostasis. Recently, it has been shown that transient receptor potential melastatin 2 (TRPM2), a non-selective calcium-permeable cation channel has been postulated to play a vital role in the neuronal death, indicating the potential of TRPM2 inhibition in CNS disease. In this study, neuroprotective potential of 2-aminoethoxydiphenyl borate (2-APB), a broad-spectrum calcium channels blocker was investigated in Aβ-induced memory deficits in rats. In addition, effect of 2-APB on TRPM2 channels gene and protein expressions and also on calcium and memory related proteins was investigated in the hippocampus. Intracerebroventricular (I.C.V.) administration of Aβ (Aβ25-35, 10 μg) markedly induced cognitive impairment and upregulation of mRNA and protein expression of TRPM2 in the hippocampus. In addition, AChE activity was also increased in the cortex of the Aβ administered animals. Three-week treatment with 2-APB led to the down-regulation of TRPM2 mRNA and protein expression in the hippocampus and also improved the cognitive functions which was evident from the behavioral parameters. Moreover, 2-APB treatment also increased the calcium and memory associated proteins namely p-CaMKII, p-GSK-3β, p-CREB and PSD-95 in the hippocampus and reduced the mRNA level of calcium buffering proteins and calcineurin A (PPP3CA) in the hippocampus. Furthermore, 2-APB treatment significantly reduced the AChE activity in the cortex. Thus, our findings suggest the neuroprotective effect of 2-APB in Aβ-induced cognitive impairment.
Collapse
Affiliation(s)
- Pavan Thapak
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), S.A.S. Nagar, Mohali, Punjab, 160062, India
| | - Pragyanshu Khare
- National Agri-Food Biotechnology Institute (NABI), S.A.S. Nagar, Mohali, Punjab, India
| | - Mahendra Bishnoi
- National Agri-Food Biotechnology Institute (NABI), S.A.S. Nagar, Mohali, Punjab, India
| | - Shyam Sunder Sharma
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), S.A.S. Nagar, Mohali, Punjab, 160062, India.
| |
Collapse
|
22
|
WEI HF, ANCHIPOLOVSKY S, VERA R, LIANG G, CHUANG DM. Potential mechanisms underlying lithium treatment for Alzheimer's disease and COVID-19. EUROPEAN REVIEW FOR MEDICAL AND PHARMACOLOGICAL SCIENCES 2022; 26:2201-2214. [PMID: 35363371 PMCID: PMC9173589 DOI: 10.26355/eurrev_202203_28369] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 02/11/2023]
Abstract
Disruption of intracellular Ca2+ homeostasis plays an important role as an upstream pathology in Alzheimer's disease (AD), and correction of Ca2+ dysregulation has been increasingly proposed as a target of future effective disease-modified drugs for treating AD. Calcium dysregulation is also an upstream pathology for the COVID-19 virus SARS-CoV-2 infection and replication, leading to host cell damage. Clinically available drugs that can inhibit the disturbed intracellular Ca2+ homeostasis have been repurposed to treat COVID-19 patients. This narrative review aims at exploring the underlying mechanism by which lithium, a first line drug for the treatment of bipolar disorder, inhibits Ca2+ dysregulation and associated downstream pathology in both AD and COVID-19. It is suggested that lithium can be repurposed to treat AD patients, especially those afflicted with COVID-19.
Collapse
Affiliation(s)
- H.-F. WEI
- Department of Anaesthesiology and Critical Care, University of Pennsylvania, Philadelphia, PA, USA
| | - S. ANCHIPOLOVSKY
- Department of Anaesthesiology and Critical Care, University of Pennsylvania, Philadelphia, PA, USA
| | - R. VERA
- Department of Anaesthesiology and Critical Care, University of Pennsylvania, Philadelphia, PA, USA
| | - G. LIANG
- Department of Anaesthesiology and Critical Care, University of Pennsylvania, Philadelphia, PA, USA
| | - D.-M. CHUANG
- Intramural Research Program, National Institute of Mental Health, NIH, Bethesda, MD, USA
| |
Collapse
|
23
|
Kieliszek M, Bano I, Zare H. A Comprehensive Review on Selenium and Its Effects on Human Health and Distribution in Middle Eastern Countries. Biol Trace Elem Res 2022; 200:971-987. [PMID: 33884538 PMCID: PMC8761138 DOI: 10.1007/s12011-021-02716-z] [Citation(s) in RCA: 74] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Accepted: 04/08/2021] [Indexed: 12/16/2022]
Abstract
Selenium (Se) is an important microelement with numerous positive effects on human health and diseases. It is important to specify that the status and consumption of Se are for a specific community as the levels of Se are extremely unpredictable between different populations and regions. Our existing paper was based on the impacts of Se on human health and disease along with data on the Se levels in Middle Eastern countries. Overall, the findings of this comprehensive review show that the consumption and levels of Se are inadequate in Middle Eastern nations. Such findings, together with the growing awareness of the importance of Se to general health, require further work primarily on creating an acceptable range of blood Se concentration or other measures to determine optimal Se consumption and, consequently, to guarantee adequate Se supplementation in populations at high risk of low Se intake.
Collapse
Affiliation(s)
- Marek Kieliszek
- Department of Food Biotechnology and Microbiology, Institute of Food Sciences, Warsaw University of Life Sciences-SGGW, Nowoursynowska 159C, 02-776 Warsaw, Poland
| | - Iqra Bano
- Department of Veterinary Physiology and Biochemistry, Shaheed Benazir Bhutto University of Veterinary & Animal Sciences Sakrand, Sindh, 67210 Pakistan
| | - Hamed Zare
- Cellular and Molecular Research Center, Birjand University of Medical Sciences, Birjand, Iran
| |
Collapse
|
24
|
Iafusco D, Zanfardino A, Piscopo A, Curto S, Troncone A, Chianese A, Rollato AS, Testa V, Iafusco F, Maione G, Pennarella A, Boccabella L, Ozen G, Palma PL, Mazzaccara C, Tinto N, Miraglia del Giudice E. Metabolic Treatment of Wolfram Syndrome. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:2755. [PMID: 35270448 PMCID: PMC8910219 DOI: 10.3390/ijerph19052755] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 02/14/2022] [Accepted: 02/21/2022] [Indexed: 12/10/2022]
Abstract
Wolfram Syndrome (WS) is a very rare genetic disorder characterized by several symptoms that occur from childhood to adulthood. Usually, the first clinical sign is non-autoimmune diabetes even if other clinical features (optic subatrophy, neurosensorial deafness, diabetes insipidus) may be present in an early state and may be diagnosed after diabetes' onset. Prognosis is poor, and the death occurs at the median age of 39 years as a consequence of progressive respiratory impairment, secondary to brain atrophy and neurological failure. The aim of this paper is the description of the metabolic treatment of the WS. We reported the experience of long treatment in patients with this syndrome diagnosed in pediatric age and followed also in adult age. It is known that there is a correlation between metabolic control of diabetes, the onset of other associated symptoms, and the progression of the neurodegenerative alterations. Therefore, a multidisciplinary approach is necessary in order to prevent, treat and carefully monitor all the comorbidities that may occur. An extensive understanding of WS from pathophysiology to novel possible therapy is fundamental and further studies are needed to better manage this devastating disease and to guarantee to patients a better quality of life and a longer life expectancy.
Collapse
Affiliation(s)
- Dario Iafusco
- Regional Center of Pediatric Diabetology “G.Stoppoloni”, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (A.Z.); (A.P.); (S.C.); (A.T.); (A.C.); (A.S.R.); (V.T.); (A.P.); (L.B.); (G.O.); (P.L.P.); (E.M.d.G.)
| | - Angela Zanfardino
- Regional Center of Pediatric Diabetology “G.Stoppoloni”, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (A.Z.); (A.P.); (S.C.); (A.T.); (A.C.); (A.S.R.); (V.T.); (A.P.); (L.B.); (G.O.); (P.L.P.); (E.M.d.G.)
| | - Alessia Piscopo
- Regional Center of Pediatric Diabetology “G.Stoppoloni”, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (A.Z.); (A.P.); (S.C.); (A.T.); (A.C.); (A.S.R.); (V.T.); (A.P.); (L.B.); (G.O.); (P.L.P.); (E.M.d.G.)
| | - Stefano Curto
- Regional Center of Pediatric Diabetology “G.Stoppoloni”, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (A.Z.); (A.P.); (S.C.); (A.T.); (A.C.); (A.S.R.); (V.T.); (A.P.); (L.B.); (G.O.); (P.L.P.); (E.M.d.G.)
| | - Alda Troncone
- Regional Center of Pediatric Diabetology “G.Stoppoloni”, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (A.Z.); (A.P.); (S.C.); (A.T.); (A.C.); (A.S.R.); (V.T.); (A.P.); (L.B.); (G.O.); (P.L.P.); (E.M.d.G.)
| | - Antonietta Chianese
- Regional Center of Pediatric Diabetology “G.Stoppoloni”, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (A.Z.); (A.P.); (S.C.); (A.T.); (A.C.); (A.S.R.); (V.T.); (A.P.); (L.B.); (G.O.); (P.L.P.); (E.M.d.G.)
| | - Assunta Serena Rollato
- Regional Center of Pediatric Diabetology “G.Stoppoloni”, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (A.Z.); (A.P.); (S.C.); (A.T.); (A.C.); (A.S.R.); (V.T.); (A.P.); (L.B.); (G.O.); (P.L.P.); (E.M.d.G.)
| | - Veronica Testa
- Regional Center of Pediatric Diabetology “G.Stoppoloni”, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (A.Z.); (A.P.); (S.C.); (A.T.); (A.C.); (A.S.R.); (V.T.); (A.P.); (L.B.); (G.O.); (P.L.P.); (E.M.d.G.)
| | - Fernanda Iafusco
- Department of Molecular Medicine and Medical Biotechnology, University of Naples “Federico II”, 80131 Naples, Italy; (F.I.); (G.M.); (C.M.); (N.T.)
- CEINGE Advanced Biotechnologies, 80131 Naples, Italy
| | - Giovanna Maione
- Department of Molecular Medicine and Medical Biotechnology, University of Naples “Federico II”, 80131 Naples, Italy; (F.I.); (G.M.); (C.M.); (N.T.)
- CEINGE Advanced Biotechnologies, 80131 Naples, Italy
| | - Alessandro Pennarella
- Regional Center of Pediatric Diabetology “G.Stoppoloni”, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (A.Z.); (A.P.); (S.C.); (A.T.); (A.C.); (A.S.R.); (V.T.); (A.P.); (L.B.); (G.O.); (P.L.P.); (E.M.d.G.)
| | - Lucia Boccabella
- Regional Center of Pediatric Diabetology “G.Stoppoloni”, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (A.Z.); (A.P.); (S.C.); (A.T.); (A.C.); (A.S.R.); (V.T.); (A.P.); (L.B.); (G.O.); (P.L.P.); (E.M.d.G.)
| | - Gulsum Ozen
- Regional Center of Pediatric Diabetology “G.Stoppoloni”, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (A.Z.); (A.P.); (S.C.); (A.T.); (A.C.); (A.S.R.); (V.T.); (A.P.); (L.B.); (G.O.); (P.L.P.); (E.M.d.G.)
| | - Pier Luigi Palma
- Regional Center of Pediatric Diabetology “G.Stoppoloni”, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (A.Z.); (A.P.); (S.C.); (A.T.); (A.C.); (A.S.R.); (V.T.); (A.P.); (L.B.); (G.O.); (P.L.P.); (E.M.d.G.)
| | - Cristina Mazzaccara
- Department of Molecular Medicine and Medical Biotechnology, University of Naples “Federico II”, 80131 Naples, Italy; (F.I.); (G.M.); (C.M.); (N.T.)
- CEINGE Advanced Biotechnologies, 80131 Naples, Italy
| | - Nadia Tinto
- Department of Molecular Medicine and Medical Biotechnology, University of Naples “Federico II”, 80131 Naples, Italy; (F.I.); (G.M.); (C.M.); (N.T.)
- CEINGE Advanced Biotechnologies, 80131 Naples, Italy
| | - Emanuele Miraglia del Giudice
- Regional Center of Pediatric Diabetology “G.Stoppoloni”, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (A.Z.); (A.P.); (S.C.); (A.T.); (A.C.); (A.S.R.); (V.T.); (A.P.); (L.B.); (G.O.); (P.L.P.); (E.M.d.G.)
| |
Collapse
|
25
|
Yan L, Guo MS, Zhang Y, Yu L, Wu JM, Tang Y, Ai W, Zhu FD, Law BYK, Chen Q, Yu CL, Wong VKW, Li H, Li M, Zhou XG, Qin DL, Wu AG. Dietary Plant Polyphenols as the Potential Drugs in Neurodegenerative Diseases: Current Evidence, Advances, and Opportunities. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:5288698. [PMID: 35237381 PMCID: PMC8885204 DOI: 10.1155/2022/5288698] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 01/10/2022] [Accepted: 01/28/2022] [Indexed: 02/07/2023]
Abstract
Neurodegenerative diseases, including Alzheimer's disease (AD), Parkinson's disease (PD), and Huntington's disease (HD), are characterized by the progressive degeneration of neurons. Although the etiology and pathogenesis of neurodegenerative diseases have been studied intensively, the mechanism is still in its infancy. In general, most neurodegenerative diseases share common molecular mechanisms, and multiple risks interact and promote the pathologic process of neurogenerative diseases. At present, most of the approved drugs only alleviate the clinical symptoms but fail to cure neurodegenerative diseases. Numerous studies indicate that dietary plant polyphenols are safe and exhibit potent neuroprotective effects in various neurodegenerative diseases. However, low bioavailability is the biggest obstacle for polyphenol that largely limits its adoption from evidence into clinical practice. In this review, we summarized the widely recognized mechanisms associated with neurodegenerative diseases, such as misfolded proteins, mitochondrial dysfunction, oxidative damage, and neuroinflammatory responses. In addition, we summarized the research advances about the neuroprotective effect of the most widely reported dietary plant polyphenols. Moreover, we discussed the current clinical study and application of polyphenols and the factors that result in low bioavailability, such as poor stability and low permeability across the blood-brain barrier (BBB). In the future, the improvement of absorption and stability, modification of structure and formulation, and the combination therapy will provide more opportunities from the laboratory into the clinic for polyphenols. Lastly, we hope that the present review will encourage further researches on natural dietary polyphenols in the treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Lu Yan
- Sichuan Key Medical Laboratory of New Drug Discovery and Druggability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, School of Pharmacy; Education Ministry Key Laboratory of Medical Electrophysiology, College of Preclinical Medicine, Southwest Medical University, Luzhou 646000, China
| | - Min-Song Guo
- Sichuan Key Medical Laboratory of New Drug Discovery and Druggability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, School of Pharmacy; Education Ministry Key Laboratory of Medical Electrophysiology, College of Preclinical Medicine, Southwest Medical University, Luzhou 646000, China
| | - Yue Zhang
- Sichuan Key Medical Laboratory of New Drug Discovery and Druggability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, School of Pharmacy; Education Ministry Key Laboratory of Medical Electrophysiology, College of Preclinical Medicine, Southwest Medical University, Luzhou 646000, China
| | - Lu Yu
- Sichuan Key Medical Laboratory of New Drug Discovery and Druggability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, School of Pharmacy; Education Ministry Key Laboratory of Medical Electrophysiology, College of Preclinical Medicine, Southwest Medical University, Luzhou 646000, China
| | - Jian-Ming Wu
- Sichuan Key Medical Laboratory of New Drug Discovery and Druggability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, School of Pharmacy; Education Ministry Key Laboratory of Medical Electrophysiology, College of Preclinical Medicine, Southwest Medical University, Luzhou 646000, China
| | - Yong Tang
- Sichuan Key Medical Laboratory of New Drug Discovery and Druggability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, School of Pharmacy; Education Ministry Key Laboratory of Medical Electrophysiology, College of Preclinical Medicine, Southwest Medical University, Luzhou 646000, China
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, Macau SAR, China
| | - Wei Ai
- Sichuan Key Medical Laboratory of New Drug Discovery and Druggability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, School of Pharmacy; Education Ministry Key Laboratory of Medical Electrophysiology, College of Preclinical Medicine, Southwest Medical University, Luzhou 646000, China
| | - Feng-Dan Zhu
- Sichuan Key Medical Laboratory of New Drug Discovery and Druggability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, School of Pharmacy; Education Ministry Key Laboratory of Medical Electrophysiology, College of Preclinical Medicine, Southwest Medical University, Luzhou 646000, China
| | - Betty Yuen-Kwan Law
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, Macau SAR, China
| | - Qi Chen
- Sichuan Key Medical Laboratory of New Drug Discovery and Druggability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, School of Pharmacy; Education Ministry Key Laboratory of Medical Electrophysiology, College of Preclinical Medicine, Southwest Medical University, Luzhou 646000, China
- Department of Nursing, Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Chong-Lin Yu
- Sichuan Key Medical Laboratory of New Drug Discovery and Druggability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, School of Pharmacy; Education Ministry Key Laboratory of Medical Electrophysiology, College of Preclinical Medicine, Southwest Medical University, Luzhou 646000, China
| | - Vincent Kam-Wai Wong
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, Macau SAR, China
| | - Hua Li
- Sichuan Key Medical Laboratory of New Drug Discovery and Druggability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, School of Pharmacy; Education Ministry Key Laboratory of Medical Electrophysiology, College of Preclinical Medicine, Southwest Medical University, Luzhou 646000, China
| | - Mao Li
- Sichuan Key Medical Laboratory of New Drug Discovery and Druggability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, School of Pharmacy; Education Ministry Key Laboratory of Medical Electrophysiology, College of Preclinical Medicine, Southwest Medical University, Luzhou 646000, China
| | - Xiao-Gang Zhou
- Sichuan Key Medical Laboratory of New Drug Discovery and Druggability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, School of Pharmacy; Education Ministry Key Laboratory of Medical Electrophysiology, College of Preclinical Medicine, Southwest Medical University, Luzhou 646000, China
| | - Da-Lian Qin
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, Macau SAR, China
| | - An-Guo Wu
- Sichuan Key Medical Laboratory of New Drug Discovery and Druggability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, School of Pharmacy; Education Ministry Key Laboratory of Medical Electrophysiology, College of Preclinical Medicine, Southwest Medical University, Luzhou 646000, China
| |
Collapse
|
26
|
Tang AS, Oskotsky T, Havaldar S, Mantyh WG, Bicak M, Solsberg CW, Woldemariam S, Zeng B, Hu Z, Oskotsky B, Dubal D, Allen IE, Glicksberg BS, Sirota M. Deep phenotyping of Alzheimer's disease leveraging electronic medical records identifies sex-specific clinical associations. Nat Commun 2022; 13:675. [PMID: 35115528 PMCID: PMC8814236 DOI: 10.1038/s41467-022-28273-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Accepted: 01/18/2022] [Indexed: 12/14/2022] Open
Abstract
Alzheimer's Disease (AD) is a neurodegenerative disorder that is still not fully understood. Sex modifies AD vulnerability, but the reasons for this are largely unknown. We utilize two independent electronic medical record (EMR) systems across 44,288 patients to perform deep clinical phenotyping and network analysis to gain insight into clinical characteristics and sex-specific clinical associations in AD. Embeddings and network representation of patient diagnoses demonstrate greater comorbidity interactions in AD in comparison to matched controls. Enrichment analysis identifies multiple known and new diagnostic, medication, and lab result associations across the whole cohort and in a sex-stratified analysis. With this data-driven method of phenotyping, we can represent AD complexity and generate hypotheses of clinical factors that can be followed-up for further diagnostic and predictive analyses, mechanistic understanding, or drug repurposing and therapeutic approaches.
Collapse
Affiliation(s)
- Alice S Tang
- Bakar Computational Health Sciences Institute, UCSF, San Francisco, CA, USA.
- Graduate Program in Bioengineering, UCSF, San Francisco, CA, USA.
- School of Medicine, UCSF, San Francisco, CA, USA.
| | - Tomiko Oskotsky
- Bakar Computational Health Sciences Institute, UCSF, San Francisco, CA, USA
- Department of Pediatrics, UCSF, San Francisco, CA, USA
| | - Shreyas Havaldar
- Hasso Plattner Institute for Digital Health at Mount Sinai, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - William G Mantyh
- Department of Neurology, University of Minnesota School of Medicine, Minneapolis, MN, USA
| | - Mesude Bicak
- Hasso Plattner Institute for Digital Health at Mount Sinai, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Caroline Warly Solsberg
- Pharmaceutical Sciences and Pharmacogenomics, UCSF, San Francisco, CA, USA
- Department of Neurology and Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, 94158, USA
- Memory and Aging Center, UCSF, San Francisco, CA, USA
| | - Sarah Woldemariam
- Bakar Computational Health Sciences Institute, UCSF, San Francisco, CA, USA
| | - Billy Zeng
- School of Medicine, UCSF, San Francisco, CA, USA
| | - Zicheng Hu
- Bakar Computational Health Sciences Institute, UCSF, San Francisco, CA, USA
| | - Boris Oskotsky
- Bakar Computational Health Sciences Institute, UCSF, San Francisco, CA, USA
| | - Dena Dubal
- Department of Neurology and Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, 94158, USA
| | - Isabel E Allen
- Department of Epidemiology and Biostatistics, UCSF, San Francisco, CA, USA
| | - Benjamin S Glicksberg
- Hasso Plattner Institute for Digital Health at Mount Sinai, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Marina Sirota
- Bakar Computational Health Sciences Institute, UCSF, San Francisco, CA, USA.
- Department of Pediatrics, UCSF, San Francisco, CA, USA.
| |
Collapse
|
27
|
Tatulian SA. Challenges and hopes for Alzheimer's disease. Drug Discov Today 2022; 27:1027-1043. [PMID: 35121174 DOI: 10.1016/j.drudis.2022.01.016] [Citation(s) in RCA: 146] [Impact Index Per Article: 48.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 09/01/2021] [Accepted: 01/27/2022] [Indexed: 12/11/2022]
Abstract
Recent drug development efforts targeting Alzheimer's disease (AD) have failed to produce effective disease-modifying agents for many reasons, including the substantial presymptomatic neuronal damage that is caused by the accumulation of the amyloid β (Aβ) peptide and tau protein abnormalities, deleterious adverse effects of drug candidates, and inadequate design of clinical trials. New molecular targets, biomarkers, and diagnostic techniques, as well as alternative nonpharmacological approaches, are sorely needed to detect and treat early pathological events. This article analyzes the successes and debacles of pharmaceutical endeavors to date, and highlights new technologies that may lead to the more effective diagnosis and treatment of the pathologies that underlie AD. The use of focused ultrasound, deep brain stimulation, stem cell therapy, and gene therapy, in parallel with pharmaceuticals and judicious lifestyle adjustments, holds promise for the deceleration, prevention, or cure of AD and other neurodegenerative disorders.
Collapse
Affiliation(s)
- Suren A Tatulian
- Department of Physics, College of Sciences, and Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32816, USA.
| |
Collapse
|
28
|
Phuna ZX, Madhavan P. A CLOSER LOOK AT THE MYCOBIOME IN ALZHEIMER'S DISEASE: FUNGAL SPECIES, PATHOGENESIS AND TRANSMISSION. Eur J Neurosci 2022; 55:1291-1321. [DOI: 10.1111/ejn.15599] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 01/17/2022] [Accepted: 01/18/2022] [Indexed: 11/30/2022]
Affiliation(s)
- Zhi Xin Phuna
- School of Medicine, Faculty of Health & Medical Sciences, Taylor’s University Malaysia Subang Jaya Selangor
| | - Priya Madhavan
- School of Medicine, Faculty of Health & Medical Sciences, Taylor’s University Malaysia Subang Jaya Selangor
| |
Collapse
|
29
|
Walia V, Kaushik D, Mittal V, Kumar K, Verma R, Parashar J, Akter R, Rahman MH, Bhatia S, Al-Harrasi A, Karthika C, Bhattacharya T, Chopra H, Ashraf GM. Delineation of Neuroprotective Effects and Possible Benefits of AntioxidantsTherapy for the Treatment of Alzheimer's Diseases by Targeting Mitochondrial-Derived Reactive Oxygen Species: Bench to Bedside. Mol Neurobiol 2021; 59:657-680. [PMID: 34751889 DOI: 10.1007/s12035-021-02617-1] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Accepted: 10/19/2021] [Indexed: 12/25/2022]
Abstract
Alzheimer's disease (AD) is considered the sixth leading cause of death in elderly patients and is characterized by progressive neuronal degeneration and impairment in memory, language, etc. AD is characterized by the deposition of senile plaque, accumulation of fibrils, and neurofibrillary tangles (NFTs) which are responsible for neuronal degeneration. Amyloid-β (Aβ) plays a key role in the process of neuronal degeneration in the case of AD. It has been reported that Aβ is responsible for the production of reactive oxygen species (ROS), depletion of endogenous antioxidants, increase in intracellular Ca2+ which further increases mitochondria dysfunctions, oxidative stress, release of pro-apoptotic factors, neuronal apoptosis, etc. Thus, oxidative stress plays a key role in the pathogenesis of AD. Antioxidants are compounds that have the ability to counteract the oxidative damage conferred by ROS. Therefore, the antioxidant therapy may provide benefits and halt the progress of AD to advance stages by counteracting neuronal degeneration. However, despite the beneficial effects imposed by the antioxidants, the findings from the clinical studies suggested inconsistent results which might be due to poor study design, selection of the wrong antioxidant, inability of the molecule to cross the blood-brain barrier (BBB), treatment in the advanced state of disease, etc. The present review insights into the neuroprotective effects and limitations of the antioxidant therapy for the treatment of AD by targeting mitochondrial-derived ROS. This particular article will certainly help the researchers to search new avenues for the treatment of AD by utilizing mitochondrial-derived ROS-targeted antioxidant therapies.
Collapse
Affiliation(s)
- Vaibhav Walia
- SGT College of Pharmacy, SGT University, Gurugram, Haryana, India
| | - Deepak Kaushik
- Department of Pharmaceutical Sciences, Maharshi Dayanand University, Rohtak, 124001, India
| | - Vineet Mittal
- Department of Pharmaceutical Sciences, Maharshi Dayanand University, Rohtak, 124001, India
| | - Kuldeep Kumar
- Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, Punjab, India
- University Institute of Pharmaceutical Sciences (UIPS), Chandigarh University, Gharuan, Mohali, Punjab, India
| | - Ravinder Verma
- Department of Pharmacy, School of Medical and Allied Sciences, G.D. Goenka University, Gurugram, 122103, India
| | - Jatin Parashar
- Department of Pharmaceutical Sciences, Maharshi Dayanand University, Rohtak, 124001, India
| | - Rokeya Akter
- Department of Pharmacy, Jagannath University, Sadarghat, Dhaka, 1100, Bangladesh
| | - Md Habibur Rahman
- Department of Pharmacy, Southeast University, Banani, Dhaka, 1213, Bangladesh.
| | - Saurabh Bhatia
- School of Health Science University of Petroleum and Energy Studies, Dehrandun, Uttarkhand, 248007, India
- Natural & Medical Sciences Research Center, University of Nizwa, 616 Birkat Al Mouz, P.O. Box 33, Nizwa, Oman
| | - Ahmed Al-Harrasi
- Natural & Medical Sciences Research Center, University of Nizwa, 616 Birkat Al Mouz, P.O. Box 33, Nizwa, Oman
| | - Chenmala Karthika
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education & Research, The Nilgiris, Ooty, 643001, Tamil Nadu, India
| | - Tanima Bhattacharya
- College of Chemistry & Chemical Engineering, Hubei University, Wuhan, 430062, China
| | - Hitesh Chopra
- Chitkara College of Pharmacy, Chitkara University, Punjab, 140401, India
| | - Ghulam Md Ashraf
- Pre-Clinical Research Unit, 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
|
30
|
Effects of Aβ-derived peptide fragments on fibrillogenesis of Aβ. Sci Rep 2021; 11:19262. [PMID: 34584131 PMCID: PMC8479085 DOI: 10.1038/s41598-021-98644-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Accepted: 09/13/2021] [Indexed: 11/08/2022] Open
Abstract
Amyloid β (Aβ) peptide aggregation plays a central role in Alzheimer's disease (AD) etiology. AD drug candidates have included small molecules or peptides directed towards inhibition of Aβ fibrillogenesis. Although some Aβ-derived peptide fragments suppress Aβ fibril growth, comprehensive analysis of inhibitory potencies of peptide fragments along the whole Aβ sequence has not been reported. The aim of this work is (a) to identify the region(s) of Aβ with highest propensities for aggregation and (b) to use those fragments to inhibit Aβ fibrillogenesis. Structural and aggregation properties of the parent Aβ1-42 peptide and seven overlapping peptide fragments have been studied, i.e. Aβ1-10 (P1), Aβ6-15 (P2), Aβ11-20 (P3), Aβ16-25 (P4), Aβ21-30 (P5), Aβ26-36 (P6), and Aβ31-42 (P7). Structural transitions of the peptides in aqueous buffer have been monitored by circular dichroism and Fourier transform infrared spectroscopy. Aggregation and fibrillogenesis were analyzed by light scattering and thioflavin-T fluorescence. The mode of peptide-peptide interactions was characterized by fluorescence resonance energy transfer. Three peptide fragments, P3, P6, and P7, exhibited exceptionally high propensity for β-sheet formation and aggregation. Remarkably, only P3 and P6 exerted strong inhibitory effect on the aggregation of Aβ1-42, whereas P7 and P2 displayed moderate inhibitory potency. It is proposed that P3 and P6 intercalate between Aβ1-42 molecules and thereby inhibit Aβ1-42 aggregation. These findings may facilitate therapeutic strategies of inhibition of Aβ fibrillogenesis by Aβ-derived peptides.
Collapse
|
31
|
The Beneficial Role of Natural Endocrine Disruptors: Phytoestrogens in Alzheimer's Disease. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:3961445. [PMID: 34527172 PMCID: PMC8437597 DOI: 10.1155/2021/3961445] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Accepted: 08/17/2021] [Indexed: 01/14/2023]
Abstract
Alzheimer's disease (AD) is the most common form of dementia with a growing incidence rate primarily among the elderly. It is a neurodegenerative, progressive disorder leading to significant cognitive loss. Despite numerous pieces of research, no cure for halting the disease has been discovered yet. Phytoestrogens are nonestradiol compounds classified as one of the endocrine-disrupting chemicals (EDCs), meaning that they can potentially disrupt hormonal balance and result in developmental and reproductive abnormalities. Importantly, phytoestrogens are structurally, chemically, and functionally akin to estrogens, which undoubtedly has the potential to be detrimental to the organism. What is intriguing, although classified as EDCs, phytoestrogens seem to have a beneficial influence on Alzheimer's disease symptoms and neuropathologies. They have been observed to act as antioxidants, improve visual-spatial memory, lower amyloid-beta production, and increase the growth, survival, and plasticity of brain cells. This review article is aimed at contributing to the collective understanding of the role of phytoestrogens in the prevention and treatment of Alzheimer's disease. Importantly, it underlines the fact that despite being EDCs, phytoestrogens and their use can be beneficial in the prevention of Alzheimer's disease.
Collapse
|
32
|
Shi Y, Zhang L, Gao X, Zhang J, Ben Abou M, Liang G, Meng Q, Hepner A, Eckenhoff MF, Wei H. Intranasal Dantrolene as a Disease-Modifying Drug in Alzheimer 5XFAD Mice. J Alzheimers Dis 2021; 76:1375-1389. [PMID: 32623395 PMCID: PMC7505009 DOI: 10.3233/jad-200227] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Background/Objective: This study compares the effectiveness and safety of intranasal versus subcutaneous administration of dantrolene in 5XFAD Alzheimer’s disease (AD) mice. Methods: 5XFAD and wild type (WT) B6SJLF1/J mice were treated with intranasal or subcutaneous dantrolene (5 mg/kg, 3×/wk), or vehicle. The early (ETG) and late (LTG) treatment groups began treatment at 2 or 6 months of age, respectively, and both treatment groups finished at12 months of age. Behavior was assessed for olfaction (buried food test), motor function (rotarod), and cognition (fear conditioning, Morris water maze). Liver histology (H & E staining) and function, synaptic proteins, and brain amyloid immunohistochemistry were examined. Plasma and brain dantrolene concentrations were determined in a separate cohort after intranasal or subcutaneous administration. Results: Intranasal dantrolene achieved higher brain and lower plasma concentrations than subcutaneous administration. Dantrolene administration at both approaches significantly improved hippocampal-dependent and -independent memory in the ETG, whereas only intranasal dantrolene improved cognition in the LTG. Dantrolene treatment had no significant change in the amyloid burden or synaptic proteins and no significant side effects on mortality, olfaction, motor, or liver functions in 5XFAD mice. Intranasal dantrolene treatment significantly ameliorated memory loss when it was started either before or after the onset of AD symptoms in 5XFAD mice. Conclusions: The long-term intranasal administration of dantrolene had therapeutic effects on memory compared to the subcutaneous approach even started after onset of AD symptoms, suggesting use as a disease-modifying drug, without significant effects on amyloid plaques, side effects, or mortality.
Collapse
Affiliation(s)
- Yun Shi
- Department of Anesthesiology and Critical Care, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.,Department of Anesthesiology, Children's Hospital of Fudan University, Shanghai, China
| | - Lei Zhang
- Department of Anesthesiology and Critical Care, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.,Department of Anesthesiology, People's Hospital of Beijing Daxing District, Beijing, China
| | - Xue Gao
- Department of Anesthesiology and Critical Care, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.,Department of Anesthesiology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Jing Zhang
- Department of Anesthesiology and Critical Care, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.,Department of Anesthesiology, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Matan Ben Abou
- Department of Anesthesiology and Critical Care, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ge Liang
- Department of Anesthesiology and Critical Care, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Qingcheng Meng
- Department of Anesthesiology and Critical Care, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Adrian Hepner
- Eagle Pharmaceuticals, Inc., Woodcliff Lake, NJ, USA
| | - Maryellen F Eckenhoff
- Department of Anesthesiology and Critical Care, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Huafeng Wei
- Department of Anesthesiology and Critical Care, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
33
|
Cheraghzadeh M, Nazeri Z, Mohammadi A, Azizidoost S, Aberomand M, Kheirollah A. Amyloid Beta sharply increases HMG-CoA reductase protein levels in astrocytes isolated from C57BL/6 mice. GENE REPORTS 2021. [DOI: 10.1016/j.genrep.2021.101070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
34
|
Cascella R, Cecchi C. Calcium Dyshomeostasis in Alzheimer's Disease Pathogenesis. Int J Mol Sci 2021; 22:ijms22094914. [PMID: 34066371 PMCID: PMC8124842 DOI: 10.3390/ijms22094914] [Citation(s) in RCA: 79] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 04/26/2021] [Accepted: 04/30/2021] [Indexed: 01/12/2023] Open
Abstract
Alzheimer’s disease (AD) is the most common age-related neurodegenerative disorder that is characterized by amyloid β-protein deposition in senile plaques, neurofibrillary tangles consisting of abnormally phosphorylated tau protein, and neuronal loss leading to cognitive decline and dementia. Despite extensive research, the exact mechanisms underlying AD remain unknown and effective treatment is not available. Many hypotheses have been proposed to explain AD pathophysiology; however, there is general consensus that the abnormal aggregation of the amyloid β peptide (Aβ) is the initial event triggering a pathogenic cascade of degenerating events in cholinergic neurons. The dysregulation of calcium homeostasis has been studied considerably to clarify the mechanisms of neurodegeneration induced by Aβ. Intracellular calcium acts as a second messenger and plays a key role in the regulation of neuronal functions, such as neural growth and differentiation, action potential, and synaptic plasticity. The calcium hypothesis of AD posits that activation of the amyloidogenic pathway affects neuronal Ca2+ homeostasis and the mechanisms responsible for learning and memory. Aβ can disrupt Ca2+ signaling through several mechanisms, by increasing the influx of Ca2+ from the extracellular space and by activating its release from intracellular stores. Here, we review the different molecular mechanisms and receptors involved in calcium dysregulation in AD and possible therapeutic strategies for improving the treatment.
Collapse
|
35
|
Garg J, Lakhani A, Dave V. Effects of the Involvement of Calcium Channels on Neuronal Hyperexcitability Related to Alzheimer’s Disease: A Computational Model. NEUROPHYSIOLOGY+ 2021. [DOI: 10.1007/s11062-021-09890-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
36
|
Behl T, Kaur G, Sehgal A, Singh S, Bhatia S, Al-Harrasi A, Zengin G, Bungau SG, Munteanu MA, Brisc MC, Andronie-Cioara FL, Brisc C. Elucidating the Multi-Targeted Role of Nutraceuticals: A Complementary Therapy to Starve Neurodegenerative Diseases. Int J Mol Sci 2021; 22:4045. [PMID: 33919895 PMCID: PMC8070907 DOI: 10.3390/ijms22084045] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 04/09/2021] [Accepted: 04/13/2021] [Indexed: 12/13/2022] Open
Abstract
The mechanisms underlying multifactorial diseases are always complex and challenging. Neurodegenerative disorders (NDs) are common around the globe, posing a critical healthcare issue and financial burden to the country. However, integrative evidence implies some common shared mechanisms and pathways in NDs, which include mitochondrial dysfunction, neuroinflammation, oxidative stress, intracellular calcium overload, protein aggregates, oxidative stress (OS), and neuronal destruction in specific regions of the brain, owing to multifaceted pathologies. The co-existence of these multiple pathways often limits the advantages of available therapies. The nutraceutical-based approach has opened the doors to target these common multifaceted pathways in a slow and more physiological manner to starve the NDs. Peer-reviewed articles were searched via MEDLINE and PubMed published to date for in-depth research and database collection. Considered to be complementary therapy with current clinical management and common drug therapy, the intake of nutraceuticals is considered safe to target multiple mechanisms of action in NDs. The current review summarizes the popular nutraceuticals showing different effects (anti-inflammatory, antioxidant, neuro-protectant, mitochondrial homeostasis, neurogenesis promotion, and autophagy regulation) on vital molecular mechanisms involved in NDs, which can be considered as complementary therapy to first-line treatment. Moreover, owing to its natural source, lower toxicity, therapeutic interventions, biocompatibility, potential nutritional effects, and presence of various anti-oxidative and neuroprotective constituents, the nutraceuticals serve as an attractive option to tackle NDs.
Collapse
Affiliation(s)
- Tapan Behl
- Department of Pharmacology, Chitkara College of Pharmacy, Chitkara University, Chandigarh 160009, India; (T.B.); (G.K.); (A.S.); (S.S.)
| | - Gagandeep Kaur
- Department of Pharmacology, Chitkara College of Pharmacy, Chitkara University, Chandigarh 160009, India; (T.B.); (G.K.); (A.S.); (S.S.)
| | - Aayush Sehgal
- Department of Pharmacology, Chitkara College of Pharmacy, Chitkara University, Chandigarh 160009, India; (T.B.); (G.K.); (A.S.); (S.S.)
| | - Sukhbir Singh
- Department of Pharmacology, Chitkara College of Pharmacy, Chitkara University, Chandigarh 160009, India; (T.B.); (G.K.); (A.S.); (S.S.)
| | - Saurabh Bhatia
- Natural and Medical Sciences Research Center, University of Nizwa, 616 Birkat Al Mauz, P.O. Box 33, Nizwa, Oman; (S.B.); (A.A.-H.)
| | - Ahmed Al-Harrasi
- Natural and Medical Sciences Research Center, University of Nizwa, 616 Birkat Al Mauz, P.O. Box 33, Nizwa, Oman; (S.B.); (A.A.-H.)
| | - Gokhan Zengin
- Department of Biology, Faculty of Science, Selcuk University Campus, Konya 42130, Turkey;
| | - Simona Gabriela Bungau
- Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, 410028 Oradea, Romania
| | - Mihai Alexandru Munteanu
- Department of Medical Disciplines, Faculty of Medicine and Pharmacy, University of Oradea, 410073 Oradea, Romania; (M.A.M.); (M.C.B.); (C.B.)
| | - Mihaela Cristina Brisc
- Department of Medical Disciplines, Faculty of Medicine and Pharmacy, University of Oradea, 410073 Oradea, Romania; (M.A.M.); (M.C.B.); (C.B.)
| | - Felicia Liana Andronie-Cioara
- Department of Psycho-Neuroscience and Recovery, Faculty of Medicine and Pharmacy, University of Oradea, 410073 Oradea, Romania;
| | - Ciprian Brisc
- Department of Medical Disciplines, Faculty of Medicine and Pharmacy, University of Oradea, 410073 Oradea, Romania; (M.A.M.); (M.C.B.); (C.B.)
| |
Collapse
|
37
|
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
|
38
|
Abedin F, Tatulian SA. Mutual structural effects of unmodified and pyroglutamylated amyloid β peptides during aggregation. J Pept Sci 2021; 27:e3312. [PMID: 33631839 DOI: 10.1002/psc.3312] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2020] [Revised: 02/05/2021] [Accepted: 02/16/2021] [Indexed: 11/09/2022]
Abstract
Amyloid β (Aβ) peptide aggregates are linked to Alzheimer's disease (AD). Posttranslationally pyroglutamylated Aβ (pEAβ) occurs in AD brains in significant quantities and is hypertoxic, but the underlying structural and aggregation properties remain poorly understood. Here, the structure and aggregation of Aβ1-40 and pEAβ3-40 are analyzed separately and in equimolar combination. Circular dichroism data show that Aβ1-40 , pEAβ3-40 , and their combination assume α-helical structure in dry state and transition to unordered structure in aqueous buffer. Aβ1-40 and the 1:1 combination gradually acquire β-sheet structure while pEAβ3-40 adopts an α-helix/β-sheet conformation. Thioflavin-T fluorescence studies suggest that the two peptides mutually inhibit fibrillogenesis. Fourier transform infrared (FTIR) spectroscopy identifies the presence of β-turn and α-helical structures in addition to β-sheet structure in peptides in aqueous buffer. The kinetics of transitions from the initial α-helical structure to β-sheet structure were resolved by slow hydration of dry peptides by D2 O vapor, coupled with isotope-edited FTIR. These data confirmed the mutual suppression of β-sheet formation by the two peptides. Remarkably, pEAβ3-40 maintained a significant fraction of α-helical structure in the combined sample, implying a reduced β-sheet propensity of pEAβ3-40 . Altogether, the data imply that the combination of unmodified and pyroglutamylated Aβ peptides resists fibrillogenesis and favors the prefibrillar state, which may underlie hypertoxicity of pEAβ.
Collapse
Affiliation(s)
- Faisal Abedin
- Physics Graduate Program, University of Central Florida, Orlando, Florida, USA
| | - Suren A Tatulian
- Department of Physics, University of Central Florida, Orlando, Florida, USA
| |
Collapse
|
39
|
Di Benedetto G, Iannucci LF, Surdo NC, Zanin S, Conca F, Grisan F, Gerbino A, Lefkimmiatis K. Compartmentalized Signaling in Aging and Neurodegeneration. Cells 2021; 10:464. [PMID: 33671541 PMCID: PMC7926881 DOI: 10.3390/cells10020464] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 02/16/2021] [Accepted: 02/17/2021] [Indexed: 12/12/2022] Open
Abstract
The cyclic AMP (cAMP) signalling cascade is necessary for cell homeostasis and plays important roles in many processes. This is particularly relevant during ageing and age-related diseases, where drastic changes, generally decreases, in cAMP levels have been associated with the progressive decline in overall cell function and, eventually, the loss of cellular integrity. The functional relevance of reduced cAMP is clearly supported by the finding that increases in cAMP levels can reverse some of the effects of ageing. Nevertheless, despite these observations, the molecular mechanisms underlying the dysregulation of cAMP signalling in ageing are not well understood. Compartmentalization is widely accepted as the modality through which cAMP achieves its functional specificity; therefore, it is important to understand whether and how this mechanism is affected during ageing and to define which is its contribution to this process. Several animal models demonstrate the importance of specific cAMP signalling components in ageing, however, how age-related changes in each of these elements affect the compartmentalization of the cAMP pathway is largely unknown. In this review, we explore the connection of single components of the cAMP signalling cascade to ageing and age-related diseases whilst elaborating the literature in the context of cAMP signalling compartmentalization.
Collapse
Affiliation(s)
- Giulietta Di Benedetto
- Neuroscience Institute, National Research Council of Italy (CNR), 35121 Padova, Italy;
- Veneto Institute of Molecular Medicine, Foundation for Advanced Biomedical Research, 35129 Padova, Italy; (L.F.I.); (S.Z.); (F.C.); (F.G.)
| | - Liliana F. Iannucci
- Veneto Institute of Molecular Medicine, Foundation for Advanced Biomedical Research, 35129 Padova, Italy; (L.F.I.); (S.Z.); (F.C.); (F.G.)
- Department of Molecular Medicine, University of Pavia, 27100 Pavia, Italy
| | - Nicoletta C. Surdo
- Neuroscience Institute, National Research Council of Italy (CNR), 35121 Padova, Italy;
- Veneto Institute of Molecular Medicine, Foundation for Advanced Biomedical Research, 35129 Padova, Italy; (L.F.I.); (S.Z.); (F.C.); (F.G.)
| | - Sofia Zanin
- Veneto Institute of Molecular Medicine, Foundation for Advanced Biomedical Research, 35129 Padova, Italy; (L.F.I.); (S.Z.); (F.C.); (F.G.)
- Department of Molecular Medicine, University of Pavia, 27100 Pavia, Italy
| | - Filippo Conca
- Veneto Institute of Molecular Medicine, Foundation for Advanced Biomedical Research, 35129 Padova, Italy; (L.F.I.); (S.Z.); (F.C.); (F.G.)
- Department of Biology, University of Padova, 35122 Padova, Italy
| | - Francesca Grisan
- Veneto Institute of Molecular Medicine, Foundation for Advanced Biomedical Research, 35129 Padova, Italy; (L.F.I.); (S.Z.); (F.C.); (F.G.)
- Department of Biology, University of Padova, 35122 Padova, Italy
| | - Andrea Gerbino
- Department of Biosciences, Biotechnology and Biopharmaceutics, University of Bari, 70121 Bari, Italy;
| | - Konstantinos Lefkimmiatis
- Veneto Institute of Molecular Medicine, Foundation for Advanced Biomedical Research, 35129 Padova, Italy; (L.F.I.); (S.Z.); (F.C.); (F.G.)
- Department of Molecular Medicine, University of Pavia, 27100 Pavia, Italy
| |
Collapse
|
40
|
Borden EA, Furey M, Gattone NJ, Hambardikar VD, Liang XH, Scoma ER, Abou Samra A, D-Gary LR, Dennis DJ, Fricker D, Garcia C, Jiang Z, Khan SA, Kumarasamy D, Kuppala H, Ringrose S, Rosenheim EJ, Van Exel K, Vudhayagiri HS, Zhang J, Zhang Z, Guitart-Mampel M, Urquiza P, Solesio ME. Is there a link between inorganic polyphosphate (polyP), mitochondria, and neurodegeneration? Pharmacol Res 2021; 163:105211. [PMID: 33010423 PMCID: PMC7855267 DOI: 10.1016/j.phrs.2020.105211] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 09/08/2020] [Accepted: 09/15/2020] [Indexed: 12/14/2022]
Abstract
Mitochondrial dysfunction - including increased apoptosis, calcium and protein dyshomeostasis within the organelle, and dysfunctional bioenergetics and oxidative status - is a common, early feature in all the major neurodegenerative diseases, including Alzheimer's Disease (AD) and Parkinson's Disease (PD). However, the exact molecular mechanisms that drive the organelle to dysfunction and ultimately to failure in these conditions are still not well described. Different authors have shown that inorganic polyphosphate (polyP), an ancient and well-conserved molecule, plays a key role in the regulation of mitochondrial physiology under basal conditions. PolyP, which is present in all studied organisms, is composed of chains of orthophosphates linked together by highly energetic phosphoanhydride bonds, similar to those found in ATP. This polymer shows a ubiquitous distribution, even if a high co-localization with mitochondria has been reported. It has been proposed that polyP might be an alternative to ATP for cellular energy storage in different organisms, as well as the implication of polyP in the regulation of many of the mitochondrial processes affected in AD and PD, including protein and calcium homeostasis. Here, we conduct a comprehensive review and discussion of the bibliography available regarding the role of polyP in the mitochondrial dysfunction present in AD and PD. Taking into account the data presented in this review, we postulate that polyP could be a valid, innovative and, plausible pharmacological target against mitochondrial dysfunction in AD and PD. However, further research should be conducted to better understand the exact role of polyP in neurodegeneration, as well as the metabolism of the polymer, and the effect of different lengths of polyP on cellular and mitochondrial physiology.
Collapse
Affiliation(s)
- Emily A Borden
- Department of Biology, College of Arts and Sciences, Rutgers University, NJ, USA
| | - Matthew Furey
- Department of Biology, College of Arts and Sciences, Rutgers University, NJ, USA
| | - Nicholas J Gattone
- Department of Biology, College of Arts and Sciences, Rutgers University, NJ, USA
| | | | - Xiao Hua Liang
- Department of Biology, College of Arts and Sciences, Rutgers University, NJ, USA
| | - Ernest R Scoma
- Department of Biology, College of Arts and Sciences, Rutgers University, NJ, USA
| | - Antonella Abou Samra
- Department of Biology, College of Arts and Sciences, Rutgers University, NJ, USA
| | - LaKeshia R D-Gary
- Department of Biology, College of Arts and Sciences, Rutgers University, NJ, USA
| | - Dayshaun J Dennis
- Department of Biology, College of Arts and Sciences, Rutgers University, NJ, USA
| | - Daniel Fricker
- Department of Biology, College of Arts and Sciences, Rutgers University, NJ, USA
| | - Cindy Garcia
- Department of Biology, College of Arts and Sciences, Rutgers University, NJ, USA
| | - ZeCheng Jiang
- Department of Biology, College of Arts and Sciences, Rutgers University, NJ, USA
| | - Shariq A Khan
- Department of Biology, College of Arts and Sciences, Rutgers University, NJ, USA
| | | | - Hasmitha Kuppala
- Department of Biology, College of Arts and Sciences, Rutgers University, NJ, USA
| | - Savannah Ringrose
- Department of Biology, College of Arts and Sciences, Rutgers University, NJ, USA
| | - Evan J Rosenheim
- Department of Biology, College of Arts and Sciences, Rutgers University, NJ, USA
| | - Kimberly Van Exel
- Center for Computational and Integrative Biology, Rutgers University, NJ, USA
| | | | - Jiarui Zhang
- Center for Computational and Integrative Biology, Rutgers University, NJ, USA
| | - Zhaowen Zhang
- Department of Biology, College of Arts and Sciences, Rutgers University, NJ, USA
| | | | - Pedro Urquiza
- Department of Biology, College of Arts and Sciences, Rutgers University, NJ, USA
| | - Maria E Solesio
- Department of Biology, College of Arts and Sciences, Rutgers University, NJ, USA; Center for Computational and Integrative Biology, Rutgers University, NJ, USA.
| |
Collapse
|
41
|
He Y, Qiang Y. Mechanism of Autonomic Exercise Improving Cognitive Function of Alzheimer's Disease by Regulating lncRNA SNHG14. Am J Alzheimers Dis Other Demen 2021; 36:15333175211027681. [PMID: 34338033 PMCID: PMC10581095 DOI: 10.1177/15333175211027681] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
This paper studied the influence of exercise on the cognitive ability of AD patients and elucidated potential mechanisms. The expression of SNHG14 was validated by qRT-PCR. The cognitive impairment of mice was examined by MWM Test. ELISA tests were applied to discover the influence of SNHG14 on inflammation. Overexpression of SNHG14 was found in AD patients and underexpression of SNHG14 was identified in these AD patients after exercise. In APP/PS1 double transgenic mice, SNHG14 reversed the protective impacts of exercise on escape latency and distance moved. The upregulation of SNHG14 also inhibited the effects of exercise on the percentage of time spent in the target quadrant and times of platform crossing. Besides, overexpression of SNHG14 reversed the repressed expression of IL-6, IL-1β, and TNF-α. In total, exercise could ameliorate cognitive disorder and inflammation activity by reducing the levels of SNHG14.
Collapse
Affiliation(s)
- Yuchen He
- Rehabilitation Department, Shuguang Hospital Affiliated to Shanghai University of Chinese Medicine, Shanghai, China
| | - Yi Qiang
- Rehabilitation Department, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
42
|
Ramachandran AK, Das S, Joseph A, Gurupur Gautham S, Alex AT, Mudgal J. Neurodegenerative Pathways in Alzheimer's Disease: A Review. Curr Neuropharmacol 2021; 19:679-692. [PMID: 32851951 PMCID: PMC8573750 DOI: 10.2174/1570159x18666200807130637] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2020] [Revised: 06/26/2020] [Accepted: 07/31/2020] [Indexed: 11/23/2022] Open
Abstract
Alzheimer's disease (AD) is a complex neurodegenerative disease that leads to insidious deterioration of brain functions and is considered the sixth leading cause of death in the world. Alzheimer's patients suffer from memory loss, cognitive deficit and behavioral changes; thus, they eventually follow a low-quality life. AD is considered as a multifactorial disorder involving different neuropathological mechanisms. Recent research has identified more than 20 pathological factors that are promoting disease progression. Three significant hypotheses are said to be the root cause of disease pathology, which include acetylcholine deficit, the formation of amyloid-beta senile plaques and tau protein hyperphosphorylation. Apart from these crucial factors, pathological factors such as apolipoprotein E (APOE), glycogen synthase kinase 3β, notch signaling pathway, Wnt signaling pathway, etc., are considered to play a role in the advancement of AD and therefore could be used as targets for drug discovery and development. As of today, there is no complete cure or effective disease altering therapies for AD. The current therapy is assuring only symptomatic relief from the disease, and progressive loss of efficacy for these symptomatic treatments warrants the discovery of newer drugs by exploring these novel drug targets. A comprehensive understanding of these therapeutic targets and their neuropathological role in AD is necessary to identify novel molecules for the treatment of AD rationally.
Collapse
Affiliation(s)
- Anu Kunnath Ramachandran
- Department of Pharmaceutical Chemistry, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka-576104, India
| | - Subham Das
- Department of Pharmaceutical Chemistry, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka-576104, India
| | - Alex Joseph
- Department of Pharmaceutical Chemistry, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka-576104, India
| | - Shenoy, Gurupur Gautham
- Department of Pharmaceutical Chemistry, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka-576104, India
| | - Angel Treasa Alex
- Department of Pharmaceutical Biotechnology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka-576104, India
| | - Jayesh Mudgal
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka-576104, India
| |
Collapse
|
43
|
Potekhina ES, Bass DY, Kelmanson IV, Fetisova ES, Ivanenko AV, Belousov VV, Bilan DS. Drug Screening with Genetically Encoded Fluorescent Sensors: Today and Tomorrow. Int J Mol Sci 2020; 22:E148. [PMID: 33375682 PMCID: PMC7794770 DOI: 10.3390/ijms22010148] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 12/18/2020] [Accepted: 12/24/2020] [Indexed: 02/07/2023] Open
Abstract
Genetically-encoded fluorescent sensors have been actively developed over the last few decades and used in live imaging and drug screening. Real-time monitoring of drug action in a specific cellular compartment, organ, or tissue type; the ability to screen at the single-cell resolution; and the elimination of false-positive results caused by low drug bioavailability that is not detected by in vitro testing methods are a few of the obvious benefits of using genetically-encoded fluorescent sensors in drug screening. In combination with high-throughput screening (HTS), some genetically-encoded fluorescent sensors may provide high reproducibility and robustness to assays. We provide a brief overview of successful, perspective, and hopeful attempts at using genetically encoded fluorescent sensors in HTS of modulators of ion channels, Ca2+ homeostasis, GPCR activity, and for screening cytotoxic, anticancer, and anti-parasitic compounds. We discuss the advantages of sensors in whole organism drug screening models and the perspectives of the combination of human disease modeling by CRISPR techniques with genetically encoded fluorescent sensors for drug screening.
Collapse
Affiliation(s)
- Ekaterina S. Potekhina
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, 117997 Moscow, Russia; (D.Y.B.); (I.V.K.); (E.S.F.); (A.V.I.); (V.V.B.)
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, 117997 Moscow, Russia
| | - Dina Y. Bass
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, 117997 Moscow, Russia; (D.Y.B.); (I.V.K.); (E.S.F.); (A.V.I.); (V.V.B.)
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, 117997 Moscow, Russia
| | - Ilya V. Kelmanson
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, 117997 Moscow, Russia; (D.Y.B.); (I.V.K.); (E.S.F.); (A.V.I.); (V.V.B.)
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, 117997 Moscow, Russia
| | - Elena S. Fetisova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, 117997 Moscow, Russia; (D.Y.B.); (I.V.K.); (E.S.F.); (A.V.I.); (V.V.B.)
| | - Alexander V. Ivanenko
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, 117997 Moscow, Russia; (D.Y.B.); (I.V.K.); (E.S.F.); (A.V.I.); (V.V.B.)
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, 117997 Moscow, Russia
| | - Vsevolod V. Belousov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, 117997 Moscow, Russia; (D.Y.B.); (I.V.K.); (E.S.F.); (A.V.I.); (V.V.B.)
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, 117997 Moscow, Russia
- Federal Center of Brain Research and Neurotechnologies of the Federal Medical Biological Agency, 117997 Moscow, Russia
| | - Dmitry S. Bilan
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, 117997 Moscow, Russia; (D.Y.B.); (I.V.K.); (E.S.F.); (A.V.I.); (V.V.B.)
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, 117997 Moscow, Russia
| |
Collapse
|
44
|
Bigi A, Loffredo G, Cascella R, Cecchi C. Targeting Pathological Amyloid Aggregates with Conformation-Sensitive Antibodies. Curr Alzheimer Res 2020; 17:722-734. [PMID: 33167834 DOI: 10.2174/1567205017666201109093848] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2020] [Revised: 08/05/2020] [Accepted: 10/01/2020] [Indexed: 01/10/2023]
Abstract
BACKGROUND The pathogenesis of Alzheimer's disease (AD) is not directly caused by the presence of senile plaques but rather by the detrimental effects exerted on neuronal cells by toxic soluble oligomers. Such species are formed early during the aggregation process of the Aβ1-42 peptide or can be released from mature fibrils. Nowadays, efficient tools for an early diagnosis, as well as pharmaceutical treatments targeting the harmful agents in samples of AD patients, are still missing. OBJECTIVE By integrating in vitro immunochemical assay with in vivo neuronal models of toxicity, we aim to understand and target the principles that drive toxicity in AD. METHODS We evaluated the specificity and sensitivity of A11 and OC conformational antibodies to target a range of pathologically relevant amyloid conformers and rescue their cytotoxic effects in neuronal culture models using a number of cellular readouts. RESULTS We demonstrated the peculiar ability of conformational antibodies to label pathologically relevant Aβ1-42 oligomers and fibrils and to prevent their detrimental effects on neuronal cells. CONCLUSION Our results substantially improve our knowledge on the role of toxic assemblies in neurodegenerative diseases, thus suggesting new and more effective diagnostic and therapeutic tools for AD.
Collapse
Affiliation(s)
- Alessandra Bigi
- Department of Experimental and Clinical Biomedical Sciences, Section of Biochemistry, University of Florence, Florence 50134, Italy
| | - Gilda Loffredo
- Department of Experimental and Clinical Biomedical Sciences, Section of Biochemistry, University of Florence, Florence 50134, Italy
| | - Roberta Cascella
- Department of Experimental and Clinical Biomedical Sciences, Section of Biochemistry, University of Florence, Florence 50134, Italy
| | - Cristina Cecchi
- Department of Experimental and Clinical Biomedical Sciences, Section of Biochemistry, University of Florence, Florence 50134, Italy
| |
Collapse
|
45
|
Dhakal S, Macreadie I. Protein Homeostasis Networks and the Use of Yeast to Guide Interventions in Alzheimer's Disease. Int J Mol Sci 2020; 21:E8014. [PMID: 33126501 PMCID: PMC7662794 DOI: 10.3390/ijms21218014] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 10/24/2020] [Accepted: 10/26/2020] [Indexed: 12/12/2022] Open
Abstract
Alzheimer's Disease (AD) is a progressive multifactorial age-related neurodegenerative disorder that causes the majority of deaths due to dementia in the elderly. Although various risk factors have been found to be associated with AD progression, the cause of the disease is still unresolved. The loss of proteostasis is one of the major causes of AD: it is evident by aggregation of misfolded proteins, lipid homeostasis disruption, accumulation of autophagic vesicles, and oxidative damage during the disease progression. Different models have been developed to study AD, one of which is a yeast model. Yeasts are simple unicellular eukaryotic cells that have provided great insights into human cell biology. Various yeast models, including unmodified and genetically modified yeasts, have been established for studying AD and have provided significant amount of information on AD pathology and potential interventions. The conservation of various human biological processes, including signal transduction, energy metabolism, protein homeostasis, stress responses, oxidative phosphorylation, vesicle trafficking, apoptosis, endocytosis, and ageing, renders yeast a fascinating, powerful model for AD. In addition, the easy manipulation of the yeast genome and availability of methods to evaluate yeast cells rapidly in high throughput technological platforms strengthen the rationale of using yeast as a model. This review focuses on the description of the proteostasis network in yeast and its comparison with the human proteostasis network. It further elaborates on the AD-associated proteostasis failure and applications of the yeast proteostasis network to understand AD pathology and its potential to guide interventions against AD.
Collapse
Affiliation(s)
| | - Ian Macreadie
- School of Science, RMIT University, Bundoora, Victoria 3083, Australia;
| |
Collapse
|
46
|
Sun L, Wei H. Ryanodine Receptors: A Potential Treatment Target in Various Neurodegenerative Disease. Cell Mol Neurobiol 2020; 41:1613-1624. [PMID: 32833122 DOI: 10.1007/s10571-020-00936-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2020] [Accepted: 08/05/2020] [Indexed: 02/07/2023]
Abstract
Progressive neuronal demise is a key contributor to the key pathogenic event implicated in many different neurodegenerative disorders (NDDs). There are several therapeutic strategies available; however, none of them are particularly effective. Targeted neuroprotective therapy is one such therapy, which seems a compelling option, yet remains challenging due to the internal heterogeneity of the mechanisms underlying various NDDs. An alternative method to treat NDDs is to exploit common modalities involving molecularly distinct subtypes and thus develop specialized drugs with broad-spectrum characteristics. There is mounting evidence which supports for the theory that dysfunctional ryanodine receptors (RyRs) disrupt intracellular Ca2+ homeostasis, contributing to NDDs significantly. This review aims to provide direct and indirect evidence on the intersection of NDDs and RyRs malfunction, and to shed light on novel strategies to treat RyRs-mediated disease, modifying pharmacological therapies such as the potential therapeutic role of dantrolene, a RyRs antagonist.
Collapse
Affiliation(s)
- Liang Sun
- Department of Anesthesiology and Critical Care, Perelman School of Medicine, University of Pennsylvania, 305 John Morgan Building, 3610 Hamilton Walk, Philadelphia, PA, 19104, USA
- Department of Anesthesiology, Peking University People's Hospital, Beijing, 100044, China
| | - Huafeng Wei
- Department of Anesthesiology and Critical Care, Perelman School of Medicine, University of Pennsylvania, 305 John Morgan Building, 3610 Hamilton Walk, Philadelphia, PA, 19104, USA.
| |
Collapse
|
47
|
Subchronic administration of auranofin reduced amyloid-β plaque pathology in a transgenic APP NL-G-F/NL-G-F mouse model. Brain Res 2020; 1746:147022. [PMID: 32707043 DOI: 10.1016/j.brainres.2020.147022] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 07/13/2020] [Accepted: 07/18/2020] [Indexed: 01/26/2023]
Abstract
Alzheimer's disease (AD) is the most common cause of dementia. Neuropathological processes, including the accumulation of amyloid-β (Aβ) plaques and neurofibrillary tangles, and neuroinflammation, lead to cognitive impairment at middle and eventually later stages of AD progression. Over the last decade, focused efforts have explored repurposed drug approaches for AD pathophysiological mechanisms. Recently, auranofin, an anti-inflammatory drug, was shown to have therapeutic potential in a number of diseases in addition to rheumatoid arthritis. Surprisingly, no data regarding the effects of auranofin on cognitive deficits in AD mice or the influence of auranofin on Aβ pathology and neuroinflammatory processes are available. In the present study, we used 14-month-old transgenic male APPNL-G-F/NL-G-F mice to assess the effects of subchronic administration of auranofin at low doses (1 and 5 mg/kg, intraperitoneal) on spatial memory, Aβ pathology and the expression of cortical and hippocampal proteins (glial fibrillary acidic protein (GFAP), ionized calcium binding adaptor molecule-1 (Iba-1)) and proteins related to synaptic plasticity (glutamic acid decarboxylase 67 (GAD67), homer proteins homologue-1 (Homer-1)). The data demonstrated that auranofin significantly decreased Aβ deposition in the hippocampus and the number of Aβ plaques in the cingulate cortex, but it did not have memory-enhancing effects or induce changes in the expression of the studied proteins. Our current results highlight the importance of considering further pre-clinical research to investigate the possible beneficial effects of auranofin on the other pathological aspects of AD.
Collapse
|
48
|
Uryash A, Flores V, Adams JA, Allen PD, Lopez JR. Memory and Learning Deficits Are Associated With Ca 2+ Dyshomeostasis in Normal Aging. Front Aging Neurosci 2020; 12:224. [PMID: 32765253 PMCID: PMC7378956 DOI: 10.3389/fnagi.2020.00224] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Accepted: 06/23/2020] [Indexed: 12/12/2022] Open
Abstract
Neuronal intracellular Ca2+ homeostasis is critical to the normal physiological functions of neurons and neuronal Ca2+ dyshomeostasis has been associated with the age-related decline of cognitive functions. Accumulated evidence indicates that the underlying mechanism for this is that abnormal intracellular Ca2+ levels stimulate the dysregulation of intracellular signaling, which subsequently induces neuronal cell death. We examined intracellular Ca2+ homeostasis in cortical (in vivo) and hippocampal (in vitro) neurons from young (3-months), middle-age (12-months), and aged (24-months) wild type C57BL6J mice. We found a progressive age-related elevation of intracellular resting calcium ([Ca2+]r) in cortical (in vivo) and hippocampal (in vitro) neurons associated with increased hippocampal neuronal calpain activity and reduced cell viability. In vitro, removal of extracellular Ca2+ or treatment with SAR7334 or dantrolene reduced [Ca2+]r in all age groups and dantrolene treatment lowered calpain activity and increased cell viability. In vivo, both middle-aged and aged mice showed cognitive deficits compared to young mice, which improved after dantrolene treatment. These findings support the hypothesis that intracellular Ca2+ dyshomeostasis is a major mechanism underlying the cognitive deficits seen in both normal aging and degenerative neurologic diseases.
Collapse
Affiliation(s)
- Arkady Uryash
- Division of Neonatology, Mount Sinai Medical Center, Miami, FL, United States
| | - Valentina Flores
- Department of Research, Mount Sinai Medical Center, Miami, FL, United States
| | - Jose A. Adams
- Division of Neonatology, Mount Sinai Medical Center, Miami, FL, United States
| | - Paul D. Allen
- Malignant Hyperthermia Investigation Unit, St James’ University Hospital, University of Leeds, Leeds, United Kingdom
| | - Jose R. Lopez
- Department of Research, Mount Sinai Medical Center, Miami, FL, United States
| |
Collapse
|
49
|
Sabbir MG. CAMKK2-CAMK4 signaling regulates transferrin trafficking, turnover, and iron homeostasis. Cell Commun Signal 2020; 18:80. [PMID: 32460794 PMCID: PMC7251913 DOI: 10.1186/s12964-020-00575-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Accepted: 04/02/2020] [Indexed: 12/20/2022] Open
Abstract
Background Circulatory iron is a hazardous biometal. Therefore, iron is transported in a redox-safe state by a serum glycoprotein - transferrin (TF). Different organs acquire iron from the systemic circulation through a tightly regulated mechanism at the blood-tissue interface which involves receptor-mediated internalization of TF. Thus, abnormal TF trafficking may lead to iron dyshomeostasis associated with several diseases including neurodegeneration. Iron -induced toxicity can cause neuronal damage to iron-sensitive brain regions. Recently, it was discovered that CAMKK2, a calcium (Ca2+)/calmodulin-activated kinase, controls receptor-mediated TF trafficking in mouse tissues, specifically in the brain. The biological function of CAMKK2 is mediated through multiple downstream effectors. Both CAMKK2 and one of its downstream kinase, CAMK4, exhibit overlapping expression in mouse brain. The role of CAMK4 in vesicular transport has been reported and loss of CAMKK2 or CAMK4 leads to cognitive defects in mouse. Therefore, it was hypothesized that CAMKK2-CAMK4 signaling regulates receptor-mediated TF trafficking and iron homeostasis which may be responsible for the neuronal malfunction observed in CAMKK2- or CAMK4-deficient mice. Methods CAMK4−/− mouse was used to study tissue-specific turnover of TF, TF-receptor (TFRC) and iron. CRISPR/Cas9-based CAMKK2 and/or CAMK4 deleted human embryonic kidney-derived HEK293 cell clones were used to study the molecular defects in receptor-mediated TF trafficking. Further, a “zero functional G protein” condition in HEK293 cell was exploited to study CAMKK2-CAMK4 signaling-mediated regulation of intracellular Ca2+ homeostasis which was linked to calcium signaling during TF trafficking. Results Loss of CAMK4 leads to abnormal post-translational modifications (PTMs) and turnover of TF in mouse cerebellum and liver which was associated with iron dyshomeostasis in these tissues. The HEK293 cell-based study revealed that the absence of CAMKK2-CAMK4 signaling altered intracellular Ca2+ homeostasis and lead to abnormal calcium signaling during TF trafficking. Also, CAMKK2-CAMK4 signaling deficiency affected the molecular interaction of TF and TF-receptor-associated protein complexes which indicated a potential failure in the recruitment of interacting proteins due to differential PTMs in TF. Conclusion Overall, this study established a novel mechanistic link between intracellular Ca2+ level, receptor-mediated TF trafficking, and iron homeostasis, all regulated by CAMKK2-CAMK4 signaling. Video Abstract
Graphical abstract ![]()
Collapse
Affiliation(s)
- Mohammad Golam Sabbir
- Canadian Centre for Agri-Food Research in Health and Medicine, St. Boniface Albrechtsen Research Centre, Room R2034 - 351 Taché Avenue, Winnipeg, MB, R2H 2A6, Canada. .,Alzo Biosciences Inc., San Diego, CA, USA.
| |
Collapse
|
50
|
Wang Y, Liang G, Liang S, Mund R, Shi Y, Wei H. Dantrolene Ameliorates Impaired Neurogenesis and Synaptogenesis in Induced Pluripotent Stem Cell Lines Derived from Patients with Alzheimer's Disease. Anesthesiology 2020; 132:1062-1079. [PMID: 32149777 PMCID: PMC7160009 DOI: 10.1097/aln.0000000000003224] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
BACKGROUND Overactivation of ryanodine receptors and the resulting impaired calcium homeostasis contribute to Alzheimer's disease-related pathophysiology. This study hypothesized that exposing neuronal progenitors derived from induced pluripotent stems cells of patients with Alzheimer's disease to dantrolene will increase survival, proliferation, neurogenesis, and synaptogenesis. METHODS Induced pluripotent stem cells obtained from skin fibroblast of healthy subjects and patients with familial and sporadic Alzheimer's disease were used. Biochemical and immunohistochemical methods were applied to determine the effects of dantrolene on the viability, proliferation, differentiation, and calcium dynamics of these cells. RESULTS Dantrolene promoted cell viability and proliferation in these two cell lines. Compared with the control, differentiation into basal forebrain cholinergic neurons significantly decreased by 10.7% (32.9 ± 3.6% vs. 22.2 ± 2.6%, N = 5, P = 0.004) and 9.2% (32.9 ± 3.6% vs. 23.7 ± 3.1%, N = 5, P = 0.017) in cell lines from sporadic and familial Alzheimer's patients, respectively, which were abolished by dantrolene. Synapse density was significantly decreased in cortical neurons generated from stem cells of sporadic Alzheimer's disease by 58.2% (237.0 ± 28.4 vs. 99.0 ± 16.6 arbitrary units, N = 4, P = 0.001) or familial Alzheimer's disease by 52.3% (237.0 ± 28.4 vs.113.0 ± 34.9 vs. arbitrary units, N = 5, P = 0.001), which was inhibited by dantrolene in the familial cell line. Compared with the control, adenosine triphosphate (30 µM) significantly increased higher peak elevation of cytosolic calcium concentrations in the cell line from sporadic Alzheimer's patients (84.1 ± 27.0% vs. 140.4 ± 40.2%, N = 5, P = 0.049), which was abolished by the pretreatment of dantrolene. Dantrolene inhibited the decrease of lysosomal vacuolar-type H-ATPase and the impairment of autophagy activity in these two cell lines from Alzheimer's disease patients. CONCLUSIONS Dantrolene ameliorated the impairment of neurogenesis and synaptogenesis, in association with restoring intracellular Ca homeostasis and physiologic autophagy, cell survival, and proliferation in induced pluripotent stem cells and their derived neurons from sporadic and familial Alzheimer's disease patients.
Collapse
Affiliation(s)
- Yong Wang
- Department of Anesthesiology and Critical Care, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Anesthesiology, the First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Ge Liang
- Department of Anesthesiology and Critical Care, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Shuqing Liang
- Department of Anesthesiology and Critical Care, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Anesthesiology, the First Affiliated Hospital of Jinan University, Guangzhou, 510632, China
| | - Rachel Mund
- Undergraduate Student, College of Art and Science, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Yun Shi
- Department of Anesthesiology and Critical Care, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Anesthesiology, Children’s hospital of Fudan University, Shanghai, 201102, China
| | - Huafeng Wei
- Department of Anesthesiology and Critical Care, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|