1
|
Polfer R, Furukawa H. Biology, function and structure of the calcium homeostasis modulator family. J Physiol 2024. [PMID: 39470434 DOI: 10.1113/jp285197] [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: 06/17/2024] [Accepted: 09/24/2024] [Indexed: 10/30/2024] Open
Abstract
Calcium homeostasis modulators (CALHMs) are the most recently discovered members of the large-pore channel family. They mediate the conductance of ions and larger molecules, such as ATP, and play critical roles in pathways related to Alzheimer's disease, neuroinflammation, neuromodulation, taste perception and innate immune responses. Since the inaugural report on CALHM1 in 2008, significant breakthroughs have revealed their biological roles, ion and ATP channel functions, and structures, positioning the field for further advancements. In this review, we discuss the overall progress and recent developments in understanding the biological roles, functions and molecular structures of CALHM proteins.
Collapse
Affiliation(s)
- Rachel Polfer
- Cold Spring Harbor Laboratory, School of Biological Science at Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA
| | - Hiro Furukawa
- Cold Spring Harbor Laboratory, School of Biological Science at Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA
| |
Collapse
|
2
|
Zhang S, Wang X, Liu S, Hu C, Meng Y. Phlorizin ameliorates cognitive and behavioral impairments via the microbiota-gut-brain axis in high-fat and high-fructose diet-induced obese male mice. Brain Behav Immun 2024; 123:193-210. [PMID: 39277023 DOI: 10.1016/j.bbi.2024.09.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 08/26/2024] [Accepted: 09/07/2024] [Indexed: 09/17/2024] Open
Abstract
The long-term high-fat, high-sugar diet exacerbates type 2 diabetes mellitus (T2DM)-related cognitive impairments. Phlorizin, a well-studied natural compound found in apples and other plants, is recognized for its bioactive properties, including modulation of glucose and lipid metabolism. Despite its established role in mitigating metabolic disorders, the neuroprotective effects of phlorizin, particularly against diabetes-related cognitive dysfunction, have not been fully elucidated. Therefore, the present study aimed to investigate the effect of dietary supplementation of phlorizin on high-fat and high-fructose diet (HFFD)-induced cognitive dysfunction and evaluate the crucial role of the microbiota-gut-brain axis. We found that dietary supplementation of phlorizin for 14 weeks effectively prevented glucolipid metabolism disorder, spatial learning impairment, and memory impairment in HFFD mice. In addition, phlorizin improved the HFFD-induced decrease in synaptic plasticity, neuroinflammation, and excessive activation of microglia in the hippocampus. Transcriptomics analysis shows that the protective effect of phlorizin on cognitive impairment was associated with increased expression of neurotransmitters and synapse-related genes in the hippocampus. Phlorizin treatment alleviated colon microbiota disturbance, mainly manifested by an increase in gut microbiota diversity and the abundance of short-chain fatty acid (SCFA)-producing bacteria. The level of microbial metabolites, including SCFA, inosine 5'-monophosphate (IMP), and D (-)-beta-hydroxybutyric acid (BHB) were also significantly increased after phlorizin treatment. Integrating multiomics analysis observed tight connections between phlorizin-regulated genes, microbiota, and metabolites. Furthermore, removal of the gut microbiota via antibiotics treatment diminished the protective effect of phlorizin against HFFD-induced cognitive impairment, underscoring the critical role of the gut microbiota in mediating cognitive behavior. Importantly, supplementation with SCFA and BHB alone mimicked the regulatory effects of phlorizin on cognitive function. Therefore, phlorizin shows promise as a potential nutritional therapy for addressing cognitive impairment associated with metabolic disorders. Further research is needed to explore its effectiveness in preventing and alleviating neurodegenerative diseases.
Collapse
Affiliation(s)
- Shuqing Zhang
- Engineering Research Center for High-Valued Utilization of Fruit Resources in Western China, Ministry of Education, National Research & Development Center of Apple Processing Technology, College of Food Engineering and Nutritional Science, Shaanxi Normal University, 620 West Changan Avenue, Xian, Shaanxi 710119, PR China; College of Food Science and Nutritional Engineering, National Engineering Research Centre for Fruit and Vegetable Processing, Key Laboratory for Fruit and Vegetable Processing, Ministry of Agriculture, Engineering Research Centre for Fruit and Vegetable Processing, Ministry of Education, China Agricultural University, Beijing 100083, China.
| | - Xiaoyu Wang
- Engineering Research Center for High-Valued Utilization of Fruit Resources in Western China, Ministry of Education, National Research & Development Center of Apple Processing Technology, College of Food Engineering and Nutritional Science, Shaanxi Normal University, 620 West Changan Avenue, Xian, Shaanxi 710119, PR China.
| | - Shenlin Liu
- Engineering Research Center for High-Valued Utilization of Fruit Resources in Western China, Ministry of Education, National Research & Development Center of Apple Processing Technology, College of Food Engineering and Nutritional Science, Shaanxi Normal University, 620 West Changan Avenue, Xian, Shaanxi 710119, PR China.
| | - Chingyuan Hu
- Engineering Research Center for High-Valued Utilization of Fruit Resources in Western China, Ministry of Education, National Research & Development Center of Apple Processing Technology, College of Food Engineering and Nutritional Science, Shaanxi Normal University, 620 West Changan Avenue, Xian, Shaanxi 710119, PR China; Department of Human Nutrition, Food and Animal Sciences, College of Tropical Agriculture and Human Resources, University of Hawaii at Manoa, 1955 East-West Road, AgSci. 415J, Honolulu, HI 96822, USA.
| | - Yonghong Meng
- Engineering Research Center for High-Valued Utilization of Fruit Resources in Western China, Ministry of Education, National Research & Development Center of Apple Processing Technology, College of Food Engineering and Nutritional Science, Shaanxi Normal University, 620 West Changan Avenue, Xian, Shaanxi 710119, PR China.
| |
Collapse
|
3
|
Puebla M, Muñoz MF, Lillo MA, Contreras JE, Figueroa XF. Control of astrocytic Ca 2+ signaling by nitric oxide-dependent S-nitrosylation of Ca 2+ homeostasis modulator 1 channels. Biol Res 2024; 57:19. [PMID: 38689353 PMCID: PMC11059852 DOI: 10.1186/s40659-024-00503-3] [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: 08/31/2023] [Accepted: 04/18/2024] [Indexed: 05/02/2024] Open
Abstract
BACKGROUND Astrocytes Ca2+ signaling play a central role in the modulation of neuronal function. Activation of metabotropic glutamate receptors (mGluR) by glutamate released during an increase in synaptic activity triggers coordinated Ca2+ signals in astrocytes. Importantly, astrocytes express the Ca2+-dependent nitric oxide (NO)-synthetizing enzymes eNOS and nNOS, which might contribute to the Ca2+ signals by triggering Ca2+ influx or ATP release through the activation of connexin 43 (Cx43) hemichannels, pannexin-1 (Panx-1) channels or Ca2+ homeostasis modulator 1 (CALHM1) channels. Hence, we aim to evaluate the participation of NO in the astrocytic Ca2+ signaling initiated by stimulation of mGluR in primary cultures of astrocytes from rat brain cortex. RESULTS Astrocytes were stimulated with glutamate or t-ACPD and NO-dependent changes in [Ca2+]i and ATP release were evaluated. In addition, the activity of Cx43 hemichannels, Panx-1 channels and CALHM1 channels was also analyzed. The expression of Cx43, Panx-1 and CALHM1 in astrocytes was confirmed by immunofluorescence analysis and both glutamate and t-ACPD induced NO-mediated activation of CALHM1 channels via direct S-nitrosylation, which was further confirmed by assessing CALHM1-mediated current using the two-electrode voltage clamp technique in Xenopus oocytes. Pharmacological blockade or siRNA-mediated inhibition of CALHM1 expression revealed that the opening of these channels provides a pathway for ATP release and the subsequent purinergic receptor-dependent activation of Cx43 hemichannels and Panx-1 channels, which further contributes to the astrocytic Ca2+ signaling. CONCLUSIONS Our findings demonstrate that activation of CALHM1 channels through NO-mediated S-nitrosylation in astrocytes in vitro is critical for the generation of glutamate-initiated astrocytic Ca2+ signaling.
Collapse
Affiliation(s)
- Mariela Puebla
- Departamento de Fisiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, 8330025, Santiago, Chile
| | - Manuel F Muñoz
- Departamento de Fisiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, 8330025, Santiago, Chile
- Department of Physiology and Membrane Biology, University of California Davis, Davis, CA, USA
| | - Mauricio A Lillo
- Department of Pharmacology, Physiology and Neuroscience, New Jersey Medical School, Rutgers University, Newark, NJ, USA
| | - Jorge E Contreras
- Department of Physiology and Membrane Biology, University of California Davis, Davis, CA, USA
- Department of Pharmacology, Physiology and Neuroscience, New Jersey Medical School, Rutgers University, Newark, NJ, USA
| | - Xavier F Figueroa
- Departamento de Fisiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, 8330025, Santiago, Chile.
| |
Collapse
|
4
|
Choi SW, Kwon JW, Kang TM, Park KS, Kim SJ. Calcium homeostasis modulator 2 (Calhm2) as slowly activating membrane current channel in mouse B cells. Biochem Biophys Res Commun 2024; 699:149561. [PMID: 38280307 DOI: 10.1016/j.bbrc.2024.149561] [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: 12/13/2023] [Revised: 12/18/2023] [Accepted: 01/20/2024] [Indexed: 01/29/2024]
Abstract
In mouse B lymphocytes, an unidentified slow-activating voltage-dependent current resembling the characteristics of the Calhm family ion channel (ICalhm-L) was investigated. RT-PCR analysis revealed the presence of Calhm2 and 6 transcripts, with subsequent whole-cell patch-clamp studies indicating that the ICalhm-L is augmented by heat, alkaline pH, and low extracellular [Ca2+]. Overexpression of Calhm2, but not Calhm6, in N2A cells recapitulated ICalhm-L. Moreover, Calhm2 knockdown in Bal-17 cells abolished ICalhm-L. We firstly identify the voltage-dependent ion channel function of the Calhm2 in the mouse immune cells. ATP release assays in primary mouse B cells suggested a significant contribution of Calhm2 for purinergic signaling at physiological temperature.
Collapse
Affiliation(s)
- Si Won Choi
- Department of Physiology, Seoul National University College of Medicine, Seoul, Republic of Korea; Department of Physiology, Sungkyunkwan University School of Medicine, Suwon, Republic of Korea; Wide River Institute of Immunology, Seoul National University College of Medicine, Hongcheon, Republic of Korea
| | - Jae-Won Kwon
- Department of Physiology, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Tong Mook Kang
- Department of Physiology, Sungkyunkwan University School of Medicine, Suwon, Republic of Korea
| | - Kyoung Sun Park
- Wide River Institute of Immunology, Seoul National University College of Medicine, Hongcheon, Republic of Korea.
| | - Sung Joon Kim
- Department of Physiology, Seoul National University College of Medicine, Seoul, Republic of Korea; Wide River Institute of Immunology, Seoul National University College of Medicine, Hongcheon, Republic of Korea.
| |
Collapse
|
5
|
Jadiya P, Kolmetzky DW, Tomar D, Thomas M, Cohen HM, Khaledi S, Garbincius JF, Hildebrand AN, Elrod JW. Genetic ablation of neuronal mitochondrial calcium uptake halts Alzheimer's disease progression. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.11.561889. [PMID: 37904949 PMCID: PMC10614731 DOI: 10.1101/2023.10.11.561889] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/01/2023]
Abstract
Alzheimer's disease (AD) is characterized by the extracellular deposition of amyloid beta, intracellular neurofibrillary tangles, synaptic dysfunction, and neuronal cell death. These phenotypes correlate with and are linked to elevated neuronal intracellular calcium ( i Ca 2+ ) levels. Recently, our group reported that mitochondrial calcium ( m Ca 2+ ) overload, due to loss of m Ca 2+ efflux capacity, contributes to AD development and progression. We also noted proteomic remodeling of the mitochondrial calcium uniporter channel (mtCU) in sporadic AD brain samples, suggestive of altered m Ca 2+ uptake in AD. Since the mtCU is the primary mechanism for Ca 2+ uptake into the mitochondrial matrix, inhibition of the mtCU has the potential to reduce or prevent m Ca 2+ overload in AD. Here, we report that neuronal-specific loss of mtCU-dependent m Ca 2+ uptake in the 3xTg-AD mouse model of AD reduced Aβ and tau-pathology, synaptic dysfunction, and cognitive decline. Knockdown of Mcu in a cellular model of AD significantly decreased matrix Ca 2+ content, oxidative stress, and cell death. These results suggest that inhibition of neuronal m Ca 2+ uptake is a novel therapeutic target to impede AD progression.
Collapse
|
6
|
Liao Y, Wang Y, Tao QQ, Yang C, Wang J, Cheng J, Ma J, Wu ZY, Pan RY, Yuan Z. CALHM2 V136G polymorphism reduces astrocytic ATP release and is associated with depressive symptoms and Alzheimer's disease risk. Alzheimers Dement 2023; 19:4407-4420. [PMID: 37493186 DOI: 10.1002/alz.13366] [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: 12/01/2022] [Revised: 05/19/2023] [Accepted: 06/04/2023] [Indexed: 07/27/2023]
Abstract
INTRODUCTION Depression is considered a prodromal state of Alzheimer's disease (AD), yet the underlying mechanism(s) by which depression increases the risk of AD are not known. METHODS Single-nucleotide polymorphism (SNP) analysis was used to determine the CALHM2 variants in AD patients. Cellular and molecular experiments were conducted to investigate the function of CALHM2 V136G mutation. We generated a new genetically engineered Calhm2 V136G mouse model and performed behavioral tests with these mice. RESULTS CALHM2 V136G mutation (rs232660) is significantly associated with AD. V136G mutation resulted in loss of the CALHM2 ATP-release function in astrocytes and impaired synaptic plasticity. Mice homozygous for the Calhm2 V136G allele displayed depressive-like behaviors that were rescued by administration of exogenous ATP. Moreover, Calhm2 V136G mutation predisposed mice to cognitive decline in old age. DISCUSSION CALHM2 dysfunction is a biologically relevant mechanism that may contribute to the observed clinical correlation between depression and AD.
Collapse
Affiliation(s)
- Yang Liao
- The Brain Science Center, Beijing Institute of Basic Medical Sciences, Beijing, China
| | - Yingyi Wang
- The Brain Science Center, Beijing Institute of Basic Medical Sciences, Beijing, China
| | - Qing-Qing Tao
- Department of Neurology in Second Affiliated Hospital, and Key Laboratory of Medical Neurobiology of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China
| | - Chaoguang Yang
- The Brain Science Center, Beijing Institute of Basic Medical Sciences, Beijing, China
| | - Jinlei Wang
- The Brain Science Center, Beijing Institute of Basic Medical Sciences, Beijing, China
| | - Jinbo Cheng
- Center on Translational Neuroscience, College of Life and Environmental Sciences, Minzu University of China, Beijing, China
| | - Jun Ma
- The Brain Science Center, Beijing Institute of Basic Medical Sciences, Beijing, China
| | - Zhi-Ying Wu
- Department of Neurology in Second Affiliated Hospital, and Key Laboratory of Medical Neurobiology of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China
| | - Rui-Yuan Pan
- Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangdong Province Key Laboratory of Psychiatric Disorders, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Zengqiang Yuan
- The Brain Science Center, Beijing Institute of Basic Medical Sciences, Beijing, China
| |
Collapse
|
7
|
Shim SS, Berglund K, Yu SP. Lithium: An Old Drug for New Therapeutic Strategy for Alzheimer's Disease and Related Dementia. NEURODEGENER DIS 2023; 23:1-12. [PMID: 37666228 DOI: 10.1159/000533797] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Accepted: 08/23/2023] [Indexed: 09/06/2023] Open
Abstract
BACKGROUND Although Alzheimer's disease (AD) is the most common form of dementia, the effective treatment of AD is not available currently. Multiple trials of drugs, which were developed based on the amyloid hypothesis of AD, have not been highly successful to improve cognitive and other symptoms in AD patients, suggesting that it is necessary to explore additional and alternative approaches for the disease-modifying treatment of AD. The diverse lines of evidence have revealed that lithium reduces amyloid and tau pathology, attenuates neuronal loss, enhances synaptic plasticity, and improves cognitive function. Clinical studies have shown that lithium reduces the risk of AD and deters the progress of mild cognitive impairment and early AD. SUMMARY Our recent study has revealed that lithium stabilizes disruptive calcium homeostasis, and subsequently, attenuates the downstream neuropathogenic processes of AD. Through these therapeutic actions, lithium produces therapeutic effects on AD with potential to modify the disease process. This review critically analyzed the preclinical and clinical studies for the therapeutic effects of lithium on AD. We suggest that disruptive calcium homeostasis is likely to be the early neuropathological mechanism of AD, and the stabilization of disruptive calcium homeostasis by lithium would be associated with its therapeutic effects on neuropathology and cognitive deficits in AD. KEY MESSAGES Lithium is likely to be efficacious for AD as a disease-modifying drug by acting on multiple neuropathological targets including disruptive calcium homeostasis.
Collapse
Affiliation(s)
- Seong Sool Shim
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, Georgia, USA
- Mental Health Service Line, Department of Veteran's Affair, Atlanta VA Medical Center, Decatur, Georgia, USA
- Department of Veteran's Affair, Center for Visual and Neurocognitive Rehabilitation, Atlanta VA Medical Center, Decatur, Georgia, USA
| | - Ken Berglund
- Department of Veteran's Affair, Center for Visual and Neurocognitive Rehabilitation, Atlanta VA Medical Center, Decatur, Georgia, USA
- Department of Neurosurgery, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Shan Ping Yu
- Department of Veteran's Affair, Center for Visual and Neurocognitive Rehabilitation, Atlanta VA Medical Center, Decatur, Georgia, USA
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, Georgia, USA
| |
Collapse
|
8
|
Zheng H, Wang Z, Jia Q. Simultaneous Profiling of Palmitoylomics and Glycomics with Photo/pH Dual-Responsive Magnetic Nanocomposites. SMALL METHODS 2023; 7:e2300254. [PMID: 37231570 DOI: 10.1002/smtd.202300254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 04/10/2023] [Indexed: 05/27/2023]
Abstract
Following an in-depth examination of a single type of protein posttranslational modification, the synergistic analysis of two or more modification types has gradually emerged as a focal point in proteomic research. Palmitoylation and glycosylation are both critical for protein, implicated in carcinogenesis and inflammation. In this study, novel dual-responsive magnetic nanocomposites that serve as an ideal platform for the sequential or simultaneous enrichment of palmitoyl and glycopeptides are reported. The nanocomposites denoted as magDVS-VBA are constructed by modifying magnetic nanoparticles with azobenzene and divinyl sulfone (DVS), and self-assembled with 4-vinylbenzeneboronic acid (VBA)-immobilized β-cyclodextrin, which responds to light. The incorporated DVS component possesses the ability to recognize palmitoyl or glycopeptides under different pH conditions, whereas the introduction of VBA enhances the affinity of the nanocomposite for glycopeptides. Notably, magDVS-VBA exhibits flexible photo-, pH-, and magnetic-responsive capabilities, enabling the simultaneous recognition of hydrophobic palmitoyl peptides and hydrophilic glycopeptides for the first time. The developed platform demonstrates high specificity for sensitive palmitoylomics and glycomics analysis of mouse liver tissue, providing an effective method for studying of their crosstalk, and potential implications in clinical applications.
Collapse
Affiliation(s)
- Haijiao Zheng
- College of Chemistry, Jilin University, Changchun, 130012, China
| | - Zirui Wang
- College of Chemistry, Jilin University, Changchun, 130012, China
| | - Qiong Jia
- College of Chemistry, Jilin University, Changchun, 130012, China
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, College of Life Sciences, Jilin University, Changchun, 130012, China
| |
Collapse
|
9
|
Chen GL, Li J, Zhang J, Zeng B. To Be or Not to Be an Ion Channel: Cryo-EM Structures Have a Say. Cells 2023; 12:1870. [PMID: 37508534 PMCID: PMC10378246 DOI: 10.3390/cells12141870] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 07/13/2023] [Accepted: 07/16/2023] [Indexed: 07/30/2023] Open
Abstract
Ion channels are the second largest class of drug targets after G protein-coupled receptors. In addition to well-recognized ones like voltage-gated Na/K/Ca channels in the heart and neurons, novel ion channels are continuously discovered in both excitable and non-excitable cells and demonstrated to play important roles in many physiological processes and diseases such as developmental disorders, neurodegenerative diseases, and cancer. However, in the field of ion channel discovery, there are an unignorable number of published studies that are unsolid and misleading. Despite being the gold standard of a functional assay for ion channels, electrophysiological recordings are often accompanied by electrical noise, leak conductance, and background currents of the membrane system. These unwanted signals, if not treated properly, lead to the mischaracterization of proteins with seemingly unusual ion-conducting properties. In the recent ten years, the technical revolution of cryo-electron microscopy (cryo-EM) has greatly advanced our understanding of the structures and gating mechanisms of various ion channels and also raised concerns about the pore-forming ability of some previously identified channel proteins. In this review, we summarize cryo-EM findings on ion channels with molecular identities recognized or disputed in recent ten years and discuss current knowledge of proposed channel proteins awaiting cryo-EM analyses. We also present a classification of ion channels according to their architectures and evolutionary relationships and discuss the possibility and strategy of identifying more ion channels by analyzing structures of transmembrane proteins of unknown function. We propose that cross-validation by electrophysiological and structural analyses should be essentially required for determining molecular identities of novel ion channels.
Collapse
Affiliation(s)
- Gui-Lan Chen
- Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou 646000, China
| | - Jian Li
- College of Pharmaceutical Sciences, Gannan Medical University, Ganzhou 341000, China
| | - Jin Zhang
- School of Basic Medical Sciences, Nanchang University, Nanchang 330031, China
| | - Bo Zeng
- Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou 646000, China
| |
Collapse
|
10
|
Syrjänen JL, Epstein M, Gómez R, Furukawa H. Structure of human CALHM1 reveals key locations for channel regulation and blockade by ruthenium red. Nat Commun 2023; 14:3821. [PMID: 37380652 PMCID: PMC10307800 DOI: 10.1038/s41467-023-39388-3] [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: 10/10/2022] [Accepted: 06/08/2023] [Indexed: 06/30/2023] Open
Abstract
Calcium homeostasis modulator 1 (CALHM1) is a voltage-dependent channel involved in neuromodulation and gustatory signaling. Despite recent progress in the structural biology of CALHM1, insights into functional regulation, pore architecture, and channel blockade remain limited. Here we present the cryo-EM structure of human CALHM1, revealing an octameric assembly pattern similar to the non-mammalian CALHM1s and the lipid-binding pocket conserved across species. We demonstrate by MD simulations that this pocket preferentially binds a phospholipid over cholesterol to stabilize its structure and regulate the channel activities. Finally, we show that residues in the amino-terminal helix form the channel pore that ruthenium red binds and blocks.
Collapse
Affiliation(s)
- Johanna L Syrjänen
- W.M. Keck Structural Biology Laboratory, Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, NY, 11724, USA
| | - Max Epstein
- W.M. Keck Structural Biology Laboratory, Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, NY, 11724, USA
| | - Ricardo Gómez
- W.M. Keck Structural Biology Laboratory, Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, NY, 11724, USA
| | - Hiro Furukawa
- W.M. Keck Structural Biology Laboratory, Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, NY, 11724, USA.
| |
Collapse
|
11
|
Sharma A, Sari E, Lee Y, Patel S, Brenner M, Marambaud P, Wang P. Extracellular CIRP Induces Calpain Activation in Neurons via PLC-IP 3-Dependent Calcium Pathway. Mol Neurobiol 2023; 60:3311-3328. [PMID: 36853429 PMCID: PMC10506840 DOI: 10.1007/s12035-023-03273-3] [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: 11/07/2022] [Accepted: 02/16/2023] [Indexed: 03/01/2023]
Abstract
Abnormal calcium homeostasis, activation of protease calpain, generation of p25 and hyperactivation of cyclin-dependent kinase 5 (Cdk5) have all been implicated in the pathogenesis of neurogenerative diseases including Alzheimer's disease. We have recently shown that extracellular cold-inducible RNA-binding protein (eCIRP) induces Cdk5 activation via p25. However, the precise molecular mechanism by which eCIRP regulates calcium signaling and calpain remains to be addressed. We hypothesized that eCIRP regulates p25 via Ca2+-dependent calpain activation. eCIRP increased calpain activity and decreased the endogenous calpain inhibitor calpastatin in Neuro 2a (N2a) cells. Calpain inhibition with calpeptin attenuated eCIRP-induced calpain activity and p25. eCIRP specifically upregulated cytosolic calpain 1, and calpain 1 silencing attenuated the eCIRP-induced increase in p25. eCIRP stimulation increased cytosolic free Ca2+, especially in hippocampal neuronal HT22 cells, which was attenuated by the eCIRP inhibitor Compound 23 (C23). Endoplasmic reticulum (ER) inositol 1,4,5-trisphosphate receptor (IP3R) inhibition using 2-aminoethoxy-diphenyl-borate or xestospongin-C (X-C), interleukin-6 receptor alpha (IL-6Rα)-neutralization, and phospholipase C (PLC) inhibition with U73122 attenuated eCIRP-induced Ca2+ increase, while Ca2+ influx across the plasma membrane remained unaffected by eCIRP. Finally, C23, IL-6Rα antibody, U73122 and X-C attenuated eCIRP-induced p25 in HT-22 cells. In conclusion, the current study uncovers eCIRP-triggered Ca2+ release from ER stores in an IL-6Rα/PLC/IP3-dependent manner as a novel molecular mechanism underlying eCIRP's induction of Cdk5 activity and potential involvement in neurodegeneration.
Collapse
Affiliation(s)
- Archna Sharma
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, 350 Community Dr, Manhasset, NY, 11030, USA
- Department of Molecular Medicine, Zucker School of Medicine at Hofstra/Northwell, Manhasset, NY, 11030, USA
| | - Ezgi Sari
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, 350 Community Dr, Manhasset, NY, 11030, USA
| | - Yongchan Lee
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, 350 Community Dr, Manhasset, NY, 11030, USA
| | - Shivani Patel
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, 350 Community Dr, Manhasset, NY, 11030, USA
| | - Max Brenner
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, 350 Community Dr, Manhasset, NY, 11030, USA
- Department of Molecular Medicine, Zucker School of Medicine at Hofstra/Northwell, Manhasset, NY, 11030, USA
- Department of Surgery, Zucker School of Medicine at Hofstra/Northwell, Manhasset, NY, 11030, USA
| | - Philippe Marambaud
- Department of Molecular Medicine, Zucker School of Medicine at Hofstra/Northwell, Manhasset, NY, 11030, USA
- The Litwin-Zucker Center for Alzheimer's Disease Research, The Feinstein Institutes for Medical Research, Manhasset, NY, 11030, USA
| | - Ping Wang
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, 350 Community Dr, Manhasset, NY, 11030, USA.
- Department of Molecular Medicine, Zucker School of Medicine at Hofstra/Northwell, Manhasset, NY, 11030, USA.
- Department of Surgery, Zucker School of Medicine at Hofstra/Northwell, Manhasset, NY, 11030, USA.
| |
Collapse
|
12
|
Peng L, Renauer PA, Ye L, Yang L, Park JJ, Chow RD, Zhang Y, Lin Q, Bai M, Sanchez A, Zhang Y, Lam SZ, Chen S. Perturbomics of tumor-infiltrating NK cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.14.532653. [PMID: 36993337 PMCID: PMC10055047 DOI: 10.1101/2023.03.14.532653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/19/2023]
Abstract
Natural killer (NK) cells are an innate immune cell type that serves at the first level of defense against pathogens and cancer. NK cells have clinical potential, however, multiple current limitations exist that naturally hinder the successful implementation of NK cell therapy against cancer, including their effector function, persistence, and tumor infiltration. To unbiasedly reveal the functional genetic landscape underlying critical NK cell characteristics against cancer, we perform perturbomics mapping of tumor infiltrating NK cells by joint in vivo AAV-CRISPR screens and single cell sequencing. We establish a strategy with AAV-SleepingBeauty(SB)- CRISPR screening leveraging a custom high-density sgRNA library targeting cell surface genes, and perform four independent in vivo tumor infiltration screens in mouse models of melanoma, breast cancer, pancreatic cancer, and glioblastoma. In parallel, we characterize single-cell transcriptomic landscapes of tumor-infiltrating NK cells, which identifies previously unexplored sub-populations of NK cells with distinct expression profiles, a shift from immature to mature NK (mNK) cells in the tumor microenvironment (TME), and decreased expression of mature marker genes in mNK cells. CALHM2, a calcium homeostasis modulator that emerges from both screen and single cell analyses, shows both in vitro and in vivo efficacy enhancement when perturbed in chimeric antigen receptor (CAR)-NK cells. Differential gene expression analysis reveals that CALHM2 knockout reshapes cytokine production, cell adhesion, and signaling pathways in CAR- NKs. These data directly and systematically map out endogenous factors that naturally limit NK cell function in the TME to offer a broad range of cellular genetic checkpoints as candidates for future engineering to enhance NK cell-based immunotherapies.
Collapse
|
13
|
Abstract
Salt taste, the taste of sodium chloride (NaCl), is mechanistically one of the most complex and puzzling among basic tastes. Sodium has essential functions in the body but causes harm in excess. Thus, animals use salt taste to ingest the right amount of salt, which fluctuates by physiological needs: typically, attraction to low salt concentrations and rejection of high salt. This concentration-valence relationship is universally observed in terrestrial animals, and research has revealed complex peripheral codes for NaCl involving multiple taste pathways of opposing valence. Sodium-dependent and -independent pathways mediate attraction and aversion to NaCl, respectively. Gustatory sensors and cells that transduce NaCl have been uncovered, along with downstream signal transduction and neurotransmission mechanisms. However, much remains unknown. This article reviews classical and recent advances in our understanding of the molecular and cellular mechanisms underlying salt taste in mammals and insects and discusses perspectives on human salt taste.
Collapse
Affiliation(s)
- Akiyuki Taruno
- Department of Molecular Cell Physiology, Kyoto Prefectural University of Medicine, Kyoto, Japan; .,Japan Science and Technology Agency, CREST, Saitama, Japan
| | - Michael D Gordon
- Department of Zoology and Life Sciences Institute, The University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
14
|
Joshi M, Joshi S, Khambete M, Degani M. Role of calcium dysregulation in Alzheimer's disease and its therapeutic implications. Chem Biol Drug Des 2023; 101:453-468. [PMID: 36373976 DOI: 10.1111/cbdd.14175] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 10/29/2022] [Accepted: 11/02/2022] [Indexed: 11/16/2022]
Abstract
The increasing incidence of Alzheimer's disease (AD) coupled with the lack of therapeutics to address the underlying pathology of the disease has necessitated the need for exploring newer targets. Calcium dysregulation represents a relatively newer target associated with AD. Ca+2 serves as an important cellular messenger in neurons. The concentration of the Ca+2 ion needs to be regulated at optimal concentrations intracellularly for normal functioning of the neurons. This is achieved with the help of mitochondria, endoplasmic reticulum, and neuronal plasma membrane channel proteins. Disruption in normal calcium homeostasis can induce formation of amyloid beta plaques, accumulation of neurofibrillary tangles, and dysfunction of synaptic plasticity, which in turn can affect calcium homeostasis further, thus forming a vicious cycle. Hence, understanding calcium dysregulation can prove to be a key to develop newer therapeutics. This review provides detailed account of physiology of calcium homeostasis and its dysregulation associated with AD. Further, with an understanding of various receptors and organelles involved in these pathways, the review also discusses various calcium channel blockers explored in AD hand in hand with some multitarget molecules addressing calcium as one of the targets.
Collapse
Affiliation(s)
- Maithili Joshi
- Department of Pharmaceutical Sciences and Technology, Institute of Chemical Technology, Mumbai, India
| | - Siddhi Joshi
- Department of Pharmaceutical Sciences and Technology, Institute of Chemical Technology, Mumbai, India
| | - Mihir Khambete
- Department of Pharmaceutical Sciences and Technology, Institute of Chemical Technology, Mumbai, India
| | - Mariam Degani
- Department of Pharmaceutical Sciences and Technology, Institute of Chemical Technology, Mumbai, India
| |
Collapse
|
15
|
de los Ríos C, Viejo L, Carretero VJ, Juárez NH, Cruz-Martins N, Hernández-Guijo JM. Promising Molecular Targets in Pharmacological Therapy for Neuronal Damage in Brain Injury. Antioxidants (Basel) 2023; 12:118. [PMID: 36670980 PMCID: PMC9854812 DOI: 10.3390/antiox12010118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Revised: 12/19/2022] [Accepted: 12/27/2022] [Indexed: 01/05/2023] Open
Abstract
The complex etiopathogenesis of brain injury associated with neurodegeneration has sparked a lot of studies in the last century. These clinical situations are incurable, and the currently available therapies merely act on symptoms or slow down the course of the diseases. Effective methods are being sought with an intent to modify the disease, directly acting on the properly studied targets, as well as to contribute to the development of effective therapeutic strategies, opening the possibility of refocusing on drug development for disease management. In this sense, this review discusses the available evidence for mitochondrial dysfunction induced by Ca2+ miscommunication in neurons, as well as how targeting phosphorylation events may be used to modulate protein phosphatase 2A (PP2A) activity in the treatment of neuronal damage. Ca2+ tends to be the catalyst for mitochondrial dysfunction, contributing to the synaptic deficiency seen in brain injury. Additionally, emerging data have shown that PP2A-activating drugs (PADs) suppress inflammatory responses by inhibiting different signaling pathways, indicating that PADs may be beneficial for the management of neuronal damage. In addition, a few bioactive compounds have also triggered the activation of PP2A-targeted drugs for this treatment, and clinical studies will help in the authentication of these compounds. If the safety profiles of PADs are proven to be satisfactory, there is a case to be made for starting clinical studies in the setting of neurological diseases as quickly as possible.
Collapse
Affiliation(s)
- Cristóbal de los Ríos
- Department of Pharmacology and Therapeutic and Teófilo Hernando Institute, Faculty of Medicine, University Autónoma de Madrid, C/. Arzobispo Morcillo 4, 28029 Madrid, Spain
- Departamento de Ciencias Básicas de la Salud, University Rey Juan Carlos, Avda. Atenas s/n, 28922 Alcorcón, Spain
| | - Lucía Viejo
- Department of Pharmacology and Therapeutic and Teófilo Hernando Institute, Faculty of Medicine, University Autónoma de Madrid, C/. Arzobispo Morcillo 4, 28029 Madrid, Spain
| | - Victoria Jiménez Carretero
- Department of Pharmacology and Therapeutic and Teófilo Hernando Institute, Faculty of Medicine, University Autónoma de Madrid, C/. Arzobispo Morcillo 4, 28029 Madrid, Spain
| | - Natalia Hernández Juárez
- Department of Pharmacology and Therapeutic and Teófilo Hernando Institute, Faculty of Medicine, University Autónoma de Madrid, C/. Arzobispo Morcillo 4, 28029 Madrid, Spain
| | - Natália Cruz-Martins
- Faculty of Medicine, Institute for Research and Innovation in Health (i3S), University of Porto, 4200-319 Porto, Portugal
- Institute for Research and Advanced Training in Health Sciences and Technologies, Rua Central de Gandra, 1317, 4585-116 Gandra, Portugal
| | - Jesús M. Hernández-Guijo
- Department of Pharmacology and Therapeutic and Teófilo Hernando Institute, Faculty of Medicine, University Autónoma de Madrid, C/. Arzobispo Morcillo 4, 28029 Madrid, Spain
- Ramón y Cajal Institute for Health Research, IRYCIS, Hospital Ramón y Cajal, Ctra. de Colmenar Viejo, Km. 9,100, 28029 Madrid, Spain
| |
Collapse
|
16
|
Bo X, Xie F, Zhang J, Gu R, Li X, Li S, Yuan Z, Cheng J. Deletion of Calhm2 alleviates MPTP-induced Parkinson's disease pathology by inhibiting EFHD2-STAT3 signaling in microglia. Theranostics 2023; 13:1809-1822. [PMID: 37064868 PMCID: PMC10091876 DOI: 10.7150/thno.83082] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Accepted: 02/28/2023] [Indexed: 04/18/2023] Open
Abstract
Background: Neuroinflammation is involved in the development of Parkinson's disease (PD). Calhm2 plays an important role in the development of microglial inflammation, but whether Calhm2 is involved in PD and its regulatory mechanisms are unclear. Methods: To study the role of Calhm2 in the development of PD, we utilized conventional Calhm2 knockout mice, microglial Calhm2 knockout mice and neuronal Calhm2 knockout mice, and established the MPTP-induced PD mice model. Moreover, a series of methods including behavioral test, immunohistochemistry, immunofluorescence, real-time polymerase chain reaction, western blot, mass spectrometry analysis and co-immunoprecipitation were utilized to study the regulatory mechanisms. Results: We found that both conventional Calhm2 knockout and microglial Calhm2 knockout significantly reduced dopaminergic neuronal loss, and decreased microglial numbers, thereby improving locomotor performance in PD model mice. Mechanistically, we found that Calhm2 interacted with EFhd2 and regulated downstream STAT3 signaling in microglia. Knockdown of Calhm2 or EFhd2 both inhibited downstream STAT3 signaling and inflammatory cytokine levels in microglia. Conclusion: We demonstrate the important role of Calhm2 in microglial activation and the pathology of PD, thus providing a potential therapeutic target for microglia-mediated neuroinflammation-related diseases.
Collapse
Affiliation(s)
- Xuena Bo
- Center on Translational Neuroscience, College of Life & Environmental Science, Minzu University of China, Beijing, 100081, China
| | - Fei Xie
- The Brain Science Center, Beijing Institute of Basic Medical Sciences, Beijing, 100850, China
- School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China
| | - Jingdan Zhang
- Center on Translational Neuroscience, College of Life & Environmental Science, Minzu University of China, Beijing, 100081, China
| | - Runze Gu
- Center on Translational Neuroscience, College of Life & Environmental Science, Minzu University of China, Beijing, 100081, China
| | - Xiaoheng Li
- The Brain Science Center, Beijing Institute of Basic Medical Sciences, Beijing, 100850, China
| | - Shuoshuo Li
- The Brain Science Center, Beijing Institute of Basic Medical Sciences, Beijing, 100850, China
| | - Zengqiang Yuan
- The Brain Science Center, Beijing Institute of Basic Medical Sciences, Beijing, 100850, China
- ✉ Corresponding authors: Jinbo Cheng, PhD. Center on Translational Neuroscience, College of Life & Environmental Science, Minzu University of China, Beijing, 100081, China. E-mail: . Zengqiang Yuan, PhD. The Brain Science Center, Beijing Institute of Basic Medical Sciences, Beijing, 100850, China. E-mail: , or
| | - Jinbo Cheng
- Center on Translational Neuroscience, College of Life & Environmental Science, Minzu University of China, Beijing, 100081, China
- The Brain Science Center, Beijing Institute of Basic Medical Sciences, Beijing, 100850, China
- ✉ Corresponding authors: Jinbo Cheng, PhD. Center on Translational Neuroscience, College of Life & Environmental Science, Minzu University of China, Beijing, 100081, China. E-mail: . Zengqiang Yuan, PhD. The Brain Science Center, Beijing Institute of Basic Medical Sciences, Beijing, 100850, China. E-mail: , or
| |
Collapse
|
17
|
Kwon JW, Jeon YK, Kim SJ. Bidirectional sensitivity of CALHM1 channel to protons from both sides of plasma membrane. Am J Physiol Cell Physiol 2023; 324:C98-C112. [PMID: 36409172 DOI: 10.1152/ajpcell.00250.2022] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Calcium homeostasis modulator 1 (CALHM1), a newly discovered voltage-dependent nonselective ion channel, has drawn attention for its role in neuronal activity and taste sensation. Its sluggish voltage-dependent activation is facilitated by lowering extracellular Ca2+ concentration ([Ca2+]e). Here, we investigated the effects of extracellular and intracellular pH (pHe and pHi) on human CALHM1. When normalized to the amplitude of the CALHM1 current (ICALHM1) under whole cell patch clamp at symmetrical pH 7.4, ICALHM1 decreased at acidic pHe or pHi, whereas it sharply increased at alkaline pHe or pHi. The effects of pH were preserved in the inside-out configuration. The voltage dependence of ICALHM1 showed leftward and rightward shifts at alkaline and acidic pHe and pHi, respectively. Site-directed mutagenesis of the water-accessible charged residues of the pore and nearby domains revealed that E17, K229, E233, D257, and E259 are nonadditively responsible for facilitation at alkaline pHi. Identification of the pHe-sensing residue was not possible because mutation of putative residues impaired membrane expression, resulting in undetectable ICALHM1. Alkaline pHe-dependent facilitation appeared gradually with depolarization, suggesting that the sensitivity to pHe might be due to H+ diffusion through the open-state CALHM1. At pHe 6.2, decreased [Ca2+]e could not recover the inhibited ICALHM1 but further augmented the increased ICALHM1 at pHe 8.6, suggesting that unidentified common residues might contribute to the [Ca2+]e and acidic pHe. This study is the first, to our knowledge, to demonstrate the remarkable pH sensitivity of CALHM1, which might contribute to the pH-dependent modulation of neuronal excitability or taste sensation.
Collapse
Affiliation(s)
- Jae Won Kwon
- Department of Physiology, Seoul National University College of Medicine, Seoul, Republic of Korea.,Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Young Keul Jeon
- Department of Physiology, Seoul National University College of Medicine, Seoul, Republic of Korea.,Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Sung Joon Kim
- Department of Physiology, Seoul National University College of Medicine, Seoul, Republic of Korea.,Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Republic of Korea.,Ischemic/Hypoxic Disease Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
18
|
Clupper M, Gill R, Elsayyid M, Touroutine D, Caplan JL, Tanis JE. Kinesin-2 motors differentially impact biogenesis of extracellular vesicle subpopulations shed from sensory cilia. iScience 2022; 25:105262. [PMID: 36304122 PMCID: PMC9593189 DOI: 10.1016/j.isci.2022.105262] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 07/13/2022] [Accepted: 09/26/2022] [Indexed: 01/21/2023] Open
Abstract
Extracellular vesicles (EVs) are bioactive lipid-bilayer enclosed particles released from nearly all cells. One specialized site for EV shedding is the primary cilium. Here, we discover the conserved ion channel CLHM-1 as a ciliary EV cargo. Imaging of EVs released from sensory neuron cilia of Caenorhabditis elegans expressing fluorescently tagged CLHM-1 and TRP polycystin-2 channel PKD-2 shows enrichment of these cargoes in distinct EV subpopulations that are differentially shed in response to mating partner availability. PKD-2 alone is present in EVs shed from the cilium distal tip, whereas CLHM-1 EVs bud from a secondary site(s), including the ciliary base. Heterotrimeric and homodimeric kinesin-2 motors have discrete impacts on PKD-2 and CLHM-1 colocalization in both cilia and EVs. Total loss of kinesin-2 activity decreases shedding of PKD-2 but not CLHM-1 EVs. Our data demonstrate that anterograde intraflagellar transport is required for selective enrichment of protein cargoes into heterogeneous EVs with different signaling potentials.
Collapse
Affiliation(s)
- Michael Clupper
- Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA
| | - Rachael Gill
- Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA
| | - Malek Elsayyid
- Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA
| | - Denis Touroutine
- Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA
| | - Jeffrey L. Caplan
- Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA
- Department of Plant and Soil Sciences, University of Delaware, Newark, DE 19716, USA
| | - Jessica E. Tanis
- Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA
| |
Collapse
|
19
|
Tholen P, Brown CN, Keil C, Bayir A, Zeng HH, Haase H, Thompson RB, Lengyel I, Yücesan G. A 2,7-dichlorofluorescein derivative to monitor microcalcifications. MOLECULAR SYSTEMS DESIGN & ENGINEERING 2022; 7:1415-1421. [PMID: 37927331 PMCID: PMC10624163 DOI: 10.1039/d2me00185c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/07/2023]
Abstract
Herein, we report the crystal structure of 2,7-dichlorofluorescein methyl ester (DCF-ME) and its fluorescence response to hydroxyapatite binding. The reported fluorophore is very selective for staining the bone matrix and provides turn-on fluorescence upon hydroxyapatite binding. The reported fluorophore can readily pass the cell membrane of the C2C12 cell line, and it is non-toxic for the cell line. The reported fluorophore DCF-ME may find applications in monitoring bone remodeling and microcalcification as an early diagnosis tool for breast cancer and age-related macular degeneration.
Collapse
Affiliation(s)
- Patrik Tholen
- Institute for Food Chemistry and Toxicology, Germany, Technische Universität Berlin, Gustav-Meyer-Allee 25, 13355 Berlin, Germany
| | - Connor N Brown
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, Belfast BT9 7BL, UK
| | - Claudia Keil
- Institute for Food Chemistry and Toxicology, Germany, Technische Universität Berlin, Gustav-Meyer-Allee 25, 13355 Berlin, Germany
| | - Ali Bayir
- The Department of Chemistry, Yıldız Technical University, 34220, Esenler, Istanbul, Turkey
| | - Hui-Hui Zeng
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, Maryland 21201, USA
| | - Hajo Haase
- Institute for Food Chemistry and Toxicology, Germany, Technische Universität Berlin, Gustav-Meyer-Allee 25, 13355 Berlin, Germany
| | - Richard B Thompson
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, Maryland 21201, USA
| | - Imre Lengyel
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, Belfast BT9 7BL, UK
| | - Gündoğ Yücesan
- Institute for Food Chemistry and Toxicology, Germany, Technische Universität Berlin, Gustav-Meyer-Allee 25, 13355 Berlin, Germany
| |
Collapse
|
20
|
Zhai Z, Xie D, Qin T, Zhong Y, Xu Y, Sun T. Effect and Mechanism of Exogenous Melatonin on Cognitive Deficits in Animal Models of Alzheimer's Disease: A Systematic review and Meta-analysis. Neuroscience 2022; 505:91-110. [PMID: 36116555 DOI: 10.1016/j.neuroscience.2022.09.012] [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: 06/14/2022] [Revised: 09/05/2022] [Accepted: 09/08/2022] [Indexed: 12/09/2022]
Abstract
Melatonin (MT) has been reported to control and prevent Alzheimer's disease (AD) in the clinic; however, the effect and mechanism of MT on AD have not been specifically described. Therefore, the main purpose of this meta-analysis was to explore the effect and mechanism of MT on AD models by studying behavioural indicators and pathological features. Seven databases were searched and 583 articles were retrieved. Finally, nine studies (13 analyses, 294 animals) were included according to pre-set criteria. Three authors independently judged the selected literature and the methodological quality. Meta-analysis showed that MT markedly ameliorated the learning ability by reducing the escape latency (EL), and the memory deficit was significantly corrected by increasing the dwell time in the target quadrant and crossings over the platform location in the Morris Water Maze (MWM). Among the pathological features, subgroup analysis found that MT may ease the symptoms of AD mainly by reducing the deposition of Aβ40 and Aβ42 in the cortex. In addition, MT exerted a superior effect on ameliorating the learning ability of senescence-related and metabolic AD models, and corrected the memory deficit of the toxin-induced AD model. The study was registered at PROSPERO (CRD42021226594).
Collapse
Affiliation(s)
- Zhenwei Zhai
- School of Medical Information Engineering, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| | - Danni Xie
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| | - Tao Qin
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| | - Yanmei Zhong
- School of Medical Information Engineering, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| | - Ying Xu
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China.
| | - Tao Sun
- School of Medical Information Engineering, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China; State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| |
Collapse
|
21
|
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: 51] [Impact Index Per Article: 25.5] [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
|
22
|
Singh D. Astrocytic and microglial cells as the modulators of neuroinflammation in Alzheimer's disease. J Neuroinflammation 2022; 19:206. [PMID: 35978311 PMCID: PMC9382837 DOI: 10.1186/s12974-022-02565-0] [Citation(s) in RCA: 127] [Impact Index Per Article: 63.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Accepted: 08/06/2022] [Indexed: 12/17/2022] Open
Abstract
Neuroinflammation is instigated by the misfiring of immune cells in the central nervous system (CNS) involving microglia and astrocytes as key cell-types. Neuroinflammation is a consequence of CNS injury, infection, toxicity, or autoimmunity. It is favorable as well as a detrimental process for neurodevelopment and associated processes. Transient activation of inflammatory response involving release of cytokines and growth factors positively affects the development and post-injury tissue. However, chronic or uncontrolled inflammatory responses may lead to various neurodegenerative diseases, including Alzheimer's disease (AD), Parkinson's disease (PD), amyotrophic lateral sclerosis, and multiple sclerosis. These diseases have variable clinical and pathological features, but are underlaid by the aggregation of misfolded proteins with a cytotoxic effect. Notably, abnormal activation of glial cells could mediate neuroinflammation, leading to the neurodegenerative condition. Microglia, a type of glial cell, a resident immune cell, form the forefront defense of the CNS immune system. Dysfunctional microglia and astrocyte, a different kind of glial cell with homeostatic function, impairs the protein aggregate (amyloid-beta plaque) clearance in AD. Studies have shown that microglia and astrocytes undergo alterations in their genetic profile, cellular and molecular responses, and thus promote dysfunctional immune cross-talk in AD. Hence, targeting microglia and astrocytes-driven molecular pathways could resolve the particular layers of neuroinflammation and set a reliable therapeutic intervention in AD progression.
Collapse
Affiliation(s)
- Deepali Singh
- National Brain Research Centre, Manesar, Haryana, 122052, India.
| |
Collapse
|
23
|
Callens M, Loncke J, Bultynck G. Dysregulated Ca 2+ Homeostasis as a Central Theme in Neurodegeneration: Lessons from Alzheimer's Disease and Wolfram Syndrome. Cells 2022; 11:cells11121963. [PMID: 35741091 PMCID: PMC9221778 DOI: 10.3390/cells11121963] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 06/06/2022] [Accepted: 06/13/2022] [Indexed: 12/12/2022] Open
Abstract
Calcium ions (Ca2+) operate as important messengers in the cell, indispensable for signaling the underlying numerous cellular processes in all of the cell types in the human body. In neurons, Ca2+ signaling is crucial for regulating synaptic transmission and for the processes of learning and memory formation. Hence, the dysregulation of intracellular Ca2+ homeostasis results in a broad range of disorders, including cancer and neurodegeneration. A major source for intracellular Ca2+ is the endoplasmic reticulum (ER), which has close contacts with other organelles, including mitochondria. In this review, we focus on the emerging role of Ca2+ signaling at the ER–mitochondrial interface in two different neurodegenerative diseases, namely Alzheimer’s disease and Wolfram syndrome. Both of these diseases share some common hallmarks in the early stages, including alterations in the ER and mitochondrial Ca2+ handling, mitochondrial dysfunction and increased Reactive oxygen species (ROS) production. This indicates that similar mechanisms may underly these two disease pathologies and suggests that both research topics might benefit from complementary research.
Collapse
|
24
|
Khare N, Maheshwari SK, Rizvi SMD, Albadrani HM, Alsagaby SA, Alturaiki W, Iqbal D, Zia Q, Villa C, Jha SK, Jha NK, Jha AK. Homology Modelling, Molecular Docking and Molecular Dynamics Simulation Studies of CALMH1 against Secondary Metabolites of Bauhinia variegata to Treat Alzheimer's Disease. Brain Sci 2022; 12:brainsci12060770. [PMID: 35741655 PMCID: PMC9220886 DOI: 10.3390/brainsci12060770] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 05/05/2022] [Accepted: 05/31/2022] [Indexed: 02/06/2023] Open
Abstract
Calcium homeostasis modulator 1 (CALHM1) is a protein responsible for causing Alzheimer’s disease. In the absence of an experimentally designed protein molecule, homology modelling was performed. Through homology modelling, different CALHM1 models were generated and validated through Rampage. To carry out further in silico studies, through molecular docking and molecular dynamics simulation experiments, various flavonoids and alkaloids from Bauhinia variegata were utilised as inhibitors to target the protein (CALHM1). The sequence of CALHM1 was retrieved from UniProt and the secondary structure prediction of CALHM1 was done through CFSSP, GOR4, and SOPMA methods. The structure was identified through LOMETS, MUSTER, and MODELLER and finally, the structures were validated through Rampage. Bauhinia variegata plant was used to check the interaction of alkaloids and flavonoids against CALHM1. The protein and protein–ligand complex were also validated through molecular dynamics simulations studies. The model generated through MODELLER software with 6VAM A was used because this model predicted the best results in the Ramachandran plot. Further molecular docking was performed, quercetin was found to be the most appropriate candidate for the protein molecule with the minimum binding energy of −12.45 kcal/mol and their ADME properties were analysed through Molsoft and Molinspiration. Molecular dynamics simulations showed that CALHM1 and CALHM1–quercetin complex became stable at 2500 ps. It may be seen through the study that quercetin may act as a good inhibitor for treatment. With the help of an in silico study, it was easier to analyse the 3D structure of the protein, which may be scrutinized for the best-predicted model. Quercetin may work as a good inhibitor for treating Alzheimer’s disease, according to in silico research using molecular docking and molecular dynamics simulations, and future in vitro and in vivo analysis may confirm its effectiveness.
Collapse
Affiliation(s)
- Noopur Khare
- Institute of Biosciences and Technology, Shri Ramswaroop Memorial University, Barabanki 225003, Uttar Pradesh, India; (N.K.); (S.K.M.)
- Department of Biotechnology, Dr. A.P.J. Abdul Kalam Technical University, Lucknow 226021, Uttar Pradesh, India
| | - Sanjiv Kumar Maheshwari
- Institute of Biosciences and Technology, Shri Ramswaroop Memorial University, Barabanki 225003, Uttar Pradesh, India; (N.K.); (S.K.M.)
| | - Syed Mohd Danish Rizvi
- Department of Pharmaceutics, College of Pharmacy, University of Hail, Hail 2240, Saudi Arabia;
| | - Hind Muteb Albadrani
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Majmaah University, Majmaah 11952, Saudi Arabia; (H.M.A.); (S.A.A.); (W.A.); (Q.Z.)
| | - Suliman A. Alsagaby
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Majmaah University, Majmaah 11952, Saudi Arabia; (H.M.A.); (S.A.A.); (W.A.); (Q.Z.)
| | - Wael Alturaiki
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Majmaah University, Majmaah 11952, Saudi Arabia; (H.M.A.); (S.A.A.); (W.A.); (Q.Z.)
| | - Danish Iqbal
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Majmaah University, Majmaah 11952, Saudi Arabia; (H.M.A.); (S.A.A.); (W.A.); (Q.Z.)
- Health and Basic Sciences Research Center, Majmaah University, Al Majmaah 15341, Saudi Arabia
- Correspondence: (D.I.); (A.K.J.)
| | - Qamar Zia
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Majmaah University, Majmaah 11952, Saudi Arabia; (H.M.A.); (S.A.A.); (W.A.); (Q.Z.)
- Health and Basic Sciences Research Center, Majmaah University, Al Majmaah 15341, Saudi Arabia
| | - Chiara Villa
- School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy;
| | - Saurabh Kumar Jha
- Department of Biotechnology, School of Engineering and Technology, Sharda University, Greater Noida 201310, Uttar Pradesh, India; (S.K.J.); (N.K.J.)
- Department of Biotechnology, School of Applied & Life Sciences (SALS), Uttaranchal University, Dehradun 248007, Uttarakhand, India
- Department of Biotechnology Engineering and Food Technology, Chandigarh University, Mohali 140413, Punjab, India
| | - Niraj Kumar Jha
- Department of Biotechnology, School of Engineering and Technology, Sharda University, Greater Noida 201310, Uttar Pradesh, India; (S.K.J.); (N.K.J.)
- Department of Biotechnology, School of Applied & Life Sciences (SALS), Uttaranchal University, Dehradun 248007, Uttarakhand, India
- Department of Biotechnology Engineering and Food Technology, Chandigarh University, Mohali 140413, Punjab, India
| | - Abhimanyu Kumar Jha
- Department of Biotechnology, School of Engineering and Technology, Sharda University, Greater Noida 201310, Uttar Pradesh, India; (S.K.J.); (N.K.J.)
- Correspondence: (D.I.); (A.K.J.)
| |
Collapse
|
25
|
Zhong W, Wu A, Berglund K, Gu X, Jiang M, Talati J, Zhao J, Wei L, Yu SP. Pathogenesis of sporadic Alzheimer's disease by deficiency of NMDA receptor subunit GluN3A. Alzheimers Dement 2022; 18:222-239. [PMID: 34151525 PMCID: PMC8685302 DOI: 10.1002/alz.12398] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Revised: 04/23/2021] [Accepted: 05/10/2021] [Indexed: 02/03/2023]
Abstract
The Ca2+ hypothesis for Alzheimer's disease (AD) conceives Ca2+ dyshomeostasis as a common mechanism of AD; the cause of Ca2+ dysregulation, however, is obscure. Meanwhile, hyperactivities of N-Methyl-D-aspartate receptors (NMDARs), the primary mediator of Ca2+ influx, are reported in AD. GluN3A (NR3A) is an NMDAR inhibitory subunit. We hypothesize that GluN3A is critical for sustained Ca2+ homeostasis and its deficiency is pathogenic for AD. Cellular, molecular, and functional changes were examined in adult/aging GluN3A knockout (KO) mice. The GluN3A KO mouse brain displayed age-dependent moderate but persistent neuronal hyperactivity, elevated intracellular Ca2+ , neuroinflammation, impaired synaptic integrity/plasticity, and neuronal loss. GluN3A KO mice developed olfactory dysfunction followed by psychological/cognitive deficits prior to amyloid-β/tau pathology. Memantine at preclinical stage prevented/attenuated AD syndromes. AD patients' brains show reduced GluN3A expression. We propose that chronic "degenerative excitotoxicity" leads to sporadic AD, while GluN3A represents a primary pathogenic factor, an early biomarker, and an amyloid-independent therapeutic target.
Collapse
Affiliation(s)
- Weiwei Zhong
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA 30322, USA
- Center for Visual and Neurocognitive Rehabilitation, Atlanta VA Medical Center, Department of Veteran’s Affair, Decatur, GA 30033, USA
| | - Anika Wu
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Ken Berglund
- Department of Neurosurgery, Emory University School of Medicine, Atlanta, GA 30322, USA
- Center for Visual and Neurocognitive Rehabilitation, Atlanta VA Medical Center, Department of Veteran’s Affair, Decatur, GA 30033, USA
| | - Xiaohuan Gu
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA 30322, USA
- Center for Visual and Neurocognitive Rehabilitation, Atlanta VA Medical Center, Department of Veteran’s Affair, Decatur, GA 30033, USA
| | - Michael Jiang
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA 30322, USA
- Center for Visual and Neurocognitive Rehabilitation, Atlanta VA Medical Center, Department of Veteran’s Affair, Decatur, GA 30033, USA
| | - Jay Talati
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Jingjie Zhao
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Ling Wei
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA 30322, USA
- Department of Neurology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Shan Ping Yu
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA 30322, USA
- Center for Visual and Neurocognitive Rehabilitation, Atlanta VA Medical Center, Department of Veteran’s Affair, Decatur, GA 30033, USA
| |
Collapse
|
26
|
Modeling Alzheimer's Disease in Caenorhabditis elegans. Biomedicines 2022; 10:biomedicines10020288. [PMID: 35203497 PMCID: PMC8869312 DOI: 10.3390/biomedicines10020288] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 01/18/2022] [Accepted: 01/24/2022] [Indexed: 02/04/2023] Open
Abstract
Alzheimer’s disease (AD) is the most frequent cause of dementia. After decades of research, we know the importance of the accumulation of protein aggregates such as β-amyloid peptide and phosphorylated tau. We also know that mutations in certain proteins generate early-onset Alzheimer’s disease (EOAD), and many other genes modulate the disease in its sporadic form. However, the precise molecular mechanisms underlying AD pathology are still unclear. Because of ethical limitations, we need to use animal models to investigate these processes. The nematode Caenorhabditis elegans has received considerable attention in the last 25 years, since the first AD models overexpressing Aβ peptide were described. We review here the main results obtained using this model to study AD. We include works studying the basic molecular mechanisms of the disease, as well as those searching for new therapeutic targets. Although this model also has important limitations, the ability of this nematode to generate knock-out or overexpression models of any gene, single or combined, and to carry out toxicity, recovery or survival studies in short timeframes with many individuals and at low cost is difficult to overcome. We can predict that its use as a model for various diseases will certainly continue to increase.
Collapse
|
27
|
Role of miRNAs in diabetic neuropathy: mechanisms and possible interventions. Mol Neurobiol 2022; 59:1836-1849. [PMID: 35023058 DOI: 10.1007/s12035-021-02662-w] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Accepted: 11/19/2021] [Indexed: 10/19/2022]
Abstract
Accelerating cases of diabetes worldwide have given rise to higher incidences of diabetic complications. MiRNAs, a much-explored class of non-coding RNAs, play a significant role in the pathogenesis of diabetes mellitus by affecting insulin release, β-cell proliferation, and dysfunction. Besides, disrupted miRNAs contribute to various complications, diabetic retinopathy, nephropathy, and neuropathy as well as severe conditions like diabetic foot. MiRNAs regulate various processes involved in diabetic complications like angiogenesis, vascularization, inflammations, and various signaling pathways like PI3K, MAPK, SMAD, and NF-KB signaling pathways. Diabetic neuropathy is the most common diabetic complication, characterized mainly by pain and numbness, especially in the legs and feet. MiRNAs implicated in diabetic neuropathy include mir-9, mir-106a, mir-146a, mir-182, miR-23a and b, miR-34a, and miR-503. The diabetic foot is the most common diabetic neuropathy, often leading to amputations. Mir-203, miR-23c, miR-145, miR-29b and c, miR-126, miR-23a and b, miR-503, and miR-34a are associated with diabetic foot. This review has been compiled to summarize miRNA involved in initiation, progression, and miRNAs affecting various signaling pathways involved in diabetic neuropathy including the diabetic foot. Besides, potential applications of miRNAs as biomarkers and therapeutic targets in this microvascular complication will also be discussed.
Collapse
|
28
|
Ge M, Zhang J, Chen S, Huang Y, Chen W, He L, Zhang Y. Role of Calcium Homeostasis in Alzheimer's Disease. Neuropsychiatr Dis Treat 2022; 18:487-498. [PMID: 35264851 PMCID: PMC8901263 DOI: 10.2147/ndt.s350939] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Accepted: 02/18/2022] [Indexed: 11/23/2022] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disease associated with senile plaques (SP) and neurofibrillary tangles (NFTs) in the brain. With aging of the population, AD has become the most common form of dementia. However, the mechanisms leading to AD are still under investigation, and there are currently no specific drugs for its treatment. Therefore, further study on the pathogenesis of AD to develop new drugs for AD treatment remains a top priority. Several studies have suggested that intracellular calcium homeostasis is dysregulated in AD, and this has been implicated in the deposition of amyloid β (Aβ), hyperphosphorylation of tau protein, abnormal synaptic plasticity, and apoptosis, all of which are involved in the occurrence and development of AD. In addition, some based on pathways linking calcium homeostasis and AD have achieved results in AD treatment. This review comprehensively explores the relationship between calcium homeostasis and the pathogenesis of AD to provide a theoretical basis for the future exploration of AD and the development of novel therapeutic drugs.
Collapse
Affiliation(s)
- Mengqian Ge
- The Second School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, People's Republic of China
| | - Jinghui Zhang
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, People's Republic of China
| | - Simiao Chen
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, People's Republic of China
| | - Yanfen Huang
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, People's Republic of China
| | - Weiyan Chen
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, People's Republic of China
| | - Lan He
- The Second School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, People's Republic of China
| | - Yuyan Zhang
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, People's Republic of China
| |
Collapse
|
29
|
Abstract
Astrocytes are an abundant subgroup of cells in the central nervous system (CNS) that play a critical role in controlling neuronal circuits involved in emotion, learning, and memory. In clinical cases, multiple chronic brain diseases may cause psychosocial and cognitive impairment, such as depression and Alzheimer's disease (AD). For years, complex pathological conditions driven by depression and AD have been widely perceived to contribute to a high risk of disability, resulting in gradual loss of self-care ability, lower life qualities, and vast burden on human society. Interestingly, correlational research on depression and AD has shown that depression might be a prodrome of progressive degenerative neurological disease. As a kind of multifunctional glial cell in the CNS, astrocytes maintain physiological function via supporting neuronal cells, modulating pathologic niche, and regulating energy metabolism. Mounting evidence has shown that astrocytic dysfunction is involved in the progression of depression and AD. We herein review the current findings on the roles and mechanisms of astrocytes in the development of depression and AD, with an implication of potential therapeutic avenue for these diseases by targeting astrocytes.
Collapse
|
30
|
Kwon JW, Jeon YK, Kim J, Kim SJ, Kim SJ. Intramolecular Disulfide Bonds for Biogenesis of CALHM1 Ion Channel Are Dispensable for Voltage-Dependent Activation. Mol Cells 2021; 44:758-769. [PMID: 34711692 PMCID: PMC8560582 DOI: 10.14348/molcells.2021.0131] [Citation(s) in RCA: 3] [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: 05/20/2021] [Revised: 08/19/2021] [Accepted: 09/06/2021] [Indexed: 11/27/2022] Open
Abstract
Calcium homeostasis modulator 1 (CALHM1) is a membrane protein with four transmembrane helices that form an octameric ion channel with voltage-dependent activation. There are four conserved cysteine (Cys) residues in the extracellular domain that form two intramolecular disulfide bonds. We investigated the roles of C42-C127 and C44-C161 in human CALHM1 channel biogenesis and the ionic current (ICALHM1). Replacing Cys with Ser or Ala abolished the membrane trafficking as well as ICALHM1. Immunoblotting analysis revealed dithiothreitol-sensitive multimeric CALHM1, which was markedly reduced in C44S and C161S, but preserved in C42S and C127S. The mixed expression of C42S and wild-type did not show a dominant-negative effect. While the heteromeric assembly of CALHM1 and CALHM3 formed active ion channels, the co-expression of C42S and CALHM3 did not produce functional channels. Despite the critical structural role of the extracellular cysteine residues, a treatment with the membrane-impermeable reducing agent tris(2-carboxyethyl) phosphine (TCEP, 2 mM) did not affect ICALHM1 for up to 30 min. Interestingly, incubation with TCEP (2 mM) for 2-6 h reduced both ICALHM1 and the surface expression of CALHM1 in a time-dependent manner. We propose that the intramolecular disulfide bonds are essential for folding, oligomerization, trafficking and maintenance of CALHM1 in the plasma membrane, but dispensable for the voltage-dependent activation once expressed on the plasma membrane.
Collapse
Affiliation(s)
- Jae Won Kwon
- Department of Physiology, Seoul National University College of Medicine, Seoul 03080, Korea
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Young Keul Jeon
- Department of Physiology, Seoul National University College of Medicine, Seoul 03080, Korea
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Jinsung Kim
- Department of Physiology, Seoul National University College of Medicine, Seoul 03080, Korea
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Sang Jeong Kim
- Department of Physiology, Seoul National University College of Medicine, Seoul 03080, Korea
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Sung Joon Kim
- Department of Physiology, Seoul National University College of Medicine, Seoul 03080, Korea
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Korea
- Ischemic/Hypoxic Disease Institute, Seoul National University College of Medicine, Seoul 03080, Korea
| |
Collapse
|
31
|
Gaete PS, Lillo MA, López W, Liu Y, Jiang W, Luo Y, Harris AL, Contreras JE. A novel voltage-clamp/dye uptake assay reveals saturable transport of molecules through CALHM1 and connexin channels. J Gen Physiol 2021; 152:211474. [PMID: 33074302 PMCID: PMC7579738 DOI: 10.1085/jgp.202012607] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Accepted: 09/23/2020] [Indexed: 12/14/2022] Open
Abstract
Large-pore channels permeable to small molecules such as ATP, in addition to atomic ions, are emerging as important regulators in health and disease. Nonetheless, their mechanisms of molecular permeation and selectivity remain mostly unexplored. Combining fluorescence microscopy and electrophysiology, we developed a novel technique that allows kinetic analysis of molecular permeation through connexin and CALHM1 channels in Xenopus oocytes rendered translucent. Using this methodology, we found that (1) molecular flux through these channels saturates at low micromolar concentrations, (2) kinetic parameters of molecular transport are sensitive to modulators of channel gating, (3) molecular transport and ionic currents can be differentially affected by mutation and gating, and (4) N-terminal regions of these channels control transport kinetics and permselectivity. Our methodology allows analysis of how human disease-causing mutations affect kinetic properties and permselectivity of molecular signaling and enables the study of molecular mechanisms, including selectivity and saturability, of molecular transport in other large-pore channels.
Collapse
Affiliation(s)
- Pablo S Gaete
- Department of Pharmacology, Physiology, and Neuroscience, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ
| | - Mauricio A Lillo
- Department of Pharmacology, Physiology, and Neuroscience, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ
| | - William López
- Department of Pharmacology, Physiology, and Neuroscience, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ
| | - Yu Liu
- Department of Pharmacology, Physiology, and Neuroscience, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ
| | - Wenjuan Jiang
- Department of Pharmaceutical Sciences, College of Pharmacy, Western University of Health Sciences, Pomona, CA
| | - Yun Luo
- Department of Pharmaceutical Sciences, College of Pharmacy, Western University of Health Sciences, Pomona, CA
| | - Andrew L Harris
- Department of Pharmacology, Physiology, and Neuroscience, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ
| | - Jorge E Contreras
- Department of Pharmacology, Physiology, and Neuroscience, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ
| |
Collapse
|
32
|
Ashrafuzzaman M. Artificial Intelligence, Machine Learning and Deep Learning in Ion Channel Bioinformatics. MEMBRANES 2021; 11:membranes11090672. [PMID: 34564489 PMCID: PMC8467682 DOI: 10.3390/membranes11090672] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 08/20/2021] [Accepted: 08/30/2021] [Indexed: 11/28/2022]
Abstract
Ion channels are linked to important cellular processes. For more than half a century, we have been learning various structural and functional aspects of ion channels using biological, physiological, biochemical, and biophysical principles and techniques. In recent days, bioinformaticians and biophysicists having the necessary expertise and interests in computer science techniques including versatile algorithms have started covering a multitude of physiological aspects including especially evolution, mutations, and genomics of functional channels and channel subunits. In these focused research areas, the use of artificial intelligence (AI), machine learning (ML), and deep learning (DL) algorithms and associated models have been found very popular. With the help of available articles and information, this review provide an introduction to this novel research trend. Ion channel understanding is usually made considering the structural and functional perspectives, gating mechanisms, transport properties, channel protein mutations, etc. Focused research on ion channels and related findings over many decades accumulated huge data which may be utilized in a specialized scientific manner to fast conclude pinpointed aspects of channels. AI, ML, and DL techniques and models may appear as helping tools. This review aims at explaining the ways we may use the bioinformatics techniques and thus draw a few lines across the avenue to let the ion channel features appear clearer.
Collapse
Affiliation(s)
- Md Ashrafuzzaman
- Department of Biochemistry, College of Science, King Saud University, Riyadh 11451, Saudi Arabia
| |
Collapse
|
33
|
Syrjanen J, Michalski K, Kawate T, Furukawa H. On the molecular nature of large-pore channels. J Mol Biol 2021; 433:166994. [PMID: 33865869 PMCID: PMC8409005 DOI: 10.1016/j.jmb.2021.166994] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 04/08/2021] [Accepted: 04/08/2021] [Indexed: 12/25/2022]
Abstract
Membrane transport is a fundamental means to control basic cellular processes such as apoptosis, inflammation, and neurodegeneration and is mediated by a number of transporters, pumps, and channels. Accumulating evidence over the last half century has shown that a type of so-called "large-pore channel" exists in various tissues and organs in gap-junctional and non-gap-junctional forms in order to flow not only ions but also metabolites such as ATP. They are formed by a number of protein families with little or no evolutionary linkages including connexin, innexin, pannexin, leucine-rich repeat-containing 8 (LRRC8), and calcium homeostasis modulator (CALHM). This review summarizes the history and concept of large-pore channels starting from connexin gap junction channels to the more recent developments in innexin, pannexin, LRRC8, and CALHM. We describe structural and functional features of large-pore channels that are crucial for their diverse functions on the basis of available structures.
Collapse
Affiliation(s)
- Johanna Syrjanen
- W.M. Keck Structural Biology Laboratory, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | - Kevin Michalski
- W.M. Keck Structural Biology Laboratory, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | - Toshimitsu Kawate
- Department of Molecular Medicine, Fields of Biochemistry, Molecular, and Cell Biology (BMCB), and Biophysics, Cornell University, Ithaca, NY 14853, USA
| | - Hiro Furukawa
- W.M. Keck Structural Biology Laboratory, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA.
| |
Collapse
|
34
|
Cheng J, Dong Y, Ma J, Pan R, Liao Y, Kong X, Li X, Li S, Chen P, Wang L, Yu Y, Yuan Z. Microglial Calhm2 regulates neuroinflammation and contributes to Alzheimer's disease pathology. SCIENCE ADVANCES 2021; 7:7/35/eabe3600. [PMID: 34433553 PMCID: PMC8386937 DOI: 10.1126/sciadv.abe3600] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Accepted: 07/06/2021] [Indexed: 06/13/2023]
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disease in the world. Neuronal calcium dysfunction and microglial-mediated neuroinflammation are closely associated with the development of AD. However, it remains unknown whether calcium dysfunction contributes to microglial activation and, in turn, AD pathology in vivo. In this study, we demonstrated that the expression of calcium homeostasis modulator family protein 2 (Calhm2) is increased in an AD mouse model. In 5×FAD mice carrying five familial AD gene mutations, both conventional knockout of Calhm2 and conditional microglial knockout of Calhm2 significantly reduced amyloid β deposition, neuroinflammation, and cognitive impairments. Mechanistically, knockout of Calhm2 inhibited microglial proinflammatory activity but increased phagocytic activity, leading to restoration of the balance between inflammation and phagocytosis. In addition, knockout of Calhm2 reduced acute LPS-induced neuroinflammation. These results highlight an important role for Calhm2 in microglial activation and provide a potential therapeutic target for diseases related to microglia-mediated neuroinflammation.
Collapse
Affiliation(s)
- Jinbo Cheng
- Center on Translational Neuroscience, College of Life and Environmental Science, Minzu University of China, Beijing 100081, China.
- The Brain Science Center, Beijing Institute of Basic Medical Sciences, Beijing 100850, China
| | - Yuan Dong
- Department of Biochemistry, Medical College, Qingdao University, Qingdao, Shandong 266071, China
| | - Jun Ma
- The Brain Science Center, Beijing Institute of Basic Medical Sciences, Beijing 100850, China
| | - Ruiyuan Pan
- The Brain Science Center, Beijing Institute of Basic Medical Sciences, Beijing 100850, China
| | - Yajin Liao
- Center on Translational Neuroscience, College of Life and Environmental Science, Minzu University of China, Beijing 100081, China
- The Brain Science Center, Beijing Institute of Basic Medical Sciences, Beijing 100850, China
| | - Xiangxi Kong
- The Brain Science Center, Beijing Institute of Basic Medical Sciences, Beijing 100850, China
| | - Xiaoheng Li
- The Brain Science Center, Beijing Institute of Basic Medical Sciences, Beijing 100850, China
| | - Shuoshuo Li
- The Brain Science Center, Beijing Institute of Basic Medical Sciences, Beijing 100850, China
| | - Pingfang Chen
- School of Medicine, Shanghai Jiao Tong University, Shanghai 200025, China
| | - Liang Wang
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Center for Influenza Research and Early-warning (CASCIRE), CAS-TWAS Center of Excellence for Emerging Infectious Diseases (CEEID), Chinese Academy of Sciences, Beijing 100101, China
| | - Ye Yu
- School of Medicine, Shanghai Jiao Tong University, Shanghai 200025, China
| | - Zengqiang Yuan
- Center on Translational Neuroscience, College of Life and Environmental Science, Minzu University of China, Beijing 100081, China.
- The Brain Science Center, Beijing Institute of Basic Medical Sciences, Beijing 100850, China
- Center of Alzheimer's Disease, Beijing Institute for Brain Disorders, Beijing 100069, China
| |
Collapse
|
35
|
Okui M, Murakami T, Sun H, Ikeshita C, Kanamura N, Taruno A. Posttranslational regulation of CALHM1/3 channel: N-linked glycosylation and S-palmitoylation. FASEB J 2021; 35:e21527. [PMID: 33788965 DOI: 10.1096/fj.202002632r] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 02/08/2021] [Accepted: 03/01/2021] [Indexed: 11/11/2022]
Abstract
Among calcium homeostasis modulator (CALHM) family members, CALHM1 and 3 together form a voltage-gated large-pore ion channel called CALHM1/3. CALHM1/3 plays an essential role in taste perception by mediating neurotransmitter release at channel synapses of taste bud cells. However, it is poorly understood how CALHM1/3 is regulated. Biochemical analyses of the two subunits following site-directed mutagenesis and pharmacological treatments established that both CALHM1 and 3 were N-glycosylated at single Asn residues in their second extracellular loops. Biochemical and electrophysiological studies revealed that N-glycan acquisition on CALHM1 and 3, respectively, controls the biosynthesis and gating kinetics of the CALHM1/3 channel. Furthermore, failure in subsequent remodeling of N-glycans decelerated the gating kinetics. Thus, the acquisition of N-glycans on both subunits and their remodeling differentially contribute to the functional expression of CALHM1/3. Meanwhile, metabolic labeling and acyl-biotin exchange assays combined with genetic modification demonstrated that CALHM3 was reversibly palmitoylated at three intracellular Cys residues. Screening of the DHHC protein acyltransferases identified DHHC3 and 15 as CALHM3 palmitoylating enzymes. The palmitoylation-deficient mutant CALHM3 showed a normal degradation rate and interaction with CALHM1. However, the same mutation markedly attenuated the channel activity but not surface localization of CALHM1/3, suggesting that CALHM3 palmitoylation is a critical determinant of CALHM1/3 activity but not its formation or forward trafficking. Overall, this study characterized N-glycosylation and S-palmitoylation of CALHM1/3 subunits and clarified their differential contributions to its functional expression, providing insights into the fine control of the CALHM1/3 channel and associated physiological processes.
Collapse
Affiliation(s)
- Motoki Okui
- Department of Molecular Cell Physiology, Kyoto Prefectural University of Medicine, Kyoto, Japan.,Department of Dental Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Tatsuro Murakami
- Department of Molecular Cell Physiology, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Hongxin Sun
- Department of Molecular Cell Physiology, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Chiaki Ikeshita
- Department of Molecular Cell Physiology, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Narisato Kanamura
- Department of Dental Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Akiyuki Taruno
- Department of Molecular Cell Physiology, Kyoto Prefectural University of Medicine, Kyoto, Japan.,Japan Science and Technology Agency, PRESTO, Kawaguchi, Japan
| |
Collapse
|
36
|
Bhat EA, Sajjad N, Banawas S, Khan J. Human CALHM5: Insight in large pore lipid gating ATP channel and associated neurological pathologies. Mol Cell Biochem 2021; 476:3711-3718. [PMID: 34089472 DOI: 10.1007/s11010-021-04198-y] [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: 01/15/2021] [Accepted: 05/27/2021] [Indexed: 10/21/2022]
Abstract
Recently calcium homeostasis modulators (CALHMs) are identified as ATP release channels play crucial role in functioning of neurons including gustatory signaling and neuronal excitability. Pathologies of Alzheimer's disease and depression have been associated with the dysfunction of CALHMs. Recently, CALHMs has been emerged as an important therapeutic research particularly in neurobiological studies. CALHM1 is most extensively studied among CALHMs and is an ATP and ion channel that is activated by membrane depolarization or removal of extracellular Ca2+. Despite the emerged role of CALHM5 shown by an recently assembled data; however, the neuronal function remains obscure until the first Cryo-EM structure of CALHM5 was recently solved by various research group which acts as a template to study the hidden functional properties of the CALHM5 protein based on structure function mechanism. It provides insight in some of the different pathophysiological roles. CALHM5 structure showed an abnormally large pore channel structure assembled as an undecamer with four transmembrane helices (TM1-TM4), an N-terminal helix (NTH), an extracellular loop region and an intracellular C-terminal domain (CTD) that consists of three α-helices CH1-3. The TM1 and NTH were always poorly defined among all CALHMs; however, these regions were well defined in CALHM5 channel structure. In this context, this review will provide insight in structure, function and mechanism to understand its significant role in pathological diseases particularly in Alzheimer's disease. Moreover, it focuses on CALHM5 structure and recent associated properties based on Cryo-EM research.
Collapse
Affiliation(s)
- Eijaz Ahmed Bhat
- Life Science Institute, Zhejiang University, Hangzhou, Zhejiang, 310058, P.R. China. .,Department of Biological Sciences and Bioengineering, Indian Institute of Technology, Kanpur, India.
| | - Nasreena Sajjad
- Department of Biochemistry, University of Kashmir, Hazratbal, Jammu and Kashmir, India
| | - Saeed Banawas
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Majmaah University, Majmaah, 11952, Kingdom of Saudi Arabia. .,Health and Basic Sciences Research Center, Majmaah University, Majmaah, 11952, Saudi Arabia. .,Departments of Biomedical Sciences, Oregon State University, Corvallis, OR, 97331, USA.
| | - Johra Khan
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Majmaah University, Majmaah, 11952, Kingdom of Saudi Arabia.,Health and Basic Sciences Research Center, Majmaah University, Majmaah, 11952, Saudi Arabia
| |
Collapse
|
37
|
Elevating the Levels of Calcium Ions Exacerbate Alzheimer's Disease via Inducing the Production and Aggregation of β-Amyloid Protein and Phosphorylated Tau. Int J Mol Sci 2021; 22:ijms22115900. [PMID: 34072743 PMCID: PMC8198078 DOI: 10.3390/ijms22115900] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 05/08/2021] [Accepted: 05/08/2021] [Indexed: 01/03/2023] Open
Abstract
Alzheimer’s disease (AD) is a neurodegenerative disease with a high incidence rate. The main pathological features of AD are β-amyloid plaques (APs), which are formed by β-amyloid protein (Aβ) deposition, and neurofibrillary tangles (NFTs), which are formed by the excessive phosphorylation of the tau protein. Although a series of studies have shown that the accumulation of metal ions, including calcium ions (Ca2+), can promote the formation of APs and NFTs, there is no systematic review of the mechanisms by which Ca2+ affects the development and progression of AD. In view of this, the current review summarizes the mechanisms by which Ca2+ is transported into and out of cells and organelles, such as the cell, endoplasmic reticulum, mitochondrial and lysosomal membranes to affect the balance of intracellular Ca2+ levels. In addition, dyshomeostasis of Ca2+ plays an important role in modulating the pathogenesis of AD by influencing the production and aggregation of Aβ peptides and tau protein phosphorylation and the ways that disrupting the metabolic balance of Ca2+ can affect the learning ability and memory of people with AD. In addition, the effects of these mechanisms on the synaptic plasticity are also discussed. Finally, the molecular network through which Ca2+ regulates the pathogenesis of AD is introduced, providing a theoretical basis for improving the clinical treatment of AD.
Collapse
|
38
|
Lemos FO, Bultynck G, Parys JB. A comprehensive overview of the complex world of the endo- and sarcoplasmic reticulum Ca 2+-leak channels. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2021; 1868:119020. [PMID: 33798602 DOI: 10.1016/j.bbamcr.2021.119020] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Revised: 03/09/2021] [Accepted: 03/13/2021] [Indexed: 12/11/2022]
Abstract
Inside cells, the endoplasmic reticulum (ER) forms the largest Ca2+ store. Ca2+ is actively pumped by the SERCA pumps in the ER, where intraluminal Ca2+-binding proteins enable the accumulation of large amount of Ca2+. IP3 receptors and the ryanodine receptors mediate the release of Ca2+ in a controlled way, thereby evoking complex spatio-temporal signals in the cell. The steady state Ca2+ concentration in the ER of about 500 μM results from the balance between SERCA-mediated Ca2+ uptake and the passive leakage of Ca2+. The passive Ca2+ leak from the ER is often ignored, but can play an important physiological role, depending on the cellular context. Moreover, excessive Ca2+ leakage significantly lowers the amount of Ca2+ stored in the ER compared to normal conditions, thereby limiting the possibility to evoke Ca2+ signals and/or causing ER stress, leading to pathological consequences. The so-called Ca2+-leak channels responsible for Ca2+ leakage from the ER are however still not well understood, despite over 20 different proteins have been proposed to contribute to it. This review has the aim to critically evaluate the available evidence about the various channels potentially involved and to draw conclusions about their relative importance.
Collapse
Affiliation(s)
- Fernanda O Lemos
- KU Leuven, Laboratory of Molecular and Cellular Signaling, Department of Cellular and Molecular Medicine and Leuven Kanker Instituut, Campus Gasthuisberg O/N-1 B-802, Herestraat 49, B-3000 Leuven, Belgium
| | - Geert Bultynck
- KU Leuven, Laboratory of Molecular and Cellular Signaling, Department of Cellular and Molecular Medicine and Leuven Kanker Instituut, Campus Gasthuisberg O/N-1 B-802, Herestraat 49, B-3000 Leuven, Belgium
| | - Jan B Parys
- KU Leuven, Laboratory of Molecular and Cellular Signaling, Department of Cellular and Molecular Medicine and Leuven Kanker Instituut, Campus Gasthuisberg O/N-1 B-802, Herestraat 49, B-3000 Leuven, Belgium.
| |
Collapse
|
39
|
Extracellular CIRP Activates the IL-6Rα/STAT3/Cdk5 Pathway in Neurons. Mol Neurobiol 2021; 58:3628-3640. [PMID: 33783711 PMCID: PMC10404139 DOI: 10.1007/s12035-021-02368-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Accepted: 03/22/2021] [Indexed: 10/21/2022]
Abstract
Extracellular cold-inducible RNA-binding protein (eCIRP) stimulates microglial inflammation causing neuronal damage during ischemic stroke and is a critical mediator of alcohol-induced cognitive impairment. However, the precise role of eCIRP in mediating neuroinflammation remains unknown. In this study, we report that eCIRP activates neurotoxic cyclin-dependent kinase-5 (Cdk5)/p25 through the induction of IL-6Rα/STAT3 pathway in neurons. Amyloid β (Aβ)-mediated neuronal stress, which is associated with Alzheimer's disease, increased the levels of eCIRP released from BV2 microglial cells. The released eCIRP levels from BV2 cells increased 3.2-fold upon stimulation with conditioned medium from Neuro-2a (N2a) cells containing Aβ compared to control N2a supernatant in a time-dependent manner. Stimulation of N2a cells and primary neurons with eCIRP upregulated the neuronal Cdk5 activator p25 expression in a dose- and time-dependent manner. eCIRP directly induced neuronal STAT3 phosphorylation and p25 increase via its novel receptor IL-6Rα. Next, we showed using surface plasmon resonance that eCIRP-derived peptide C23 inhibited the binding of eCIRP to IL-6Rα at 25 μM, with a 40-fold increase in equilibrium dissociation constant (Kd) value (from 8.08 × 10-8 M to 3.43 × 10-6 M), and completely abrogated the binding at 50 μM. Finally, C23 reversed the eCIRP-induced increase in neuronal STAT3 phosphorylation and p25 levels. In conclusion, the current study demonstrates that the upregulation of neuronal IL-6Rα/STAT3/Cdk5 pathway is a key mechanism of eCIRP's role in neuroinflammation and that C23 as a potent inhibitor of this pathway has translational potential in neurodegenerative pathologies controlled by eCIRP.
Collapse
|
40
|
Furukawa H, Simorowski N, Michalski K. Effective production of oligomeric membrane proteins by EarlyBac-insect cell system. Methods Enzymol 2021; 653:3-19. [PMID: 34099177 DOI: 10.1016/bs.mie.2020.12.019] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Despite major advances in methodologies for membrane protein production over the last two decades, there remain challenging protein complexes that are technically difficult to yield by conventional recombinant expression methods. A large number of these proteins are multimeric membrane proteins from eukaryotic species, which are required to pass through stringent quality control mechanisms of host cells for proper folding and complex assembly. Here, we describe the development procedure to improve the production efficiency of multi-oligomeric membrane protein complexes in insect cells and recombinant baculovirus, which involves screening of promoters, enhancers, and untranslated regions for expression levels, using calcium homeostasis modulator (CALHM) and N-methyl-d-aspartate receptor (NMDAR) proteins as examples. We demonstrate that our insect cell expression strategy is effective in expression of both multi-homomeric CALHM proteins and multi-heteromeric NMDARs.
Collapse
Affiliation(s)
- Hiro Furukawa
- WM Keck Structural Biology Laboratory, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, United States.
| | - Noriko Simorowski
- WM Keck Structural Biology Laboratory, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, United States
| | - Kevin Michalski
- WM Keck Structural Biology Laboratory, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, United States
| |
Collapse
|
41
|
Riegerová P, Brejcha J, Bezděková D, Chum T, Mašínová E, Čermáková N, Ovsepian SV, Cebecauer M, Štefl M. Expression and Localization of AβPP in SH-SY5Y Cells Depends on Differentiation State. J Alzheimers Dis 2021; 82:485-491. [PMID: 34057078 PMCID: PMC8385523 DOI: 10.3233/jad-201409] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/21/2021] [Indexed: 11/27/2022]
Abstract
Neuroblastoma cell line SH-SY5Y, due to its capacity to differentiate into neurons, easy handling, and low cost, is a common experimental model to study molecular events leading to Alzheimer's disease (AD). However, it is prevalently used in its undifferentiated state, which does not resemble neurons affected by the disease. Here, we show that the expression and localization of amyloid-β protein precursor (AβPP), one of the key molecules involved in AD pathogenesis, is dramatically altered in SH-SY5Y cells fully differentiated by combined treatment with retinoic acid and BDNF. We show that insufficient differentiation of SH-SY5Y cells results in AβPP mislocalization.
Collapse
Affiliation(s)
- Petra Riegerová
- Department of Biophysical Chemistry, J. Heyrovský Institute of Physical Chemistry of the Czech Academy of Sciences, Prague, Czech Republic
| | - Jindřich Brejcha
- Department of Biophysical Chemistry, J. Heyrovský Institute of Physical Chemistry of the Czech Academy of Sciences, Prague, Czech Republic
- Department of Philosophy and History of Science, Faculty of Science, Charles University, Prague, Czech Republic
| | - Dagmar Bezděková
- Department of Experimental Neurobiology, National Institute of Mental Health, Klecany, Czech Republic
- Department of Psychiatry and Medical Psychology, Third Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Tomáš Chum
- Department of Biophysical Chemistry, J. Heyrovský Institute of Physical Chemistry of the Czech Academy of Sciences, Prague, Czech Republic
| | - Eva Mašínová
- Department of Biophysical Chemistry, J. Heyrovský Institute of Physical Chemistry of the Czech Academy of Sciences, Prague, Czech Republic
| | - Nikola Čermáková
- Department of Biophysical Chemistry, J. Heyrovský Institute of Physical Chemistry of the Czech Academy of Sciences, Prague, Czech Republic
| | - Saak V. Ovsepian
- Department of Experimental Neurobiology, National Institute of Mental Health, Klecany, Czech Republic
- Department of Psychiatry and Medical Psychology, Third Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Marek Cebecauer
- Department of Biophysical Chemistry, J. Heyrovský Institute of Physical Chemistry of the Czech Academy of Sciences, Prague, Czech Republic
| | - Martin Štefl
- Department of Biophysical Chemistry, J. Heyrovský Institute of Physical Chemistry of the Czech Academy of Sciences, Prague, Czech Republic
| |
Collapse
|
42
|
Therapeutic Strategies to Target Calcium Dysregulation in Alzheimer's Disease. Cells 2020; 9:cells9112513. [PMID: 33233678 PMCID: PMC7699688 DOI: 10.3390/cells9112513] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 11/17/2020] [Accepted: 11/18/2020] [Indexed: 12/31/2022] Open
Abstract
Alzheimer’s disease (AD) is the most common form of dementia, affecting millions of people worldwide. Unfortunately, none of the current treatments are effective at improving cognitive function in AD patients and, therefore, there is an urgent need for the development of new therapies that target the early cause(s) of AD. Intracellular calcium (Ca2+) regulation is critical for proper cellular and neuronal function. It has been suggested that Ca2+ dyshomeostasis is an upstream factor of many neurodegenerative diseases, including AD. For this reason, chemical agents or small molecules aimed at targeting or correcting this Ca2+ dysregulation might serve as therapeutic strategies to prevent the development of AD. Moreover, neurons are not alone in exhibiting Ca2+ dyshomeostasis, since Ca2+ disruption is observed in other cell types in the brain in AD. In this review, we examine the distinct Ca2+ channels and compartments involved in the disease mechanisms that could be potential targets in AD.
Collapse
|
43
|
Jeon YK, Choi SW, Kwon JW, Woo J, Choi SW, Kim SJ, Kim SJ. Thermosensitivity of the voltage-dependent activation of calcium homeostasis modulator 1 (calhm1) ion channel. Biochem Biophys Res Commun 2020; 534:590-596. [PMID: 33199024 DOI: 10.1016/j.bbrc.2020.11.035] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Accepted: 11/08/2020] [Indexed: 01/08/2023]
Abstract
Calcium homeostasis modulator 1 (calhm1) proteins form an outwardly rectifying nonselective ion channel having exceedingly slow kinetics and low sensitivity to voltage that is shifted by lowering extracellular Ca2+ ([Ca2+]e). Here we found that physiological temperature dramatically facilitates the voltage-dependent activation of the calhm1 current (Icalhm1); increased amplitude (Q10, 7-15) and fastened speed of activation. Also, the leftward shift of the half-activation voltage (V1/2) was similary observed in the normal and lower [Ca2+]e. Since calhm1 is highly expressed in the brain and taste cells, the thermosensitivity should be considered in their electrophysiology.
Collapse
Affiliation(s)
- Young Keul Jeon
- Department of Physiology, Seoul National University College of Medicine, Republic of Korea
| | - Si Won Choi
- Department of Physiology, Seoul National University College of Medicine, Republic of Korea
| | - Jae Won Kwon
- Department of Physiology, Seoul National University College of Medicine, Republic of Korea
| | - Joohan Woo
- Department of Physiology and Ion Channel Disease Research Center, Dongguk University College of Medicine, Seoul, Republic of Korea
| | - Seong Woo Choi
- Department of Physiology, Seoul National University College of Medicine, Republic of Korea; Ischemic/Hypoxic Disease Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Sang Jeong Kim
- Department of Physiology, Seoul National University College of Medicine, Republic of Korea
| | - Sung Joon Kim
- Department of Physiology, Seoul National University College of Medicine, Republic of Korea; Ischemic/Hypoxic Disease Institute, Seoul National University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
44
|
Cryo-EM structures of human calcium homeostasis modulator 5. Cell Discov 2020; 6:81. [PMID: 33298887 PMCID: PMC7652935 DOI: 10.1038/s41421-020-00228-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Accepted: 10/09/2020] [Indexed: 01/03/2023] Open
|
45
|
Calvo-Rodriguez M, Bacskai BJ. Mitochondria and Calcium in Alzheimer's Disease: From Cell Signaling to Neuronal Cell Death. Trends Neurosci 2020; 44:136-151. [PMID: 33160650 DOI: 10.1016/j.tins.2020.10.004] [Citation(s) in RCA: 132] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 09/03/2020] [Accepted: 10/07/2020] [Indexed: 02/06/2023]
Abstract
Mitochondrial dysfunction has been implicated in the pathogenesis of almost all neurological diseases, including Alzheimer's disease (AD). Historically, a primary focus in this context has been the link between mitochondrial dynamics and amyloid β toxicity. Recent evidence suggests that dysregulation of mitochondrial calcium homeostasis is also related to tau and other risk factors in AD, although an ongoing challenge in the field is that data collected from different models or experimental settings have not always been consistent. We examine recent literature on mitochondrial dysregulation in AD, with special emphasis on mitochondrial calcium. We include data from in vitro systems, genetic animal models, and AD-derived human tissue, and discuss whether mitochondrial calcium transporters should be proposed as therapeutic candidates for the development of neuroprotective drugs against AD.
Collapse
Affiliation(s)
- Maria Calvo-Rodriguez
- Alzheimer Research Unit, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, 114 16th Street, Charlestown, MA 02129, USA
| | - Brian J Bacskai
- Alzheimer Research Unit, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, 114 16th Street, Charlestown, MA 02129, USA.
| |
Collapse
|
46
|
Thakran S, Guin D, Singh P, Singh P, Kukal S, Rawat C, Yadav S, Kushwaha SS, Srivastava AK, Hasija Y, Saso L, Ramachandran S, Kukreti R. Genetic Landscape of Common Epilepsies: Advancing towards Precision in Treatment. Int J Mol Sci 2020; 21:E7784. [PMID: 33096746 PMCID: PMC7589654 DOI: 10.3390/ijms21207784] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 10/09/2020] [Accepted: 10/14/2020] [Indexed: 12/15/2022] Open
Abstract
Epilepsy, a neurological disease characterized by recurrent seizures, is highly heterogeneous in nature. Based on the prevalence, epilepsy is classified into two types: common and rare epilepsies. Common epilepsies affecting nearly 95% people with epilepsy, comprise generalized epilepsy which encompass idiopathic generalized epilepsy like childhood absence epilepsy, juvenile myoclonic epilepsy, juvenile absence epilepsy and epilepsy with generalized tonic-clonic seizure on awakening and focal epilepsy like temporal lobe epilepsy and cryptogenic focal epilepsy. In 70% of the epilepsy cases, genetic factors are responsible either as single genetic variant in rare epilepsies or multiple genetic variants acting along with different environmental factors as in common epilepsies. Genetic testing and precision treatment have been developed for a few rare epilepsies and is lacking for common epilepsies due to their complex nature of inheritance. Precision medicine for common epilepsies require a panoramic approach that incorporates polygenic background and other non-genetic factors like microbiome, diet, age at disease onset, optimal time for treatment and other lifestyle factors which influence seizure threshold. This review aims to comprehensively present a state-of-art review of all the genes and their genetic variants that are associated with all common epilepsy subtypes. It also encompasses the basis of these genes in the epileptogenesis. Here, we discussed the current status of the common epilepsy genetics and address the clinical application so far on evidence-based markers in prognosis, diagnosis, and treatment management. In addition, we assessed the diagnostic predictability of a few genetic markers used for disease risk prediction in individuals. A combination of deeper endo-phenotyping including pharmaco-response data, electro-clinical imaging, and other clinical measurements along with genetics may be used to diagnose common epilepsies and this marks a step ahead in precision medicine in common epilepsies management.
Collapse
Affiliation(s)
- Sarita Thakran
- Genomics and Molecular Medicine Unit, Institute of Genomics and Integrative Biology (IGIB), Council of Scientific and Industrial Research (CSIR), Delhi 110007, India; (S.T.); (D.G.); (P.S.); (P.S.); (S.K.); (C.R.); (S.Y.)
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India;
| | - Debleena Guin
- Genomics and Molecular Medicine Unit, Institute of Genomics and Integrative Biology (IGIB), Council of Scientific and Industrial Research (CSIR), Delhi 110007, India; (S.T.); (D.G.); (P.S.); (P.S.); (S.K.); (C.R.); (S.Y.)
- Department of Bioinformatics, Delhi Technological University, Shahbad Daulatpur, Main Bawana Road, Delhi 110042, India;
| | - Pooja Singh
- Genomics and Molecular Medicine Unit, Institute of Genomics and Integrative Biology (IGIB), Council of Scientific and Industrial Research (CSIR), Delhi 110007, India; (S.T.); (D.G.); (P.S.); (P.S.); (S.K.); (C.R.); (S.Y.)
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India;
| | - Priyanka Singh
- Genomics and Molecular Medicine Unit, Institute of Genomics and Integrative Biology (IGIB), Council of Scientific and Industrial Research (CSIR), Delhi 110007, India; (S.T.); (D.G.); (P.S.); (P.S.); (S.K.); (C.R.); (S.Y.)
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India;
| | - Samiksha Kukal
- Genomics and Molecular Medicine Unit, Institute of Genomics and Integrative Biology (IGIB), Council of Scientific and Industrial Research (CSIR), Delhi 110007, India; (S.T.); (D.G.); (P.S.); (P.S.); (S.K.); (C.R.); (S.Y.)
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India;
| | - Chitra Rawat
- Genomics and Molecular Medicine Unit, Institute of Genomics and Integrative Biology (IGIB), Council of Scientific and Industrial Research (CSIR), Delhi 110007, India; (S.T.); (D.G.); (P.S.); (P.S.); (S.K.); (C.R.); (S.Y.)
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India;
| | - Saroj Yadav
- Genomics and Molecular Medicine Unit, Institute of Genomics and Integrative Biology (IGIB), Council of Scientific and Industrial Research (CSIR), Delhi 110007, India; (S.T.); (D.G.); (P.S.); (P.S.); (S.K.); (C.R.); (S.Y.)
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India;
| | - Suman S. Kushwaha
- Department of Neurology, Institute of Human Behaviour and Allied Sciences, Dilshad Garden, Delhi 110095, India;
| | - Achal K. Srivastava
- Department of Neurology, All India Institute of Medical Sciences, Ansari Nagar, New Delhi 110029, India;
| | - Yasha Hasija
- Department of Bioinformatics, Delhi Technological University, Shahbad Daulatpur, Main Bawana Road, Delhi 110042, India;
| | - Luciano Saso
- Department of Physiology and Pharmacology “Vittorio Erspamer”, Sapienza University of Rome, P. le Aldo Moro 5, 00185 Rome, Italy;
| | - Srinivasan Ramachandran
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India;
- G N Ramachandran Knowledge Centre, Council of Scientific and Industrial Research (CSIR)—Institute of Genomics and Integrative Biology (IGIB), New Delhi 110007, India
| | - Ritushree Kukreti
- Genomics and Molecular Medicine Unit, Institute of Genomics and Integrative Biology (IGIB), Council of Scientific and Industrial Research (CSIR), Delhi 110007, India; (S.T.); (D.G.); (P.S.); (P.S.); (S.K.); (C.R.); (S.Y.)
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India;
| |
Collapse
|
47
|
Silva A, Pereira M, Carrascal MA, Brites G, Neves B, Moreira P, Resende R, Silva MM, Santos AE, Pereira C, Cruz MT. Calcium Modulation, Anti-Oxidant and Anti-Inflammatory Effect of Skin Allergens Targeting the Nrf2 Signaling Pathway in Alzheimer's Disease Cellular Models. Int J Mol Sci 2020; 21:ijms21207791. [PMID: 33096789 PMCID: PMC7594024 DOI: 10.3390/ijms21207791] [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: 09/10/2020] [Revised: 10/06/2020] [Accepted: 10/18/2020] [Indexed: 01/13/2023] Open
Abstract
Experimental evidence highlights nuclear factor (erythroid-derived 2)-like 2 (Nrf2) as a molecular target in Alzheimer's disease (AD). The well-known effect of electrophilic cysteine-reactive skin allergens on Nrf2-activation led to the hypothesis that these compounds could have a therapeutic role in AD. This was further supported by the neuroprotective activity of the skin allergen dimethyl fumarate (DMF), demonstrated in in vivo models of neurodegenerative diseases. We evaluated the effect of the cysteine-reactive allergens 1,4-phenylenediamine (PPD) and methyl heptine carbonate (MHC) on (1) neuronal redox imbalance and calcium dyshomeostasis using N2a wild-type (N2a-wt) and human APP-overexpressing neuronal cells (wild-type, N2a-APPwt) and (2) on neuroinflammation, using microglia BV-2 cells exposed to LPS (lipopolysaccharide). Phthalic anhydride (PA, mainly lysine-reactive), was used as a negative control. DMF, PPD and MHC increased Hmox1 gene and HMOX1 protein levels in N2a-APPwt cells suggesting Nrf2-dependent antioxidant activity. MHC, but also PA, rescued N2a-APPwt mitochondrial membrane potential and calcium levels in a Nrf2-independent pathway. All the chemicals showed anti-inflammatory activity by decreasing iNOS protein in microglia. This work highlights the potential neuroprotective and anti-inflammatory role of the selected skin allergens in in vitro models of AD, and supports further studies envisaging the validation of the results using in vivo AD models.
Collapse
Affiliation(s)
- Ana Silva
- Center for Neuroscience and Cell Biology and Institute for Biomedical Imaging and Life Sciences, University of Coimbra, 3000-548 Coimbra, Portugal; (P.M.); (R.R.); (M.M.S.); (A.E.S.); (C.P.)
- Correspondence: (A.S.); (M.T.C.)
| | - Marta Pereira
- Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal;
| | | | - Gonçalo Brites
- Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal;
| | - Bruno Neves
- Department of Medical Sciences and Institute for Biomedicine (iBiMED), University of Aveiro, 3810-193 Aveiro, Portugal;
| | - Patrícia Moreira
- Center for Neuroscience and Cell Biology and Institute for Biomedical Imaging and Life Sciences, University of Coimbra, 3000-548 Coimbra, Portugal; (P.M.); (R.R.); (M.M.S.); (A.E.S.); (C.P.)
| | - Rosa Resende
- Center for Neuroscience and Cell Biology and Institute for Biomedical Imaging and Life Sciences, University of Coimbra, 3000-548 Coimbra, Portugal; (P.M.); (R.R.); (M.M.S.); (A.E.S.); (C.P.)
- University of Coimbra, Institute for Interdisciplinary Research (IIIUC), 3030-789 Coimbra, Portugal
| | - Maria Manuel Silva
- Center for Neuroscience and Cell Biology and Institute for Biomedical Imaging and Life Sciences, University of Coimbra, 3000-548 Coimbra, Portugal; (P.M.); (R.R.); (M.M.S.); (A.E.S.); (C.P.)
- Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal;
| | - Armanda E. Santos
- Center for Neuroscience and Cell Biology and Institute for Biomedical Imaging and Life Sciences, University of Coimbra, 3000-548 Coimbra, Portugal; (P.M.); (R.R.); (M.M.S.); (A.E.S.); (C.P.)
- Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal;
| | - Cláudia Pereira
- Center for Neuroscience and Cell Biology and Institute for Biomedical Imaging and Life Sciences, University of Coimbra, 3000-548 Coimbra, Portugal; (P.M.); (R.R.); (M.M.S.); (A.E.S.); (C.P.)
- Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal;
| | - Maria Teresa Cruz
- Center for Neuroscience and Cell Biology and Institute for Biomedical Imaging and Life Sciences, University of Coimbra, 3000-548 Coimbra, Portugal; (P.M.); (R.R.); (M.M.S.); (A.E.S.); (C.P.)
- Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal;
- Correspondence: (A.S.); (M.T.C.)
| |
Collapse
|
48
|
Taruno A, Nomura K, Kusakizako T, Ma Z, Nureki O, Foskett JK. Taste transduction and channel synapses in taste buds. Pflugers Arch 2020; 473:3-13. [PMID: 32936320 DOI: 10.1007/s00424-020-02464-4] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 07/29/2020] [Accepted: 09/07/2020] [Indexed: 12/31/2022]
Abstract
The variety of taste sensations, including sweet, umami, bitter, sour, and salty, arises from diverse taste cells, each of which expresses specific taste sensor molecules and associated components for downstream signal transduction cascades. Recent years have witnessed major advances in our understanding of the molecular mechanisms underlying transduction of basic tastes in taste buds, including the identification of the bona fide sour sensor H+ channel OTOP1, and elucidation of transduction of the amiloride-sensitive component of salty taste (the taste of sodium) and the TAS1R-independent component of sweet taste (the taste of sugar). Studies have also discovered an unconventional chemical synapse termed "channel synapse" which employs an action potential-activated CALHM1/3 ion channel instead of exocytosis of synaptic vesicles as the conduit for neurotransmitter release that links taste cells to afferent neurons. New images of the channel synapse and determinations of the structures of CALHM channels have provided structural and functional insights into this unique synapse. In this review, we discuss the current view of taste transduction and neurotransmission with emphasis on recent advances in the field.
Collapse
Affiliation(s)
- Akiyuki Taruno
- Department of Molecular Cell Physiology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan. .,Japan Science and Technology Agency, PRESTO, Kawaguchi, Saitama, Japan.
| | - Kengo Nomura
- Department of Molecular Cell Physiology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Tsukasa Kusakizako
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, Tokyo, Japan
| | - Zhongming Ma
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Osamu Nureki
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, Tokyo, Japan
| | - J Kevin Foskett
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.,Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
49
|
Whiten DR, Brownjohn PW, Moore S, De S, Strano A, Zuo Y, Haneklaus M, Klenerman D, Livesey FJ. Tumour necrosis factor induces increased production of extracellular amyloid-β- and α-synuclein-containing aggregates by human Alzheimer's disease neurons. Brain Commun 2020; 2:fcaa146. [PMID: 33543132 PMCID: PMC7850285 DOI: 10.1093/braincomms/fcaa146] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2020] [Revised: 07/21/2020] [Accepted: 07/27/2020] [Indexed: 01/24/2023] Open
Abstract
In addition to increased aberrant protein aggregation, inflammation has been proposed as a key element in the pathogenesis and progression of Alzheimer’s disease. How inflammation interacts with other disease pathways and how protein aggregation increases during disease are not clear. We used single-molecule imaging approaches and membrane permeabilization assays to determine the effect of chronic exposure to tumour necrosis factor, a master proinflammatory cytokine, on protein aggregation in human-induced pluripotent stem cell-derived neurons harbouring monogenic Alzheimer’s disease mutations. We report that exposure of Alzheimer’s disease neurons, but not control neurons, to tumour necrosis factor induces substantial production of extracellular protein aggregates. Aggregates from Alzheimer’s disease neurons are composed of amyloid-β and α-synuclein and induce significant permeabilization of lipid membranes in an assay of pathogenicity. These findings provide support for a causal relationship between two crucial processes in Alzheimer’s disease pathogenesis and suggest that targeting inflammation, particularly tumour necrosis factor, may have beneficial downstream effects on ameliorating aberrant protein aggregation and accumulation.
Collapse
Affiliation(s)
- Daniel R Whiten
- Department of Chemistry, University of Cambridge, Cambridge CB2 1EW, UK
| | - Philip W Brownjohn
- Zayed Centre for Research into Rare Disease in Children, UCL Great Ormond Street Institute of Child Health, London WC1N 1DZ, UK
| | - Steven Moore
- Zayed Centre for Research into Rare Disease in Children, UCL Great Ormond Street Institute of Child Health, London WC1N 1DZ, UK
| | - Suman De
- Department of Chemistry, University of Cambridge, Cambridge CB2 1EW, UK
| | - Alessio Strano
- Zayed Centre for Research into Rare Disease in Children, UCL Great Ormond Street Institute of Child Health, London WC1N 1DZ, UK
| | - Yukun Zuo
- Department of Chemistry, University of Cambridge, Cambridge CB2 1EW, UK
| | - Moritz Haneklaus
- Zayed Centre for Research into Rare Disease in Children, UCL Great Ormond Street Institute of Child Health, London WC1N 1DZ, UK
| | - David Klenerman
- Department of Chemistry, University of Cambridge, Cambridge CB2 1EW, UK.,UK Dementia Research Institute at University of Cambridge, Cambridge CB2 0XY, UK
| | - Frederick J Livesey
- Zayed Centre for Research into Rare Disease in Children, UCL Great Ormond Street Institute of Child Health, London WC1N 1DZ, UK
| |
Collapse
|
50
|
Yang W, Wang Y, Guo J, He L, Zhou Y, Zheng H, Liu Z, Zhu P, Zhang XC. Cryo-electron microscopy structure of CLHM1 ion channel from Caenorhabditis elegans. Protein Sci 2020; 29:1803-1815. [PMID: 32557855 PMCID: PMC7380676 DOI: 10.1002/pro.3904] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 06/15/2020] [Accepted: 06/16/2020] [Indexed: 02/02/2023]
Abstract
Calcium homeostasis modulators (CALHMs/CLHMs) comprise a family of pore-forming protein complexes assembling into voltage-gated, Ca2+ -sensitive, nonselective channels. These complexes contain an ion-conduction pore sufficiently wide to permit the passing of ATP molecules serving as neurotransmitters. While their function and structure information is accumulating, the precise mechanisms of these channel complexes remain to be full understood. Here, we present the structure of the Caenorhabditis elegans CLHM1 channel in its open state solved through single-particle cryo-electron microscopy at 3.7-Å resolution. The transmembrane region of the channel structure of the dominant class shows an assembly of 10-fold rotational symmetry in one layer, and its cytoplasmic region is involved in additional twofold symmetrical packing in a tail-to-tail manner. Furthermore, we identified a series of amino acid residues critical for the regulation of CeCLHM1 channel using functional assays, electrophysiological analyses as well as structural-based analysis. Our structure and function analyses provide new insights into the mechanisms of CALHM channels.
Collapse
Affiliation(s)
- Weixin Yang
- National Laboratory of Biomacromolecules, CAS Center for Excellence in BiomacromoleculesInstitute of Biophysics, Chinese Academy of SciencesBeijingChina
- College of Life SciencesUniversity of Chinese Academy of SciencesBeijingChina
| | - Youwang Wang
- National Laboratory of Biomacromolecules, CAS Center for Excellence in BiomacromoleculesInstitute of Biophysics, Chinese Academy of SciencesBeijingChina
- College of Life SciencesUniversity of Chinese Academy of SciencesBeijingChina
| | - Jianli Guo
- National Laboratory of Biomacromolecules, CAS Center for Excellence in BiomacromoleculesInstitute of Biophysics, Chinese Academy of SciencesBeijingChina
| | - Lingli He
- National Laboratory of Biomacromolecules, CAS Center for Excellence in BiomacromoleculesInstitute of Biophysics, Chinese Academy of SciencesBeijingChina
| | - Ye Zhou
- National Laboratory of Biomacromolecules, CAS Center for Excellence in BiomacromoleculesInstitute of Biophysics, Chinese Academy of SciencesBeijingChina
| | - Hui Zheng
- National Laboratory of Biomacromolecules, CAS Center for Excellence in BiomacromoleculesInstitute of Biophysics, Chinese Academy of SciencesBeijingChina
| | - Zhenfeng Liu
- National Laboratory of Biomacromolecules, CAS Center for Excellence in BiomacromoleculesInstitute of Biophysics, Chinese Academy of SciencesBeijingChina
- College of Life SciencesUniversity of Chinese Academy of SciencesBeijingChina
| | - Ping Zhu
- National Laboratory of Biomacromolecules, CAS Center for Excellence in BiomacromoleculesInstitute of Biophysics, Chinese Academy of SciencesBeijingChina
- College of Life SciencesUniversity of Chinese Academy of SciencesBeijingChina
| | - Xuejun C. Zhang
- National Laboratory of Biomacromolecules, CAS Center for Excellence in BiomacromoleculesInstitute of Biophysics, Chinese Academy of SciencesBeijingChina
- College of Life SciencesUniversity of Chinese Academy of SciencesBeijingChina
| |
Collapse
|