1
|
Kim YK, Jo D, Choi S, Song J. High-fat diet triggers transcriptomic changes in the olfactory bulb. Heliyon 2025; 11:e42196. [PMID: 39927144 PMCID: PMC11804815 DOI: 10.1016/j.heliyon.2025.e42196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 01/09/2025] [Accepted: 01/21/2025] [Indexed: 02/11/2025] Open
Abstract
Metabolic imbalance contributes to cognitive impairment, anxiety, depressive behavior, and impaired olfactory perception. Recent studies have focused on olfactory dysfunction in patients with obesity and diabetes accompanied by cognitive dysfunction, considering that the synaptic signal from the olfactory bulb is directly transmitted to memory consolidation-related brain regions. This study investigated transcriptomic changes in the olfactory bulb in high-fat diet (HFD)-fed mice compared to that in normal-diet-fed mice. We sampled olfactory bulbs from HFD-fed mice, performed RNA sequencing, and measured mRNA levels in olfactory bulb tissue. Additionally, we assessed plasma cytokine levels in HFD-fed mice. We found differences in the expression of protein-coding and non-coding RNAs involved in insulin, lipid metabolism, neurogenesis, serotonin, dopamine, and gamma-aminobutyric acid-related signaling in the olfactory bulb of HFD-fed mice compared to control mice. Thus, our findings suggest potential therapeutic targets for treating olfactory dysfunction and related neural disorders in individuals with metabolic syndrome.
Collapse
Affiliation(s)
- Young-Kook Kim
- Biomedical Science Graduate Program (BMSGP), Chonnam National University, Hwasun, 58128, Republic of Korea
- Department of Biochemistry, Chonnam National University Medical School, Hwasun, 58128, Republic of Korea
| | - Danbi Jo
- Biomedical Science Graduate Program (BMSGP), Chonnam National University, Hwasun, 58128, Republic of Korea
- Department of Anatomy, Chonnam National University Medical School, Hwasun, 58128, Republic of Korea
| | - Seoyoon Choi
- Biomedical Science Graduate Program (BMSGP), Chonnam National University, Hwasun, 58128, Republic of Korea
- Department of Anatomy, Chonnam National University Medical School, Hwasun, 58128, Republic of Korea
| | - Juhyun Song
- Biomedical Science Graduate Program (BMSGP), Chonnam National University, Hwasun, 58128, Republic of Korea
- Department of Anatomy, Chonnam National University Medical School, Hwasun, 58128, Republic of Korea
| |
Collapse
|
2
|
Qu Y, Gu J, Li L, Yan Y, Yan C, Zhang T. Guilu Erxian Jiao remodels dendritic spine morphology through activation of the hippocampal TRPC6-CaMKIV-CREB signaling pathway and suppresses fear memory generalization in rats with post-traumatic stress disorder. JOURNAL OF ETHNOPHARMACOLOGY 2025; 340:119252. [PMID: 39681200 DOI: 10.1016/j.jep.2024.119252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Revised: 12/09/2024] [Accepted: 12/13/2024] [Indexed: 12/18/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Guilu Erxian Jiao (GLEXJ) is a renowned traditional Chinese herbal formula used to tonify the kidney. It is employed to treat psychiatric disorders, and alleviate memory impairment, cognitive dysfunction, and behavioral disorders. Modern pharmacological studies have demonstrated GLEXJ's ability to significantly inhibit the fear response in post-traumatic stress disorder (PTSD) and facilitate the extinction of fear memory. However, the underlying pharmacological mechanisms remain elusive. AIM OF THE STUDY Fear memory generalization, a fundamental characteristic of PTSD, remains poorly understood, and optimal pharmacological treatments are lacking. This study aimed to investigate GLEXJ's inhibitory effects on fear memory generalization in PTSD rats and elucidate its underlying mechanisms. MATERIALS AND METHODS PTSD rats were induced using the single prolonged stress and electrical stimulation (SPS&S) protocol and treated with GLEXJ or paroxetine (PRX). Fear memory generalization was assessed using a contextual fear memory test. Hippocampal dendritic spine morphology was analyzed using Golgi-Cox staining. The chemical composition of GLEXJ was determined using ultra-high performance liquid chromatography-tandem mass spectrometry (UHPLC-MS/MS). Network pharmacology was employed to predict GLEXJ's therapeutic mechanism in PTSD treatment. Western blotting and immunofluorescence were used to measure indicators of the transient receptor potential channel 6 (TRPC6)-mediated calcium/calmodulin-dependent protein kinase IV-cAMP response element-binding protein (CaMKIV-CREB) signaling pathway. In vitro, TRPC6 was suppressed in rat adrenal pheochromocytoma (PC12) cells using lentiviral vectors, and phalloidin staining was employed to examine changes in Fibros actin (F-actin), elucidating the mechanistic effects of GLEXJ-containing serum. RESULTS GLEXJ significantly mitigated fear memory generalization in PTSD rats, even with repeated stress exposure. It also alleviated abnormal hippocampal dendritic spine morphology. Network pharmacology analysis confirmed that GLEXJ was closely related to the Ca2+ signaling pathway in PTSD treatment. PTSD rats exhibited disrupted TRPC6-mediated CaMKIV-CREB signaling and impaired synaptic plasticity. GLEXJ upregulated TRPC6 expression, reactivated the CaMKIV-CREB pathway, and promoted synaptic remodeling. In vitro studies confirmed that TRPC6 suppression reduced F-actin levels while GLEXJ-containing serum increased TRPC6 expression and F-actin content. CONCLUSIONS GLEXJ activates CaMKIV-CREB signaling by upregulating TRPC6 in the hippocampus of PTSD rats, leading to the positive modulation of dendritic spine morphology and synaptic remodeling. This mechanism contributes to the attenuation of fear memory generalization. Given the limitations of current PTSD treatments, these findings offer potential avenues for developing more effective therapeutic strategies.
Collapse
Affiliation(s)
- Yue Qu
- Integrative Medicine Research Center, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Outer Ring East Road No. 232, Higher Education Mega Center, Guangzhou, 510006, China.
| | - Jingna Gu
- Integrative Medicine Research Center, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Outer Ring East Road No. 232, Higher Education Mega Center, Guangzhou, 510006, China.
| | - Lanxin Li
- Integrative Medicine Research Center, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Outer Ring East Road No. 232, Higher Education Mega Center, Guangzhou, 510006, China.
| | - Yuqi Yan
- Integrative Medicine Research Center, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Outer Ring East Road No. 232, Higher Education Mega Center, Guangzhou, 510006, China.
| | - Can Yan
- Integrative Medicine Research Center, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Outer Ring East Road No. 232, Higher Education Mega Center, Guangzhou, 510006, China.
| | - Tiange Zhang
- Integrative Medicine Research Center, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Outer Ring East Road No. 232, Higher Education Mega Center, Guangzhou, 510006, China.
| |
Collapse
|
3
|
Wang J, Zhang S, Boda VK, Chen H, Park H, Parmar K, Ma D, Miller DD, Meibohm B, Du J, Liao FF, Wu Z, Li W. Discovery of a potent and selective TRPC3 antagonist with neuroprotective effects. Bioorg Med Chem 2025; 117:118021. [PMID: 39612770 DOI: 10.1016/j.bmc.2024.118021] [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: 08/12/2024] [Revised: 11/07/2024] [Accepted: 11/22/2024] [Indexed: 12/01/2024]
Abstract
The TRPC3 protein plays a pivotal role in calcium signaling, influencing cell function. Aberrant TRPC3 expression is implicated in various pathologies, including cardiovascular diseases, tumors, and neurodegeneration. Despite its functional similarities with TRPC6 and TRPC7, TRPC3 exhibits distinct roles in disease contexts. Therefore, it is of paramount importance to develop a potent and selective TRPC3 antagonist with favorable drug-like properties. We employed extensive medicinal chemistry synthesis and structure-activity relationships (SARs) study. Thirty-one novel TRPC3 antagonists were designed and synthesized using the lead compound JW-65 as the scaffold. Compound 60a exhibits a 4-fold improvement in potency and displays exceptional selectivity. With favorable drug-like properties, this compound shows a heightened in vitro neuronal protective effect. Molecular modeling suggests possible modes of action between the TRPC3 protein and its antagonists. In summary, 60a holds significant promise for clinical development in conditions associated with TRPC3 dysregulation.
Collapse
Affiliation(s)
- Jiaxing Wang
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, TN 38163, United States
| | - Sicheng Zhang
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, TN 38163, United States
| | - Vijay K Boda
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, TN 38163, United States
| | - Hao Chen
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, TN 38163, United States
| | - Hyunseo Park
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, TN 38163, United States
| | - Keyur Parmar
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, TN 38163, United States
| | - Dejian Ma
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, TN 38163, United States
| | - Duane D Miller
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, TN 38163, United States
| | - Bernd Meibohm
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, TN 38163, United States
| | - Jianyang Du
- Department of Anatomy and Neurobiology, University of Tennessee Health Science Center, Memphis, TN 38163, United States; Neuroscience Institute, University of Tennessee Health Science Center, Memphis, TN 38163, United States
| | - Francesca-Fang Liao
- Department of Pharmacology, University of Tennessee Health Science Center, Memphis, TN 38163, United States
| | - Zhongzhi Wu
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, TN 38163, United States.
| | - Wei Li
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, TN 38163, United States.
| |
Collapse
|
4
|
Wang J, Chen L, Wang Z, Zhang S, Ding D, Lin G, Zhang H, Boda VK, Kong D, Ortyl TC, Wang X, Lu L, Zhou FM, Bezprozvanny I, Du J, Wu Z, Li W, Liao FF. TRPC3 suppression ameliorates synaptic dysfunctions and memory deficits in Alzheimer's disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.16.611061. [PMID: 39345364 PMCID: PMC11430068 DOI: 10.1101/2024.09.16.611061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
Transient receptor potential canonical (TRPC) channels are widely expressed in the brain; however, their precise roles in neurodegeneration, such as Alzheimer's disease (AD) remain elusive. Bioinformatic analysis of the published single-cell RNA-seq data collected from AD patient cohorts indicates that the Trpc3 gene is uniquely upregulated in excitatory neurons. TRPC3 expression is also upregulated in post-mortem AD brains, and in both acute and chronic mouse models of AD. Functional screening of TRPC3 antagonists resulted in a lead inhibitor JW-65, which completely rescued Aβ-induced neurotoxicity, impaired synaptic plasticity (e.g., LTP), and learning memory in acute and chronic experimental AD models. In cultured rat hippocampal neurons, we found that treatment with soluble β-amyloid oligomers (AβOs) induces rapid and sustained upregulation of the TRPC3 expression selectively in excitatory neurons. This aberrantly upregulated TRPC3 contributes to AβOs-induced Ca 2+ overload through the calcium entry and store-release mechanisms. The neuroprotective action of JW-65 is primarily mediated via restoring AβOs-impaired Ca 2+ /calmodulin-mediated signaling pathways, including calmodulin kinases CaMKII/IV and calcineurin (CaN). The synaptic protective mechanism via TRPC3 inhibition was further supported by hippocampal RNA-seq data from the symptomatic 5xFAD mice after chronic treatment with JW-65. Overall, these findings not only validate TRPC3 as a novel therapeutic target for treating synaptic dysfunction of AD but most importantly, disclose a distinct role of upregulated TRPC3 in AD pathogenesis in mediating Ca 2+ dyshomeostasis.
Collapse
|
5
|
Hayato R, Matsumoto T, Higure Y. Ca2+ Depletion in the ER Causes Store-Operated Ca2+ Entry via the TRPC6 Channel in Mouse Brown Adipocytes. Physiol Res 2024; 73:69-80. [PMID: 38466006 PMCID: PMC11019620 DOI: 10.33549/physiolres.935071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Accepted: 10/31/2023] [Indexed: 04/26/2024] Open
Abstract
beta3-adrenergic activation causes Ca2+ release from the mitochondria and subsequent Ca2+ release from the endoplasmic reticulum (ER), evoking store-operated Ca2+ entry (SOCE) due to Ca2+ depletion from the ER in mouse brown adipocytes. In this study, we investigated how Ca2+ depletion from the ER elicits SOCE in mouse brown adipocytes using fluorometry of intracellular Ca2+ concentration ([Ca2+]i). The administration of cyclopiazonic acid (CPA), a reversible sarcoplasmic/endoplasmic reticulum calcium ATPase (SERCA) pump blocker in the ER, caused an increase in [Ca2+]i. Moreover, CPA induced SOCE was suppressed by the administration of a Ca2+ free Krebs solution and the transient receptor potential canonical 6 (TRPC6) selective blockers 2-APB, ML-9 and GsMTx-4 but not Pico145, which blocks TRPC1/4/5. Administration of TRPC6 channel agonist 1-oleoyl-2-acetyl-sn-glycerol (OAG) and flufenamic acid elicited Ca2+ entry. Moreover, our RT-PCR analyses detected mRNAs for TRPC6 in brown adipose tissues. In addition, western blot analyses showed the expression of the TRPC6 protein. Thus, TRPC6 is one of the Ca2+ pathways involved in SOCE. These modes of Ca2+ entry provide the basis for heat production via activation of Ca2+-dependent dehydrogenase and the expression of uncoupling protein 1 (UCP1). Enhancing thermogenic metabolism in brown adipocytes may serve as broad therapeutic utility to reduce obesity and metabolic syndrome.
Collapse
Affiliation(s)
- R Hayato
- Laboratory of Anatomy and Physiology, School of Nutritional Sciences, Nagoya University of Arts and Sciences, Takenoyama, Nissin-City, Aichi, Japan.
| | | | | |
Collapse
|
6
|
Hamad MIK, Emerald BS, Kumar KK, Ibrahim MF, Ali BR, Bataineh MF. Extracellular molecular signals shaping dendrite architecture during brain development. Front Cell Dev Biol 2023; 11:1254589. [PMID: 38155836 PMCID: PMC10754048 DOI: 10.3389/fcell.2023.1254589] [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: 07/10/2023] [Accepted: 11/28/2023] [Indexed: 12/30/2023] Open
Abstract
Proper growth and branching of dendrites are crucial for adequate central nervous system (CNS) functioning. The neuronal dendritic geometry determines the mode and quality of information processing. Any defects in dendrite development will disrupt neuronal circuit formation, affecting brain function. Besides cell-intrinsic programmes, extrinsic factors regulate various aspects of dendritic development. Among these extrinsic factors are extracellular molecular signals which can shape the dendrite architecture during early development. This review will focus on extrinsic factors regulating dendritic growth during early neuronal development, including neurotransmitters, neurotrophins, extracellular matrix proteins, contact-mediated ligands, and secreted and diffusible cues. How these extracellular molecular signals contribute to dendritic growth has been investigated in developing nervous systems using different species, different areas within the CNS, and different neuronal types. The response of the dendritic tree to these extracellular molecular signals can result in growth-promoting or growth-limiting effects, and it depends on the receptor subtype, receptor quantity, receptor efficiency, the animal model used, the developmental time windows, and finally, the targeted signal cascade. This article reviews our current understanding of the role of various extracellular signals in the establishment of the architecture of the dendrites.
Collapse
Affiliation(s)
- Mohammad I. K. Hamad
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Bright Starling Emerald
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Kukkala K. Kumar
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Marwa F. Ibrahim
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Bassam R. Ali
- Department of Genetics and Genomics, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Mo’ath F. Bataineh
- Department of Nutrition and Health, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| |
Collapse
|
7
|
Zernov N, Popugaeva E. Role of Neuronal TRPC6 Channels in Synapse Development, Memory Formation and Animal Behavior. Int J Mol Sci 2023; 24:15415. [PMID: 37895105 PMCID: PMC10607207 DOI: 10.3390/ijms242015415] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Revised: 10/11/2023] [Accepted: 10/16/2023] [Indexed: 10/29/2023] Open
Abstract
The transient receptor potential cation channel, subfamily C, member 6 (TRPC6), has been believed to adjust the formation of an excitatory synapse. The positive regulation of TRPC6 engenders synapse enlargement and improved learning and memory in animal models. TRPC6 is involved in different synaptoprotective signaling pathways, including antagonism of N-methyl-D-aspartate receptor (NMDAR), activation of brain-derived neurotrophic factor (BDNF) and postsynaptic store-operated calcium entry. Positive regulation of TRPC6 channels has been repeatedly shown to be good for memory formation and storage. TRPC6 is mainly expressed in the hippocampus, particularly in the dentate granule cells, cornu Ammonis 3 (CA3) pyramidal cells and gamma-aminobutyric acid (GABA)ergic interneurons. It has been observed that TRPC6 agonists have a great influence on animal behavior including memory formation and storage The purpose of this review is to collect the available information on the role of TRPC6 in memory formation in various parts of the brain to understand how TRPC6-specific pharmaceutical agents will affect memory in distinct parts of the central nervous system (CNS).
Collapse
Affiliation(s)
| | - Elena Popugaeva
- Laboratory of Molecular Neurodegeneration, Peter the Great St. Petersburg Polytechnic University, 195251 St. Petersburg, Russia
| |
Collapse
|
8
|
Zhao Y, Liu J, Liu S, Yang P, Liang Y, Ma J, Mao S, Sun C, Yang Y. Fibroblast exosomal TFAP2C induced by chitosan oligosaccharides promotes peripheral axon regeneration via the miR-132-5p/CAMKK1 axis. Bioact Mater 2023; 26:249-263. [PMID: 36936807 PMCID: PMC10020534 DOI: 10.1016/j.bioactmat.2023.03.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Revised: 03/03/2023] [Accepted: 03/03/2023] [Indexed: 03/11/2023] Open
Abstract
Chitosan and its degradation product, oligosaccharides, have been shown to facilitate peripheral nerve regeneration. However, the underlying mechanisms are not well understood. In this study, we analyzed the protein expression profiles in sciatic nerves after injury using proteomics. A group of proteins related to exosome packaging and transport is up-regulated by chitosan oligosaccharides (COS), implying that exosomes are involved in COS-induced peripheral nerve regeneration. In fact, exosomes derived from fibroblasts (f-EXOs) treated with COS significantly promoted axon extension and regeneration. Exosomal protein identification and functional studies, revealed that TFAP2C is a key factor in neurite outgrowth induced by COS-f-EXOs. Furthermore, we showed that TFAP2C targets the pri-miRNA-132 gene and represses miR-132-5p expression in dorsal root ganglion neurons. Camkk1 is a downstream substrate of miR-132-5p that positively affects axon extension. In rats, miR-132-5p antagomir stimulates CAMKK1 expression and improves axon regeneration and functional recovery in sciatic nerves after injury. Our data reveal the mechanism for COS in axon regeneration, that is COS induce fibroblasts to produce TFAP2C-enriched EXOs, which are then transferred into axons to promote axon regeneration via miR-132-5p/CAMKK1. Moreover, these results show a new facet of fibroblasts in axon regeneration in peripheral nerves.
Collapse
Affiliation(s)
- Yahong Zhao
- Key Laboratory of Neuroregeneration of Jiangsu Province and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, 19 Qixiu Road, Nantong, Jiangsu, 226001, China
| | - Jina Liu
- Key Laboratory of Neuroregeneration of Jiangsu Province and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, 19 Qixiu Road, Nantong, Jiangsu, 226001, China
| | - Sha Liu
- Key Laboratory of Neuroregeneration of Jiangsu Province and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, 19 Qixiu Road, Nantong, Jiangsu, 226001, China
| | - Panpan Yang
- School of Medicine, Nantong University, 19 Qixiu Road, Nantong, Jiangsu, 226001, China
| | - Yunyun Liang
- Key Laboratory of Neuroregeneration of Jiangsu Province and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, 19 Qixiu Road, Nantong, Jiangsu, 226001, China
| | - Jinyu Ma
- Key Laboratory of Neuroregeneration of Jiangsu Province and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, 19 Qixiu Road, Nantong, Jiangsu, 226001, China
| | - Susu Mao
- Key Laboratory of Neuroregeneration of Jiangsu Province and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, 19 Qixiu Road, Nantong, Jiangsu, 226001, China
| | - Cheng Sun
- Key Laboratory of Neuroregeneration of Jiangsu Province and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, 19 Qixiu Road, Nantong, Jiangsu, 226001, China
| | - Yumin Yang
- Key Laboratory of Neuroregeneration of Jiangsu Province and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, 19 Qixiu Road, Nantong, Jiangsu, 226001, China
| |
Collapse
|
9
|
Baracaldo-Santamaría D, Avendaño-Lopez SS, Ariza-Salamanca DF, Rodriguez-Giraldo M, Calderon-Ospina CA, González-Reyes RE, Nava-Mesa MO. Role of Calcium Modulation in the Pathophysiology and Treatment of Alzheimer's Disease. Int J Mol Sci 2023; 24:ijms24109067. [PMID: 37240413 DOI: 10.3390/ijms24109067] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 05/02/2023] [Accepted: 05/05/2023] [Indexed: 05/28/2023] Open
Abstract
Alzheimer's disease (AD) is a chronic neurodegenerative disease and the most frequent cause of progressive dementia in senior adults. It is characterized by memory loss and cognitive impairment secondary to cholinergic dysfunction and N-methyl-D-aspartate (NMDA)-mediated neurotoxicity. Intracellular neurofibrillary tangles, extracellular plaques composed of amyloid-β (Aβ), and selective neurodegeneration are the anatomopathological hallmarks of this disease. The dysregulation of calcium may be present in all the stages of AD, and it is associated with other pathophysiological mechanisms, such as mitochondrial failure, oxidative stress, and chronic neuroinflammation. Although the cytosolic calcium alterations in AD are not completely elucidated, some calcium-permeable channels, transporters, pumps, and receptors have been shown to be involved at the neuronal and glial levels. In particular, the relationship between glutamatergic NMDA receptor (NMDAR) activity and amyloidosis has been widely documented. Other pathophysiological mechanisms involved in calcium dyshomeostasis include the activation of L-type voltage-dependent calcium channels, transient receptor potential channels, and ryanodine receptors, among many others. This review aims to update the calcium-dysregulation mechanisms in AD and discuss targets and molecules with therapeutic potential based on their modulation.
Collapse
Affiliation(s)
- Daniela Baracaldo-Santamaría
- Pharmacology Unit, Department of Biomedical Sciences, School of Medicine and Health Sciences, Universidad del Rosario, Bogotá 111221, Colombia
| | - Sara Sofia Avendaño-Lopez
- Pharmacology Unit, Department of Biomedical Sciences, School of Medicine and Health Sciences, Universidad del Rosario, Bogotá 111221, Colombia
| | - Daniel Felipe Ariza-Salamanca
- Medical and Health Sciences Education Research Group, School of Medicine and Health Sciences, Universidad del Rosario, Bogotá 111221, Colombia
| | - Mateo Rodriguez-Giraldo
- Grupo de Investigación en Neurociencias (NeURos), Centro de Neurociencias Neurovitae-UR, Instituto de Medicina Traslacional (IMT), Escuela de Medicina y Ciencias de la Salud, Universidad del Rosario, Bogotá 111221, Colombia
| | - Carlos A Calderon-Ospina
- Pharmacology Unit, Department of Biomedical Sciences, School of Medicine and Health Sciences, Universidad del Rosario, Bogotá 111221, Colombia
- Grupo de Investigación en Ciencias Biomédicas Aplicadas (UR Biomed), School of Medicine and Health Sciences, Universidad del Rosario, Bogotá 111221, Colombia
| | - Rodrigo E González-Reyes
- Grupo de Investigación en Neurociencias (NeURos), Centro de Neurociencias Neurovitae-UR, Instituto de Medicina Traslacional (IMT), Escuela de Medicina y Ciencias de la Salud, Universidad del Rosario, Bogotá 111221, Colombia
| | - Mauricio O Nava-Mesa
- Grupo de Investigación en Neurociencias (NeURos), Centro de Neurociencias Neurovitae-UR, Instituto de Medicina Traslacional (IMT), Escuela de Medicina y Ciencias de la Salud, Universidad del Rosario, Bogotá 111221, Colombia
| |
Collapse
|
10
|
Hong DK, Kho AR, Lee SH, Kang BS, Park MK, Choi BY, Suh SW. Pathophysiological Roles of Transient Receptor Potential (Trp) Channels and Zinc Toxicity in Brain Disease. Int J Mol Sci 2023; 24:ijms24076665. [PMID: 37047637 PMCID: PMC10094935 DOI: 10.3390/ijms24076665] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 03/30/2023] [Accepted: 04/01/2023] [Indexed: 04/05/2023] Open
Abstract
Maintaining the correct ionic gradient from extracellular to intracellular space via several membrane-bound transporters is critical for maintaining overall cellular homeostasis. One of these transporters is the transient receptor potential (TRP) channel family that consists of six putative transmembrane segments systemically expressed in mammalian tissues. Upon the activation of TRP channels by brain disease, several cations are translocated through TRP channels. Brain disease, especially ischemic stroke, epilepsy, and traumatic brain injury, triggers the dysregulation of ionic gradients and promotes the excessive release of neuro-transmitters and zinc. The divalent metal cation zinc is highly distributed in the brain and is specifically located in the pre-synaptic vesicles as free ions, usually existing in cytoplasm bound with metallothionein. Although adequate zinc is essential for regulating diverse physiological functions, the brain-disease-induced excessive release and translocation of zinc causes cell damage, including oxidative stress, apoptotic cascades, and disturbances in energy metabolism. Therefore, the regulation of zinc homeostasis following brain disease is critical for the prevention of brain damage. In this review, we summarize recent experimental research findings regarding how TRP channels (mainly TRPC and TRPM) and zinc are regulated in animal brain-disease models of global cerebral ischemia, epilepsy, and traumatic brain injury. The blockade of zinc translocation via the inhibition of TRPC and TRPM channels using known channel antagonists, was shown to be neuroprotective in brain disease. The regulation of both zinc and TRP channels may serve as targets for treating and preventing neuronal death.
Collapse
Affiliation(s)
- Dae Ki Hong
- Department of Physiology, College of Medicine, Hallym University, Chuncheon 24252, Republic of Korea
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - A Ra Kho
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, College of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Song Hee Lee
- Department of Physiology, College of Medicine, Hallym University, Chuncheon 24252, Republic of Korea
| | - Beom Seok Kang
- Department of Physiology, College of Medicine, Hallym University, Chuncheon 24252, Republic of Korea
| | - Min Kyu Park
- Department of Physiology, College of Medicine, Hallym University, Chuncheon 24252, Republic of Korea
| | - Bo Young Choi
- Department of Physical Education, Hallym University, Chuncheon 24252, Republic of Korea
- Institute of Sport Science, Hallym University, Chuncheon 24252, Republic of Korea
| | - Sang Won Suh
- Department of Physiology, College of Medicine, Hallym University, Chuncheon 24252, Republic of Korea
| |
Collapse
|
11
|
Liu H, Zhang X, Shi P, Yuan J, Jia Q, Pi C, Chen T, Xiong L, Chen J, Tang J, Yue R, Liu Z, Shen H, Zuo Y, Wei Y, Zhao L. α7 Nicotinic acetylcholine receptor: a key receptor in the cholinergic anti-inflammatory pathway exerting an antidepressant effect. J Neuroinflammation 2023; 20:84. [PMID: 36973813 PMCID: PMC10041767 DOI: 10.1186/s12974-023-02768-z] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2023] [Accepted: 03/17/2023] [Indexed: 03/28/2023] Open
Abstract
Depression is a common mental illness, which is related to monoamine neurotransmitters and the dysfunction of the cholinergic, immune, glutamatergic, and neuroendocrine systems. The hypothesis of monoamine neurotransmitters is one of the commonly recognized pathogenic mechanisms of depression; however, the drugs designed based on this hypothesis have not achieved good clinical results. A recent study demonstrated that depression and inflammation were strongly correlated, and the activation of alpha7 nicotinic acetylcholine receptor (α7 nAChR)-mediated cholinergic anti-inflammatory pathway (CAP) in the cholinergic system exhibited good therapeutic effects against depression. Therefore, anti-inflammation might be a potential direction for the treatment of depression. Moreover, it is also necessary to further reveal the key role of inflammation and α7 nAChR in the pathogenesis of depression. This review focused on the correlations between inflammation and depression as well-discussed the crucial role of α7 nAChR in the CAP.
Collapse
Affiliation(s)
- Huiyang Liu
- grid.410578.f0000 0001 1114 4286Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy of Southwest Medical University, Luzhou, 646000 People’s Republic of China
- grid.488387.8Key Laboratory of Medical Electrophysiology, Ministry of Education, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, No. 182, Chunhui Road, Longmatan District, Luzhou, 646000 Sichuan People’s Republic of China
- grid.410578.f0000 0001 1114 4286Central Nervous System Drug Key Laboratory of Sichuan Province, School of Pharmacy of Southwest Medical University, Luzhou, 646000 Sichuan People’s Republic of China
- grid.488387.8Luzhou Key Laboratory of Traditional Chinese Medicine for Chronic Diseases Jointly Built by Sichuan and Chongqing, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, 646000 Sichuan People’s Republic of China
| | - Xiaomei Zhang
- grid.469520.c0000 0004 1757 8917Luzhou Key Laboratory of Traditional Chinese Medicine for Chronic Diseases Jointly Built by Sichuan and Chongqing, Institute of Medicinal Chemistry of Chinese Medicine, Chongqing Academy of Chinese Materia Medica, Chongqing, 400065 People’s Republic of China
| | - Peng Shi
- grid.410578.f0000 0001 1114 4286Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy of Southwest Medical University, Luzhou, 646000 People’s Republic of China
- grid.488387.8Key Laboratory of Medical Electrophysiology, Ministry of Education, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, No. 182, Chunhui Road, Longmatan District, Luzhou, 646000 Sichuan People’s Republic of China
- grid.410578.f0000 0001 1114 4286Central Nervous System Drug Key Laboratory of Sichuan Province, School of Pharmacy of Southwest Medical University, Luzhou, 646000 Sichuan People’s Republic of China
- grid.488387.8Luzhou Key Laboratory of Traditional Chinese Medicine for Chronic Diseases Jointly Built by Sichuan and Chongqing, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, 646000 Sichuan People’s Republic of China
| | - Jiyuan Yuan
- grid.488387.8Clinical Trial Center, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, 646000 Sichuan People’s Republic of China
| | - Qiang Jia
- grid.488387.8Ethics Committee Office, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, 646000 Sichuan China
| | - Chao Pi
- grid.410578.f0000 0001 1114 4286Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy of Southwest Medical University, Luzhou, 646000 People’s Republic of China
| | - Tao Chen
- grid.410578.f0000 0001 1114 4286Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy of Southwest Medical University, Luzhou, 646000 People’s Republic of China
- grid.488387.8Key Laboratory of Medical Electrophysiology, Ministry of Education, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, No. 182, Chunhui Road, Longmatan District, Luzhou, 646000 Sichuan People’s Republic of China
- grid.410578.f0000 0001 1114 4286Central Nervous System Drug Key Laboratory of Sichuan Province, School of Pharmacy of Southwest Medical University, Luzhou, 646000 Sichuan People’s Republic of China
- grid.488387.8Luzhou Key Laboratory of Traditional Chinese Medicine for Chronic Diseases Jointly Built by Sichuan and Chongqing, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, 646000 Sichuan People’s Republic of China
| | - Linjin Xiong
- grid.410578.f0000 0001 1114 4286Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy of Southwest Medical University, Luzhou, 646000 People’s Republic of China
- grid.488387.8Key Laboratory of Medical Electrophysiology, Ministry of Education, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, No. 182, Chunhui Road, Longmatan District, Luzhou, 646000 Sichuan People’s Republic of China
- grid.410578.f0000 0001 1114 4286Central Nervous System Drug Key Laboratory of Sichuan Province, School of Pharmacy of Southwest Medical University, Luzhou, 646000 Sichuan People’s Republic of China
- grid.488387.8Luzhou Key Laboratory of Traditional Chinese Medicine for Chronic Diseases Jointly Built by Sichuan and Chongqing, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, 646000 Sichuan People’s Republic of China
| | - Jinglin Chen
- grid.410578.f0000 0001 1114 4286Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy of Southwest Medical University, Luzhou, 646000 People’s Republic of China
- grid.488387.8Key Laboratory of Medical Electrophysiology, Ministry of Education, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, No. 182, Chunhui Road, Longmatan District, Luzhou, 646000 Sichuan People’s Republic of China
- grid.410578.f0000 0001 1114 4286Central Nervous System Drug Key Laboratory of Sichuan Province, School of Pharmacy of Southwest Medical University, Luzhou, 646000 Sichuan People’s Republic of China
- grid.488387.8Luzhou Key Laboratory of Traditional Chinese Medicine for Chronic Diseases Jointly Built by Sichuan and Chongqing, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, 646000 Sichuan People’s Republic of China
| | - Jia Tang
- grid.410578.f0000 0001 1114 4286Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy of Southwest Medical University, Luzhou, 646000 People’s Republic of China
- grid.488387.8Key Laboratory of Medical Electrophysiology, Ministry of Education, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, No. 182, Chunhui Road, Longmatan District, Luzhou, 646000 Sichuan People’s Republic of China
- grid.410578.f0000 0001 1114 4286Central Nervous System Drug Key Laboratory of Sichuan Province, School of Pharmacy of Southwest Medical University, Luzhou, 646000 Sichuan People’s Republic of China
- grid.488387.8Luzhou Key Laboratory of Traditional Chinese Medicine for Chronic Diseases Jointly Built by Sichuan and Chongqing, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, 646000 Sichuan People’s Republic of China
| | - Ruxu Yue
- grid.410578.f0000 0001 1114 4286Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy of Southwest Medical University, Luzhou, 646000 People’s Republic of China
- grid.488387.8Key Laboratory of Medical Electrophysiology, Ministry of Education, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, No. 182, Chunhui Road, Longmatan District, Luzhou, 646000 Sichuan People’s Republic of China
- grid.410578.f0000 0001 1114 4286Central Nervous System Drug Key Laboratory of Sichuan Province, School of Pharmacy of Southwest Medical University, Luzhou, 646000 Sichuan People’s Republic of China
- grid.488387.8Luzhou Key Laboratory of Traditional Chinese Medicine for Chronic Diseases Jointly Built by Sichuan and Chongqing, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, 646000 Sichuan People’s Republic of China
| | - Zerong Liu
- Central Nervous System Drug Key Laboratory of Sichuan Province, Sichuan Credit Pharmaceutical CO., Ltd., Luzhou, 646000 Sichuan China
- grid.190737.b0000 0001 0154 0904Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400030 China
| | - Hongping Shen
- grid.488387.8Clinical Trial Center, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, 646000 Sichuan People’s Republic of China
| | - Ying Zuo
- grid.488387.8Department of Comprehensive Medicine, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, No. 182, Chunhui Road, Longmatan District, Luzhou, 646000 Sichuan China
| | - Yumeng Wei
- grid.410578.f0000 0001 1114 4286Central Nervous System Drug Key Laboratory of Sichuan Province, School of Pharmacy of Southwest Medical University, Luzhou, 646000 Sichuan People’s Republic of China
| | - Ling Zhao
- grid.488387.8Key Laboratory of Medical Electrophysiology, Ministry of Education, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, No. 182, Chunhui Road, Longmatan District, Luzhou, 646000 Sichuan People’s Republic of China
- grid.410578.f0000 0001 1114 4286Central Nervous System Drug Key Laboratory of Sichuan Province, School of Pharmacy of Southwest Medical University, Luzhou, 646000 Sichuan People’s Republic of China
- grid.488387.8Luzhou Key Laboratory of Traditional Chinese Medicine for Chronic Diseases Jointly Built by Sichuan and Chongqing, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, 646000 Sichuan People’s Republic of China
| |
Collapse
|
12
|
Xu Q, Zou Y, Miao Z, Jiang L, Zhao X. Transient receptor potential ion channels and cerebral stroke. Brain Behav 2023; 13:e2843. [PMID: 36527242 PMCID: PMC9847613 DOI: 10.1002/brb3.2843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2022] [Accepted: 11/23/2022] [Indexed: 12/23/2022] Open
Abstract
METHODS The databases Pubmed, and the National Library of Medicine were searched for literature. All papers on celebral stroke and transient receptor potential ion channels were considered. RESULTS Stroke is the second leading cause of death and disability, with an increasing incidence in developing countries. About 75 per cent of strokes are caused by occlusion of cerebral arteries, and substantial advances have been made in elucidating mechanisms how stroke affects the brain. Transient receptor potential (TRP) ion channels are calcium-permeable channels highly expressed in brain that drives Ca2+ entry into multiple cellular compartments. TRPC1/3/4/6, TRPV1/2/4, and TRPM2/4/7 channels have been implicated in stroke pathophysiology. CONCLUSIONS Although the precise mechanism of transient receptor potential ion channels in cerebral stroke is still unclear, it has the potential to be a therapeutic target for patients with stroke if developed appropriately. Hence, more research is needed to prove its efficacy in this context.
Collapse
Affiliation(s)
- Qin'yi Xu
- Department of Neurosurgery, The Affiliated Wuxi No. 2 Hospital of Nanjing Medical University, Wuxi, China
| | - Yan Zou
- Department of Neurosurgery, The Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Zeng'li Miao
- Department of Neurosurgery, The Affiliated Wuxi No. 2 Hospital of Nanjing Medical University, Wuxi, China
| | - Lei Jiang
- Department of Neurosurgery, The Affiliated Wuxi No. 2 Hospital of Nanjing Medical University, Wuxi, China
| | - Xu'dong Zhao
- Department of Neurosurgery, The Affiliated Wuxi No. 2 Hospital of Nanjing Medical University, Wuxi, China
| |
Collapse
|
13
|
Nayak M, Das D, Pradhan J, Ahmed R, Laureano-Melo R, Dandapat J. Epigenetic signature in neural plasticity: the journey so far and journey ahead. Heliyon 2022; 8:e12292. [PMID: 36590572 PMCID: PMC9798197 DOI: 10.1016/j.heliyon.2022.e12292] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 10/31/2022] [Accepted: 12/05/2022] [Indexed: 12/23/2022] Open
Abstract
Neural plasticity is a remarkable characteristic of the brain which allows neurons to rewire their structure in response to internal and external stimuli. Many external stimuli collectively referred to as 'epigenetic factors' strongly influence structural and functional reorganization of the brain, thereby acting as a potential driver of neural plasticity. DNA methylation and demethylation, histone acetylation, and deacetylation are some of the frontline epigenetic mechanisms behind neural plasticity. Epigenetic signature molecules (mostly proteins) play a pivotal role in epigenetic reprogramming. Though neuro-epigenetics is an incredibly important field of emerging research, the critical role of signature proteins associated with epigenetic alteration and their involvement in neural plasticity needs further attention. This study gives an integrated and systematic overview of the current state of knowledge with a clear idea of types of neural plasticity and the context-dependent role of epigenetic signature molecules and their modulation by some natural bioactive compounds.
Collapse
Affiliation(s)
- Madhusmita Nayak
- Post-Graduate Department of Biotechnology, Utkal University, Bhubaneswar 751004, Odisha, India,Centre of Excellence in Integrated Omics and Computational Biology, Utkal University, Bhubaneswar 751004, Odisha, India
| | - Diptimayee Das
- Post-Graduate Department of Biotechnology, Utkal University, Bhubaneswar 751004, Odisha, India,Faculty of Allied Health Science, Chettinad Academy of Research and Education, Chettinad Hospital and Research Institute, Chennai India
| | - Jyotsnarani Pradhan
- Post-Graduate Department of Biotechnology, Utkal University, Bhubaneswar 751004, Odisha, India,Corresponding author.
| | - R.G. Ahmed
- Division of Anatomy and Embryology, Zoology Department, Faculty of Science, Beni-Suef University, Beni-Suef, Egypt
| | - Roberto Laureano-Melo
- Barra Mansa University Center, R. Ver. Pinho de Carvalho, 267, 27330-550, Barra Mansa, Rio de Janeiro, Brazil
| | - Jagneshwar Dandapat
- Post-Graduate Department of Biotechnology, Utkal University, Bhubaneswar 751004, Odisha, India,Centre of Excellence in Integrated Omics and Computational Biology, Utkal University, Bhubaneswar 751004, Odisha, India,Corresponding author.
| |
Collapse
|
14
|
Song Q, Huang W, Ye W, Yan H, Wang L, Yang Y, Cheng X, Zhang W, Zheng J, He P, He Y, Fang D, Han X. Neuroprotective Effects of Estrogen Through BDNF-Transient Receptor Potential Channels 6 Signaling Pathway in the Hippocampus in a Rat Model of Perimenopausal Depression. Front Aging Neurosci 2022; 14:869274. [PMID: 35875795 PMCID: PMC9305198 DOI: 10.3389/fnagi.2022.869274] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Accepted: 06/07/2022] [Indexed: 12/18/2022] Open
Abstract
Estradiol (E2) has been proven to be effective in treating perimenopausal depression (PD); however, the downstream signaling pathways have not been fully elucidated. Transient receptor potential channels 6 (TRPC6) plays a vital role in promoting neuronal development and the formation of excitatory synapses. At present, we found that the serum levels of E2 and brain-derived neurotrophic factor (BDNF) declined significantly in the women with PD compared to perimenopausal women, which was accompanied by a clear reduction in TRPC6 levels. To further reveal the effects of TRPC6 on neuronal survival and excitability, the PD-like rat model was established by the total removal of left ovary and 80% removal of right ovary followed by 21 days of the chronic unpredictable mild stress. Intragastric administration of E2 (2 mg/kg), intraperitoneal injection of BDNF/TrB signaling pathway inhibitor (K252a, 100 μg/kg) and TRPC6 agonist (OAG, 0.6 mg/kg), and intracerebroventricular infusion of anti-BDNF antibody for blocking BDNF (0.5 μg/24 μl/rat) daily for 21 days were conducted. The levels of BDNF and TRPC6 in rat serum were lower in PD rats compared to the control rats; the depression-like behavior was induced, the neuronal death rate in the hippocampus increased, and the thickness of postsynaptic density (PSD) and the number of asymmetric synapses decreased significantly in the PD group. E2 treatment greatly upregulated the serum levels of BDNF and TRPC6, the neuronal excitability indicated by an elevation in the PSD thickness and the numbers of asymmetric synapses, and these actions were reversed by K252a; co-administration of TRPC6 agonist and K252a improved neuronal degeneration and increased the neuronal excitability induced in the E2-treated PD rats. K252a or anti-BDNF antibody inhibited the increased neuronal BDNF and TRPC6 expression in E2-treated PD rats; co-treatment of TRPC6 agonist and anti-BDNF antibody reduced neuronal death and increased the BDNF and TRPC6 expression in the hippocampal CA1 neurons in the E2-treated PD rats. These results suggest that the neuroprotective role of E2 in PD is closely related to enhance the activity of BDNF/TRPC6 pathway and is helpful to provide new prevention and strategies.
Collapse
|
15
|
Wang Z, do Carmo JM, da Silva AA, Fu Y, Jaynes LT, Sears J, Li X, Mouton AJ, Omoto ACM, Xu BP, Hall JE. Transient receptor potential cation channel 6 (TRPC6) deficiency leads to increased body weight and metabolic dysfunction. Am J Physiol Regul Integr Comp Physiol 2022; 323:R81-R97. [PMID: 35537100 DOI: 10.1152/ajpregu.00097.2021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
TRPC6, a member of the TRPC family, is expressed in the hypothalamus and modulates cell Ca2+ influx. However, the role of TRPC6 in controlling metabolic and cardiovascular functions under normal conditions has not been previously determined. Thus, the impacts of TRPC6 deletion on energy balance, metabolic and cardiovascular regulation as well as the anorexic responses to leptin and melanocortin 3/4 receptor (MC3/4R) activation were investigated in this study. Extensive cardiometabolic phenotyping was conducted in male and female TRPC6 knock out (KO) and control mice from 6 to 24 weeks of age to assess mechanisms by which TRPC6 influences regulation of energy balance and blood pressure (BP). We found that TRPC6 KO mice are heavier with greater adiposity, hyperphagic, and have reduced energy expenditure, impaired glucose tolerance, hyperinsulinemia, and increased liver fat compared to controls. TRPC6 KO mice also have smaller brains, reduced POMC mRNA levels in the hypothalamus, and impaired anorexic response to leptin but not to MC3/4R activation. BP and heart rate, assessed by telemetry, were similar in TRPC6 KO and control mice, and BP responses to air-jet stress were attenuated in TRPC6 KO mice despite increased body weight and metabolic disorders that normally raise BP and increase BP responses to stress. Our results provide evidence for a novel and important role of TRPC6 in controlling energy balance, adiposity, and glucose homeostasis, which suggests that normal TRPC6 function may be necessary to link weight gain and hyperleptinemia with BP responses to acute stress.
Collapse
Affiliation(s)
- Zhen Wang
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS, United States.,Mississippi Center for Obesity Research, University of Mississippi Medical Center, Jackson, MS, United States
| | - Jussara M do Carmo
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS, United States.,Mississippi Center for Obesity Research, University of Mississippi Medical Center, Jackson, MS, United States
| | - Alexandre A da Silva
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS, United States.,Mississippi Center for Obesity Research, University of Mississippi Medical Center, Jackson, MS, United States
| | - Yiling Fu
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS, United States.,Mississippi Center for Obesity Research, University of Mississippi Medical Center, Jackson, MS, United States
| | - Lance T Jaynes
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS, United States.,Mississippi Center for Obesity Research, University of Mississippi Medical Center, Jackson, MS, United States
| | - Jaylan Sears
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS, United States.,Mississippi Center for Obesity Research, University of Mississippi Medical Center, Jackson, MS, United States
| | - Xuan Li
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS, United States.,Mississippi Center for Obesity Research, University of Mississippi Medical Center, Jackson, MS, United States
| | - Alan J Mouton
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS, United States.,Mississippi Center for Obesity Research, University of Mississippi Medical Center, Jackson, MS, United States
| | - Ana C M Omoto
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS, United States.,Mississippi Center for Obesity Research, University of Mississippi Medical Center, Jackson, MS, United States
| | - Brittney P Xu
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS, United States.,Mississippi Center for Obesity Research, University of Mississippi Medical Center, Jackson, MS, United States
| | - John E Hall
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS, United States.,Mississippi Center for Obesity Research, University of Mississippi Medical Center, Jackson, MS, United States
| |
Collapse
|
16
|
Hou P, Sun Y, Yang W, Wu H, Sun L, Xiu X, Xiu C, Zhang X, Zhang W. Magnesium promotes osteogenesis via increasing OPN expression and activating CaM/CaMKIV/CREB1 pathway. J Biomed Mater Res B Appl Biomater 2022; 110:1594-1603. [PMID: 35106922 DOI: 10.1002/jbm.b.35020] [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: 11/04/2021] [Revised: 01/05/2022] [Accepted: 01/18/2022] [Indexed: 11/11/2022]
Abstract
Magnesium (Mg) based alloy has been used as a biodegradable implant for fracture repair with considerable efficacy, and it has been proved that magnesium ion (Mg2+ ), one of the degradation products, could stimulate osteogenesis. Here, we investigated the osteogenesis property of magnesium both in vitro and in vivo, and to identify the cellular and molecular mechanisms that mediate these effects. Results showed that magnesium exerts a dose-dependent increase in the proliferation of MC3T3 and MG63 cells, and in the expression of osteopontin (OPN), a promising biomarker of osteogenesis. Subsequently, the protein-protein interaction (PPI) network analysis showed the interactions between calmodulin (CaM) and calmodulin-dependent protein kinase (CaMK) and CREB1. The ratio of p-CaMKIV/CaMKIV and p-CREB1/CREB were increased at protein level in MC3T3 and MG63 cells after treatment with Mg2+ . Dual-luciferase reporter gene assay showed that p-CREB1 could directly bind to OPN promoter and up-regulate the transcription of OPN after nuclear entry. Meanwhile, the expression of OPN and p-CREB1, which increased after Mg2+ treatment, was down-regulated by sh-CaMKIV or sh-CREB1. Moreover, the mineralized deposit and expression of OPN were reduced after treatment with an inhibitor of CaMKIV, KN93. In addition, massive cavities in the cortical bone around the Mg screw were showed in vivo after injection of KN93. These data indicated that the osteogenic effect of Mg is related to the activation OPN through CaM/CaMKIV/CREB1 signaling pathway.
Collapse
Affiliation(s)
- Peng Hou
- Orthopaedic Department, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Yu Sun
- State Key Laboratory of Metal Matrix Composites, School of Materials Science and Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Weichao Yang
- Orthopaedic Department, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Hongliu Wu
- State Key Laboratory of Metal Matrix Composites, School of Materials Science and Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Luyuan Sun
- Orthopaedic Department, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Xinjie Xiu
- College of Food Science and Engineering of Qingdao Agricultural University, Qingdao, China
| | - Chaoyang Xiu
- College of Food Science and Engineering of Qingdao Agricultural University, Qingdao, China
| | - Xiaonong Zhang
- State Key Laboratory of Metal Matrix Composites, School of Materials Science and Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Wen Zhang
- Orthopaedic Department, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| |
Collapse
|
17
|
Mutations in trpγ, the homologue of TRPC6 autism candidate gene, causes autism-like behavioral deficits in Drosophila. Mol Psychiatry 2022; 27:3328-3342. [PMID: 35501408 PMCID: PMC9708601 DOI: 10.1038/s41380-022-01555-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 03/15/2022] [Accepted: 03/29/2022] [Indexed: 11/09/2022]
Abstract
Autism Spectrum Disorder (ASD) is characterized by impaired social communication, restricted interests, and repetitive and stereotyped behaviors. The TRPC6 (transient receptor potential channel 6) represents an ASD candidate gene under an oligogenic/multifactorial model based on the initial description and cellular characterization of an individual with ASD bearing a de novo heterozygous mutation disrupting TRPC6, together with the enrichment of disruptive TRPC6 variants in ASD cases as compared to controls. Here, we perform a clinical re-evaluation of the initial non-verbal patient, and also present eight newly reported individuals ascertained for ASD and bearing predicted loss-of-function mutations in TRPC6. In order to understand the consequences of mutations in TRPC6 on nervous system function, we used the fruit fly, Drosophila melanogaster, to show that null mutations in transient receptor gamma (trpγ; the fly gene most similar to TRPC6), cause a number of behavioral defects that mirror features seen in ASD patients, including deficits in social interactions (based on courtship behavior), impaired sleep homeostasis (without affecting the circadian control of sleep), hyperactivity in both young and old flies, and defects in learning and memory. Some defects, most notably in sleep, differed in severity between males and females and became normal with age. Interestingly, hyperforin, a TRPC6 agonist and the primary active component of the St. John's wort antidepressant, attenuated many of the deficits expressed by trpγ mutant flies. In summary, our results provide further evidence that the TRPC6 gene is a risk factor for ASD. In addition, they show that the behavioral defects caused by mutations in TRPC6 can be modeled in Drosophila, thereby establishing a paradigm to examine the impact of mutations in other candidate genes.
Collapse
|
18
|
Structure-function relationships of the disease-linked A218T oxytocin receptor variant. Mol Psychiatry 2022; 27:907-917. [PMID: 34980886 PMCID: PMC9054668 DOI: 10.1038/s41380-021-01241-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Revised: 06/24/2021] [Accepted: 07/15/2021] [Indexed: 12/20/2022]
Abstract
Various single nucleotide polymorphisms (SNPs) in the oxytocin receptor (OXTR) gene have been associated with behavioral traits, autism spectrum disorder (ASD) and other diseases. The non-synonymous SNP rs4686302 results in the OXTR variant A218T and has been linked to core characteristics of ASD, trait empathy and preterm birth. However, the molecular and intracellular mechanisms underlying those associations are still elusive. Here, we uncovered the molecular and intracellular consequences of this mutation that may affect the psychological or behavioral outcome of oxytocin (OXT)-treatment regimens in clinical studies, and provide a mechanistic explanation for an altered receptor function. We created two monoclonal HEK293 cell lines, stably expressing either the wild-type or A218T OXTR. We detected an increased OXTR protein stability, accompanied by a shift in Ca2+ dynamics and reduced MAPK pathway activation in the A218T cells. Combined whole-genome and RNA sequencing analyses in OXT-treated cells revealed 7823 differentially regulated genes in A218T compared to wild-type cells, including 429 genes being associated with ASD. Furthermore, computational modeling provided a molecular basis for the observed change in OXTR stability suggesting that the OXTR mutation affects downstream events by altering receptor activation and signaling, in agreement with our in vitro results. In summary, our study provides the cellular mechanism that links the OXTR rs4686302 SNP with genetic dysregulations associated with aspects of ASD.
Collapse
|
19
|
Roy P, Martinelli I, Moruzzi M, Maggi F, Amantini C, Micioni Di Bonaventura MV, Cifani C, Amenta F, Tayebati SK, Tomassoni D. Ion channels alterations in the forebrain of high-fat diet fed rats. Eur J Histochem 2021; 65:3305. [PMID: 34814650 PMCID: PMC8636841 DOI: 10.4081/ejh.2021.3305] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Accepted: 10/27/2021] [Indexed: 12/22/2022] Open
Abstract
Evidence suggests that transient receptor potential (TRP) ion channels dysfunction significantly contributes to the physiopathology of metabolic and neurological disorders. Dysregulation in functions and expression in genes encoding the TRP channels cause several inherited diseases in humans (the so-called 'TRP channelopathies'), which affect the cardiovascular, renal, skeletal, and nervous systems. This study aimed to evaluate the expression of ion channels in the forebrain of rats with diet-induced obesity (DIO). DIO rats were studied after 17 weeks under a hypercaloric diet (high-fat diet, HFD) and were compared to the control rats with a standard diet (CHOW). To determine the systemic effects of HFD exposure, we examined food intake, fat mass content, fasting glycemia, insulin levels, cholesterol, and triglycerides. qRT-PCR, Western blot, and immunochemistry analysis were performed in the frontal cortex (FC) and hippocampus (HIP). After 17 weeks of HFD, DIO rats increased their body weight significantly compared to the CHOW rats. In DIO rats, TRPC1 and TRPC6 were upregulated in the HIP, while they were downregulated in the FC. In the case of TRPM2 expression, instead was increased both in the HIP and in the FC. These could be related to the increase of proteins and nucleic acid oxidation. TRPV1 and TRPV2 gene expression showed no differences both in the FC and HIP. In general, qRT-PCR analyses were confirmed by Western blot analysis. Immunohistochemical procedures highlighted the expression of the channels in the cell body of neurons and axons, particularly for the TRPC1 and TRPC6. The alterations of TRP channel expression could be related to the activation of glial cells or the neurodegenerative process presented in the brain of the DIO rat highlighted with post synaptic protein (PSD 95) alterations. The availability of suitable animal models may be useful for studying possible pharmacological treatments to counter obesity-induced brain injury. The identified changes in DIO rats may represent the first insight to characterize the neuronal alterations occurring in obesity. Further investigations are necessary to characterize the role of TRP channels in the regulation of synaptic plasticity and obesity-related cognitive decline.
Collapse
Affiliation(s)
- Proshanta Roy
- School of Biosciences and Veterinary Medicine, University of Camerino.
| | | | | | - Federica Maggi
- Department of Molecular Medicine, La Sapienza University of Rome.
| | - Consuelo Amantini
- School of Biosciences and Veterinary Medicine, University of Camerino.
| | | | | | | | | | - Daniele Tomassoni
- School of Biosciences and Veterinary Medicine, University of Camerino.
| |
Collapse
|
20
|
Xie R, Wang Z, Liu T, Xiao R, Lv K, Wu C, Luo Y, Cai Y, Fan X. AAV Delivery of shRNA Against TRPC6 in Mouse Hippocampus Impairs Cognitive Function. Front Cell Dev Biol 2021; 9:688655. [PMID: 34327201 PMCID: PMC8313999 DOI: 10.3389/fcell.2021.688655] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 06/22/2021] [Indexed: 11/13/2022] Open
Abstract
Transient Receptor Potential Canonical 6 (TRPC6) has been suggested to be involved in synapse function and contribute to hippocampal-dependent cognitive processes. Gene silencing of TRPC6 was performed by injecting adeno-associated virus (AAV) expressing TRPC6-specific shRNA (shRNA-TRPC6) into the hippocampal dentate gyrus (DG). Spatial learning, working memory and social recognition memory were impaired in the shRNA-TRPC6 treated mice compared to control mice after 4 weeks. In addition, gene ontology (GO) analysis of RNA-sequencing revealed that viral intervention of TRPC6 expression in DG resulted in the enrichment of the process of synaptic transmission and cellular compartment of synaptic structure. KEGG analysis showed PI3K-Akt signaling pathway were significantly down-regulated. Furthermore, the shRNA-TRPC6 treatment reduced dendritic spines of DG granule neurons, in terms of spine loss, the thin and mushroom types predominated. Accompanying the spine loss, the levels of PSD95, pAkt and CREB in the hippocampus were decreased in the shRNA-TRPC6 treated animals. Taken together, our results suggest that knocking down TRPC6 in the DG have a disadvantageous effect on cognitive processes.
Collapse
Affiliation(s)
- Ruxin Xie
- Department of Military Cognitive Psychology, School of Psychology, Third Military Medical University (Army Medical University), Chongqing, China
| | - Zhongke Wang
- Department of Military Cognitive Psychology, School of Psychology, Third Military Medical University (Army Medical University), Chongqing, China
| | - Tianyao Liu
- Department of Military Cognitive Psychology, School of Psychology, Third Military Medical University (Army Medical University), Chongqing, China
| | - Rui Xiao
- Department of Military Cognitive Psychology, School of Psychology, Third Military Medical University (Army Medical University), Chongqing, China
| | - Keyi Lv
- Department of Military Cognitive Psychology, School of Psychology, Third Military Medical University (Army Medical University), Chongqing, China
| | - Chuan Wu
- Department of Military Cognitive Psychology, School of Psychology, Third Military Medical University (Army Medical University), Chongqing, China
| | - Yi Luo
- Department of Military Cognitive Psychology, School of Psychology, Third Military Medical University (Army Medical University), Chongqing, China
| | - Yun Cai
- Department of Military Cognitive Psychology, School of Psychology, Third Military Medical University (Army Medical University), Chongqing, China
| | - Xiaotang Fan
- Department of Military Cognitive Psychology, School of Psychology, Third Military Medical University (Army Medical University), Chongqing, China
| |
Collapse
|
21
|
Congenital hypothyroidism impairs spine growth of dentate granule cells by downregulation of CaMKIV. Cell Death Discov 2021; 7:143. [PMID: 34127648 PMCID: PMC8203692 DOI: 10.1038/s41420-021-00530-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 05/23/2021] [Indexed: 12/13/2022] Open
Abstract
Congenital hypothyroidism (CH), a common neonatal endocrine disorder, can result in cognitive deficits if delay in diagnose and treatment. Dentate gyrus (DG) is the severely affected subregion of the hippocampus by the CH, where the dentate granule cells (DGCs) reside in. However, how CH impairs the cognitive function via affecting DGCs and the underlying mechanisms are not fully elucidated. In the present study, the CH model of rat pups was successfully established, and the aberrant dendrite growth of the DGCs and the impaired cognitive behaviors were observed in the offspring. Transcriptome analysis of hippocampal tissues following rat CH successfully identified that calcium/calmodulin-dependent protein kinase IV (CaMKIV) was the prominent regulator involved in mediating deficient growth of DGC dendrites. CaMKIV was shown to be dynamically regulated in the DG subregion of the rats following drug-induced CH. Interference of CaMKIV expression in the primary DGCs significantly reduced the spine density of dendrites, while addition of T3 to the primary DGCs isolated from CH pups could facilitate the spine growth of dendrites. Insights into relevant mechanisms revealed that CH-mediated CaMKIV deficiency resulted in the significant decrease of phosphorylated CREB in DGCs, in association with the abnormality of dendrites. Our results have provided a distinct cell type in hippocampus that is affected by CH, which would be beneficial for the treatment of CH-induced cognitive deficiency.
Collapse
|
22
|
Meng C, Zeng W, Lv J, Wang Y, Gao M, Chang R, Li Q, Wang X. 1,8-cineole ameliorates ischaemic brain damage via TRPC6/CREB pathways in rats. J Pharm Pharmacol 2021; 73:979-985. [PMID: 33877307 DOI: 10.1093/jpp/rgab035] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Accepted: 04/12/2021] [Indexed: 11/12/2022]
Abstract
OBJECTIVES A previous in vitro study reported that the monoterpene oxide 1,8-cineole (cineole) attenuates neuronal caused by oxygen-glucose deprivation/reoxygenation in culture. However, to date, there is no in vivo evidence showing neuroprotective effects of cineole against stroke. This study aimed to investigate whether cineole attenuates cerebral ischaemic damage in rats. METHODS A rat model of middle cerebral artery occlusion (MCAO) followed by 24 h reperfusion was applied. Male rats were treated with oral cineole (100 mg/kg) for 7 consecutive days, then subjected to MCAO surgery. Infarct volume, neurologic deficits, apoptosis and expression levels of all-spectrin breakdown products of 145 kDa (SBDP145), transient receptor potential canonical (subtype) 6 (TRPC6) and phosphorylated CREB (p-CREB) were measured in ischaemic brain tissues. KEY FINDINGS Cineole treatment significantly reduced infarct volume, neurological dysfunction, neuronal apoptosis, SBDP145 formation and TRPC6 degradation and enhanced p-CREB expression in MCAO rats compared with vehicle treatment. These neuroprotective effects were markedly suppressed by pharmacological inhibition of MEK or CaMKIV signalling. CONCLUSIONS Our study provides in vivo evidence demonstrating that cineole pretreatment attenuates ischaemic stroke-induced brain damage, mainly through blocking calpain-induced TRPC6 degradation and activating CREB via MEK/CREB and CaMKIV/CREB signalling pathways.
Collapse
Affiliation(s)
- Chen Meng
- Department of Anesthesiology, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China
- Institute of Anesthesiology, Hubei University of Medicine, Shiyan, Hubei, China
| | - Wenjing Zeng
- Department of Anesthesiology, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China
- Institute of Anesthesiology, Hubei University of Medicine, Shiyan, Hubei, China
| | - Jing Lv
- Department of Anesthesiology, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China
- Institute of Anesthesiology, Hubei University of Medicine, Shiyan, Hubei, China
| | - Yu Wang
- Department of Anesthesiology, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China
| | - Meiling Gao
- Department of Anesthesiology, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China
- Institute of Anesthesiology, Hubei University of Medicine, Shiyan, Hubei, China
| | - Ruijie Chang
- Department of Anesthesiology, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China
| | - Qing Li
- Department of Anesthesiology, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China
- Institute of Anesthesiology, Hubei University of Medicine, Shiyan, Hubei, China
| | - Xianyu Wang
- Department of Anesthesiology, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China
- Institute of Anesthesiology, Hubei University of Medicine, Shiyan, Hubei, China
| |
Collapse
|
23
|
The Calcium/Calmodulin-Dependent Kinases II and IV as Therapeutic Targets in Neurodegenerative and Neuropsychiatric Disorders. Int J Mol Sci 2021; 22:ijms22094307. [PMID: 33919163 PMCID: PMC8122486 DOI: 10.3390/ijms22094307] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 04/13/2021] [Accepted: 04/17/2021] [Indexed: 12/14/2022] Open
Abstract
CaMKII and CaMKIV are calcium/calmodulin-dependent kinases playing a rudimentary role in many regulatory processes in the organism. These kinases attract increasing interest due to their involvement primarily in memory and plasticity and various cellular functions. Although CaMKII and CaMKIV are mostly recognized as the important cogs in a memory machine, little is known about their effect on mood and role in neuropsychiatric diseases etiology. Here, we aimed to review the structure and functions of CaMKII and CaMKIV, as well as how these kinases modulate the animals’ behavior to promote antidepressant-like, anxiolytic-like, and procognitive effects. The review will help in the understanding of the roles of the above kinases in the selected neurodegenerative and neuropsychiatric disorders, and this knowledge can be used in future drug design.
Collapse
|
24
|
Chinigò G, Castel H, Chever O, Gkika D. TRP Channels in Brain Tumors. Front Cell Dev Biol 2021; 9:617801. [PMID: 33928077 PMCID: PMC8076903 DOI: 10.3389/fcell.2021.617801] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Accepted: 03/17/2021] [Indexed: 12/21/2022] Open
Abstract
Malignant glioma including glioblastoma (GBM) is the most common group of primary brain tumors. Despite standard optimized treatment consisting of extensive resection followed by radiotherapy/concomitant and adjuvant therapy, GBM remains one of the most aggressive human cancers. GBM is a typical example of intra-heterogeneity modeled by different micro-environmental situations, one of the main causes of resistance to conventional treatments. The resistance to treatment is associated with angiogenesis, hypoxic and necrotic tumor areas while heterogeneity would accumulate during glioma cell invasion, supporting recurrence. These complex mechanisms require a focus on potential new molecular actors to consider new treatment options for gliomas. Among emerging and underexplored targets, transient receptor potential (TRP) channels belonging to a superfamily of non-selective cation channels which play critical roles in the responses to a number of external stimuli from the external environment were found to be related to cancer development, including glioma. Here, we discuss the potential as biological markers of diagnosis and prognosis of TRPC6, TRPM8, TRPV4, or TRPV1/V2 being associated with glioma patient overall survival. TRPs-inducing common or distinct mechanisms associated with their Ca2+-channel permeability and/or kinase function were detailed as involving miRNA or secondary effector signaling cascades in turn controlling proliferation, cell cycle, apoptotic pathways, DNA repair, resistance to treatment as well as migration/invasion. These recent observations of the key role played by TRPs such as TRPC6 in GBM growth and invasiveness, TRPV2 in proliferation and glioma-stem cell differentiation and TRPM2 as channel carriers of cytotoxic chemotherapy within glioma cells, should offer new directions for innovation in treatment strategies of high-grade glioma as GBM to overcome high resistance and recurrence.
Collapse
Affiliation(s)
- Giorgia Chinigò
- Laboratory of Cell Physiology, Department of Life Sciences, Univ. Lille, Inserm, U1003 - PHYCEL, University of Lille, Lille, France.,Laboratory of Cellular and Molecular Angiogenesis, Department of Life Sciences and Systems Biology, University of Torino, Turin, Italy
| | - Hélène Castel
- UNIROUEN, Inserm U1239, DC2N, Normandie Université, Rouen, France.,Institute for Research and Innovation in Biomedicine, Rouen, France
| | - Oana Chever
- UNIROUEN, Inserm U1239, DC2N, Normandie Université, Rouen, France.,Institute for Research and Innovation in Biomedicine, Rouen, France
| | - Dimitra Gkika
- CNRS, Inserm, CHU Lille, Centre Oscar Lambret, UMR 9020-UMR 1277-Canther-Cancer Heterogeneity, Plasticity and Resistance to Therapies, University of Lille, Lille, France.,Institut Universitaire de France, Paris, France
| |
Collapse
|
25
|
Kinoshita PF, Orellana AMM, Nakao VW, de Souza Port's NM, Quintas LEM, Kawamoto EM, Scavone C. The Janus face of ouabain in Na + /K + -ATPase and calcium signalling in neurons. Br J Pharmacol 2021; 179:1512-1524. [PMID: 33644859 DOI: 10.1111/bph.15419] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 02/03/2021] [Accepted: 02/14/2021] [Indexed: 12/20/2022] Open
Abstract
Na+ /K+ -ATPase, a transmembrane protein essential for maintaining the electrochemical gradient across the plasma membrane, acts as a receptor for cardiotonic steroids such as ouabain. Cardiotonic steroids binding to Na+ /K+ -ATPase triggers signalling pathways or inhibits Na+ /K+ -ATPas activity in a concentration-dependent manner, resulting in a modulation of Ca2+ levels, which are essential for homeostasis in neurons. However, most of the pharmacological strategies for avoiding neuronal death do not target Na+ /K+ -ATPase activity due to its complexity and the poor understanding of the mechanisms involved in Na+ /K+ -ATPase modulation. The present review aims to discuss two points regarding the interplay between Na+ /K+ -ATPase and Ca2+ signalling in the brain. One, Na+ /K+ -ATPase impairment causing illness and neuronal death due to Ca2+ signalling and two, benefits to the brain by modulating Na+ /K+ -ATPase activity. These interactions play an essential role in neuronal cell fate determination and are relevant to find new targets for the treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Paula Fernanda Kinoshita
- Laboratory of Molecular Neuropharmacology, Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Ana Maria Marques Orellana
- Laboratory of Molecular Neuropharmacology, Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil.,Laboratory of Molecular and Functional Neurobiology, Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Vinicius Watanabe Nakao
- Laboratory of Molecular Neuropharmacology, Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Natacha Medeiros de Souza Port's
- Laboratory of Molecular Neuropharmacology, Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Luis Eduardo Menezes Quintas
- Laboratory of Biochemical and Molecular Pharmacology, Institute of Biomedical Sciences, Health Sciences Centre Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
| | - Elisa Mitiko Kawamoto
- Laboratory of Molecular and Functional Neurobiology, Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Cristoforo Scavone
- Laboratory of Molecular Neuropharmacology, Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
26
|
Abstract
Already for centuries, humankind is driven to understand the physiological and pathological mechanisms that occur in our brains. Today, we know that ion channels play an essential role in the regulation of neural processes and control many functions of the central nervous system. Ion channels present a diverse group of membrane-spanning proteins that allow ions to penetrate the insulating cell membrane upon opening of their channel pores. This regulated ion permeation results in different electrical and chemical signals that are necessary to maintain physiological excitatory and inhibitory processes in the brain. Therefore, it is no surprise that disturbances in the functions of cerebral ion channels can result in a plethora of neurological disorders, which present a tremendous health care burden for our current society. The identification of ion channel-related brain disorders also fuel the research into the roles of ion channel proteins in various brain states. In the last decade, mounting evidence has been collected that indicates a pivotal role for transient receptor potential (TRP) ion channels in the development and various physiological functions of the central nervous system. For instance, TRP channels modulate neurite growth, synaptic plasticity and integration, and are required for neuronal survival. Moreover, TRP channels are involved in numerous neurological disorders. TRPM3 belongs to the melastatin subfamily of TRP channels and represents a non-selective cation channel that can be activated by several different stimuli, including the neurosteroid pregnenolone sulfate, osmotic pressures and heat. The channel is best known as a peripheral nociceptive ion channel that participates in heat sensation. However, recent research identifies TRPM3 as an emerging new player in the brain. In this review, we summarize the available data regarding the roles of TRPM3 in the brain, and correlate these data with the neuropathological processes in which this ion channel may be involved.
Collapse
Affiliation(s)
- Katharina Held
- Laboratory of Endometrium, Endometriosis and Reproductive Medicine, Department of Development and Regeneration, KU Leuven, Leuven, Belgium
- Laboratory of Ion Channel Research, Department of Cellular and Molecular Medicine and VIB-KU Leuven Center for Brain and Disease Research, KU Leuven, Leuven, Belgium
| | - Balázs István Tóth
- Laboratory of Cellular and Molecular Physiology, Department of Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| |
Collapse
|
27
|
Promotion of Dendritic Differentiation of Cerebellar Purkinje Cells by Ca 2+/calmodulin-dependent Protein Kinase IIα, IIβ and IV and Possible Involvement of CREB Phosphorylation. Neuroscience 2021; 458:87-98. [PMID: 33493619 DOI: 10.1016/j.neuroscience.2021.01.024] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 01/12/2021] [Accepted: 01/13/2021] [Indexed: 11/20/2022]
Abstract
Cerebellar Purkinje cells develop the most elaborate dendritic trees among neurons in the brain. To examine the role of Ca2+/calmodulin-dependent protein kinase (CaMK) IIα, IIβ and IV in the dendritic differentiation of Purkinje cells, we introduced siRNA against these CaMKs into Purkinje cells in cerebellar cell cultures using a single-cell electroporation technique. Single-cell electroporation enables us to transfer siRNA into specific cells within a heterogeneous cell population. In addition, we can easily and reliably transfer multiple types of siRNA into a cell simply by loading them together in one micropipette. Any one of the siRNA against CaMKIIα, IIβ and IV (single knockdown) or any combinations of two of the siRNA against these CaMKs (double knockdown) had no significant effects on the dendritic differentiation of Purkinje cells. However, the combination of all three siRNA against these CaMKs (triple knockdown) inhibited the branching of Purkinje cell dendrites. Furthermore, the triple knockdown reduced the phosphorylation of CREB in Purkinje cells. These findings suggest the promotion of dendritic differentiation of Purkinje cells by CaMKIIα, IIβ and IV and the possible involvement of phosphorylation of CREB as a common substrate of these CaMKs.
Collapse
|
28
|
Zeitler S, Schumacher F, Monti J, Anni D, Guhathakurta D, Kleuser B, Friedland K, Fejtová A, Kornhuber J, Rhein C. Acid Sphingomyelinase Impacts Canonical Transient Receptor Potential Channels 6 (TRPC6) Activity in Primary Neuronal Systems. Cells 2020; 9:E2502. [PMID: 33218173 PMCID: PMC7698877 DOI: 10.3390/cells9112502] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Revised: 10/16/2020] [Accepted: 11/13/2020] [Indexed: 12/30/2022] Open
Abstract
: The acid sphingomyelinase (ASM)/ceramide system exhibits a crucial role in the pathology of major depressive disorder (MDD). ASM hydrolyzes the abundant membrane lipid sphingomyelin to ceramide that regulates the clustering of membrane proteins via microdomain and lipid raft organization. Several commonly used antidepressants, such as fluoxetine, rely on the functional inhibition of ASM in terms of their antidepressive pharmacological effects. Transient receptor potential canonical 6 (TRPC6) ion channels are located in the plasma membrane of neurons and serve as receptors for hyperforin, a phytochemical constituent of the antidepressive herbal remedy St. John's wort. TRPC6 channels are involved in the regulation of neuronal plasticity, which likely contributes to their antidepressant effect. In this work, we investigated the impact of reduced ASM activity on the TRPC6 function in neurons. A lipidomic analysis of cortical brain tissue of ASM deficient mice revealed a decrease in ceramide/sphingomyelin molar ratio and an increase in sphingosine. In neurons with ASM deletion, hyperforin-mediated Ca2+-influx via TRPC6 was decreased. Consequently, downstream activation of nuclear phospho-cAMP response element-binding protein (pCREB) was changed, a transcriptional factor involved in neuronal plasticity. Our study underlines the importance of balanced ASM activity, as well as sphingolipidome composition for optimal TRPC6 function. A better understanding of the interaction of the ASM/ceramide and TRPC6 systems could help to draw conclusions about the pathology of MDD.
Collapse
Affiliation(s)
- Stefanie Zeitler
- Department of Psychiatry and Psychotherapy, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany; (S.Z.); (J.M.); (D.A.); (D.G.); (A.F.); (J.K.)
| | - Fabian Schumacher
- Department of Toxicology, University of Potsdam, 14558 Nuthetal, Germany;
- Department of Pharmacology & Toxicology, Institute of Pharmacy, Freie Universität Berlin, 14195 Berlin, Germany;
- Institute of Molecular Biology, University of Duisburg-Essen, 45147 Essen, Germany
| | - Juliana Monti
- Department of Psychiatry and Psychotherapy, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany; (S.Z.); (J.M.); (D.A.); (D.G.); (A.F.); (J.K.)
| | - Daniela Anni
- Department of Psychiatry and Psychotherapy, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany; (S.Z.); (J.M.); (D.A.); (D.G.); (A.F.); (J.K.)
| | - Debarpan Guhathakurta
- Department of Psychiatry and Psychotherapy, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany; (S.Z.); (J.M.); (D.A.); (D.G.); (A.F.); (J.K.)
| | - Burkhard Kleuser
- Department of Pharmacology & Toxicology, Institute of Pharmacy, Freie Universität Berlin, 14195 Berlin, Germany;
| | - Kristina Friedland
- Institute for Pharmacy and Biochemistry, Johannes-Gutenberg Universität Mainz, 55128 Mainz, Germany;
| | - Anna Fejtová
- Department of Psychiatry and Psychotherapy, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany; (S.Z.); (J.M.); (D.A.); (D.G.); (A.F.); (J.K.)
| | - Johannes Kornhuber
- Department of Psychiatry and Psychotherapy, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany; (S.Z.); (J.M.); (D.A.); (D.G.); (A.F.); (J.K.)
| | - Cosima Rhein
- Department of Psychiatry and Psychotherapy, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany; (S.Z.); (J.M.); (D.A.); (D.G.); (A.F.); (J.K.)
- Department of Psychosomatic Medicine and Psychotherapy, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany
| |
Collapse
|
29
|
Lavanderos B, Silva I, Cruz P, Orellana-Serradell O, Saldías MP, Cerda O. TRP Channels Regulation of Rho GTPases in Brain Context and Diseases. Front Cell Dev Biol 2020; 8:582975. [PMID: 33240883 PMCID: PMC7683514 DOI: 10.3389/fcell.2020.582975] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Accepted: 10/05/2020] [Indexed: 12/13/2022] Open
Abstract
Neurological and neuropsychiatric disorders are mediated by several pathophysiological mechanisms, including developmental and degenerative abnormalities caused primarily by disturbances in cell migration, structural plasticity of the synapse, and blood-vessel barrier function. In this context, critical pathways involved in the pathogenesis of these diseases are related to structural, scaffolding, and enzymatic activity-bearing proteins, which participate in Ca2+- and Ras Homologs (Rho) GTPases-mediated signaling. Rho GTPases are GDP/GTP binding proteins that regulate the cytoskeletal structure, cellular protrusion, and migration. These proteins cycle between GTP-bound (active) and GDP-bound (inactive) states due to their intrinsic GTPase activity and their dynamic regulation by GEFs, GAPs, and GDIs. One of the most important upstream inputs that modulate Rho GTPases activity is Ca2+ signaling, positioning ion channels as pivotal molecular entities for Rho GTPases regulation. Multiple non-selective cationic channels belonging to the Transient Receptor Potential (TRP) family participate in cytoskeletal-dependent processes through Ca2+-mediated modulation of Rho GTPases. Moreover, these ion channels have a role in several neuropathological events such as neuronal cell death, brain tumor progression and strokes. Although Rho GTPases-dependent pathways have been extensively studied, how they converge with TRP channels in the development or progression of neuropathologies is poorly understood. Herein, we review recent evidence and insights that link TRP channels activity to downstream Rho GTPase signaling or modulation. Moreover, using the TRIP database, we establish associations between possible mediators of Rho GTPase signaling with TRP ion channels. As such, we propose mechanisms that might explain the TRP-dependent modulation of Rho GTPases as possible pathways participating in the emergence or maintenance of neuropathological conditions.
Collapse
Affiliation(s)
- Boris Lavanderos
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile.,Millennium Nucleus of Ion Channel-Associated Diseases (MiNICAD), Santiago, Chile
| | - Ian Silva
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile.,Millennium Nucleus of Ion Channel-Associated Diseases (MiNICAD), Santiago, Chile
| | - Pablo Cruz
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile.,Millennium Nucleus of Ion Channel-Associated Diseases (MiNICAD), Santiago, Chile
| | - Octavio Orellana-Serradell
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile.,Millennium Nucleus of Ion Channel-Associated Diseases (MiNICAD), Santiago, Chile
| | - María Paz Saldías
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile.,Millennium Nucleus of Ion Channel-Associated Diseases (MiNICAD), Santiago, Chile
| | - Oscar Cerda
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile.,Millennium Nucleus of Ion Channel-Associated Diseases (MiNICAD), Santiago, Chile.,The Wound Repair, Treatment and Health (WoRTH) Initiative, Santiago, Chile
| |
Collapse
|
30
|
Prikhodko V, Chernyuk D, Sysoev Y, Zernov N, Okovityi S, Popugaeva E. Potential Drug Candidates to Treat TRPC6 Channel Deficiencies in the Pathophysiology of Alzheimer's Disease and Brain Ischemia. Cells 2020; 9:cells9112351. [PMID: 33114455 PMCID: PMC7692306 DOI: 10.3390/cells9112351] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 09/28/2020] [Accepted: 10/20/2020] [Indexed: 12/16/2022] Open
Abstract
Alzheimer’s disease and cerebral ischemia are among the many causative neurodegenerative diseases that lead to disabilities in the middle-aged and elderly population. There are no effective disease-preventing therapies for these pathologies. Recent in vitro and in vivo studies have revealed the TRPC6 channel to be a promising molecular target for the development of neuroprotective agents. TRPC6 channel is a non-selective cation plasma membrane channel that is permeable to Ca2+. Its Ca2+-dependent pharmacological effect is associated with the stabilization and protection of excitatory synapses. Downregulation as well as upregulation of TRPC6 channel functions have been observed in Alzheimer’s disease and brain ischemia models. Thus, in order to protect neurons from Alzheimer’s disease and cerebral ischemia, proper TRPC6 channels modulators have to be used. TRPC6 channels modulators are an emerging research field. New chemical structures modulating the activity of TRPC6 channels are being currently discovered. The recent publication of the cryo-EM structure of TRPC6 channels should speed up the discovery process even more. This review summarizes the currently available information about potential drug candidates that may be used as basic structures to develop selective, highly potent TRPC6 channel modulators to treat neurodegenerative disorders, such as Alzheimer’s disease and cerebral ischemia.
Collapse
Affiliation(s)
- Veronika Prikhodko
- Laboratory of Molecular Neurodegeneration, Peter the Great St. Petersburg Polytechnic University, 195251 St. Petersburg, Russia; (V.P.); (D.C.); (Y.S.); (N.Z.)
- Department of Pharmacology and Clinical Pharmacology, Saint Petersburg State Chemical Pharmaceutical University, 197022 St. Petersburg, Russia;
- N.P. Bechtereva Institute of the Human Brain of the Russian Academy of Sciences, 197376 St. Petersburg, Russia
| | - Daria Chernyuk
- Laboratory of Molecular Neurodegeneration, Peter the Great St. Petersburg Polytechnic University, 195251 St. Petersburg, Russia; (V.P.); (D.C.); (Y.S.); (N.Z.)
| | - Yurii Sysoev
- Laboratory of Molecular Neurodegeneration, Peter the Great St. Petersburg Polytechnic University, 195251 St. Petersburg, Russia; (V.P.); (D.C.); (Y.S.); (N.Z.)
- Department of Pharmacology and Clinical Pharmacology, Saint Petersburg State Chemical Pharmaceutical University, 197022 St. Petersburg, Russia;
- N.P. Bechtereva Institute of the Human Brain of the Russian Academy of Sciences, 197376 St. Petersburg, Russia
- Institute of Translational Biomedicine, Saint Petersburg State University, 199034 St. Petersburg, Russia
| | - Nikita Zernov
- Laboratory of Molecular Neurodegeneration, Peter the Great St. Petersburg Polytechnic University, 195251 St. Petersburg, Russia; (V.P.); (D.C.); (Y.S.); (N.Z.)
| | - Sergey Okovityi
- Department of Pharmacology and Clinical Pharmacology, Saint Petersburg State Chemical Pharmaceutical University, 197022 St. Petersburg, Russia;
- N.P. Bechtereva Institute of the Human Brain of the Russian Academy of Sciences, 197376 St. Petersburg, Russia
| | - Elena Popugaeva
- Laboratory of Molecular Neurodegeneration, Peter the Great St. Petersburg Polytechnic University, 195251 St. Petersburg, Russia; (V.P.); (D.C.); (Y.S.); (N.Z.)
- Correspondence:
| |
Collapse
|
31
|
He C, Gao P, Cui Y, Li Q, Li Y, Lu Z, Ma H, Zhao Y, Li L, Sun F, Chen X, Jia H, Liu D, Yang G, Zheng H, Zhu Z. Low-glucose-sensitive TRPC6 dysfunction drives hypoglycemia-induced cognitive impairment in diabetes. Clin Transl Med 2020; 10:e205. [PMID: 33135341 PMCID: PMC7568851 DOI: 10.1002/ctm2.205] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 09/28/2020] [Accepted: 09/28/2020] [Indexed: 12/15/2022] Open
Abstract
Background Recurrent moderate hypoglycemia (RH), a major adverse effect of hypoglycemic therapy in diabetic patients, is one of the main risk factors for cognitive impairment and dementia. Transient receptor potential canonical channel 6 (TRPC6) is a potential therapeutic target for Alzheimer's disease (AD) and its expression is highly regulated by glucose concentration. Objective To investigate whether RH regulates the expression of TRPC6 in brain and whether TRPC6 dysfunction can drive hypoglycemia‐associated cognitive impairment in diabetes, and reveal the underlying mechanism. Methods Histological staining, in vivo two‐photon Ca2+ imaging, and behavioral tests were used to measure neuronal death, brain network activity, and cognitive function in mice, respectively. High‐resolution respirometry and transmission electron microscope were used to assess mitochondrial structure and function. Intracellular calcium measurement and molecular biology techniques were conducted to uncover the underlying mechanism. Results Here, we report that the expression of TRPC6 in hippocampus was specifically repressed by RH in streptozocin‐induced type 1 diabetic mice, but not in nondiabetic mice. TRPC6 knockout directly leads to neuron loss, neuronal activity, and cognitive function impairment under diabetic condition, the degree of which is similar to that of RH. Activation of TRPC6 with hyperforin substantially improved RH‐induced cognitive impairment. Mechanistically, TRPC6 inhibited mitochondrial fission in the hippocampus of diabetic mice undergoing RH episodes by activating adenosine 5‘‐monophosphate‐activated protein kinase, and TRPC6‐mediated cytosolic calcium influx was required for this process. Clinically, dysfunction of TRPC6 was closely associated with cognitive impairment in type 2 diabetic patients with RH. Conclusions Our results indicate that TRPC6 is a critical sensitive cation channel to hypoglycemia and is a promising target to prevent RH‐induced cognitive impairment by properly orchestrating the mitochondrial dynamics in diabetic patients.
Collapse
Affiliation(s)
- Chengkang He
- Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital, Chongqing Institute of Hypertension, Army Medical University, Chongqing, China
| | - Peng Gao
- Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital, Chongqing Institute of Hypertension, Army Medical University, Chongqing, China
| | - Yuanting Cui
- Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital, Chongqing Institute of Hypertension, Army Medical University, Chongqing, China
| | - Qiang Li
- Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital, Chongqing Institute of Hypertension, Army Medical University, Chongqing, China
| | - Yingsha Li
- Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital, Chongqing Institute of Hypertension, Army Medical University, Chongqing, China
| | - Zongshi Lu
- Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital, Chongqing Institute of Hypertension, Army Medical University, Chongqing, China
| | - Huan Ma
- Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital, Chongqing Institute of Hypertension, Army Medical University, Chongqing, China
| | - Yu Zhao
- Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital, Chongqing Institute of Hypertension, Army Medical University, Chongqing, China
| | - Li Li
- Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital, Chongqing Institute of Hypertension, Army Medical University, Chongqing, China
| | - Fang Sun
- Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital, Chongqing Institute of Hypertension, Army Medical University, Chongqing, China
| | - Xiaowei Chen
- Brain Research Center, Army Medical University, Chongqing, China
| | - Hongbo Jia
- Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou, China
| | - Daoyan Liu
- Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital, Chongqing Institute of Hypertension, Army Medical University, Chongqing, China
| | - Gangyi Yang
- Endocrine Department, Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Hongting Zheng
- Department of Endocrinology, Translational Research Key Laboratory for Diabetes, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Zhiming Zhu
- Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital, Chongqing Institute of Hypertension, Army Medical University, Chongqing, China
| |
Collapse
|
32
|
Maria-Ferreira D, de Oliveira NMT, da Silva LCM, Fernandes ES. Evidence of a Role for the TRPC Subfamily in Mediating Oxidative Stress in Parkinson's Disease. Front Physiol 2020; 11:332. [PMID: 32457638 PMCID: PMC7225354 DOI: 10.3389/fphys.2020.00332] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Accepted: 03/23/2020] [Indexed: 12/20/2022] Open
Abstract
Parkinson's disease (PD) represents one of the most common multifactorial neurodegenerative disorders affecting the elderly population. It is associated with the aggregation of α-synuclein protein and the loss of dopaminergic neurons in the substantia nigra pars compacta of the brain. The disease is mainly represented by motor symptoms, such as resting tremors, postural instability, rigidity, and bradykinesia, that develop slowly over time. Parkinson's disease can also manifest as disturbances in non-motor functions. Although the pathology of PD has not yet been fully understood, it has been suggested that the disruption of the cellular redox status may contribute to cellular oxidative stress and, thus, to cell death. The generation of reactive oxygen species and reactive nitrogen intermediates, as well as the dysfunction of dopamine metabolism, play important roles in the degeneration of dopaminergic neurons. In this context, the transient receptor potential channel canonical (TRPC) sub-family plays an important role in neuronal degeneration. Additionally, PD gene products, including DJ-1, SNCA, UCH-L1, PINK-1, and Parkin, also interfere with mitochondrial function leading to reactive oxygen species production and dopaminergic neuronal vulnerability to oxidative stress. Herein, we discuss the interplay between these various biochemical and molecular events that ultimately lead to dopaminergic signaling disruption, highlighting the recently identified roles of TRPC in PD.
Collapse
Affiliation(s)
- Daniele Maria-Ferreira
- Faculdades Pequeno Príncipe, Programa de Pós-graduação em Biotecnologia Aplicada à Saúde da Criança e do Adolescente, Curitiba, Brazil
- Instituto de Pesquisa Pelé Pequeno Príncipe, Curitiba, Brazil
| | - Natalia Mulinari Turin de Oliveira
- Faculdades Pequeno Príncipe, Programa de Pós-graduação em Biotecnologia Aplicada à Saúde da Criança e do Adolescente, Curitiba, Brazil
- Instituto de Pesquisa Pelé Pequeno Príncipe, Curitiba, Brazil
| | - Liziane Cristine Malaquias da Silva
- Faculdades Pequeno Príncipe, Programa de Pós-graduação em Biotecnologia Aplicada à Saúde da Criança e do Adolescente, Curitiba, Brazil
- Instituto de Pesquisa Pelé Pequeno Príncipe, Curitiba, Brazil
| | - Elizabeth Soares Fernandes
- Faculdades Pequeno Príncipe, Programa de Pós-graduação em Biotecnologia Aplicada à Saúde da Criança e do Adolescente, Curitiba, Brazil
- Instituto de Pesquisa Pelé Pequeno Príncipe, Curitiba, Brazil
| |
Collapse
|
33
|
Liu Z, Du Z, Li K, Han Y, Ren G, Yang Z. TRPC6-Mediated Ca 2+ Entry Essential for the Regulation of Nano-ZnO Induced Autophagy in SH-SY5Y Cells. Neurochem Res 2020; 45:1602-1613. [PMID: 32274628 DOI: 10.1007/s11064-020-03025-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Revised: 04/02/2020] [Accepted: 04/04/2020] [Indexed: 02/06/2023]
Abstract
Recently, possible applications of zinc oxide nanoparticles (nano-ZnO) have been extensively studied owing to their ease of synthesis. However, the effect of nano-ZnO on the nervous system remains unclear. This study investigates the action of nano-ZnO on SH-SY5Y neuroblastoma cells. We found that nano-ZnO (0-50 µg/mL) induced a significant decrease in cell survival rate in a dose-dependent manner, and increased LC3 puncta formation. However, the apoptosis was not affected by nano-ZnO, because the protein levels of cytochrome c, caspase-3, Bcl-xL, and BAX were not varied by the nano-ZnO treatment. Nano-ZnO increased Ca2+ entry and the expression of TRPC6.The results suggested that nano-ZnO increased [Ca2+] through the TRPC-dependent Ca2+ influx, since Ca2+ influx can be prevented by the TRPC inhibitor. Furthermore, cells on nano-ZnO-treatment groups displayed loss of F-actin in a dose dependent manner, which also could be prevented by TRPC inhibitor. Herein, we demonstrated that the nano-ZnO activated TRPC6 channel, thereby increasing the Ca2+ flux and resulting in increased autophagy. Nano-ZnO could have possible anticancer effects in neuroblastoma by inhibiting the proliferation of tumor cells. However, we should also pay attention toward the biosecurity of nano materials.
Collapse
Affiliation(s)
- Zhaowei Liu
- College of Medicine, State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials Ministry of Education, Nankai University, No.94, Weijin Road, Nankai District, Tianjin, 300071, China.
| | - Zhanqiang Du
- College of Medicine, State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials Ministry of Education, Nankai University, No.94, Weijin Road, Nankai District, Tianjin, 300071, China
| | - Kai Li
- College of Medicine, State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials Ministry of Education, Nankai University, No.94, Weijin Road, Nankai District, Tianjin, 300071, China
| | - Yangguang Han
- School of precision instrument and optoelectronic engineering, Tianjin University, Tianjin, 300072, China
| | - Guogang Ren
- Science and Technology Research Institute, University of Hertfordshire, Hatfield, Herts, AL10 9AB, UK
| | - Zhuo Yang
- College of Medicine, State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials Ministry of Education, Nankai University, No.94, Weijin Road, Nankai District, Tianjin, 300071, China.
| |
Collapse
|
34
|
Xiao R, Zhong H, Li X, Ma Y, Zhang R, Wang L, Zang Z, Fan X. Abnormal Cerebellar Development Is Involved in Dystonia-Like Behaviors and Motor Dysfunction of Autistic BTBR Mice. Front Cell Dev Biol 2020; 8:231. [PMID: 32318573 PMCID: PMC7154340 DOI: 10.3389/fcell.2020.00231] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Accepted: 03/18/2020] [Indexed: 12/12/2022] Open
Abstract
Motor control and learning impairments are common complications in individuals with autism spectrum disorder (ASD). Abnormal cerebellar development during critical phases may disrupt these motor functions and lead to autistic motor dysfunction. However, the underlying mechanisms behind these impairments are not clear. Here, we utilized BTBR T+ Itprtf/J (BTBR) mice, an animal model of autism, to investigate the involvement of abnormal cerebellar development in motor performance. We found BTBR mice exhibited severe dystonia-like behavior and motor coordination or motor learning impairments. The onset of these abnormal movements coincided with the increased proliferation of granule neurons and enhanced foliation, and Purkinje cells displayed morphological hypotrophy with increased dendritic spine formation but suppressed maturation. The migration of granule neurons seemed unaffected. Transcriptional analyses confirmed the differential expression of genes involved in abnormal neurogenesis and revealed TRPC as a critical regulator in proliferation and synaptic formation. Taken together, these findings indicate that abnormal cerebellar development is closely related to dystonia-like behavior and motor dysfunction of BTBR mice and that TRPC may be a novel risk gene for ASD that may participate in the pathological process of autistic movement disorders.
Collapse
Affiliation(s)
- Rui Xiao
- Department of Military Cognitive Psychology, School of Psychology, Army Medical University, Chongqing, China
| | - Hongyu Zhong
- Department of Military Cognitive Psychology, School of Psychology, Army Medical University, Chongqing, China
| | - Xin Li
- Department of Military Cognitive Psychology, School of Psychology, Army Medical University, Chongqing, China
| | - Yuanyuan Ma
- Department of Military Cognitive Psychology, School of Psychology, Army Medical University, Chongqing, China.,Department of Basic Nursing, School of Nursing, Army Medical University, Chongqing, China
| | - Ruiyu Zhang
- Department of Military Cognitive Psychology, School of Psychology, Army Medical University, Chongqing, China
| | - Lian Wang
- Department of Military Cognitive Psychology, School of Psychology, Army Medical University, Chongqing, China
| | - Zhenle Zang
- Department of Military Cognitive Psychology, School of Psychology, Army Medical University, Chongqing, China
| | - Xiaotang Fan
- Department of Military Cognitive Psychology, School of Psychology, Army Medical University, Chongqing, China
| |
Collapse
|
35
|
Liu L, Gu L, Chen M, Zheng Y, Xiong X, Zhu S. Novel Targets for Stroke Therapy: Special Focus on TRPC Channels and TRPC6. Front Aging Neurosci 2020; 12:70. [PMID: 32256338 PMCID: PMC7093711 DOI: 10.3389/fnagi.2020.00070] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Accepted: 02/26/2020] [Indexed: 12/14/2022] Open
Abstract
Stroke remains a leading cause of death, disability, and medical care burden worldwide. However, transformation from laboratory findings toward effective pharmacological interventions for clinical stroke has been unsatisfactory. Novel evidence has been gained on the underlying mechanisms and therapeutic potential related to the transient receptor potential (TRP) channels in several disorders. The TRP superfamily consists of a diverse group of Ca2+ permeable non-selective cation channels. In particular, the members of TRP subfamilies, TRP canonical (TRPC) channels and TRPC6, have been found in different cell types in the whole body and have high levels of expression in the central nervous system (CNS). Notably, the TRPCs and TRPC6 channel have been implicated in neurite outgrowth and neuronal survival during normal development and in a range of CNS pathological conditions. Recent studies have shown that suppression of TRPC6 channel degradation prevents ischemic neuronal cell death in experimental stroke. Accumulating evidence supports the important functions of TRPC6 in brain ischemia. We have highlighted some crucial advancement that points toward an important involvement of TRPCs and TRPC6 in ischemic stroke. This review will make an overview of the TRP and TRPC channels due to their roles as targets for clinical trials and CNS disorders. Besides, the primary goal is to discuss and update the critical role of TRPC6 channels in stroke and provide a promising target for stroke prevention and therapy.
Collapse
Affiliation(s)
- Lu Liu
- Department of Anesthesiology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Lijuan Gu
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, China
| | - Manli Chen
- Department of Anesthesiology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Yueying Zheng
- Department of Anesthesiology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Xiaoxing Xiong
- Department of Anesthesiology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China.,Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, China
| | - Shengmei Zhu
- Department of Anesthesiology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
36
|
Ma M, Zhao S, Zhang J, Sun T, Fan Y, Zheng Z. High Glucose-Induced TRPC6 Channel Activation Decreases Glutamate Uptake in Rat Retinal Müller Cells. Front Pharmacol 2020; 10:1668. [PMID: 32116675 PMCID: PMC7033573 DOI: 10.3389/fphar.2019.01668] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Accepted: 12/20/2019] [Indexed: 12/15/2022] Open
Abstract
High glucose (HG) increases the production of reactive oxygen species (ROS), leading to decreased glutamate uptake in Müller cells. The transient receptor potential cation channel 6 (TRPC6) channel, an oxidative stress-sensitive Ca2+-permeable cationic channel, is readily detected in Müller cells and highly expressed under HG conditions. Yet, the effect of high glucose-induced TRPC6 channel activation in Müller cells is poorly understood. We hypothesized that TRPC6 channel activation mediates high glucose-induced decreases in Müller cell glutamate uptake. We found RNA interference (RNAi) of the TRPC6 channel abolished HG-induced decreases in glutamate uptake and cell death. HG also decreased the expression of the glutamate-aspartate transporter (GLAST), which is the most important transporter involved in glutamate uptake. The mRNA level of ciliary neurotrophic factor (CNTF) in rMC-1 cells and the release of CNTF in the culture media was decreased, but the mRNA levels of IL-6 and vascular endothelial growth factor (VEGF) were increased under HG conditions. After RNAi silencing in rMC-1 cells, the mRNA levels of CNTF increased, but IL-6 and VEGF levels decreased. Furthermore, TRPC6 knockdown (KD) decreased expression of glial fibrillary acidic protein (GFAP) and increased expression of Kir4.1, pointing to inhibition of HG-induced gliosis in rMC-1 cells. ROS and intracellular Ca2+ levels decreased after TRPC6 knockdown. Exposure to Hyp9 (10 μM), a highly selective TRPC6 channel agonist, can aggravate HG-induced pathological changes. Collectively, our results suggest TRPC6 channel activation is involved in HG-induced decreases in glutamate uptake in rMC-1 cells. These findings provide novel insights into the role of TRPC6 in HG-induced retinal neurovasculopathy and suggest TRPC6 is a promising target for drug development for diabetic retinopathy (DR).
Collapse
Affiliation(s)
- Mingming Ma
- Department of Ophthalmology, Shanghai General Hospital, Shanghai, China.,National Clinical Research Center for Eye Diseases, Shanghai, China.,Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai Jiao Tong University, Shanghai, China.,Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai, China.,Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai, China
| | - Shuzhi Zhao
- Department of Ophthalmology, Shanghai General Hospital, Shanghai, China.,National Clinical Research Center for Eye Diseases, Shanghai, China.,Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai Jiao Tong University, Shanghai, China.,Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai, China.,Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai, China
| | - Jian Zhang
- Department of Ophthalmology, Shanghai General Hospital, Shanghai, China.,National Clinical Research Center for Eye Diseases, Shanghai, China.,Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai Jiao Tong University, Shanghai, China.,Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai, China.,Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai, China
| | - Tao Sun
- Department of Ophthalmology, Shanghai General Hospital, Shanghai, China.,National Clinical Research Center for Eye Diseases, Shanghai, China.,Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai Jiao Tong University, Shanghai, China.,Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai, China.,Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai, China
| | - Ying Fan
- Department of Ophthalmology, Shanghai General Hospital, Shanghai, China.,National Clinical Research Center for Eye Diseases, Shanghai, China.,Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai Jiao Tong University, Shanghai, China.,Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai, China.,Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai, China
| | - Zhi Zheng
- Department of Ophthalmology, Shanghai General Hospital, Shanghai, China.,National Clinical Research Center for Eye Diseases, Shanghai, China.,Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai Jiao Tong University, Shanghai, China.,Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai, China.,Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai, China
| |
Collapse
|
37
|
Jurek B, Meyer M. Anxiolytic and Anxiogenic? How the Transcription Factor MEF2 Might Explain the Manifold Behavioral Effects of Oxytocin. Front Endocrinol (Lausanne) 2020; 11:186. [PMID: 32322239 PMCID: PMC7156634 DOI: 10.3389/fendo.2020.00186] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Accepted: 03/17/2020] [Indexed: 01/01/2023] Open
Abstract
The neuromodulator oxytocin, since its first synthesis by du Vigneaud in 1953, has mainly been associated with beneficial physiological effects, as well as positive social and emotional behaviors. This overall positive picture of oxytocin as the "love-, cuddle-, or bonding-hormone" has repeatedly been challenged since then. Oxytocin-induced effects that would be perceived as negative by the individual, such as increased anxiety or potentiation of stress-induced ACTH release, as well as the regulation of negative approach-related emotions, such as envy and schadenfreude (gloating) have been described. The general consent is that oxytocin, instead of acting unidirectional, induces changes in the salience network to shift the emphasis of emotional contexts, and therefore can, e.g., produce both anxiolytic as well as anxiogenic behavioral outcomes. However, the underlying mechanisms leading to alterations in the salience network are still unclear. With the aim to understand the manifold effects of oxytocin on a cellular/molecular level, a set of oxytocin receptor-coupled signaling cascades and downstream effectors regulating transcription and translation has been identified. Those oxytocin-driven effectors, such as MEF2 and CREB, are known modulators of the neuronal and glial cytoarchitecture. We hypothesize that, by determining cellular morphology and connectivity, MEF2 is one of the key factors that might contribute to the diverse behavioral effects of oxytocin.
Collapse
|
38
|
Li W, Yang F, Gao J, Tang Y, Wang J, Pan Y. Over-Expression of TRPC6 via CRISPR Based Synergistic Activation Mediator in BMSCs Ameliorates Brain Injury in a Rat Model of Cerebral Ischemia/Reperfusion. Neuroscience 2019; 415:147-160. [PMID: 31369718 DOI: 10.1016/j.neuroscience.2019.06.041] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2018] [Revised: 06/26/2019] [Accepted: 06/27/2019] [Indexed: 12/23/2022]
Abstract
Stroke is a major life-threatening and disabling disease with a restricted therapeutic approach. Bone marrow stromal cells (BMSCs) possess proliferative ability and a multi-directional differentiation potential, and secrete a range of trophic/growth factors that can protect neurons after cerebral ischemia/reperfusion. Transient receptor potential canonical (TRPC) is a family of non-selective channels permeable to Ca2+, with several functions including neuronal survival. Over-expression of TRPC6, a subtype of the TRPC family, was shown to protect neurons against cerebral ischemia/reperfusion injury. However, it remains unclear whether over-expression of TRPC6 in BMSCs can further reduce brain injury after ischemia/reperfusion. In the present study, we report that over-expression of TRPC6 via a CRISPR-based synergistic activation mediator in BMSCs provided a greater reduction of brain injury in a rat model of ischemia/reperfusion. Further, the improved neurofunctional outcomes were associated with increased TRPC6 and brain derived neurotrophic factor expression levels. Overall, these data suggest that TRPC6 over-expressing BMSCs may be a promising therapeutic agent for ischemic stroke.
Collapse
Affiliation(s)
- Wenbin Li
- Department of Neurology, First Clinical College of Harbin Medical University, Room 501, Building 3, 23 Youzheng Street, Harbin, Heilongjiang Province, 150001, People's Republic of China
| | - Fan Yang
- Department of Neurology, First Clinical College of Harbin Medical University, Room 501, Building 3, 23 Youzheng Street, Harbin, Heilongjiang Province, 150001, People's Republic of China
| | - Jinxing Gao
- Department of Neurology, First Clinical College of Harbin Medical University, Room 501, Building 3, 23 Youzheng Street, Harbin, Heilongjiang Province, 150001, People's Republic of China
| | - Yushi Tang
- Department of Neurology, First Clinical College of Harbin Medical University, Room 501, Building 3, 23 Youzheng Street, Harbin, Heilongjiang Province, 150001, People's Republic of China
| | - Jing Wang
- Department of Neurology, First Clinical College of Harbin Medical University, Room 501, Building 3, 23 Youzheng Street, Harbin, Heilongjiang Province, 150001, People's Republic of China
| | - Yujun Pan
- Department of Neurology, First Clinical College of Harbin Medical University, Room 501, Building 3, 23 Youzheng Street, Harbin, Heilongjiang Province, 150001, People's Republic of China.
| |
Collapse
|
39
|
Talbot BE, Vandorpe DH, Stotter BR, Alper SL, Schlondorff JS. Transmembrane insertases and N-glycosylation critically determine synthesis, trafficking, and activity of the nonselective cation channel TRPC6. J Biol Chem 2019; 294:12655-12669. [PMID: 31266804 PMCID: PMC6709635 DOI: 10.1074/jbc.ra119.008299] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Revised: 06/28/2019] [Indexed: 12/12/2022] Open
Abstract
Transient receptor potential cation channel subfamily C member 6 (TRPC6) is a widely expressed ion channel. Gain-of-function mutations in the human TRPC6 channel cause autosomal-dominant focal segmental glomerulosclerosis, but the molecular components involved in disease development remain unclear. Here, we found that overexpression of gain-of-function TRPC6 channel variants is cytotoxic in cultured cells. Exploiting this phenotype in a genome-wide CRISPR/Cas screen for genes whose inactivation rescues cells from TRPC6-associated cytotoxicity, we identified several proteins essential for TRPC6 protein expression, including the endoplasmic reticulum (ER) membrane protein complex transmembrane insertase. We also identified transmembrane protein 208 (TMEM208), a putative component of a signal recognition particle-independent (SND) ER protein-targeting pathway, as being necessary for expression of TRPC6 and several other ion channels and transporters. TRPC6 expression was also diminished by loss of the previously uncharacterized WD repeat domain 83 opposite strand (WDR83OS), which interacted with both TRPC6 and TMEM208. Additionally enriched among the screen hits were genes involved in N-linked protein glycosylation. Deletion of the mannosyl (α-1,3-)-glycoprotein β-1,2-N-acetylglucosaminyltransferase (MGAT1), necessary for the generation of complex N-linked glycans, abrogated TRPC6 gain-of-function variant-mediated Ca2+ influx and extracellular signal-regulated kinase activation in HEK cells, but failed to diminish cytotoxicity in cultured podocytes. However, mutating the two TRPC6 N-glycosylation sites abrogated the cytotoxicity of mutant TRPC6 and reduced its surface expression. These results expand the targets of TMEM208-mediated ER translocation to include multipass transmembrane proteins and suggest that TRPC6 N-glycosylation plays multiple roles in modulating channel trafficking and activity.
Collapse
Affiliation(s)
- Brianna E Talbot
- Division of Nephrology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts 02215
| | - David H Vandorpe
- Division of Nephrology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts 02215
| | - Brian R Stotter
- Division of Nephrology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts 02215
- Division of Nephrology, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts 02115
| | - Seth L Alper
- Division of Nephrology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts 02215
| | - Johannes S Schlondorff
- Division of Nephrology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts 02215
| |
Collapse
|
40
|
Zeitler S, Ye L, Andreyeva A, Schumacher F, Monti J, Nürnberg B, Nowak G, Kleuser B, Reichel M, Fejtová A, Kornhuber J, Rhein C, Friedland K. Acid sphingomyelinase - a regulator of canonical transient receptor potential channel 6 (TRPC6) activity. J Neurochem 2019; 150:678-690. [PMID: 31310676 DOI: 10.1111/jnc.14823] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Revised: 07/02/2019] [Accepted: 07/04/2019] [Indexed: 12/28/2022]
Abstract
Recent investigations propose the acid sphingomyelinase (ASM)/ceramide system as a novel target for antidepressant action. ASM catalyzes the breakdown of the abundant membrane lipid sphingomyelin to the lipid messenger ceramide. This ASM-induced lipid modification induces a local shift in membrane properties, which influences receptor clustering and downstream signaling. Canonical transient receptor potential channels 6 (TRPC6) are non-selective cation channels located in the cell membrane that play an important role in dendritic growth, synaptic plasticity and cognition in the brain. They can be activated by hyperforin, an ingredient of the herbal remedy St. John's wort for treatment of depression disorders. Because of their role in the context of major depression, we investigated the crosstalk between the ASM/ceramide system and TRPC6 ion channels in a pheochromocytoma cell line 12 neuronal cell model (PC12 rat pheochromocytoma cell line). Ca2+ imaging experiments indicated that hyperforin-induced Ca2+ influx through TRPC6 channels is modulated by ASM activity. While antidepressants, known as functional inhibitors of ASM activity, reduced TRPC6-mediated Ca2+ influx, extracellular application of bacterial sphingomyelinase rebalanced TRPC6 activity in a concentration-related way. This effect was confirmed in whole-cell patch clamp electrophysiology recordings. Lipidomic analyses revealed a decrease in very long chain ceramide/sphingomyelin molar ratio after ASM inhibition, which was connected with changes in the abundance of TRPC6 channels in flotillin-1-positive lipid rafts as visualized by western blotting. Our data provide evidence that the ASM/ceramide system regulates TRPC6 channels likely by controlling their recruitment to specific lipid subdomains and thereby fine-tuning their physical properties.
Collapse
Affiliation(s)
- Stefanie Zeitler
- Department of Psychiatry and Psychotherapy, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany.,Department of Chemistry and Pharmacy, Molecular and Clinical Pharmacy, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Lian Ye
- Department of Chemistry and Pharmacy, Molecular and Clinical Pharmacy, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Aksana Andreyeva
- Department of Chemistry and Pharmacy, Molecular and Clinical Pharmacy, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Fabian Schumacher
- Department of Toxicology, Faculty of Mathematics and Natural Science, Institute of Nutritional Science, University of Potsdam, Nuthetal, Germany.,Department of Molecular Biology, University Clinic, University of Duisburg-Essen, Essen, Germany
| | - Juliana Monti
- Department of Psychiatry and Psychotherapy, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Bernd Nürnberg
- Department of Pharmacology and Experimental Therapy, Institute of Experimental and Clinical Pharmacology and Toxicology, Eberhard Karls University Hospitals and Clinics and Interfaculty Center of Pharmacogenomics and Drug Research (ICePhA), Tübingen, Germany
| | - Gabriel Nowak
- Department of Pharmacobiology, Jagiellonian University Medical College, Kraków, Poland.,Institute of Pharmacology, Polish Academy of Sciences, Kraków, Poland
| | - Burkhard Kleuser
- Department of Toxicology, Faculty of Mathematics and Natural Science, Institute of Nutritional Science, University of Potsdam, Nuthetal, Germany
| | - Martin Reichel
- Department of Psychiatry and Psychotherapy, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Anna Fejtová
- Department of Psychiatry and Psychotherapy, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Johannes Kornhuber
- Department of Psychiatry and Psychotherapy, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Cosima Rhein
- Department of Psychiatry and Psychotherapy, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Kristina Friedland
- Department of Chemistry and Pharmacy, Molecular and Clinical Pharmacy, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany.,Institute for Pharmacy and Biochemistry, Pharmacology and Toxicology, Johannes-Gutenberg University Mainz, Mainz, Germany
| |
Collapse
|
41
|
Landeira BS, Santana TTDS, Araújo JADM, Tabet EI, Tannous BA, Schroeder T, Costa MR. Activity-Independent Effects of CREB on Neuronal Survival and Differentiation during Mouse Cerebral Cortex Development. Cereb Cortex 2019; 28:538-548. [PMID: 27999124 DOI: 10.1093/cercor/bhw387] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2015] [Accepted: 12/01/2016] [Indexed: 11/14/2022] Open
Abstract
Neuronal survival and morphological maturation depends on the action of the transcription factor calcium responsive element binding protein (CREB), which regulates expression of several target genes in an activity-dependent manner. However, it remains largely unknown whether CREB-mediated transcription could play a role at early stages of neuronal differentiation, prior to the establishment of functional synaptic contacts. Here, we show that CREB is phosphorylated at very early stages of neuronal differentiation in vivo and in vitro, even in the absence of depolarizing agents. Using genetic tools, we also show that inhibition of CREB-signaling affects neuronal growth and survival in vitro without affecting cell proliferation and neurogenesis. Expression of A-CREB or M-CREB, 2 dominant-negative inhibitors of CREB, decreases cell survival and the complexity of neuronal arborization. Similar changes are observed in neurons treated with protein kinase A (PKA) and Ca2+/calmodulin-dependent protein kinase II (CaMKII) inhibitors, which also show decreased levels of pCREBSer133. Notably, expression of CREB-FY, a Tyr134Phe CREB mutant with a lower Km for phosphorylation, partly rescues the effects of PKA and CaMKII inhibition. Our data indicate that CREB-mediated signaling play important roles at early stages of cortical neuron differentiation, prior to the establishment of fully functional synaptic contacts.
Collapse
Affiliation(s)
| | | | | | - Elie I Tabet
- Experimental Therapeutics and Molecular Imaging Laboratory, Neuroscience Center, Department of Neurology, Massachusetts General Hospital, Boston, Massachusetts, USA.,Program in Neuroscience, Harvard Medical School, Boston, Massachusetts, USA
| | - Bakhos A Tannous
- Experimental Therapeutics and Molecular Imaging Laboratory, Neuroscience Center, Department of Neurology, Massachusetts General Hospital, Boston, Massachusetts, USA.,Program in Neuroscience, Harvard Medical School, Boston, Massachusetts, USA
| | - Timm Schroeder
- Department of Biosystems Science and Engineering, ETH Zurich, Mattenstr. 26, 4058 Basel, Switzerland
| | - Marcos R Costa
- Brain Institute, Federal University of Rio Grande do Norte, Natal 59056-450, Brazil
| |
Collapse
|
42
|
Ma R, Xu Y, Zhou H, Zhang D, Yao D, Song L, Liu Y. Participation of the AngII/TRPC6/NFAT axis in the pathogenesis of podocyte injury in rats with type 2 diabetes. Mol Med Rep 2019; 19:2421-2430. [PMID: 30664212 DOI: 10.3892/mmr.2019.9871] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2018] [Accepted: 12/19/2018] [Indexed: 11/06/2022] Open
Abstract
The canonical transient receptor potential channel 6 ion channel is expressed in podocytes and is an important component of the glomerular slit diaphragm. Focal segmental glomerulosclerosis is closely associated with TRPC6 gene mutations, and TRPC6 mediates podocyte injury induced by high glucose. Angiotensin II (AngII) has been revealed to enhance TRPC6 currents in certain types of cells, including podocytes and ventricular myocytes. It has been reported that glucose regulated TRPC6 expression in an AngII‑dependent manner in podocytes and that this pathway is critical in diabetic nephropathy. In the present study, the role of TRPC6 detected by western blotting and reverse transcription‑quantitative polymerase chain reaction in AngII‑mediated podocyte injury was evaluated in rats with type 2 diabetes induced by high‑calorie diets and streptozotocin. The results demonstrated that urinary albumin excretion was elevated, and morphological changes, including glomerular basement membrane thickening and podocyte process effacement, were observed. There was increased expression of AngII and TRPC6 in diabetic rats. The angiotensin receptor blocker valsartan significantly reduced TRPC6 and nuclear factor of activated T‑cells (NFAT) overexpression in diabetic rats. These results in vivo were confirmed by studies in vitro, which demonstrated that inhibition of TRPC6 ameliorated high glucose‑induced podocyte injury by decreasing NFAT mRNA levels. Taken together, the present results suggested that the AngII/TRPC6/NFAT axis may be a crucial signaling pathway in podocytes that is necessary for maintaining the integrity of the glomerular filtration barrier. In addition, TRPC6 may represent a potential therapeutic target for diabetic nephropathy.
Collapse
Affiliation(s)
- Ruixia Ma
- Department of Nephrology, Affiliated Hospital of Qingdao University, Qingdao, Shandong 266000, P.R. China
| | - Yan Xu
- Department of Nephrology, Affiliated Hospital of Qingdao University, Qingdao, Shandong 266000, P.R. China
| | - Hanyan Zhou
- Department of Nephrology, Affiliated Hospital of Qingdao University, Qingdao, Shandong 266000, P.R. China
| | - Di Zhang
- Department of Special Medicine, School of Medicine, Qingdao University, Qingdao, Shandong 266071, P.R. China
| | - Dandan Yao
- Department of Nephrology, Affiliated Hospital of Qingdao University, Qingdao, Shandong 266000, P.R. China
| | - Limin Song
- Department of Special Medicine, School of Medicine, Qingdao University, Qingdao, Shandong 266071, P.R. China
| | - Yuan Liu
- Department of Special Medicine, School of Medicine, Qingdao University, Qingdao, Shandong 266071, P.R. China
| |
Collapse
|
43
|
Popugaeva E, Pchitskaya E, Bezprozvanny I. Dysregulation of Intracellular Calcium Signaling in Alzheimer's Disease. Antioxid Redox Signal 2018; 29:1176-1188. [PMID: 29890840 PMCID: PMC6157344 DOI: 10.1089/ars.2018.7506] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
SIGNIFICANCE Calcium (Ca2+) hypothesis of Alzheimer's disease (AD) gains popularity. It points to new signaling pathways that may underlie AD pathogenesis. Based on calcium hypothesis, novel targets for the development of potential AD therapies are identified. Recent Advances: Recently, the key role of neuronal store-operated calcium entry (nSOCE) in the development of AD has been described. Correct regulation of nSOCE is necessary for the stability of postsynaptic contacts to preserve the memory formation. Molecular identity of hippocampal nSOCE is defined. Perspective nSOCE-activating molecule, prototype of future anti-AD drugs, is described. CRITICAL ISSUES Endoplasmic reticulum Ca2+ overload happens in many but not in all AD models. The nSOCE targeting therapy described in this review may not be universally applicable. FUTURE DIRECTIONS There is a need to determine whether AD is a syndrome with one critical signaling pathway that initiates pathology, or it is a disorder with many different signaling pathways that are disrupted simultaneously or one after each other. It is necessary to validate applicability of nSOCE-activating therapy for the development of anti-AD medication. There is an experimental correlation between downregulated nSOCE and disrupted postsynaptic contacts in AD mouse models. Signaling mechanisms downstream of nSOCE which are responsible for the regulation of stability of postsynaptic contacts have to be discovered. That will bring new targets for the development of AD-preventing therapies. Antioxid. Redox Signal. 29, 1176-1188.
Collapse
Affiliation(s)
- Elena Popugaeva
- 1 Laboratory of Molecular Neurodegeneration, Department of Medical Physics, Peter the Great St.Petersburg Polytechnic University , St.Petersburg, Russian Federation
| | - Ekaterina Pchitskaya
- 1 Laboratory of Molecular Neurodegeneration, Department of Medical Physics, Peter the Great St.Petersburg Polytechnic University , St.Petersburg, Russian Federation
| | - Ilya Bezprozvanny
- 1 Laboratory of Molecular Neurodegeneration, Department of Medical Physics, Peter the Great St.Petersburg Polytechnic University , St.Petersburg, Russian Federation.,2 Department of Physiology, UT Southwestern Medical Center at Dallas , Dallas, Texas
| |
Collapse
|
44
|
Actin cytoskeleton dynamics in stem cells from autistic individuals. Sci Rep 2018; 8:11138. [PMID: 30042445 PMCID: PMC6057935 DOI: 10.1038/s41598-018-29309-6] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Accepted: 07/03/2018] [Indexed: 11/15/2022] Open
Abstract
Several lines of indirect evidence, such as mutations or dysregulated expression of genes related to cytoskeleton, have suggested that cytoskeletal dynamics, a process essential for axons and dendrites development, is compromised in autism spectrum disorders (ASD). However, no study has yet examined whether cytoskeleton dynamics is functionally altered in cells from ASD patients. Here we investigated the regulation of actin cytoskeleton dynamics in stem cells from human exfoliated deciduous teeth (SHEDs) of 13 ASD patients and 8 control individuals by inducing actin filament depolymerization and then measuing their reconstruction upon activation of the RhoGTPases Rac, Cdc42 or RhoA. We observed that stem cells from seven ASD individuals (53%) presented altered dymanics of filament reconstruction, including a patient recently studied by our group whose iPSC-derived neuronal cells show shorten and less arborized neurites. We also report potentially pathogenic genetic variants that might be related to the alterations in actin repolymerization dynamics observed in some patient-derived cells. Our results suggest that, at least for a subgroup of ASD patients, the dynamics of actin polymerization is impaired, which might be ultimately leading to neuronal abnormalities.
Collapse
|
45
|
Wang Q, He Q, Chen Y, Shao W, Yuan C, Wang Y. JNK-mediated microglial DICER degradation potentiates inflammatory responses to induce dopaminergic neuron loss. J Neuroinflammation 2018; 15:184. [PMID: 29907159 PMCID: PMC6003208 DOI: 10.1186/s12974-018-1218-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Accepted: 05/29/2018] [Indexed: 12/12/2022] Open
Abstract
Background Amplified inflammation is important for the progression of Parkinson’s disease (PD). However, how this enhanced inflammation is regulated remains largely unknown. Deletion of DICER leads to progressive dopamine neuronal loss and induces gliosis. We hypothesized that the homeostasis of microglial DICER would be responsible for the amplified inflammation in the mouse model of PD. Methods The microglia or C57BL/6 mice were treated or injected with l-methyl-4-phenyl-l,2,3,6-tetrahydropyridine (MPTP) or 1-methyl-4-phenylpyridinium (MPP+), respectively, for the model establishment. Microglia and astrocytes sorted by fluorescence-activated cell sorter (FACS) were assayed by quantitative real-time PCR, Western blotting, immunoprecipitation, enzyme-linked immunosorbent assay (ELISA), immunohistofluorescence, and mass spectrometry. Results Microglial DICER was phosphorylated at serine 1456 by c-jun N-terminal kinase (JNK) and downregulated in response to 1-methyl-4-phenylpyridinium (MPP+), a causative agent in PD. Inhibition of JNK phosphorylation of DICER at serine 1456 rescued the MPP+-induced DICER degradation, suppressed microglial inflammatory process, and prevented the loss of tyrosine hydroxylase-expressing neurons in the mouse MPTP model. Conclusions JNK-mediated microglial DICER degradation potentiates inflammation to induce dopaminergic neuronal loss. Thus, preventing microglial DICER degradation could be a novel strategy for controlling neuroinflammation in PD. Electronic supplementary material The online version of this article (10.1186/s12974-018-1218-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Qing Wang
- Center of Cognition and Brain Science, Beijing Institute of Medical Sciences, Beijing, 100000, People's Republic of China
| | - Qian He
- Center of Cognition and Brain Science, Beijing Institute of Medical Sciences, Beijing, 100000, People's Republic of China
| | - Yifei Chen
- Center of Cognition and Brain Science, Beijing Institute of Medical Sciences, Beijing, 100000, People's Republic of China
| | - Wei Shao
- Center of Cognition and Brain Science, Beijing Institute of Medical Sciences, Beijing, 100000, People's Republic of China
| | - Chao Yuan
- Center of Cognition and Brain Science, Beijing Institute of Medical Sciences, Beijing, 100000, People's Republic of China
| | - Yizheng Wang
- National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, 12 Middle Wulumuqi Road, Shanghai, 200040, People's Republic of China.
| |
Collapse
|
46
|
Schulz C, Fritz N, Sommer T, Krofta K, Friedland K, Pischetsrieder M. Activation of membrane-located Ca2+ channels by hop beta acids and their tricyclic transformation products. Food Chem 2018; 252:215-227. [DOI: 10.1016/j.foodchem.2018.01.073] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Revised: 12/11/2017] [Accepted: 01/09/2018] [Indexed: 12/23/2022]
|
47
|
Hu HJ, Song M. Disrupted Ionic Homeostasis in Ischemic Stroke and New Therapeutic Targets. J Stroke Cerebrovasc Dis 2017; 26:2706-2719. [PMID: 29054733 DOI: 10.1016/j.jstrokecerebrovasdis.2017.09.011] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Revised: 08/30/2017] [Accepted: 09/06/2017] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Stroke is a leading cause of long-term disability. All neuroprotectants targeting excitotoxicity have failed to become stroke medications. In order to explore and identify new therapeutic targets for stroke, we here reviewed present studies of ionic transporters and channels that are involved in ischemic brain damage. METHOD We surveyed recent literature from animal experiments and clinical reports in the databases of PubMed and Elsevier ScienceDirect to analyze ionic mechanisms underlying ischemic cell damage and suggest promising ideas for stroke therapy. RESULTS Dysfunction of ionic transporters and disrupted ionic homeostasis are most early changes that underlie ischemic brain injury, thus receiving sustained attention in translational stroke research. The Na+/K+-ATPase, Na+/Ca2+ Exchanger, ionotropic glutamate receptor, acid-sensing ion channels (ASICs), sulfonylurea receptor isoform 1 (SUR1)-regulated NCCa-ATP channels, and transient receptor potential (TRP) channels are critically involved in ischemia-induced cellular degenerating processes such as cytotoxic edema, excitotoxicity, necrosis, apoptosis, and autophagic cell death. Some ionic transporters/channels also act as signalosomes to regulate cell death signaling. For acute stroke treatment, glutamate-mediated excitotoxicity must be interfered within 2 hours after stroke. The SUR1-regulated NCCa-ATP channels, Na+/K+-ATPase, ASICs, and TRP channels have a much longer therapeutic window, providing new therapeutic targets for developing feasible pharmacological treatments toward acute ischemic stroke. CONCLUSION The next generation of stroke therapy can apply a polypharmacology strategy for which drugs are designed to target multiple ion transporters/channels or their interaction with neurotoxic signaling pathways. But a successful translation of neuroprotectants relies on in-depth analyses of cell death mechanisms and suitable animal models resembling human stroke.
Collapse
Affiliation(s)
- Hui-Jie Hu
- Department of Pharmacology, Institute of Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Mingke Song
- Department of Pharmacology, Institute of Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
48
|
Xiao X, Liu HX, Shen K, Cao W, Li XQ. Canonical Transient Receptor Potential Channels and Their Link with Cardio/Cerebro-Vascular Diseases. Biomol Ther (Seoul) 2017; 25:471-481. [PMID: 28274093 PMCID: PMC5590790 DOI: 10.4062/biomolther.2016.096] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Revised: 12/04/2016] [Accepted: 12/27/2016] [Indexed: 12/29/2022] Open
Abstract
The canonical transient receptor potential channels (TRPCs) constitute a series of nonselective cation channels with variable degrees of Ca2+ selectivity. TRPCs consist of seven mammalian members, TRPC1, TRPC2, TRPC3, TRPC4, TRPC5, TRPC6, and TRPC7, which are further divided into four subtypes, TRPC1, TRPC2, TRPC4/5, and TRPC3/6/7. These channels take charge of various essential cell functions such as contraction, relaxation, proliferation, and dysfunction. This review, organized into seven main sections, will provide an overview of current knowledge about the underlying pathogenesis of TRPCs in cardio/cerebrovascular diseases, including hypertension, pulmonary arterial hypertension, cardiac hypertrophy, atherosclerosis, arrhythmia, and cerebrovascular ischemia reperfusion injury. Collectively, TRPCs could become a group of drug targets with important physiological functions for the therapy of human cardio/cerebro-vascular diseases.
Collapse
Affiliation(s)
- Xiong Xiao
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an 710032, China
| | - Hui-Xia Liu
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an 710032, China.,Cadet Brigade, Fourth Military Medical University, Xi'an 710032, China
| | - Kuo Shen
- Cadet Brigade, Fourth Military Medical University, Xi'an 710032, China
| | - Wei Cao
- Department of Natural Medicine & Institute of Materia Medica, School of Pharmacy, Fourth Military Medical University, Xi'an 710032, China
| | - Xiao-Qiang Li
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an 710032, China
| |
Collapse
|
49
|
Thiel G, Lesch A, Rubil S, Backes TM, Rössler OG. Regulation of Gene Transcription Following Stimulation of Transient Receptor Potential (TRP) Channels. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2017; 335:167-189. [PMID: 29305012 DOI: 10.1016/bs.ircmb.2017.07.010] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Transient receptor potential (TRP) channels belong to a heterogeneous superfamily of cation channels that are involved in the regulation of numerous biological functions, including regulation of Ca2+ and glucose homeostasis, tumorigenesis, temperature, and pain sensation. To understand the functions of TRP channels, their associated intracellular signaling pathways and molecular targets have to be identified on the cellular level. Stimulation of TRP channels frequently induces an influx of Ca2+ ions into the cells and the subsequent activation of protein kinases. These intracellular signal transduction pathways ultimately induce changes in the gene expression pattern of the cells. Here, we review the effects of TRPC6, TRPM3, and TRPV1 channel stimulation on the activation of the stimulus-responsive transcription factors AP-1, CREB, Egr-1, Elk-1, and NFAT. Following activation, these transcription factors induce the transcription of delayed response genes. We propose that many biological functions of TRP channels can be explained by the activation of stimulus-responsive transcription factors and their delayed response genes. The proteins encoded by those delayed response genes may be responsible for the biochemical and physiological changes following TRP channel activation.
Collapse
Affiliation(s)
- Gerald Thiel
- Saarland University Medical Faculty, Homburg, Germany.
| | - Andrea Lesch
- Saarland University Medical Faculty, Homburg, Germany
| | - Sandra Rubil
- Saarland University Medical Faculty, Homburg, Germany
| | | | | |
Collapse
|
50
|
Store-Operated Calcium Channel Complex in Postsynaptic Spines: A New Therapeutic Target for Alzheimer's Disease Treatment. J Neurosci 2017; 36:11837-11850. [PMID: 27881772 DOI: 10.1523/jneurosci.1188-16.2016] [Citation(s) in RCA: 98] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Revised: 09/08/2016] [Accepted: 09/11/2016] [Indexed: 12/11/2022] Open
Abstract
Mushroom dendritic spine structures are essential for memory storage and the loss of mushroom spines may explain memory defects in aging and Alzheimer's disease (AD). The stability of mushroom spines depends on stromal interaction molecule 2 (STIM2)-mediated neuronal-store-operated Ca2+ influx (nSOC) pathway, which is compromised in AD mouse models, in aging neurons, and in sporadic AD patients. Here, we demonstrate that the Transient Receptor Potential Canonical 6 (TRPC6) and Orai2 channels form a STIM2-regulated nSOC Ca2+ channel complex in hippocampal mushroom spines. We further demonstrate that a known TRPC6 activator, hyperforin, and a novel nSOC positive modulator, NSN21778 (NSN), can stimulate activity of nSOC pathway in the spines and rescue mushroom spine loss in both presenilin and APP knock-in mouse models of AD. We further show that NSN rescues hippocampal long-term potentiation impairment in APP knock-in mouse model. We conclude that the STIM2-regulated TRPC6/Orai2 nSOC channel complex in dendritic mushroom spines is a new therapeutic target for the treatment of memory loss in aging and AD and that NSN is a potential candidate molecule for therapeutic intervention in brain aging and AD. SIGNIFICANCE STATEMENT Mushroom dendritic spine structures are essential for memory storage and the loss of mushroom spines may explain memory defects in Alzheimer's disease (AD). This study demonstrated that Transient Receptor Potential Canonical 6 (TRPC6) and Orai2 form stromal interaction molecule 2 (STIM2)-regulated neuronal-store-operated Ca2+ influx (nSOC) channel complex in hippocampal synapse and the resulting Ca2+ influx is critical for long-term maintenance of mushroom spines in hippocampal neurons. A novel nSOC-positive modulator, NSN21778 (NSN), rescues mushroom spine loss and synaptic plasticity impairment in AD mice models. The TRPC6/Orai2 nSOC channel complex is a new therapeutic target and NSN is a potential candidate molecule for therapeutic intervention in brain aging and AD.
Collapse
|