1
|
Gideon S, Boyd B, Ramirez Sierra B, Arenas D, Ochoa P, Eme J, Kolosov D. Voltage-gated ion channels in cultured gill epithelia of rainbow trout, Oncorhynchus mykiss, change in transcript abundance with exposure to freshwater. Comp Biochem Physiol A Mol Integr Physiol 2025; 304:111835. [PMID: 40074167 DOI: 10.1016/j.cbpa.2025.111835] [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: 11/25/2024] [Revised: 03/05/2025] [Accepted: 03/07/2025] [Indexed: 03/14/2025]
Abstract
Salmonid fishes are well adapted to transition between salinities as part of a diadromid lifestyle, and many species are both economically and environmentally important. Ion-transporting gill epithelium helps fishes maintain ion balance during salinity transition. Recent transcriptomic surveys suggest that voltage-gated ion channels (VGICs) are present in gill epithelium of fishes. However, fish gill epithelia are architecturally complex and structurally heterogeneous (which includes layers of excitable tissues), which necessitates a model to study isolated gill epithelial cells. In the present study, we isolated gill epithelial cells, used them to reconstruct primary cultured gill epithelium model, and exposed the reconstructed epithelia to apical freshwater (FW). Using RNAseq and molecular biology we demonstrate that multiple VGICs are expressed in cultured gill epithelia of a salmonid, rainbow trout Oncorhynchus mykiss. Following apical exposure to FW, multiple subunits of voltage-gated calcium (CaV) channels, as well as KCNE2 were upregulated in mRNA abundance. Using a custom-made antibody, we demonstrated that CaV1.3 immunolocalized to the apical membrane of epithelia in intact trout gill, as well as in the cultured gill epithelium. Pharmacological inhibition of CaV1 in FW-exposed cultured epithelia led to increased transepithelial resistance. Therefore, we propose that VGICs are present in gill epithelia of fishes, and may rapidly and autonomously respond to environmental salinity changes to help the fish maintain salt and water balance, where CaV1 specifically may play a particularly important role in rapid adjustment of gill epithelia barrier properties and resistivity and potentially in responding to regulatory cell volume decrease in vitro.
Collapse
Affiliation(s)
- Siaje Gideon
- Department of Biological Sciences, California State University San Marcos, CA 92096, USA
| | - Brendan Boyd
- Department of Biological Sciences, California State University San Marcos, CA 92096, USA
| | - Brandon Ramirez Sierra
- Department of Biological Sciences, California State University San Marcos, CA 92096, USA
| | - Dennise Arenas
- Department of Biological Sciences, California State University San Marcos, CA 92096, USA
| | - Perla Ochoa
- Department of Biological Sciences, California State University San Marcos, CA 92096, USA
| | - John Eme
- Department of Biological Sciences, California State University San Marcos, CA 92096, USA
| | - Dennis Kolosov
- Department of Biological Sciences, California State University San Marcos, CA 92096, USA.
| |
Collapse
|
2
|
Limpitikul WB, Dick IE. Inactivation of CaV1 and CaV2 channels. J Gen Physiol 2025; 157:e202313531. [PMID: 39883005 PMCID: PMC11781272 DOI: 10.1085/jgp.202313531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Revised: 01/02/2025] [Accepted: 01/15/2025] [Indexed: 01/31/2025] Open
Abstract
Voltage-gated Ca2+ channels (VGCCs) are highly expressed throughout numerous biological systems and play critical roles in synaptic transmission, cardiac excitation, and muscle contraction. To perform these various functions, VGCCs are highly regulated. Inactivation comprises a critical mechanism controlling the entry of Ca2+ through these channels and constitutes an important means to regulate cellular excitability, shape action potentials, control intracellular Ca2+ levels, and contribute to long-term potentiation and depression. For CaV1 and CaV2 channel families, inactivation proceeds via two distinct processes. Voltage-dependent inactivation (VDI) reduces Ca2+ entry through the channel in response to sustained or repetitive depolarization, while Ca2+-dependent inactivation (CDI) occurs in response to elevations in intracellular Ca2+ levels. These processes are critical for physiological function and undergo exquisite fine-tuning through multiple mechanisms. Here, we review known determinants and modulatory features of these two critical forms of channel regulation and their role in normal physiology and pathophysiology.
Collapse
Affiliation(s)
| | - Ivy E. Dick
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD, USA
| |
Collapse
|
3
|
Liu Y, Tang Q, Tao Q, Dong H, Shi Z, Zhou L. Low-frequency magnetic field therapy for glioblastoma: Current advances, mechanisms, challenges and future perspectives. J Adv Res 2025; 69:531-543. [PMID: 38565404 PMCID: PMC11954840 DOI: 10.1016/j.jare.2024.03.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Revised: 03/10/2024] [Accepted: 03/29/2024] [Indexed: 04/04/2024] Open
Abstract
BACKGROUND Glioblastoma (GBM) is the most common malignant tumour of the central nervous system. Despite recent advances in multimodal GBM therapy incorporating surgery, radiotherapy, systemic therapy (chemotherapy, targeted therapy), and supportive care, the overall survival (OS) remains poor, and long-term survival is rare. Currently, the primary obstacles hindering the effectiveness of GBM treatment are still the blood-brain barrier and tumor heterogeneity. In light of its substantial advantages over conventional therapies, such as strong penetrative ability and minimal side effects, low-frequency magnetic fields (LF-MFs) therapy has gradually caught the attention of scientists. AIM OF REVIEW In this review, we shed the light on the current status of applying LF-MFs in the treatment of GBM. We specifically emphasize our current understanding of the mechanisms by which LF-MFs mediate anticancer effects and the challenges faced by LF-MFs in treating GBM cells. Furthermore, we discuss the prospective applications of magnetic field therapy in the future treatment of GBM. Key scientific concepts of review: The review explores the current progress on the use of LF-MFs in the treatment of GBM with a special focus on the potential underlying mechanisms of LF-MFs in anticancer effects. Additionally, we also discussed the complex magnetic field features and biological characteristics related to magnetic bioeffects. Finally, we proposed a promising magnetic field treatment strategy for future applications in GBM therapy.
Collapse
Affiliation(s)
- Yinlong Liu
- Department of Neurosurgery, Huashan Hospital, Fudan University, China
| | - Qisheng Tang
- Department of Neurosurgery, Huashan Hospital, Fudan University, China; National Center for Neurological Disorders, China; Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, China; Neurosurgical Institute of Fudan University, Shanghai, China; Shanghai Clinical Medical Center of Neurosurgery, China
| | - Quan Tao
- Shanghai Institute of Microsystem and Information Technology, China
| | - Hui Dong
- Shanghai Institute of Microsystem and Information Technology, China
| | - Zhifeng Shi
- Department of Neurosurgery, Huashan Hospital, Fudan University, China; National Center for Neurological Disorders, China; Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, China; Neurosurgical Institute of Fudan University, Shanghai, China; Shanghai Clinical Medical Center of Neurosurgery, China.
| | - Liangfu Zhou
- Department of Neurosurgery, Huashan Hospital, Fudan University, China; National Center for Neurological Disorders, China; Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, China; Neurosurgical Institute of Fudan University, Shanghai, China; Shanghai Clinical Medical Center of Neurosurgery, China.
| |
Collapse
|
4
|
Sun Y, Geng J, Fan Y, Li Y, Zhong Y, Cai J, Liu X, Wang S, Gong Y, Chang C, Yang Y, Fan C. A Non-Invasive and DNA-free Approach to Upregulate Mammalian Voltage-Gated Calcium Channels and Neuronal Calcium Signaling via Terahertz Stimulation. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2405436. [PMID: 39435751 DOI: 10.1002/advs.202405436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/18/2024] [Revised: 09/22/2024] [Indexed: 10/23/2024]
Abstract
Mammalian voltage-gated calcium channels (CaV) play critical roles in cardiac excitability, synaptic transmission, and gene transcription. Dysfunctions in CaV are implicated in a variety of cardiac and neurodevelopmental disorders. Current pharmacological approaches to enhance CaV activity are limited by off-target effects, drug metabolism issues, cytotoxicity, and imprecise modulation. Additionally, genetically-encoded channel activators and optogenetic tools are restricted by gene delivery challenges and biosafety concerns. Here a novel terahertz (THz) wave-based method to upregulate CaV1.2, a key subtype of CaV, and boost CaV1-mediated Ca2+ signaling in neurons without introducing exogenous DNA is presented. Using molecular dynamics simulations, it is shown that 42.5 THz (7.05 µm, 1418 cm-1) waves enhance Ca2+ conductance in CaV1.2 by resonating with the stretching mode of the -COO- group in the selectivity filter. Electrophysiological recordings and Ca2+ imaging confirm that these waves rapidly, reversibly, and non-thermally increase calcium influx of CaV1.2 in HEK293 cells and induce acute Ca2+ signals in neurons. Furthermore, this irradiation upregulates critical CaV1 signals, including CREB phosphorylation and c-Fos expression, in vitro and in vivo, without raising significant biosafety risks. This DNA-free, non-invasive approach offers a promising approach for modulating CaV gating and Ca2+ signaling and treating diseases characterized by deficits in CaV functions.
Collapse
Affiliation(s)
- Yuankun Sun
- School of Electronic Science and Engineering, University of Electronic Science and Technology of China, Chengdu, 611731, P. R. China
| | - Jinli Geng
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University Beijing, Beijing, 100191, P. R. China
| | - Yu Fan
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University Beijing, Beijing, 100191, P. R. China
| | - Yangmei Li
- Innovation Laboratory of Terahertz Biophysics, National Innovation Institute of Defense Technology, Beijing, 100072, P. R. China
| | - Yuan Zhong
- Innovation Laboratory of Terahertz Biophysics, National Innovation Institute of Defense Technology, Beijing, 100072, P. R. China
| | - Jing Cai
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University Beijing, Beijing, 100191, P. R. China
| | - Xiaodong Liu
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University Beijing, Beijing, 100191, P. R. China
| | - Shaomeng Wang
- School of Electronic Science and Engineering, University of Electronic Science and Technology of China, Chengdu, 611731, P. R. China
| | - Yubin Gong
- School of Electronic Science and Engineering, University of Electronic Science and Technology of China, Chengdu, 611731, P. R. China
| | - Chao Chang
- Innovation Laboratory of Terahertz Biophysics, National Innovation Institute of Defense Technology, Beijing, 100072, P. R. China
- School of Physics, Peking University, Beijing, 100871, P. R. China
| | - Yaxiong Yang
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University Beijing, Beijing, 100191, P. R. China
| | - Chunhai Fan
- School of Chemistry and Chemical Engineering, Institute of Molecular Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200240, P. R. China
| |
Collapse
|
5
|
Timothy KW, Bauer R, Larkin KA, Walsh EP, Abrams DJ, Gonzalez Corcia C, Valsamakis A, Pitt GS, Dick IE, Golden A. A Natural History Study of Timothy Syndrome. Orphanet J Rare Dis 2024; 19:433. [PMID: 39580446 PMCID: PMC11585941 DOI: 10.1186/s13023-024-03445-x] [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: 05/15/2024] [Accepted: 11/11/2024] [Indexed: 11/25/2024] Open
Abstract
BACKGROUND Timothy syndrome (OMIM #601005) is a rare disease caused by variants in the gene CACNA1C. Initially, Timothy syndrome was characterized by a cardiac presentation of long QT syndrome and syndactyly of the fingers and/or toes, all associated with the CACNA1C variant, Gly406Arg. However, subsequent identification of diverse variants in CACNA1C has expanded the clinical spectrum, revealing various cardiac and extra-cardiac manifestations. It remains underexplored whether individuals with the canonical Gly406Arg variants in mutually exclusive exon 8A (Timothy syndrome 1) or exon 8 (Timothy syndrome 2) exhibit overlapping symptoms. Moreover, case reports have indicated that some CACNA1C variants may produce a cardiac-selective form of Timothy syndrome often referred to as non-syndromic long QT type 8 or cardiac-only Timothy syndrome, however few reports follow up on these patients to confirm the cardiac selectivity of the phenotype over time. METHODS A survey was administered to the parents of patients with Timothy syndrome, querying a broad range of symptoms and clinical features. Study participants were organized into 5 separate categories based on genotype and initial diagnosis, enabling comparison between groups of patients which have been described differentially in the literature. RESULTS Our findings reveal that Timothy syndrome patients commonly exhibit both cardiac and extra-cardiac features, with long QT syndrome, neurodevelopmental impairments, hypoglycemia, and respiratory issues being frequently reported. Notably, the incidence of these features was similar across all patient categories, including those diagnosed with non-syndromic long QT type 8, suggesting that the 'non-syndromic' classification may be incomplete. CONCLUSIONS This study represents the first Natural History Study of Timothy syndrome, offering a comprehensive overview of the disease's clinical manifestations. We demonstrate that both cardiac and extra-cardiac features are prevalent across all patient groups, underscoring the syndromic nature of CACNA1C variants. While the critical role of long QT syndrome and cardiac arrhythmias in Timothy syndrome has been well recognized, our findings indicate that hypoglycemia and respiratory dysfunction also pose significant life-threatening risks, emphasizing the need for comprehensive therapeutic management of affected individuals.
Collapse
Affiliation(s)
- Katherine W Timothy
- The Timothy Syndrome Foundation, Charitable Organization, Brigham City, UT, USA
| | - Rosemary Bauer
- Laboratory of Biochemistry and Genetics, National Institute of Diabetes, Digestive, and Kidney Diseases, National Institute of Health, Bethesda, MD, USA
- Division of Endocrinology, Metabolism, and Molecular Medicine, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Kerry A Larkin
- Laboratory of Biochemistry and Genetics, National Institute of Diabetes, Digestive, and Kidney Diseases, National Institute of Health, Bethesda, MD, USA
- Department of Cell Biology, Yale School of Medicine, 295 Congress Ave, New Haven, CT, USA
| | - Edward P Walsh
- Department of Cardiology, Harvard Medical School, Boston Children's Hospital, Boston, MA, USA
| | - Dominic J Abrams
- Department of Cardiology, Harvard Medical School, Boston Children's Hospital, Boston, MA, USA
| | | | - Alexandra Valsamakis
- Clinical Development and Medical Affairs, Roche Diagnostics Solutions, Pleasanton, CA, USA
| | - Geoffrey S Pitt
- Cardiovascular Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Ivy E Dick
- Department of Physiology, School of Medicine, University of Maryland, Baltimore, MD, USA.
| | - Andy Golden
- Laboratory of Biochemistry and Genetics, National Institute of Diabetes, Digestive, and Kidney Diseases, National Institute of Health, Bethesda, MD, USA
| |
Collapse
|
6
|
Wang S, Qin M, Fan X, Jiang C, Hou Q, Ye Z, Zhang X, Yang Y, Xiao J, Wallace K, Rastegar-Kashkooli Y, Peng Q, Jin D, Wang J, Wang M, Ding R, Tao J, Kim YT, Bhawal UK, Wang J, Chen X, Wang J. The role of metal ions in stroke: Current evidence and future perspectives. Ageing Res Rev 2024; 101:102498. [PMID: 39243890 DOI: 10.1016/j.arr.2024.102498] [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: 03/18/2024] [Revised: 07/24/2024] [Accepted: 09/04/2024] [Indexed: 09/09/2024]
Abstract
Metal ions play a pivotal role in maintaining optimal brain function within the human body. Nevertheless, the accumulation of these ions can result in irregularities that lead to brain damage and dysfunction. Disruptions of metal ion homeostasis can result in various pathologies, including inflammation, redox dysregulation, and blood-brain barrier disruption. While research on metal ions has chiefly focused on neurodegenerative diseases, little attention has been given to their involvement in the onset and progression of stroke. Recent studies have identified cuproptosis and confirmed ferroptosis as significant factors in stroke pathology, underscoring the importance of metal ions in stroke pathology, including abnormal ion transport, neurotoxicity, blood-brain barrier damage, and cell death. Additionally, it provides an overview of contemporary metal ion chelators and detection techniques, which may offer novel approaches to stroke treatment.
Collapse
Affiliation(s)
- Shaoshuai Wang
- Department of Pain Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450000, China; Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China; Non-commissioned Officer School of Army Medical University, Shijiazhuang, Hebei 050000, China
| | - Mengzhe Qin
- Department of Pain Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450000, China
| | - Xiaochong Fan
- Department of Pain Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450000, China
| | - Chao Jiang
- Department of Neurology, People's Hospital of Zhengzhou University, Zhengzhou, Henan 450000, China
| | - Qingchuan Hou
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Ziyi Ye
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Xinru Zhang
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Yunfan Yang
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Jingyu Xiao
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Kevin Wallace
- College of Mathematical and Natural Sciences, University of Maryland, College Park, MD 20742, USA
| | - Yousef Rastegar-Kashkooli
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China; School of International Education, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Qinfeng Peng
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Dongqi Jin
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Junyang Wang
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Menglu Wang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Ruoqi Ding
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Jin Tao
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Yun Tai Kim
- Division of Functional Food Research, Korea Food Research Institute, 245, Nongsaengmyeong-ro, Iseo-myeon, Wanju-gun, Jeollabuk-do 55365, Republic of Korea; Department of Food Biotechnology, Korea University of Science & Technology, 217 Gajeong-ro, Yuseong-gu, Daejeon 34113, Republic of Korea
| | - Ujjal K Bhawal
- Center for Global Health Research, Saveetha Dental College, Saveetha Institute of Medical and Technical Sciences, Chennai, Tamil Nadu 600077, India; Research Institute of Oral Science, Nihon University School of Dentistry at Matsudo, Chiba 271-8587, Japan
| | - Junmin Wang
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China.
| | - Xuemei Chen
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China.
| | - Jian Wang
- Department of Pain Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450000, China; Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China.
| |
Collapse
|
7
|
del Rivero Morfin PJ, Chavez DS, Jayaraman S, Yang L, Geisler SM, Kochiss AL, Tuluc P, Colecraft HM, Marx SO, Liu XS, Rajadhyaksha AM, Ben-Johny M. A genetically encoded actuator boosts L-type calcium channel function in diverse physiological settings. SCIENCE ADVANCES 2024; 10:eadq3374. [PMID: 39475605 PMCID: PMC11524184 DOI: 10.1126/sciadv.adq3374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Accepted: 09/24/2024] [Indexed: 11/02/2024]
Abstract
L-type Ca2+ channels (CaV1.2/1.3) convey influx of calcium ions that orchestrate a bevy of biological responses including muscle contraction, neuronal function, and gene transcription. Deficits in CaV1 function play a vital role in cardiac and neurodevelopmental disorders. Here, we develop a genetically encoded enhancer of CaV1.2/1.3 channels (GeeCL) to manipulate Ca2+ entry in distinct physiological settings. We functionalized a nanobody that targets the CaV complex by attaching a minimal effector domain from an endogenous CaV modulator-leucine-rich repeat containing protein 10 (Lrrc10). In cardiomyocytes, GeeCL selectively increased L-type current amplitude. In neurons in vitro and in vivo, GeeCL augmented excitation-transcription (E-T) coupling. In all, GeeCL represents a powerful strategy to boost CaV1.2/1.3 function and lays the groundwork to illuminate insights on neuronal and cardiac physiology and disease.
Collapse
Affiliation(s)
| | - Diego Scala Chavez
- Pediatric Neurology, Department of Pediatrics, Weill Cornell Medicine, New York, NY, USA
| | - Srinidhi Jayaraman
- Department of Physiology and Cellular Biophysics, Columbia University, New York, NY, USA
| | - Lin Yang
- Division of Cardiology, Department of Medicine, Columbia University, Vagelos College of Physicians and Surgeons, New York, NY, USA
| | - Stefanie M. Geisler
- Department of Pharmacology and Toxicology, University of Innsbruck, Innsbruck, Tyrol, Austria
| | - Audrey L. Kochiss
- Department of Physiology and Cellular Biophysics, Columbia University, New York, NY, USA
| | - Petronel Tuluc
- Department of Pharmacology and Toxicology, University of Innsbruck, Innsbruck, Tyrol, Austria
| | - Henry M. Colecraft
- Department of Physiology and Cellular Biophysics, Columbia University, New York, NY, USA
- Department of Molecular Pharmacology and Therapeutics, Columbia University, New York, NY, USA
| | - Steven O. Marx
- Division of Cardiology, Department of Medicine, Columbia University, Vagelos College of Physicians and Surgeons, New York, NY, USA
- Department of Molecular Pharmacology and Therapeutics, Columbia University, New York, NY, USA
| | - X. Shawn Liu
- Department of Physiology and Cellular Biophysics, Columbia University, New York, NY, USA
- Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, NY, USA
| | - Anjali M. Rajadhyaksha
- Pediatric Neurology, Department of Pediatrics, Weill Cornell Medicine, New York, NY, USA
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
- Weill Cornell Autism Research Program, Weill Cornell Medicine, New York, NY, USA
| | - Manu Ben-Johny
- Department of Physiology and Cellular Biophysics, Columbia University, New York, NY, USA
| |
Collapse
|
8
|
Matsui M, Lynch LE, Distefano I, Galante A, Gade AR, Wang HG, Gómez-Banoy N, Towers P, Sinden DS, Wei EQ, Barnett AS, Johnson K, Lima R, Rubio-Navarro A, Li AK, Marx SO, McGraw TE, Thornton PS, Timothy KW, Lo JC, Pitt GS. Multiple beta cell-independent mechanisms drive hypoglycemia in Timothy syndrome. Nat Commun 2024; 15:8980. [PMID: 39420001 PMCID: PMC11487186 DOI: 10.1038/s41467-024-52885-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Accepted: 09/25/2024] [Indexed: 10/19/2024] Open
Abstract
The canonical G406R mutation that increases Ca2+ influx through the CACNA1C-encoded CaV1.2 Ca2+ channel underlies the multisystem disorder Timothy syndrome (TS), characterized by life-threatening arrhythmias. Severe episodic hypoglycemia is among the poorly characterized non-cardiac TS pathologies. While hypothesized from increased Ca2+ influx in pancreatic beta cells and consequent hyperinsulinism, this hypoglycemia mechanism is undemonstrated because of limited clinical data and lack of animal models. We generated a CaV1.2 G406R knockin mouse model that recapitulates key TS features, including hypoglycemia. Unexpectedly, these mice do not show hyperactive beta cells or hyperinsulinism in the setting of normal intrinsic beta cell function, suggesting dysregulated glucose homeostasis. Patient data confirm the absence of hyperinsulinism. We discover multiple alternative contributors, including perturbed counterregulatory hormone responses with defects in glucagon secretion and abnormal hypothalamic control of glucose homeostasis. These data provide new insights into contributions of CaV1.2 channels and reveal integrated consequences of the mutant channels driving life-threatening events in TS.
Collapse
Affiliation(s)
- Maiko Matsui
- Cardiovascular Research Institute, Weill Cornell Medicine, 413 E. 69th St., New York, NY, 10021, USA
| | - Lauren E Lynch
- Cardiovascular Research Institute, Weill Cornell Medicine, 413 E. 69th St., New York, NY, 10021, USA
| | - Isabella Distefano
- Cardiovascular Research Institute, Weill Cornell Medicine, 413 E. 69th St., New York, NY, 10021, USA
| | - Allison Galante
- Cardiovascular Research Institute, Weill Cornell Medicine, 413 E. 69th St., New York, NY, 10021, USA
| | - Aravind R Gade
- Cardiovascular Research Institute, Weill Cornell Medicine, 413 E. 69th St., New York, NY, 10021, USA
| | - Hong-Gang Wang
- Cardiovascular Research Institute, Weill Cornell Medicine, 413 E. 69th St., New York, NY, 10021, USA
| | - Nicolas Gómez-Banoy
- Cardiovascular Research Institute, Weill Cornell Medicine, 413 E. 69th St., New York, NY, 10021, USA
- Weill Center for Metabolic Health, Weill Cornell Medicine, 413 E. 69th St., New York, NY, 10021, USA
| | - Patrick Towers
- Cardiovascular Research Institute, Weill Cornell Medicine, 413 E. 69th St., New York, NY, 10021, USA
| | - Daniel S Sinden
- Cardiovascular Research Institute, Weill Cornell Medicine, 413 E. 69th St., New York, NY, 10021, USA
| | - Eric Q Wei
- Department of Medicine, MSRB II, 2 Genome Ct, Duke University Medical Center, Durham, NC, 27710, USA
| | - Adam S Barnett
- Department of Medicine, MSRB II, 2 Genome Ct, Duke University Medical Center, Durham, NC, 27710, USA
| | - Kenneth Johnson
- Cardiovascular Research Institute, Weill Cornell Medicine, 413 E. 69th St., New York, NY, 10021, USA
| | - Renan Lima
- Weill Center for Metabolic Health, Weill Cornell Medicine, 413 E. 69th St., New York, NY, 10021, USA
| | - Alfonso Rubio-Navarro
- Cardiovascular Research Institute, Weill Cornell Medicine, 413 E. 69th St., New York, NY, 10021, USA
- Weill Center for Metabolic Health, Weill Cornell Medicine, 413 E. 69th St., New York, NY, 10021, USA
| | - Ang K Li
- Cardiovascular Research Institute, Weill Cornell Medicine, 413 E. 69th St., New York, NY, 10021, USA
- Weill Center for Metabolic Health, Weill Cornell Medicine, 413 E. 69th St., New York, NY, 10021, USA
| | - Steven O Marx
- Division of Cardiology, Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University, 622 W 168th St, PH-3 Center, New York, NY, USA
- Department of Pharmacology, Vagelos College of Physicians and Surgeons, Columbia University, 622 W 168th St, PH-3 Center, New York, NY, USA
| | - Timothy E McGraw
- Department of Biochemistry, Weill Cornell Medical College, 1300 York Ave, New York, NY, 10065, USA
| | - Paul S Thornton
- Division of Endocrinology and Diabetes, Cook Children's Medical Center, 801 7th Ave, Fort Worth, TX, 76104, USA
| | - Katherine W Timothy
- Children's Hospital Boston, Harvard Medical School, 300 Longwood Ave., Boston, MA, 02115, USA
| | - James C Lo
- Cardiovascular Research Institute, Weill Cornell Medicine, 413 E. 69th St., New York, NY, 10021, USA
- Weill Center for Metabolic Health, Weill Cornell Medicine, 413 E. 69th St., New York, NY, 10021, USA
- Division of Cardiology, Department of Medicine, Weill Cornell Medicine, 413 E. 69th St, New York, NY, 10021, USA
| | - Geoffrey S Pitt
- Cardiovascular Research Institute, Weill Cornell Medicine, 413 E. 69th St., New York, NY, 10021, USA.
| |
Collapse
|
9
|
Chen Q, Pan Y, Hu Y, Chen G, Chen X, Xie Y, Wang M, Li Z, Huang J, Shi Y, Huang H, Zhang T, Wang M, Zeng P, Wang S, Chen R, Zheng Y, Zhong L, Yang H, Liang D. An L-type calcium channel blocker nimodipine exerts anti-fibrotic effects by attenuating TGF-β1 induced calcium response in an in vitro model of thyroid eye disease. EYE AND VISION (LONDON, ENGLAND) 2024; 11:37. [PMID: 39237996 PMCID: PMC11378575 DOI: 10.1186/s40662-024-00401-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Accepted: 08/02/2024] [Indexed: 09/07/2024]
Abstract
BACKGROUND Thyroid eye disease (TED) is a vision-threatening autoimmune disorder. Orbital tissue fibrosis leading to intractable complications remains a troublesome issue in TED management. Exploration of novel therapeutic targets and agents to ameliorate tissue fibrosis is crucial for TED. Recent work suggests that Ca2+ signaling participates in tissue fibrosis. However, whether an alteration of Ca2+ signaling has a role in fibrogenesis during TED remains unclear. In this study, we aimed to investigate the role of Ca2+ signaling in the fibrogenesis process during TED and the potential therapeutic effects of a highly selective inhibitor of the L-type calcium channel (LTCC), nimodipine, through a TGF-β1 induced in vitro TED model. METHODS Primary culture of orbital fibroblasts (OFs) were established from orbital adipose connective tissues of patients with TED and healthy control donors. Real-time quantitative polymerase chain reaction (RT-qPCR) and RNA sequencing were used to assess the genes expression associated with LTCC in OFs. Flow cytometry, RT-qPCR, 5-ethynyl-2'-deoxyuridine (EdU) proliferation assay, wound healing assay and Western blot (WB) were used to assess the intracellular Ca2+ response on TGF-β1 stimulation, and to evaluate the potential therapeutic effects of nimodipine in the TGF-β1 induced in vitro TED model. The roles of Ca2+/calmodulin-dependent protein kinase II (CaMKII) and signal transducer and activator of transcription 1 (STAT1) in fibrogenesis during TED were determined by immunohistochemistry, WB, flow cytometry and co-immunoprecipitation assay. Selective inhibitors were used to explore the downstream signaling pathways. RESULTS LTCC inhibitor nimodipine blocked the TGF-β1 induced intracellular Ca2+ response and further reduced the expression of alpha-smooth muscle actin (α-SMA), collagen type I alpha 1 (Col1A1) and collagen type I alpha 2 (Col1A2) in OFs. Besides, nimodipine inhibited cell proliferation and migration of OFs. Moreover, our results provided evidence that activation of the CaMKII/STAT1 signaling pathway was involved in fibrogenesis during TED, and nimodipine inhibited the pro-fibrotic functions of OFs by down-regulating the CaMKII/STAT1 signaling pathway. CONCLUSIONS TGF-β1 induces an LTCC-mediated Ca2+ response, followed by activation of CaMKII/STAT1 signaling pathway, which promotes the pro-fibrotic functions of OFs and participates in fibrogenesis during TED. Nimodipine exerts potent anti-fibrotic benefits in vitro by suppressing the CaMKII/STAT1 signaling pathway. Our work deepens our understanding of the fibrogenesis process during TED and provides potential therapeutic targets and alternative candidate for TED.
Collapse
Affiliation(s)
- Qian Chen
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology Visual Science, Guangzhou, 510060, China
- Department of Ophthalmology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510630, China
| | - Yuan Pan
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology Visual Science, Guangzhou, 510060, China
| | - Yunwei Hu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology Visual Science, Guangzhou, 510060, China
- Ophthalmic Center, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
| | - Guanyu Chen
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology Visual Science, Guangzhou, 510060, China
| | - Xiaoqing Chen
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology Visual Science, Guangzhou, 510060, China
| | - Yanyan Xie
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology Visual Science, Guangzhou, 510060, China
| | - Minzhen Wang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology Visual Science, Guangzhou, 510060, China
| | - Zhuang Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology Visual Science, Guangzhou, 510060, China
| | - Jun Huang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology Visual Science, Guangzhou, 510060, China
- Ophthalmic Center, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
| | - Yuxun Shi
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology Visual Science, Guangzhou, 510060, China
| | - Haixiang Huang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology Visual Science, Guangzhou, 510060, China
| | - Te Zhang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology Visual Science, Guangzhou, 510060, China
| | - Mei Wang
- Department of Ophthalmology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China
| | - Peng Zeng
- Department of Ophthalmology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China
| | - Sha Wang
- Eye Center of Xiangya Hospital, Central South University, Hunan Key Laboratory of Ophthalmology, Changsha, 410008, China
| | - Rongxin Chen
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology Visual Science, Guangzhou, 510060, China
| | - Yongxin Zheng
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology Visual Science, Guangzhou, 510060, China
| | - Liuxueying Zhong
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology Visual Science, Guangzhou, 510060, China
| | - Huasheng Yang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology Visual Science, Guangzhou, 510060, China
| | - Dan Liang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology Visual Science, Guangzhou, 510060, China.
| |
Collapse
|
10
|
Li T, Liu M, Yu F, Yang S, Bu W, Liu K, Yang J, Ni H, Yang M, Yin H, Hong R, Li D, Zhao H, Zhou J. Pathologically relevant aldoses and environmental aldehydes cause cilium disassembly via formyl group-mediated mechanisms. J Mol Cell Biol 2024; 16:mjad079. [PMID: 38059869 PMCID: PMC11245732 DOI: 10.1093/jmcb/mjad079] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 10/23/2023] [Accepted: 12/05/2023] [Indexed: 12/08/2023] Open
Abstract
Carbohydrate metabolism disorders (CMDs), such as diabetes, galactosemia, and mannosidosis, cause ciliopathy-like multiorgan defects. However, the mechanistic link of cilia to CMD complications is still poorly understood. Herein, we describe significant cilium disassembly upon treatment of cells with pathologically relevant aldoses rather than the corresponding sugar alcohols. Moreover, environmental aldehydes are able to trigger cilium disassembly by the steric hindrance effect of their formyl groups. Mechanistic studies reveal that aldehydes stimulate extracellular calcium influx across the plasma membrane, which subsequently activates the calmodulin-Aurora A-histone deacetylase 6 pathway to deacetylate axonemal microtubules and triggers cilium disassembly. In vivo experiments further show that Hdac6 knockout mice are resistant to aldehyde-induced disassembly of tracheal cilia and sperm flagella. These findings reveal a previously unrecognized role for formyl group-mediated cilium disassembly in the complications of CMDs.
Collapse
Affiliation(s)
- Te Li
- Haihe Laboratory of Cell Ecosystem, State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Science, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Min Liu
- Laboratory of Tissue Homeostasis, Haihe Laboratory of Cell Ecosystem, Tianjin 300462, China
| | - Fan Yu
- Haihe Laboratory of Cell Ecosystem, State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Science, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Song Yang
- Haihe Laboratory of Cell Ecosystem, State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Science, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Weiwen Bu
- Haihe Laboratory of Cell Ecosystem, State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Science, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Kai Liu
- Haihe Laboratory of Cell Ecosystem, State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Science, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Jia Yang
- Haihe Laboratory of Cell Ecosystem, State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Science, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Hua Ni
- Haihe Laboratory of Cell Ecosystem, State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Science, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Mulin Yang
- Haihe Laboratory of Cell Ecosystem, State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Science, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Hanxiao Yin
- Haihe Laboratory of Cell Ecosystem, State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Science, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Renjie Hong
- Haihe Laboratory of Cell Ecosystem, State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Science, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Dengwen Li
- Haihe Laboratory of Cell Ecosystem, State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Science, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Huijie Zhao
- Center for Cell Structure and Function, Collaborative Innovation Center of Cell Biology in Universities of Shandong, Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Sciences, Shandong Normal University, Jinan 250014, China
| | - Jun Zhou
- Haihe Laboratory of Cell Ecosystem, State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Science, College of Life Sciences, Nankai University, Tianjin 300071, China
- Center for Cell Structure and Function, Collaborative Innovation Center of Cell Biology in Universities of Shandong, Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Sciences, Shandong Normal University, Jinan 250014, China
| |
Collapse
|
11
|
Schwenzer N, Teiwes NK, Kohl T, Pohl C, Giller MJ, Lehnart SE, Steinem C. Ca V1.3 channel clusters characterized by live-cell and isolated plasma membrane nanoscopy. Commun Biol 2024; 7:620. [PMID: 38783117 PMCID: PMC11116533 DOI: 10.1038/s42003-024-06313-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Accepted: 05/08/2024] [Indexed: 05/25/2024] Open
Abstract
A key player of excitable cells in the heart and brain is the L-type calcium channel CaV1.3. In the heart, it is required for voltage-dependent Ca2+-signaling, i.e., for controlling and modulating atrial cardiomyocyte excitation-contraction coupling. The clustering of CaV1.3 in functionally relevant channel multimers has not been addressed due to a lack of stoichiometric labeling combined with high-resolution imaging. Here, we developed a HaloTag-labeling strategy to visualize and quantify CaV1.3 clusters using STED nanoscopy to address the questions of cluster size and intra-cluster channel density. Channel clusters were identified in the plasma membrane of transfected live HEK293 cells as well as in giant plasma membrane vesicles derived from these cells that were spread on modified glass support to obtain supported plasma membrane bilayers (SPMBs). A small fraction of the channel clusters was colocalized with early and recycling endosomes at the membranes. STED nanoscopy in conjunction with live-cell and SPMB imaging enabled us to quantify CaV1.3 cluster sizes and their molecular density revealing significantly lower channel densities than expected for dense channel packing. CaV1.3 channel cluster size and molecular density were increased in SPMBs after treatment of the cells with the sympathomimetic compound isoprenaline, suggesting a regulated channel cluster condensation mechanism.
Collapse
Affiliation(s)
- Niko Schwenzer
- Department of Cardiology and Pneumology, University Medical Center Göttingen, Robert-Koch-Str. 40, 37075, Göttingen, Germany
- Cellular Biophysics and Translational Cardiology Section, Heart Research Center Göttingen, University Medical Center Göttingen, Robert‑Koch‑Str. 42a, 37075, Göttingen, Germany
- Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC 2067), University of Göttingen, 37073, Göttingen, Germany
| | - Nikolas K Teiwes
- Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC 2067), University of Göttingen, 37073, Göttingen, Germany
- Georg-August Universität, Institut für Organische und Biomolekulare Chemie, Tammannstr. 2, 37077, Göttingen, Germany
| | - Tobias Kohl
- Department of Cardiology and Pneumology, University Medical Center Göttingen, Robert-Koch-Str. 40, 37075, Göttingen, Germany
- Cellular Biophysics and Translational Cardiology Section, Heart Research Center Göttingen, University Medical Center Göttingen, Robert‑Koch‑Str. 42a, 37075, Göttingen, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Göttingen, Robert-Koch-Str. 40, 37075, Göttingen, Germany
| | - Celine Pohl
- Georg-August Universität, Institut für Organische und Biomolekulare Chemie, Tammannstr. 2, 37077, Göttingen, Germany
| | - Michelle J Giller
- Georg-August Universität, Institut für Organische und Biomolekulare Chemie, Tammannstr. 2, 37077, Göttingen, Germany
| | - Stephan E Lehnart
- Department of Cardiology and Pneumology, University Medical Center Göttingen, Robert-Koch-Str. 40, 37075, Göttingen, Germany.
- Cellular Biophysics and Translational Cardiology Section, Heart Research Center Göttingen, University Medical Center Göttingen, Robert‑Koch‑Str. 42a, 37075, Göttingen, Germany.
- Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC 2067), University of Göttingen, 37073, Göttingen, Germany.
- DZHK (German Centre for Cardiovascular Research), partner site Göttingen, Robert-Koch-Str. 40, 37075, Göttingen, Germany.
- Collaborative Research Center SFB 1190 "Compartmental Gates and Contact Sites in Cells", University of Göttingen, Humboldtallee 23, 37073, Göttingen, Germany.
| | - Claudia Steinem
- Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC 2067), University of Göttingen, 37073, Göttingen, Germany.
- Georg-August Universität, Institut für Organische und Biomolekulare Chemie, Tammannstr. 2, 37077, Göttingen, Germany.
- Max-Planck-Institut für Dynamik und Selbstorganisation, Am Fassberg 17, 37077, Göttingen, Germany.
| |
Collapse
|
12
|
Dates J, Kolosov D. Voltage-gated ion channels as novel regulators of epithelial ion transport in the osmoregulatory organs of insects. FRONTIERS IN INSECT SCIENCE 2024; 4:1385895. [PMID: 38835480 PMCID: PMC11148248 DOI: 10.3389/finsc.2024.1385895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Accepted: 05/01/2024] [Indexed: 06/06/2024]
Abstract
Voltage-gated ion channels (VGICs) respond to changes in membrane potential (Vm) and typically exhibit fast kinetic properties. They play an important role in signal detection and propagation in excitable tissues. In contrast, the role of VGICs in non-excitable tissues like epithelia is less studied and less clear. Studies in epithelia of vertebrates and invertebrates demonstrate wide expression of VGICs in epithelia of animals. Recently, VGICs have emerged as regulators of ion transport in the Malpighian tubules (MTs) and other osmoregulatory organs of insects. This mini-review aims to concisely summarize which VGICs have been implicated in the regulation of ion transport in the osmoregulatory epithelia of insects to date, and highlight select groups for further study. We have also speculated on the roles VGICs may potentially play in regulating processes connected directly to ion transport in insects (e.g., acid-base balance, desiccation, thermal tolerance). This review is not meant to be exhaustive but should rather serve as a thought-provoking collection of select existing highlights on VGICs, and to emphasize how understudied this mechanism of ion transport regulation is in insect epithelia.
Collapse
Affiliation(s)
- Jocelyne Dates
- Department of Biological Sciences, California State University San Marcos, San Marcos, CA, United States
| | - Dennis Kolosov
- Department of Biological Sciences, California State University San Marcos, San Marcos, CA, United States
| |
Collapse
|
13
|
Rühl P, Nair AG, Gawande N, Dehiwalage SNCW, Münster L, Schönherr R, Heinemann SH. An Ultrasensitive Genetically Encoded Voltage Indicator Uncovers the Electrical Activity of Non-Excitable Cells. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2307938. [PMID: 38526185 PMCID: PMC11132041 DOI: 10.1002/advs.202307938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 02/10/2024] [Indexed: 03/26/2024]
Abstract
Most animal cell types are classified as non-excitable because they do not generate action potentials observed in excitable cells, such as neurons and muscle cells. Thus, resolving voltage signals in non-excitable cells demands sensors with exceptionally high voltage sensitivity. In this study, the ultrabright, ultrasensitive, and calibratable genetically encoded voltage sensor rEstus is developed using structure-guided engineering. rEstus is most sensitive in the resting voltage range of non-excitable cells and offers a 3.6-fold improvement in brightness change for fast voltage spikes over its precursor ASAP3. Using rEstus, it is uncovered that the membrane voltage in several non-excitable cell lines (A375, HEK293T, MCF7) undergoes spontaneous endogenous alterations on a second to millisecond timescale. Correlation analysis of these optically recorded voltage alterations provides a direct, real-time readout of electrical cell-cell coupling, showing that visually connected A375 and HEK293T cells are also largely electrically connected, while MCF7 cells are only weakly coupled. The presented work provides enhanced tools and methods for non-invasive voltage imaging in living cells and demonstrates that spontaneous endogenous membrane voltage alterations are not limited to excitable cells but also occur in a variety of non-excitable cell types.
Collapse
Affiliation(s)
- Philipp Rühl
- Center for Molecular Biomedicine, Department of BiophysicsFriedrich Schiller University Jena and Jena University HospitalD‐07745JenaGermany
| | - Anagha G. Nair
- Center for Molecular Biomedicine, Department of BiophysicsFriedrich Schiller University Jena and Jena University HospitalD‐07745JenaGermany
| | - Namrata Gawande
- Center for Molecular Biomedicine, Department of BiophysicsFriedrich Schiller University Jena and Jena University HospitalD‐07745JenaGermany
| | - Sassrika N. C. W. Dehiwalage
- Center for Molecular Biomedicine, Department of BiophysicsFriedrich Schiller University Jena and Jena University HospitalD‐07745JenaGermany
| | - Lukas Münster
- Center for Molecular Biomedicine, Department of BiophysicsFriedrich Schiller University Jena and Jena University HospitalD‐07745JenaGermany
| | - Roland Schönherr
- Center for Molecular Biomedicine, Department of BiophysicsFriedrich Schiller University Jena and Jena University HospitalD‐07745JenaGermany
| | - Stefan H. Heinemann
- Center for Molecular Biomedicine, Department of BiophysicsFriedrich Schiller University Jena and Jena University HospitalD‐07745JenaGermany
| |
Collapse
|
14
|
Hashimoto H, Mandai S, Shikuma S, Kimura M, Toma H, Sakaguchi Y, Shiraishi S, Toshima N, Hoshino M, Kimura M, Ota J, Horiuchi S, Adachi S, Uchida S. The Effect of Antihypertensive Therapy on Skeletal Muscle Mass and Bone Mineral Density in Patients With End-Stage Kidney Disease. J Ren Nutr 2024; 34:223-234. [PMID: 37918643 DOI: 10.1053/j.jrn.2023.10.008] [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: 01/24/2023] [Revised: 09/12/2023] [Accepted: 10/18/2023] [Indexed: 11/04/2023] Open
Abstract
OBJECTIVE Sarcopenia and osteoporosis substantially influence health and lifespan. However, the variables affecting skeletal muscle mass (SMM) or bone mineral density (BMD) remain unknown. DESIGN AND METHODS From August 1, 2018 to July 31, 2019, we conducted a single-center, observational cohort study with 291 Japanese adult patients on maintenance hemodialysis due to end-stage kidney disease, who had their femoral neck BMD measured using dual-energy X-ray absorptiometry. After 1-year follow-up, we measured annual changes of BMD (ΔBMD) and SMM (ΔSMM), which were calculated through a modified creatinine index (mg/kg/day) using age, sex, serum creatinine, and single-pooled Kt/V for urea. The factors associated with ΔSMM/ΔBMD or progressive loss of SMM/BMD, defined as ΔSMM/ΔBMD < 0 per year, respectively, were analyzed with multivariable, linear regression or logistic regression models. RESULTS The median age of the patients was 66 years and 33% were female. Dialysis vintage and β-blocker-use were inversely correlated to ΔSMM. In comparison to nonusers, β-blockers users had 2.5-fold higher SMM loss odd ratios [95% confidence interval, 1.3-4.8]. The risk for SMM loss caused by β-blockers was not increased in users of renin-angiotensin system inhibitors. The ΔBMD was negatively correlated to the usage of calcium channel blockers. The risk of developing osteosarcopenia, which was defined as annual loss of both SMM and BMD, increased in calcium channel blockers users. CONCLUSIONS The use of β-blockers is associated with an elevated risk of developing sarcopenia, whereas renin-angiotensin system inhibitors may minimize this effect in patients with end-stage kidney disease. Use of calcium channel blocker therapy was associated with a faster decline of BMD.
Collapse
Affiliation(s)
- Hiroko Hashimoto
- Department of Nephrology, Shuuwa General Hospital, Kasukabe, Saitama, Japan; Department of Nephrology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Bunkyo, Tokyo, Japan
| | - Shintaro Mandai
- Department of Nephrology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Bunkyo, Tokyo, Japan.
| | - Satomi Shikuma
- Department of Nephrology, Shuuwa General Hospital, Kasukabe, Saitama, Japan
| | - Mai Kimura
- Department of Nephrology, Shuuwa General Hospital, Kasukabe, Saitama, Japan
| | - Hayato Toma
- Department of Nephrology, Shuuwa General Hospital, Kasukabe, Saitama, Japan
| | - Yuki Sakaguchi
- Department of Nephrology, Shuuwa General Hospital, Kasukabe, Saitama, Japan
| | - Sayuka Shiraishi
- Department of Nephrology, Shuuwa General Hospital, Kasukabe, Saitama, Japan
| | - Noriyuki Toshima
- Department of Nephrology, Shuuwa General Hospital, Kasukabe, Saitama, Japan
| | - Motoki Hoshino
- Department of Nephrology, Shuuwa General Hospital, Kasukabe, Saitama, Japan
| | - Moe Kimura
- Department of Nephrology, Shuuwa General Hospital, Kasukabe, Saitama, Japan
| | - Jun Ota
- Department of Nephrology, Shuuwa General Hospital, Kasukabe, Saitama, Japan
| | - Susumu Horiuchi
- Department of Urology, Shuuwa General Hospital, Kasukabe, Saitama, Japan
| | - Susumu Adachi
- Department of Cardiology, Shuuwa General Hospital, Kasukabe, Saitama, Japan
| | - Shinichi Uchida
- Department of Nephrology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Bunkyo, Tokyo, Japan
| |
Collapse
|
15
|
Cipriano L, Piscopo R, Aiello C, Novelli A, Iolascon A, Piscopo C. Expanding the Phenotype of the CACNA1C-Associated Neurological Disorders in Children: Systematic Literature Review and Description of a Novel Mutation. CHILDREN (BASEL, SWITZERLAND) 2024; 11:541. [PMID: 38790536 PMCID: PMC11119747 DOI: 10.3390/children11050541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Revised: 04/23/2024] [Accepted: 04/29/2024] [Indexed: 05/26/2024]
Abstract
Background: CACNA1C gene encodes the alpha 1 subunit of the CaV1.2 L-type Ca2+ channel. Pathogenic variants in this gene have been associated with cardiac rhythm disorders such as long QT syndrome, Brugada syndrome and Timothy syndrome. Recent evidence has suggested the possible association between CACNA1C mutations and neurologically-isolated (in absence of cardiac involvement) phenotypes in children, giving birth to a wider spectrum of CACNA1C-related clinical presentations. However, to date, little is known about the variety of both neurological and non-neurological signs/symptoms in the neurologically-predominant phenotypes. Methods and Results: We conducted a systematic review of neurologically-predominant presentations without cardiac conduction defects, associated with CACNA1C mutations. We also reported a novel de novo missense pathogenic variant in the CACNA1C gene of a children patient presenting with constructional, dressing and oro-buccal apraxia associated with behavioral abnormalities, mild intellectual disability, dental anomalies, gingival hyperplasia and mild musculoskeletal defects, without cardiac conduction defects. Conclusions: The present study highlights the importance of considering the investigation of the CACNA1C gene in children's neurological isolated syndromes, and expands the phenotype of the CACNA1C related conditions. In addition, the present study highlights that, even in absence of cardiac conduction defects, nuanced clinical manifestations of the Timothy syndrome (e.g., dental and gingival defects) could be found. These findings suggest the high variable expressivity of the CACNA1C gene and remark that the absence of cardiac involvement should not mislead the diagnosis of a CACNA1C related disorder.
Collapse
Affiliation(s)
- Lorenzo Cipriano
- Department of Molecular Medicine and Medical Biotechnology, University Federico II, 80131 Naples, Italy; (L.C.); (A.I.)
| | - Raffaele Piscopo
- Department of Neuroscience, Reproductive and Odontostomatological Sciences, University Federico II, 80131 Naples, Italy;
| | - Chiara Aiello
- Laboratory of Medical Genetics, Translational Cytogenomics Research Unit, Bambino Gesù Children Hospital, IRCCS, 00146 Rome, Italy; (C.A.); (A.N.)
| | - Antonio Novelli
- Laboratory of Medical Genetics, Translational Cytogenomics Research Unit, Bambino Gesù Children Hospital, IRCCS, 00146 Rome, Italy; (C.A.); (A.N.)
| | - Achille Iolascon
- Department of Molecular Medicine and Medical Biotechnology, University Federico II, 80131 Naples, Italy; (L.C.); (A.I.)
| | - Carmelo Piscopo
- Medical and Laboratory Genetics Unit, A.O.R.N. “Antonio Cardarelli”, 80131 Naples, Italy
| |
Collapse
|
16
|
Scala M, Khan K, Beneteau C, Fox RG, von Hardenberg S, Khan A, Joubert M, Fievet L, Musquer M, Le Vaillant C, Holsclaw JK, Lim D, Berking AC, Accogli A, Giacomini T, Nobili L, Striano P, Zara F, Torella A, Nigro V, Cogné B, Salick MR, Kaykas A, Eggan K, Capra V, Bézieau S, Davis EE, Wells MF. Biallelic loss-of-function variants in CACHD1 cause a novel neurodevelopmental syndrome with facial dysmorphism and multisystem congenital abnormalities. Genet Med 2024; 26:101057. [PMID: 38158856 PMCID: PMC11910193 DOI: 10.1016/j.gim.2023.101057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2023] [Revised: 12/22/2023] [Accepted: 12/22/2023] [Indexed: 01/03/2024] Open
Abstract
PURPOSE We established the genetic etiology of a syndromic neurodevelopmental condition characterized by variable cognitive impairment, recognizable facial dysmorphism, and a constellation of extra-neurological manifestations. METHODS We performed phenotypic characterization of 6 participants from 4 unrelated families presenting with a neurodevelopmental syndrome and used exome sequencing to investigate the underlying genetic cause. To probe relevance to the neurodevelopmental phenotype and craniofacial dysmorphism, we established two- and three-dimensional human stem cell-derived neural models and generated a stable cachd1 zebrafish mutant on a transgenic cartilage reporter line. RESULTS Affected individuals showed mild cognitive impairment, dysmorphism featuring oculo-auriculo abnormalities, and developmental defects involving genitourinary and digestive tracts. Exome sequencing revealed biallelic putative loss-of-function variants in CACHD1 segregating with disease in all pedigrees. RNA sequencing in CACHD1-depleted neural progenitors revealed abnormal expression of genes with key roles in Wnt signaling, neurodevelopment, and organ morphogenesis. CACHD1 depletion in neural progenitors resulted in reduced percentages of post-mitotic neurons and enlargement of 3D neurospheres. Homozygous cachd1 mutant larvae showed mandibular patterning defects mimicking human facial dysmorphism. CONCLUSION Our findings support the role of loss-of-function variants in CACHD1 as the cause of a rare neurodevelopmental syndrome with facial dysmorphism and multisystem abnormalities.
Collapse
Affiliation(s)
- Marcello Scala
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genoa, Genoa, Italy; Pediatric Neurology and Muscular Diseases Unit, IRCCS Istituto Giannina Gaslini, University of Genoa, Genoa, Italy; Medical Genetics Unit, IRCCS Giannina Gaslini Institute, Genoa, Italy
| | - Kamal Khan
- Stanley Manne Children's Research Institute, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL
| | - Claire Beneteau
- CHU Nantes, Department of Medical Genetics, CHU Nantes, 9 quai Moncousu, Nantes, France; CHU Nantes, UF of Fœtopathology and Genetics, Nantes, France; CHU de Bordeaux, Service de Génétique Médicale, Bordeaux, France
| | - Rachel G Fox
- Department of Human Genetics, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA
| | | | - Ayaz Khan
- Stanley Manne Children's Research Institute, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL
| | - Madeleine Joubert
- CHU Nantes, UF of Fœtopathology and Genetics, Nantes, France; CHU Nantes, Department of Anatomical Pathology, Nantes, France
| | - Lorraine Fievet
- Center for Human Disease Modeling, Duke University Medical Center, Durham, NC
| | - Marie Musquer
- CHU Nantes, UF of Fœtopathology and Genetics, Nantes, France; CHU Nantes, Department of Anatomical Pathology, Nantes, France
| | | | | | - Derek Lim
- Department of Clinical Genetics, Birmingham Women's and Children's NHS Foundation Trust and Birmingham Health Partners, Birmingham, United Kingdom; Department of Medicine, University of Birmingham, Birmingham, United Kingdom
| | | | - Andrea Accogli
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genoa, Genoa, Italy
| | - Thea Giacomini
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genoa, Genoa, Italy; Child Neuropsychiatry Unit, IRCCS G. Gaslini Institute, Genoa, Italy
| | - Lino Nobili
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genoa, Genoa, Italy; Child Neuropsychiatry Unit, IRCCS G. Gaslini Institute, Genoa, Italy
| | - Pasquale Striano
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genoa, Genoa, Italy; Pediatric Neurology and Muscular Diseases Unit, IRCCS Istituto Giannina Gaslini, University of Genoa, Genoa, Italy
| | - Federico Zara
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genoa, Genoa, Italy; Medical Genetics Unit, IRCCS Giannina Gaslini Institute, Genoa, Italy
| | - Annalaura Torella
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli," Naples, Italy; Telethon Institute of Genetics and Medicine, Pozzuoli, Italy
| | - Vincenzo Nigro
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli," Naples, Italy; Telethon Institute of Genetics and Medicine, Pozzuoli, Italy
| | - Benjamin Cogné
- CHU Nantes, Department of Medical Genetics, CHU Nantes, 9 quai Moncousu, Nantes, France; Nantes Université, CHU de Nantes, CNRS, INSERM, l'institut du thorax, Nantes, France
| | | | | | - Kevin Eggan
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA
| | - Valeria Capra
- Medical Genetics Unit, IRCCS Giannina Gaslini Institute, Genoa, Italy
| | - Stéphane Bézieau
- CHU Nantes, Department of Medical Genetics, CHU Nantes, 9 quai Moncousu, Nantes, France; Nantes Université, CHU de Nantes, CNRS, INSERM, l'institut du thorax, Nantes, France
| | - Erica E Davis
- Stanley Manne Children's Research Institute, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL; Department of Pediatrics and Department of Cell and Developmental Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL.
| | - Michael F Wells
- Department of Human Genetics, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA; Molecular Biology Institute, University of California Los Angeles, Los Angeles, CA.
| |
Collapse
|
17
|
Farrell S, Dates J, Ramirez N, Hausknecht-Buss H, Kolosov D. Voltage-gated ion channels are expressed in the Malpighian tubules and anal papillae of the yellow fever mosquito (Aedes aegypti), and may regulate ion transport during salt and water imbalance. J Exp Biol 2024; 227:jeb246486. [PMID: 38197515 PMCID: PMC10912814 DOI: 10.1242/jeb.246486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Accepted: 12/22/2023] [Indexed: 01/11/2024]
Abstract
Vectors of infectious disease include several species of Aedes mosquitoes. The life cycle of Aedes aegypti, the yellow fever mosquito, consists of a terrestrial adult and an aquatic larval life stage. Developing in coastal waters can expose larvae to fluctuating salinity, causing salt and water imbalance, which is addressed by two prime osmoregulatory organs - the Malpighian tubules (MTs) and anal papillae (AP). Voltage-gated ion channels (VGICs) have recently been implicated in the regulation of ion transport in the osmoregulatory epithelia of insects. In the current study, we: (i) generated MT transcriptomes of freshwater-acclimated and brackish water-exposed larvae of Ae. aegypti, (ii) detected expression of several voltage-gated Ca2+, K+, Na+ and non-ion-selective ion channels in the MTs and AP using transcriptomics, PCR and gel electrophoresis, (iii) demonstrated that mRNA abundance of many altered significantly following brackish water exposure, and (iv) immunolocalized CaV1, NALCN, TRP/Painless and KCNH8 in the MTs and AP of larvae using custom-made antibodies. We found CaV1 to be expressed in the apical membrane of MTs of both larvae and adults, and its inhibition to alter membrane potentials of this osmoregulatory epithelium. Our data demonstrate that multiple VGICs are expressed in osmoregulatory epithelia of Ae. aegypti and may play an important role in the autonomous regulation of ion transport.
Collapse
Affiliation(s)
- Serena Farrell
- Department of Biological Sciences, California State University San Marcos, 333 S. Twin Oaks Valley Road, San Marcos, CA 92096, USA
| | - Jocelyne Dates
- Department of Biological Sciences, California State University San Marcos, 333 S. Twin Oaks Valley Road, San Marcos, CA 92096, USA
| | - Nancy Ramirez
- Department of Biological Sciences, California State University San Marcos, 333 S. Twin Oaks Valley Road, San Marcos, CA 92096, USA
| | - Hannah Hausknecht-Buss
- Department of Biological Sciences, California State University San Marcos, 333 S. Twin Oaks Valley Road, San Marcos, CA 92096, USA
| | - Dennis Kolosov
- Department of Biological Sciences, California State University San Marcos, 333 S. Twin Oaks Valley Road, San Marcos, CA 92096, USA
| |
Collapse
|
18
|
Min Q, Gao Y, Wang Y. Bioelectricity in dental medicine: a narrative review. Biomed Eng Online 2024; 23:3. [PMID: 38172866 PMCID: PMC10765628 DOI: 10.1186/s12938-023-01189-6] [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: 09/07/2023] [Accepted: 12/05/2023] [Indexed: 01/05/2024] Open
Abstract
BACKGROUND Bioelectric signals, whether exogenous or endogenous, play crucial roles in the life processes of organisms. Recently, the significance of bioelectricity in the field of dentistry is steadily gaining greater attention. OBJECTIVE This narrative review aims to comprehensively outline the theory, physiological effects, and practical applications of bioelectricity in dental medicine and to offer insights into its potential future direction. It attempts to provide dental clinicians and researchers with an electrophysiological perspective to enhance their clinical practice or fundamental research endeavors. METHODS An online computer search for relevant literature was performed in PubMed, Web of Science and Cochrane Library, with the keywords "bioelectricity, endogenous electric signal, electric stimulation, dental medicine." RESULTS Eventually, 288 documents were included for review. The variance in ion concentration between the interior and exterior of the cell membrane, referred to as transmembrane potential, forms the fundamental basis of bioelectricity. Transmembrane potential has been established as an essential regulator of intercellular communication, mechanotransduction, migration, proliferation, and immune responses. Thus, exogenous electric stimulation can significantly alter cellular action by affecting transmembrane potential. In the field of dental medicine, electric stimulation has proven useful for assessing pulp condition, locating root apices, improving the properties of dental biomaterials, expediting orthodontic tooth movement, facilitating implant osteointegration, addressing maxillofacial malignancies, and managing neuromuscular dysfunction. Furthermore, the reprogramming of bioelectric signals holds promise as a means to guide organism development and intervene in disease processes. Besides, the development of high-throughput electrophysiological tools will be imperative for identifying ion channel targets and precisely modulating bioelectricity in the future. CONCLUSIONS Bioelectricity has found application in various concepts of dental medicine but large-scale, standardized, randomized controlled clinical trials are still necessary in the future. In addition, the precise, repeatable and predictable measurement and modulation methods of bioelectric signal patterns are essential research direction.
Collapse
Affiliation(s)
- Qingqing Min
- Department of Endodontics, Wuxi Stomatology Hospital, Wuxi, 214000, China
| | - Yajun Gao
- Department of Endodontics, Wuxi Stomatology Hospital, Wuxi, 214000, China
| | - Yao Wang
- Department of Implantology, Wuxi Stomatology Hospital, Wuxi, 214000, China.
| |
Collapse
|
19
|
Harraz OF, Delpire E. Recent insights into channelopathies. Physiol Rev 2024; 104:23-31. [PMID: 37561136 DOI: 10.1152/physrev.00022.2023] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 07/12/2023] [Accepted: 08/05/2023] [Indexed: 08/11/2023] Open
Affiliation(s)
- Osama F Harraz
- Department of Pharmacology, Larner College of Medicine, Vermont Center for Cardiovascular and Brain Health, University of Vermont, Burlington, Vermont, United States
| | - Eric Delpire
- Department of Anesthesiology, Vanderbilt University Medical School, Nashville, Tennessee, United States
- Department of Molecular Physiology and Biophysics, Vanderbilt University Medical School, Nashville, Tennessee, United States
| |
Collapse
|
20
|
Porta-Sánchez A, Mazzanti A, Tarifa C, Kukavica D, Trancuccio A, Mohsin M, Zanfrini E, Perota A, Duchi R, Hernandez-Lopez K, Jáuregui-Abularach ME, Pergola V, Fernandez E, Bongianino R, Tavazzani E, Gambelli P, Memmi M, Scacchi S, Pavarino LF, Franzone PC, Lentini G, Filgueiras-Rama D, Galli C, Santiago DJ, Priori SG. Unexpected impairment of INa underpins reentrant arrhythmias in a knock-in swine model of Timothy syndrome. NATURE CARDIOVASCULAR RESEARCH 2023; 2:1291-1309. [PMID: 38665938 PMCID: PMC11041658 DOI: 10.1038/s44161-023-00393-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Accepted: 11/15/2023] [Indexed: 04/28/2024]
Abstract
Timothy syndrome 1 (TS1) is a multi-organ form of long QT syndrome associated with life-threatening cardiac arrhythmias, the organ-level dynamics of which remain unclear. In this study, we developed and characterized a novel porcine model of TS1 carrying the causative p.Gly406Arg mutation in CACNA1C, known to impair CaV1.2 channel inactivation. Our model fully recapitulated the human disease with prolonged QT interval and arrhythmic mortality. Electroanatomical mapping revealed the presence of a functional substrate vulnerable to reentry, stemming from an unforeseen constitutional slowing of cardiac activation. This signature substrate of TS1 was reliably identified using the reentry vulnerability index, which, we further demonstrate, can be used as a benchmark for assessing treatment efficacy, as shown by testing of multiple clinical and preclinical anti-arrhythmic compounds. Notably, in vitro experiments showed that TS1 cardiomyocytes display Ca2+ overload and decreased peak INa current, providing a rationale for the arrhythmogenic slowing of impulse propagation in vivo.
Collapse
Affiliation(s)
- Andreu Porta-Sánchez
- Novel Arrhythmogenic Mechanism Program, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Andrea Mazzanti
- Novel Arrhythmogenic Mechanism Program, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
- Molecular Cardiology, IRCCS Istituti Clinici Scientifici Maugeri, Pavia, Italy
- Department of Molecular Medicine, University of Pavia, Pavia, Italy
| | - Carmen Tarifa
- Novel Arrhythmogenic Mechanism Program, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Deni Kukavica
- Novel Arrhythmogenic Mechanism Program, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
- Molecular Cardiology, IRCCS Istituti Clinici Scientifici Maugeri, Pavia, Italy
- Department of Molecular Medicine, University of Pavia, Pavia, Italy
| | - Alessandro Trancuccio
- Novel Arrhythmogenic Mechanism Program, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
- Molecular Cardiology, IRCCS Istituti Clinici Scientifici Maugeri, Pavia, Italy
- Department of Molecular Medicine, University of Pavia, Pavia, Italy
| | - Muhammad Mohsin
- Molecular Cardiology, IRCCS Istituti Clinici Scientifici Maugeri, Pavia, Italy
| | | | | | | | - Kevin Hernandez-Lopez
- Novel Arrhythmogenic Mechanism Program, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | | | - Valerio Pergola
- Novel Arrhythmogenic Mechanism Program, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
- Department of Advanced Biomedical Sciences, University of Naples Federico II, Naples, Italy
| | - Eugenio Fernandez
- Novel Arrhythmogenic Mechanism Program, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Rossana Bongianino
- Novel Arrhythmogenic Mechanism Program, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
- Molecular Cardiology, IRCCS Istituti Clinici Scientifici Maugeri, Pavia, Italy
| | - Elisa Tavazzani
- Molecular Cardiology, IRCCS Istituti Clinici Scientifici Maugeri, Pavia, Italy
| | - Patrick Gambelli
- Molecular Cardiology, IRCCS Istituti Clinici Scientifici Maugeri, Pavia, Italy
| | - Mirella Memmi
- Molecular Cardiology, IRCCS Istituti Clinici Scientifici Maugeri, Pavia, Italy
| | - Simone Scacchi
- Department of Mathematics, University of Milan, Milano, Italy
| | | | - Piero Colli Franzone
- Novel Arrhythmogenic Mechanism Program, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
- Molecular Cardiology, IRCCS Istituti Clinici Scientifici Maugeri, Pavia, Italy
- Department of Molecular Medicine, University of Pavia, Pavia, Italy
- AVANTEA, Cremona, Italy
- Department of Advanced Biomedical Sciences, University of Naples Federico II, Naples, Italy
- Department of Mathematics, University of Milan, Milano, Italy
- Department of Mathematics, University of Pavia, Pavia, Italy
- Department of Pharmacology, University of Bari, Bari, Italy
- Cardiovascular Institute, Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares, Madrid, Spain
| | | | - David Filgueiras-Rama
- Novel Arrhythmogenic Mechanism Program, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
- Cardiovascular Institute, Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares, Madrid, Spain
| | | | - Demetrio Julián Santiago
- Novel Arrhythmogenic Mechanism Program, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Silvia G. Priori
- Novel Arrhythmogenic Mechanism Program, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
- Molecular Cardiology, IRCCS Istituti Clinici Scientifici Maugeri, Pavia, Italy
- Department of Molecular Medicine, University of Pavia, Pavia, Italy
| |
Collapse
|
21
|
Del Rivero Morfin PJ, Chavez DS, Jayaraman S, Yang L, Kochiss AL, Colecraft HM, Liu XS, Marx SO, Rajadhyaksha AM, Ben-Johny M. A Genetically Encoded Actuator Selectively Boosts L-type Calcium Channels in Diverse Physiological Settings. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.22.558856. [PMID: 37790372 PMCID: PMC10542531 DOI: 10.1101/2023.09.22.558856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/05/2023]
Abstract
L-type Ca 2+ channels (Ca V 1.2/1.3) convey influx of calcium ions (Ca 2+ ) that orchestrate a bevy of biological responses including muscle contraction and gene transcription. Deficits in Ca V 1 function play a vital role in cardiac and neurodevelopmental disorders. Yet conventional pharmacological approaches to upregulate Ca V 1 are limited, as excessive Ca 2+ influx leads to cytotoxicity. Here, we develop a genetically encoded enhancer of Ca V 1.2/1.3 channels (GeeC) to manipulate Ca 2+ entry in distinct physiological settings. Specifically, we functionalized a nanobody that targets the Ca V macromolecular complex by attaching a minimal effector domain from a Ca V enhancer-leucine rich repeat containing protein 10 (Lrrc10). In cardiomyocytes, GeeC evoked a 3-fold increase in L-type current amplitude. In neurons, GeeC augmented excitation-transcription (E-T) coupling. In all, GeeC represents a powerful strategy to boost Ca V 1.2/1.3 function in distinct physiological settings and, in so doing, lays the groundwork to illuminate new insights on neuronal and cardiac physiology and disease.
Collapse
|
22
|
Li H, Korcari A, Ciufo D, Mendias CL, Rodeo SA, Buckley MR, Loiselle AE, Pitt GS, Cao C. Increased Ca 2+ signaling through Ca V 1.2 induces tendon hypertrophy with increased collagen fibrillogenesis and biomechanical properties. FASEB J 2023; 37:e23007. [PMID: 37261735 PMCID: PMC10254118 DOI: 10.1096/fj.202300607r] [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: 03/30/2023] [Revised: 05/03/2023] [Accepted: 05/17/2023] [Indexed: 06/02/2023]
Abstract
Tendons are tension-bearing tissues transmitting force from muscle to bone for body movement. This mechanical loading is essential for tendon development, homeostasis, and healing after injury. While Ca2+ signaling has been studied extensively for its roles in mechanotransduction, regulating muscle, bone, and cartilage development and homeostasis, knowledge about Ca2+ signaling and the source of Ca2+ signals in tendon fibroblast biology are largely unknown. Here, we investigated the function of Ca2+ signaling through CaV 1.2 voltage-gated Ca2+ channel in tendon formation. Using a reporter mouse, we found that CaV 1.2 is highly expressed in tendon during development and downregulated in adult homeostasis. To assess its function, we generated ScxCre;CaV 1.2TS mice that express a gain-of-function mutant CaV 1.2 in tendon. We found that mutant tendons were hypertrophic, with more tendon fibroblasts but decreased cell density. TEM analyses demonstrated increased collagen fibrillogenesis in the hypertrophic tendons. Biomechanical testing revealed that the hypertrophic tendons display higher peak load and stiffness, with no changes in peak stress and elastic modulus. Proteomic analysis showed no significant difference in the abundance of type I and III collagens, but mutant tendons had about two-fold increase in other ECM proteins such as tenascin C, tenomodulin, periostin, type XIV and type VIII collagens, around 11-fold increase in the growth factor myostatin, and significant elevation of matrix remodeling proteins including Mmp14, Mmp2, and cathepsin K. Taken together, these data highlight roles for increased Ca2+ signaling through CaV 1.2 on regulating expression of myostatin growth factor and ECM proteins for tendon collagen fibrillogenesis during tendon formation.
Collapse
Affiliation(s)
- Haiyin Li
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA
- Department of Orthopeadics, University of Rochester Medical Center, Rochester, NY, USA
| | - Antonion Korcari
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA
- Department of Biomedical Engineering, University of Rochester Medical Center, Rochester, NY, USA
| | - David Ciufo
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA
- Department of Orthopeadics, University of Rochester Medical Center, Rochester, NY, USA
| | | | - Scott A. Rodeo
- Sports Medicine and Shoulder Service, Hospital for Special Surgery, New York, NY, USA
| | - Mark R. Buckley
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA
- Department of Biomedical Engineering, University of Rochester Medical Center, Rochester, NY, USA
| | - Alayna E. Loiselle
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA
- Department of Orthopeadics, University of Rochester Medical Center, Rochester, NY, USA
| | - Geoffrey S. Pitt
- Cardiovascular Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Chike Cao
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA
- Department of Orthopeadics, University of Rochester Medical Center, Rochester, NY, USA
| |
Collapse
|
23
|
Newmyer S, Ssemadaali MA, Radhakrishnan H, Javitz HS, Bhatnagar P. Electrically regulated cell-based intervention for viral infections. Bioeng Transl Med 2023; 8:e10434. [PMID: 36925710 PMCID: PMC10013824 DOI: 10.1002/btm2.10434] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 09/09/2022] [Accepted: 10/10/2022] [Indexed: 11/17/2022] Open
Abstract
This work reports on an engineered cell that-when electrically stimulated-synthesizes a desired protein, that is, ES-Biofactory. The platform has been used to express interferon (IFN)-β as a universal antiviral protein. Compelling evidence indicates the inevitability of new pandemics and drives the need for a pan-viral intervention that may be quickly deployed while more specific vaccines are in development. Toward this goal, a fast-growing mammalian cell (Chassis) has been engineered with multiple synthetic elements. These include-(1) a voltage-gated Ca2+ channel (Voltage-Sensor) that, upon sensing the electric field, activates the (2) Ca2+-mediated signaling pathway (Actuator) to upregulate (3) IFN-β, via an engineered antiviral transgene (Effector), that is, ES-Biofactory➔IFN-β. The antiviral effects of the ES-Biofactory➔IFN-β have been validated on severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)-infected cells. The irradiated ES-Biofactory, that does not exhibit oncogenic capacity, continues to exert antiviral effect. The resulting ES-Biofactory➔IFN-β uses a novel signaling pathway that, unlike the natural IFN synthesis pathway, is not subject to viral interference. Once clinically validated, the ES-Biofactory will be a universal antiviral cell therapy that can be immediately deployed in the event of an outbreak. The platform may also be useful in treating other diseases including cancer and autoimmune disorders.
Collapse
Affiliation(s)
- Sherri Newmyer
- Biosciences DivisionSRI InternationalMenlo ParkCaliforniaUSA
| | | | | | | | | |
Collapse
|
24
|
Li H, Korcari A, Ciufo D, Mendias CL, Rodeo SA, Buckley MR, Loiselle AE, Pitt GS, Cao C. Increased Ca 2+ signaling through Ca V 1.2 induces tendon hypertrophy with increased collagen fibrillogenesis and biomechanical properties. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.24.525119. [PMID: 36747837 PMCID: PMC9900778 DOI: 10.1101/2023.01.24.525119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Tendons are tension-bearing tissues transmitting force from muscle to bone for body movement. This mechanical loading is essential for tendon development, homeostasis, and healing after injury. While Ca 2+ signaling has been studied extensively for its roles in mechanotransduction, regulating muscle, bone and cartilage development and homeostasis, knowledge about Ca 2+ signaling and the source of Ca 2+ signals in tendon fibroblast biology are largely unknown. Here, we investigated the function of Ca 2+ signaling through Ca V 1.2 voltage-gated Ca 2+ channel in tendon formation. Using a reporter mouse, we found that Ca V 1.2 is highly expressed in tendon during development and downregulated in adult homeostasis. To assess its function, we generated ScxCre;Ca V 1.2 TS mice that express a gain-of-function mutant Ca V 1.2 channel (Ca V 1.2 TS ) in tendon. We found that tendons in the mutant mice were approximately 2/3 larger and had more tendon fibroblasts, but the cell density of the mutant mice decreased by around 22%. TEM analyses demonstrated increased collagen fibrillogenesis in the hypertrophic tendon. Biomechanical testing revealed that the hypertrophic Achilles tendons display higher peak load and stiffness, with no changes in peak stress and elastic modulus. Proteomics analysis reveals no significant difference in the abundance of major extracellular matrix (ECM) type I and III collagens, but mutant mice had about 2-fold increase in other ECM proteins such as tenascin C, tenomodulin, periostin, type XIV and type VIII collagens, around 11-fold increase in the growth factor of TGF-β family myostatin, and significant elevation of matrix remodeling proteins including Mmp14, Mmp2 and cathepsin K. Taken together, these data highlight roles for increased Ca 2+ signaling through Ca V 1.2 on regulating expression of myostatin growth factor and ECM proteins for tendon collagen fibrillogenesis during tendon formation.
Collapse
|
25
|
Xing C, Bao L, Li W, Fan H. Progress on role of ion channels of cardiac fibroblasts in fibrosis. Front Physiol 2023; 14:1138306. [PMID: 36969589 PMCID: PMC10033868 DOI: 10.3389/fphys.2023.1138306] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Accepted: 02/27/2023] [Indexed: 03/29/2023] Open
Abstract
Cardiac fibrosis is defined as excessive deposition of extracellular matrix (ECM) in pathological conditions. Cardiac fibroblasts (CFs) activated by injury or inflammation differentiate into myofibroblasts (MFs) with secretory and contractile functions. In the fibrotic heart, MFs produce ECM which is composed mainly of collagen and is initially involved in maintaining tissue integrity. However, persistent fibrosis disrupts the coordination of excitatory contractile coupling, leading to systolic and diastolic dysfunction, and ultimately heart failure. Numerous studies have demonstrated that both voltage- and non-voltage-gated ion channels alter intracellular ion levels and cellular activity, contributing to myofibroblast proliferation, contraction, and secretory function. However, an effective treatment strategy for myocardial fibrosis has not been established. Therefore, this review describes the progress made in research related to transient receptor potential (TRP) channels, Piezo1, Ca2+ release-activated Ca2+ (CRAC) channels, voltage-gated Ca2+ channels (VGCCs), sodium channels, and potassium channels in myocardial fibroblasts with the aim of providing new ideas for treating myocardial fibrosis.
Collapse
|
26
|
Abstract
The CACNA1C gene encodes the pore-forming subunit of the CaV1.2 L-type Ca2+ channel, a critical component of membrane physiology in multiple tissues, including the heart, brain, and immune system. As such, mutations altering the function of these channels have the potential to impact a wide array of cellular functions. The first mutations identified within CACNA1C were shown to cause a severe, multisystem disorder known as Timothy syndrome (TS), which is characterized by neurodevelopmental deficits, long-QT syndrome, life-threatening cardiac arrhythmias, craniofacial abnormalities, and immune deficits. Since this initial description, the number and variety of disease-associated mutations identified in CACNA1C have grown tremendously, expanding the range of phenotypes observed in affected patients. CACNA1C channelopathies are now known to encompass multisystem phenotypes as described in TS, as well as more selective phenotypes where patients may exhibit predominantly cardiac or neurological symptoms. Here, we review the impact of genetic mutations on CaV1.2 function and the resultant physiological consequences.
Collapse
Affiliation(s)
- Kevin G Herold
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - John W Hussey
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Ivy E Dick
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
27
|
Walker V, Vuister GW. Biochemistry and pathophysiology of the Transient Potential Receptor Vanilloid 6 (TRPV6) calcium channel. Adv Clin Chem 2023; 113:43-100. [PMID: 36858649 DOI: 10.1016/bs.acc.2022.11.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
TRPV6 is a Transient Receptor Potential Vanilloid (TRPV) cation channel with high selectivity for Ca2+ ions. First identified in 1999 in a search for the gene which mediates intestinal Ca2+ absorption, its far more extensive repertoire as a guardian of intracellular Ca2+ has since become apparent. Studies on TRPV6-deficient mice demonstrated additional important roles in placental Ca2+ transport, fetal bone development and male fertility. The first reports of inherited deficiency in newborn babies appeared in 2018, revealing its physiological importance in humans. There is currently strong evidence that TRPV6 also contributes to the pathogenesis of some common cancers. The recently reported association of TRPV6 deficiency with non-alcoholic chronic pancreatitis suggests a role in normal pancreatic function. Over time and with greater awareness of TRPV6, other disease-associations are likely to emerge. Powerful analytical tools have provided invaluable insights into the structure and operation of TRPV6. Its roles in Ca2+ signaling and carcinogenesis, and the use of channel inhibitors in cancer treatment are being intensively investigated. This review first briefly describes the biochemistry and physiology of the channel, and analytical methods used to investigate these. The focus subsequently shifts to the clinical disorders associated with abnormal expression and the underlying pathophysiology. The aims of this review are to increase awareness of this channel, and to draw together findings from a wide range of sources which may help to formulate new ideas for further studies.
Collapse
Affiliation(s)
- Valerie Walker
- Department of Clinical Biochemistry, University Hospital Southampton NHS Foundation Trust, Southampton General Hospital, Southampton, United Kingdom.
| | - Geerten W Vuister
- Department of Molecular and Cell Biology, Leicester Institute of Structural and Chemical Biology, University of Leicester, Leicester, United Kingdom
| |
Collapse
|
28
|
Zhou K, Luo W, Liu T, Ni Y, Qin Z. Neurotoxins Acting at Synaptic Sites: A Brief Review on Mechanisms and Clinical Applications. Toxins (Basel) 2022; 15:18. [PMID: 36668838 PMCID: PMC9865788 DOI: 10.3390/toxins15010018] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Revised: 12/20/2022] [Accepted: 12/21/2022] [Indexed: 12/29/2022] Open
Abstract
Neurotoxins generally inhibit or promote the release of neurotransmitters or bind to receptors that are located in the pre- or post-synaptic membranes, thereby affecting physiological functions of synapses and affecting biological processes. With more and more research on the toxins of various origins, many neurotoxins are now widely used in clinical treatment and have demonstrated good therapeutic outcomes. This review summarizes the structural properties and potential pharmacological effects of neurotoxins acting on different components of the synapse, as well as their important clinical applications, thus could be a useful reference for researchers and clinicians in the study of neurotoxins.
Collapse
Affiliation(s)
- Kunming Zhou
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases, College of Pharmaceutical Sciences, Suzhou Medical College of Soochow University, Suzhou 215123, China
| | - Weifeng Luo
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou 215004, China
| | - Tong Liu
- Institute of Pain Medicine and Special Environmental Medicine, Nantong University, Nantong 226019, China
| | - Yong Ni
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou 215004, China
| | - Zhenghong Qin
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases, College of Pharmaceutical Sciences, Suzhou Medical College of Soochow University, Suzhou 215123, China
| |
Collapse
|
29
|
Autism associated mutations in β 2 subunit of voltage-gated calcium channels constitutively activate gene expression. Cell Calcium 2022; 108:102672. [PMID: 36427431 DOI: 10.1016/j.ceca.2022.102672] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 10/23/2022] [Accepted: 11/08/2022] [Indexed: 11/13/2022]
Abstract
Membrane depolarization triggers gene expression through voltage-gated calcium channels (VGCC) in a process called Excitation-transcription (ET) coupling. Mutations in the channel subunits α11.2, or β2d, are associated with neurodevelopmental disorders such as ASD. Here, we found that two mutations S143F and G113S within the rat Cavβ2a corresponding to autistic related mutations Cavβ2dS197F and Cavβ2dG167S in the human Cavβ2d, activate ET-coupling via the RAS/ERK/CREB pathway. Membrane depolarization of HEK293 cells co-expressing α11.2 and α2δ with Cavβ2aS143F or Cavβ2aG113S triggers constitutive transcriptional activation, which is correlated with facilitated channel activity. Similar to the Timothy-associated autistic mutation α11.2G406R, constitutive gene activation is attributed to a hyperpolarizing shift in the activation kinetics of Cav1.2. Pulldown of RasGRF2 and RhoGEF by wt and the Cavβ2a autistic mutants is consistent with Cavβ2/Ras activation in ET coupling and implicates Rho signaling as yet another molecular pathway activated by Cavα11.2/Cavβ2 . Facilitated spontaneous channel activity preceding enhanced gene activation via the Ras/ERK/CREB pathway, appears a general molecular mechanism for Ca2+ channel mediated ASD and other neurodevelopmental disorders.
Collapse
|
30
|
Harrison PJ, Husain SM, Lee H, Los Angeles AD, Colbourne L, Mould A, Hall NAL, Haerty W, Tunbridge EM. CACNA1C (Ca V1.2) and other L-type calcium channels in the pathophysiology and treatment of psychiatric disorders: Advances from functional genomics and pharmacoepidemiology. Neuropharmacology 2022; 220:109262. [PMID: 36154842 DOI: 10.1016/j.neuropharm.2022.109262] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 08/09/2022] [Accepted: 09/17/2022] [Indexed: 11/17/2022]
Abstract
A role for voltage-gated calcium channels (VGCCs) in psychiatric disorders has long been postulated as part of a broader involvement of intracellular calcium signalling. However, the data were inconclusive and hard to interpret. We review three areas of research that have markedly advanced the field. First, there is now robust genomic evidence that common variants in VGCC subunit genes, notably CACNA1C which encodes the L-type calcium channel (LTCC) CaV1.2 subunit, are trans-diagnostically associated with psychiatric disorders including schizophrenia and bipolar disorder. Rare variants in these genes also contribute to the risk. Second, pharmacoepidemiological evidence supports the possibility that calcium channel blockers, which target LTCCs, might have beneficial effects on the onset or course of these disorders. This is especially true for calcium channel blockers that are brain penetrant. Third, long-range sequencing is revealing the repertoire of full-length LTCC transcript isoforms. Many novel and abundant CACNA1C isoforms have been identified in human and mouse brain, including some which are enriched compared to heart or aorta, and predicted to encode channels with differing functional and pharmacological properties. These isoforms may contribute to the molecular mechanisms of genetic association to psychiatric disorders. They may also enable development of therapeutic agents that can preferentially target brain LTCC isoforms and be of potential value for psychiatric indications.
Collapse
Affiliation(s)
- Paul J Harrison
- Department of Psychiatry, University of Oxford, Warneford Hospital, Oxford, OX3 7JX, UK; Oxford Health NHS Foundation Trust, Warneford Hospital, Oxford, OX3 7JX, UK.
| | - Syed M Husain
- Department of Psychiatry, University of Oxford, Warneford Hospital, Oxford, OX3 7JX, UK
| | - Hami Lee
- Department of Psychiatry, University of Oxford, Warneford Hospital, Oxford, OX3 7JX, UK
| | | | - Lucy Colbourne
- Department of Psychiatry, University of Oxford, Warneford Hospital, Oxford, OX3 7JX, UK; Oxford Health NHS Foundation Trust, Warneford Hospital, Oxford, OX3 7JX, UK
| | - Arne Mould
- Department of Psychiatry, University of Oxford, Warneford Hospital, Oxford, OX3 7JX, UK; Oxford Health NHS Foundation Trust, Warneford Hospital, Oxford, OX3 7JX, UK
| | - Nicola A L Hall
- Department of Psychiatry, University of Oxford, Warneford Hospital, Oxford, OX3 7JX, UK
| | - Wilfried Haerty
- Earlham Institute, Norwich Research Park, Norwich, NR4 7UZ, UK; School of Biological Sciences, University of East Anglia, Norwich, UK
| | - Elizabeth M Tunbridge
- Department of Psychiatry, University of Oxford, Warneford Hospital, Oxford, OX3 7JX, UK; Oxford Health NHS Foundation Trust, Warneford Hospital, Oxford, OX3 7JX, UK
| |
Collapse
|
31
|
Pellegrino M, Ricci E, Ceraldi R, Nigro A, Bonofiglio D, Lanzino M, Morelli C. From HDAC to Voltage-Gated Ion Channels: What's Next? The Long Road of Antiepileptic Drugs Repositioning in Cancer. Cancers (Basel) 2022; 14:cancers14184401. [PMID: 36139561 PMCID: PMC9497059 DOI: 10.3390/cancers14184401] [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: 08/04/2022] [Revised: 09/02/2022] [Accepted: 09/03/2022] [Indexed: 11/16/2022] Open
Abstract
Simple Summary Although in the last decades the clinical outcome of cancer patients considerably improved, the major drawbacks still associated with chemotherapy are the unwanted side effects and the development of drug resistance. Therefore, a continuous effort in trying to discover new tumor markers, possibly of diagnostic, prognostic and therapeutic value, is being made. This review is aimed at highlighting the anti-tumor activity that several antiepileptic drugs (AEDs) exert in breast, prostate and other types of cancers, mainly focusing on their ability to block the voltage-gated Na+ and Ca++ channels, as well as to inhibit the activity of histone deacetylases (HDACs), all well-documented tumor markers and/or molecular targets. The existence of additional AEDs molecular targets is highly suspected. Therefore, the repurposing of already available drugs as adjuvants in cancer treatment would have several advantages, such as reductions in dose-related toxicity CVs will be sent in a separate mail to the indicated address of combined treatments, lower production costs, and faster approval for clinical use. Abstract Cancer is a major health burden worldwide. Although the plethora of molecular targets identified in the last decades and the deriving developed treatments, which significantly improved patients’ outcome, the occurrence of resistance to therapies remains the major cause of relapse and mortality. Thus, efforts in identifying new markers to be exploited as molecular targets in cancer therapy are needed. This review will first give a glance on the diagnostic and therapeutic significance of histone deacetylase (HDAC) and voltage gated ion channels (VGICs) in cancer. Nevertheless, HDAC and VGICs have also been reported as molecular targets through which antiepileptic drugs (AEDs) seem to exert their anticancer activity. This should be claimed as a great advantage. Indeed, due to the slowness of drug approval procedures, the attempt to turn to off-label use of already approved medicines would be highly preferable. Therefore, an updated and accurate overview of both preclinical and clinical data of commonly prescribed AEDs (mainly valproic acid, lamotrigine, carbamazepine, phenytoin and gabapentin) in breast, prostate, brain and other cancers will follow. Finally, a glance at the emerging attempt to administer AEDs by means of opportunely designed drug delivery systems (DDSs), so to limit toxicity and improve bioavailability, is also given.
Collapse
Affiliation(s)
| | | | | | | | | | - Marilena Lanzino
- Correspondence: (M.L.); (C.M.); Tel.: +39-0984-496206 (M.L.); +39-0984-496211 (C.M.)
| | - Catia Morelli
- Correspondence: (M.L.); (C.M.); Tel.: +39-0984-496206 (M.L.); +39-0984-496211 (C.M.)
| |
Collapse
|
32
|
McClenaghan C, Nichols CG. Kir6.1 and SUR2B in Cantú syndrome. Am J Physiol Cell Physiol 2022; 323:C920-C935. [PMID: 35876283 PMCID: PMC9467476 DOI: 10.1152/ajpcell.00154.2022] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 07/07/2022] [Accepted: 07/07/2022] [Indexed: 12/25/2022]
Abstract
Kir6.1 and SUR2 are subunits of ATP-sensitive potassium (KATP) channels expressed in a wide range of tissues. Extensive study has implicated roles of these channel subunits in diverse physiological functions. Together they generate the predominant KATP conductance in vascular smooth muscle and are the target of vasodilatory drugs. Roles for Kir6.1/SUR2 dysfunction in disease have been suggested based on studies of animal models and human genetic discoveries. In recent years, it has become clear that gain-of-function (GoF) mutations in both genes result in Cantú syndrome (CS)-a complex, multisystem disorder. There is currently no targeted therapy for CS, but studies of mouse models of the disease reveal that pharmacological reversibility of cardiovascular and gastrointestinal pathologies can be achieved by administration of the KATP channel inhibitor, glibenclamide. Here we review the function, structure, and physiological and pathological roles of Kir6.1/SUR2B channels, with a focus on CS. Recent studies have led to much improved understanding of the underlying pathologies and the potential for treatment, but important questions remain: Can the study of genetically defined CS reveal new insights into Kir6.1/SUR2 function? Do these reveal new pathophysiological mechanisms that may be important in more common diseases? And is our pharmacological armory adequately stocked?
Collapse
Affiliation(s)
- Conor McClenaghan
- Department of Cell Biology and Physiology, Center for the Investigation of Membrane Excitability Diseases (CIMED), Washington University, St. Louis, Missouri
| | - Colin G Nichols
- Department of Cell Biology and Physiology, Center for the Investigation of Membrane Excitability Diseases (CIMED), Washington University, St. Louis, Missouri
| |
Collapse
|
33
|
Loh D, Reiter RJ. Melatonin: Regulation of Viral Phase Separation and Epitranscriptomics in Post-Acute Sequelae of COVID-19. Int J Mol Sci 2022; 23:8122. [PMID: 35897696 PMCID: PMC9368024 DOI: 10.3390/ijms23158122] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 07/09/2022] [Accepted: 07/20/2022] [Indexed: 01/27/2023] Open
Abstract
The relentless, protracted evolution of the SARS-CoV-2 virus imposes tremendous pressure on herd immunity and demands versatile adaptations by the human host genome to counter transcriptomic and epitranscriptomic alterations associated with a wide range of short- and long-term manifestations during acute infection and post-acute recovery, respectively. To promote viral replication during active infection and viral persistence, the SARS-CoV-2 envelope protein regulates host cell microenvironment including pH and ion concentrations to maintain a high oxidative environment that supports template switching, causing extensive mitochondrial damage and activation of pro-inflammatory cytokine signaling cascades. Oxidative stress and mitochondrial distress induce dynamic changes to both the host and viral RNA m6A methylome, and can trigger the derepression of long interspersed nuclear element 1 (LINE1), resulting in global hypomethylation, epigenetic changes, and genomic instability. The timely application of melatonin during early infection enhances host innate antiviral immune responses by preventing the formation of "viral factories" by nucleocapsid liquid-liquid phase separation that effectively blockades viral genome transcription and packaging, the disassembly of stress granules, and the sequestration of DEAD-box RNA helicases, including DDX3X, vital to immune signaling. Melatonin prevents membrane depolarization and protects cristae morphology to suppress glycolysis via antioxidant-dependent and -independent mechanisms. By restraining the derepression of LINE1 via multifaceted strategies, and maintaining the balance in m6A RNA modifications, melatonin could be the quintessential ancient molecule that significantly influences the outcome of the constant struggle between virus and host to gain transcriptomic and epitranscriptomic dominance over the host genome during acute infection and PASC.
Collapse
Affiliation(s)
- Doris Loh
- Independent Researcher, Marble Falls, TX 78654, USA;
| | - Russel J. Reiter
- Department of Cell Systems and Anatomy, UT Health San Antonio, San Antonio, TX 78229, USA
| |
Collapse
|
34
|
Chen D, Cheng H, Liu S, Al-Sheikh U, Fan Y, Duan D, Zou W, Zhu L, Kang L. The Voltage-Gated Calcium Channel EGL-19 Acts on Glia to Drive Olfactory Adaptation. Front Mol Neurosci 2022; 15:907064. [PMID: 35782381 PMCID: PMC9247319 DOI: 10.3389/fnmol.2022.907064] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Accepted: 05/11/2022] [Indexed: 11/13/2022] Open
Abstract
Calcium channelopathies have been strongly linked to cardiovascular, muscular, neurological and psychiatric disorders. The voltage-gated calcium channels (VGCC) are vital transducers of membrane potential changes to facilitate the dynamics of calcium ions and release of neurotransmitter. Whether these channels function in the glial cell to mediate calcium variations and regulate behavioral outputs, is poorly understood. Our results showed that odorant and mechanical stimuli evoked robust calcium increases in the amphid sheath (AMsh) glia from C. elegans, which were largely dependent on the L-Type VGCC EGL-19. Moreover, EGL-19 modulates the morphologies of both ASH sensory neurons and AMsh glia. Tissue-specific knock-down of EGL-19 in AMsh glia regulated sensory adaptability of ASH neurons and promoted olfactory adaptation. Our results reveal a novel role of glial L-Type VGCC EGL-19 on olfaction, lead to improved understanding of the functions of VGCCs in sensory transduction.
Collapse
Affiliation(s)
- Du Chen
- Department of Neurobiology and Department of Neurology of the Fourth Affiliated Hospital, Zhejiang University, School of Medicine, Hangzhou, China
- NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, China
| | - Hankui Cheng
- Department of Neurobiology and Department of Neurology of the Fourth Affiliated Hospital, Zhejiang University, School of Medicine, Hangzhou, China
- NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, China
| | - Siyan Liu
- Department of Neurobiology and Department of Neurology of the Fourth Affiliated Hospital, Zhejiang University, School of Medicine, Hangzhou, China
- NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, China
| | - Umar Al-Sheikh
- Department of Neurobiology and Department of Neurology of the Fourth Affiliated Hospital, Zhejiang University, School of Medicine, Hangzhou, China
- NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, China
| | - Yuedan Fan
- Department of Neurobiology and Department of Neurology of the Fourth Affiliated Hospital, Zhejiang University, School of Medicine, Hangzhou, China
- NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, China
| | - Duo Duan
- Department of Neurobiology and Department of Neurology of the Fourth Affiliated Hospital, Zhejiang University, School of Medicine, Hangzhou, China
- NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, China
| | - Wenjuan Zou
- Department of Neurobiology and Department of Neurology of the Fourth Affiliated Hospital, Zhejiang University, School of Medicine, Hangzhou, China
- NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, China
| | - Linhui Zhu
- Department of Neurobiology and Department of Neurology of the Fourth Affiliated Hospital, Zhejiang University, School of Medicine, Hangzhou, China
- NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, China
| | - Lijun Kang
- Department of Neurobiology and Department of Neurology of the Fourth Affiliated Hospital, Zhejiang University, School of Medicine, Hangzhou, China
- NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, China
- *Correspondence: Lijun Kang
| |
Collapse
|
35
|
Kolosov D, O'Donnell MJ. Blending physiology and RNAseq to provide new insights into regulation of epithelial transport: switching between ion secretion and reabsorption. J Exp Biol 2022; 225:274251. [PMID: 35119072 DOI: 10.1242/jeb.243293] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
This Review addresses the means by which epithelia change the direction of vectorial ion transport. Recent studies have revealed that insect Malpighian (renal) tubules can switch from secreting to reabsorbing K+. When the gut of larval lepidopterans is empty (during the moult cycle) or when the larvae are reared on K+-deficient diet, the distal ileac plexus segment of the tubule secretes K+ from the haemolymph into the tubule lumen. By contrast, in larvae reared on K+-rich diet, ions and fluid are reabsorbed from the rectal lumen into the perinephric space surrounding the cryptonephridial tubules of the rectal complex. Ions and fluid are then transported from the perinephric space into the lumen of the cryptonephridial tubules, thus supplying the free segments of the tubule downstream. Under these conditions, some of the K+ and water in the tubule lumen is reabsorbed across the cells of the distal ileac plexus, allowing for expansion of haemolymph volume in the rapidly growing larvae, as well as recycling of K+ and base equivalents. RNA sequencing data reveal large-scale changes in gene transcription that are associated with the switch between ion secretion and ion reabsorption by the distal ileac plexus. An unexpected finding is the presence of voltage-gated, ligand-gated and mechanosensitive ion channels, normally seen in excitable cells, in Malpighian tubules. Transcriptomic surveys indicate that these types of channels are also present in multiple other types of vertebrate and invertebrate epithelia, suggesting that they may play novel roles in epithelial cell signalling and regulation of epithelial ion transport.
Collapse
Affiliation(s)
- Dennis Kolosov
- Department of Biological Sciences, California State University San Marcos, 333 S Twin Oaks Valley Road, San Marcos, CA 92096, USA
| | - Michael J O'Donnell
- Department of Biology, McMaster University, 1280 Main Street West, Hamilton, ON, Canada, L8S 4K1
| |
Collapse
|
36
|
Matsui M, Bouchareb R, Storto M, Hussain Y, Gregg A, Marx SO, Pitt GS. Increased Ca2+ influx through CaV1.2 drives aortic valve calcification. JCI Insight 2022; 7:155569. [PMID: 35104251 PMCID: PMC8983132 DOI: 10.1172/jci.insight.155569] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Accepted: 01/28/2022] [Indexed: 11/17/2022] Open
Abstract
Calcific aortic valve disease (CAVD) is heritable as revealed by recent genome wide association studies. While polymorphisms linked to increased expression of CACNA1C, encoding the CaV1.2 L-type voltage-gated Ca2+ channel, and increased Ca2+ signaling are associated with CAVD, whether increased Ca2+ influx through the druggable CaV1.2 is causal for calcific aortic valve disease is unknown. With surgically removed aortic valves from patients, we confirmed the association between increased CaV1.2 expression and CAVD. We extended our studies with a transgenic mouse model that mimics increased CaV1.2 expression in within aortic valve interstitial cells (VICs). In young mice maintained on normal chow, we observed dystrophic valve lesions that mimic changes found in pre-symptomatic CAVD, and showed activation of chondrogenic and osteogenic transcriptional regulators within these valve lesions. Chronic administration of verapamil, a clinically used CaV1.2 antagonist, slowed the progression of lesion development in vivo. Exploiting VIC cultures we demonstrated that increased Ca2+ influx through CaV1.2 drives signaling programs that lead to myofibroblast activation of VICs and upregulation of genes associated with aortic valve calcification. Our data support a causal role for Ca2+ influx through CaV1.2 in CAVD and suggest that early treatment with Ca2+ channel blockers is an effective therapeutic strategy.
Collapse
Affiliation(s)
- Maiko Matsui
- Cardiovascular Research Institute, Weill Cornell Medicine, New York, United States of America
| | - Rihab Bouchareb
- Zena and Michael A. Wiener Cardiovascular Institute, Icahn School of Medicine at Mount Sinai, New York, United States of America
| | - Mara Storto
- Cardiovascular Research Institute, Weill Cornell Medicine, New York, United States of America
| | - Yasin Hussain
- Cardiovascular Research Institute, Weill Cornell Medicine, New York, United States of America
| | - Andrew Gregg
- Cardiovascular Research Institute, Weill Cornell Medicine, New York, United States of America
| | - Steven O Marx
- Division of Cardiology, Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University, New York, United States of America
| | - Geoffrey S Pitt
- Cardiovascular Research Institute, Weill Cornell Medicine, New York, United States of America
| |
Collapse
|
37
|
CaV1.3 enhanced store operated calcium promotes resistance to androgen deprivation in prostate cancer. Cell Calcium 2022; 103:102554. [DOI: 10.1016/j.ceca.2022.102554] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 01/24/2022] [Accepted: 02/06/2022] [Indexed: 01/01/2023]
|
38
|
Nichols CG, McClenaghan C. Bridging Personal and Population in Excitability Diseases: Will Studies of Rare Diseases Bring Generalizable Mechanisms From Monogenic Channelopathies? FUNCTION (OXFORD, ENGLAND) 2022; 3:zqab072. [PMID: 35402918 PMCID: PMC8991026 DOI: 10.1093/function/zqab072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 12/29/2021] [Accepted: 12/21/2021] [Indexed: 01/07/2023]
Affiliation(s)
| | - Conor McClenaghan
- Center for the Investigation of Membrane Excitability Diseases, and Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| |
Collapse
|
39
|
Dixon RE, Navedo MF, Binder MD, Santana LF. Mechanisms and Physiological Implications of Cooperative Gating of Ion Channels Clusters. Physiol Rev 2021; 102:1159-1210. [PMID: 34927454 DOI: 10.1152/physrev.00022.2021] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Ion channels play a central role in the regulation of nearly every cellular process. Dating back to the classic 1952 Hodgkin-Huxley model of the generation of the action potential, ion channels have always been thought of as independent agents. A myriad of recent experimental findings exploiting advances in electrophysiology, structural biology, and imaging techniques, however, have posed a serious challenge to this long-held axiom as several classes of ion channels appear to open and close in a coordinated, cooperative manner. Ion channel cooperativity ranges from variable-sized oligomeric cooperative gating in voltage-gated, dihydropyridine-sensitive Cav1.2 and Cav1.3 channels to obligatory dimeric assembly and gating of voltage-gated Nav1.5 channels. Potassium channels, transient receptor potential channels, hyperpolarization cyclic nucleotide-activated channels, ryanodine receptors (RyRs), and inositol trisphosphate receptors (IP3Rs) have also been shown to gate cooperatively. The implications of cooperative gating of these ion channels range from fine tuning excitation-contraction coupling in muscle cells to regulating cardiac function and vascular tone, to modulation of action potential and conduction velocity in neurons and cardiac cells, and to control of pace-making activity in the heart. In this review, we discuss the mechanisms leading to cooperative gating of ion channels, their physiological consequences and how alterations in cooperative gating of ion channels may induce a range of clinically significant pathologies.
Collapse
Affiliation(s)
- Rose Ellen Dixon
- Department of Physiology and Membrane Biology, University of California, Davis, CA, United States
| | - Manuel F Navedo
- Department of Pharmacology, University of California, Davis, CA, United States
| | - Marc D Binder
- Department of Physiology and Biophysics, University of Washington, Seattle, WA, United States
| | - L Fernando Santana
- Department of Physiology and Membrane Biology, University of California, Davis, CA, United States
| |
Collapse
|
40
|
Maltan L, Najjar H, Tiffner A, Derler I. Deciphering Molecular Mechanisms and Intervening in Physiological and Pathophysiological Processes of Ca 2+ Signaling Mechanisms Using Optogenetic Tools. Cells 2021; 10:3340. [PMID: 34943850 PMCID: PMC8699489 DOI: 10.3390/cells10123340] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 11/17/2021] [Accepted: 11/22/2021] [Indexed: 11/16/2022] Open
Abstract
Calcium ion channels are involved in numerous biological functions such as lymphocyte activation, muscle contraction, neurotransmission, excitation, hormone secretion, gene expression, cell migration, memory, and aging. Therefore, their dysfunction can lead to a wide range of cellular abnormalities and, subsequently, to diseases. To date various conventional techniques have provided valuable insights into the roles of Ca2+ signaling. However, their limited spatiotemporal resolution and lack of reversibility pose significant obstacles in the detailed understanding of the structure-function relationship of ion channels. These drawbacks could be partially overcome by the use of optogenetics, which allows for the remote and well-defined manipulation of Ca2+-signaling. Here, we review the various optogenetic tools that have been used to achieve precise control over different Ca2+-permeable ion channels and receptors and associated downstream signaling cascades. We highlight the achievements of optogenetics as well as the still-open questions regarding the resolution of ion channel working mechanisms. In addition, we summarize the successes of optogenetics in manipulating many Ca2+-dependent biological processes both in vitro and in vivo. In summary, optogenetics has significantly advanced our understanding of Ca2+ signaling proteins and the used tools provide an essential basis for potential future therapeutic application.
Collapse
Affiliation(s)
| | | | | | - Isabella Derler
- Institute of Biophysics, JKU Life Science Center, Johannes Kepler University Linz, A-4020 Linz, Austria; (L.M.); (H.N.); (A.T.)
| |
Collapse
|
41
|
Kapoor D, Khan A, O'Donnell MJ, Kolosov D. Novel mechanisms of epithelial ion transport: insights from the cryptonephridial system of lepidopteran larvae. CURRENT OPINION IN INSECT SCIENCE 2021; 47:53-61. [PMID: 33866042 DOI: 10.1016/j.cois.2021.04.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 04/01/2021] [Accepted: 04/05/2021] [Indexed: 06/12/2023]
Abstract
Lepidopterans are among the most widespread and easily recognized insects. Whereas adult lepidopterans are known for their beauty and ecological importance as pollinators and sources of food for other animals, larvae are economically important pests of forests and agricultural crops. In the larval body, rapid growth while feeding on plant-based diet is associated with extreme alkalinity (up to pH = 11) of the midgut lumen that helps digest plant proteins. Additionally, the presence of plant secondary metabolites which serve as anti-herbivory agents requires uninterrupted excretory function, accomplished primarily by the Malpighian tubules (MTs). The so-called cryptonephridial condition, along with extreme regional heterogeneity of the MTs, and the ability to rapidly and reversibly alter the direction of epithelial ion transport are features that allow uninterrupted MT functioning and recycling of base equivalents. Studies of MTs in lepidopteran larvae have revealed that rapid adjustments in epithelial ion transport include unexpected roles for voltage-gated, ligand-gated and mechanosensitive ion channels, as well as gap junctions. These molecular components are present in epithelia of a variety of vertebrates and invertebrates and thus are likely to constitute a universal epithelial toolkit for rapid autonomous regulation of epithelial function.
Collapse
Affiliation(s)
| | - Aliyyah Khan
- Department of Biology, McMaster University, Hamilton, Canada
| | | | - Dennis Kolosov
- Department of Biological Sciences, California State University San Marcos, San Marcos, USA.
| |
Collapse
|
42
|
Şterbuleac D. Molecular dynamics: a powerful tool for studying the medicinal chemistry of ion channel modulators. RSC Med Chem 2021; 12:1503-1518. [PMID: 34671734 PMCID: PMC8459385 DOI: 10.1039/d1md00140j] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2021] [Accepted: 07/21/2021] [Indexed: 01/10/2023] Open
Abstract
Molecular dynamics (MD) simulations allow researchers to investigate the behavior of desired biological targets at ever-decreasing costs with ever-increasing precision. Among the biological macromolecules, ion channels are remarkable transmembrane proteins, capable of performing special biological processes and revealing a complex regulatory matrix, including modulation by small molecules, either endogenous or exogenous. Recently, given the developments in ion channel structure determination and accessibility of bio-computational techniques, MD and related tools are becoming increasingly popular in the intense research area regarding ligand-channel interactions. This review synthesizes and presents the most important fields of MD involvement in investigating channel-molecule interactions, including, but not limited to, deciphering the binding modes of ligands to their ion channel targets and the mechanisms through which chemical compounds exert their effect on channel function. Special attention is devoted to the importance of more elaborate methods, such as free energy calculations, while principles regarding drug design and discovery are highlighted. Several technical aspects involving the creation and simulation of channel-molecule MD systems (ligand parameterization, proper membrane setup, system building, etc.) are also presented.
Collapse
Affiliation(s)
- Daniel Şterbuleac
- Department of Health and Human Development, "Ştefan cel Mare" University of Suceava Str. Universităţii 13, 720229, E Building Suceava Romania
- Department of Forestry and Environmental Protection, "Ştefan cel Mare" University of Suceava Str. Universităţii 13, 720229, E Building Suceava Romania
- Integrated Center for Research, Development and Innovation in Advanced Materials, Nanotechnologies and Distributed Systems for Fabrication and Control (MANSiD), "Ştefan cel Mare" University of Suceava Str. Universităţii 13 720229 Suceava Romania
| |
Collapse
|
43
|
Bauer R, Timothy KW, Golden A. Update on the Molecular Genetics of Timothy Syndrome. Front Pediatr 2021; 9:668546. [PMID: 34079780 PMCID: PMC8165229 DOI: 10.3389/fped.2021.668546] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Accepted: 04/19/2021] [Indexed: 12/11/2022] Open
Abstract
Timothy Syndrome (TS) (OMIM #601005) is a rare autosomal dominant syndrome caused by variants in CACNA1C, which encodes the α1C subunit of the voltage-gated calcium channel Cav1.2. TS is classically caused by only a few different genetic changes and characterized by prolonged QT interval, syndactyly, and neurodevelopmental delay; however, the number of identified TS-causing variants is growing, and the resulting symptom profiles are incredibly complex and variable. Here, we aim to review the genetic and clinical findings of all published case reports of TS to date. We discuss multiple possible mechanisms for the variability seen in clinical features across these cases, including mosaicism, genetic background, isoform complexity of CACNA1C and differential expression of transcripts, and biophysical changes in mutant CACNA1C channels. Finally, we propose future research directions such as variant validation, in vivo modeling, and natural history characterization.
Collapse
Affiliation(s)
- Rosemary Bauer
- Laboratory of Biochemistry and Genetics, National Institute of Diabetes, Digestive, and Kidney Diseases, National Institutes of Health, Bethesda, MD, United States
| | | | - Andy Golden
- Laboratory of Biochemistry and Genetics, National Institute of Diabetes, Digestive, and Kidney Diseases, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|