1
|
Ganglberger M, Koschak A. Exploring the potential for gene therapy in Cav1.4-related retinal channelopathies. Channels (Austin) 2025; 19:2480089. [PMID: 40129245 PMCID: PMC11938310 DOI: 10.1080/19336950.2025.2480089] [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: 01/24/2025] [Revised: 03/07/2025] [Accepted: 03/11/2025] [Indexed: 03/26/2025] Open
Abstract
The visual process begins with photon detection in photoreceptor outer segments within the retina, which processes light signals before transmission to the thalamus and visual cortex. Cav1.4 L-type calcium channels play a crucial role in this process, and dysfunction of these channels due to pathogenic variants in corresponding genes leads to specific manifestations in visual impairments. This review explores the journey from basic research on Cav1.4 L-type calcium channel complexes in retinal physiology and pathophysiology to their potential as gene therapy targets. Moreover, we provide a concise overview of key findings from studies using different animal models to investigate retinal diseases. It will critically examine the constraints these models present when attempting to elucidate retinal channelopathies. Additionally, the paper will explore potential strategies for addressing Cav1.4 channel dysfunction and discuss the current challenges facing gene therapy approaches in this area of research.
Collapse
Affiliation(s)
- Matthias Ganglberger
- Pharmacology and Toxicology, Institute of Pharmacy, University of Innsbruck, Innsbruck, Austria
| | - Alexandra Koschak
- Pharmacology and Toxicology, Institute of Pharmacy, University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
2
|
Yang L, Liu L, Zhang P, Pan W, Huang H, Qi Y, Wang Y, Zhang R, Zhou P. Rigid and soft substrates respectively promote the myocardial differentiation and maturation of human embryonic stem cells using elastic PDMS with thick synthetic coating. Colloids Surf B Biointerfaces 2025; 250:114540. [PMID: 39904141 DOI: 10.1016/j.colsurfb.2025.114540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 01/16/2025] [Accepted: 01/26/2025] [Indexed: 02/06/2025]
Abstract
Cardiovascular disease is the predominant cause of mortality and severe disability. Cardiomyocytes (CMs) derived from human embryonic stem cells (hESCs) have good application prospects for treating this disease. Unfortunately, CMs generated via current methods are relatively immature, as proven by defects such as sarcomer-like structures, calcium processing capacity and mitochondrial maturity. Therefore, in this study, tunable PDMS substrates that modified with sufficiently thick synthetic coatings were prepared to regulate both the myocardial differentiation of hESCs and subsequent maturation. Surprisingly, the effect of substrate elasticity on the critical attachment of hESCs and hESC-CMs vanished when common Matrigel coatings were used, but apparent differences were detected in the synthetic group. Rigid substrates promoted the adhesion of hESCs but not hESC-CMs. Moreover, the PDMS substrates with the highest hardness remarkably promoted the myocardial differentiation of hESCs, which was even better than that of the rigid plate group. The softest PDMS achieved the best performance among the groups in terms of the maturation of hESC-CMs, as confirmed by enhanced functional, metabolic, and ultrastructural maturation. This study reveals the real impact of an elastic substrate on the adhesion, differentiation, and maturation of hESC-CMs, which has value for accelerating the development of clinically applicable mature hESC-CMs with high induction efficiency.
Collapse
Affiliation(s)
- Ling Yang
- Key Laboratory of Dental Maxillofacial Reconstruction and Biological Intelligence Manufacturing, School and Hospital of Stomatology, Lanzhou University, Lanzhou, Gansu Province 730000, China
| | - Lu Liu
- The Second Hospital of Lanzhou University, Lanzhou University, Lanzhou, Gansu Province 730030, China
| | - Pengxia Zhang
- Key Laboratory of Dental Maxillofacial Reconstruction and Biological Intelligence Manufacturing, School and Hospital of Stomatology, Lanzhou University, Lanzhou, Gansu Province 730000, China
| | - Wen Pan
- School of Life Sciences, Lanzhou University, Lanzhou, Gansu Province 730000, China
| | - Hongxin Huang
- Key Laboratory of Dental Maxillofacial Reconstruction and Biological Intelligence Manufacturing, School and Hospital of Stomatology, Lanzhou University, Lanzhou, Gansu Province 730000, China
| | - Yongmei Qi
- School of Life Sciences, Lanzhou University, Lanzhou, Gansu Province 730000, China
| | - Yingbin Wang
- The Second Hospital of Lanzhou University, Lanzhou University, Lanzhou, Gansu Province 730030, China
| | - Rongzhi Zhang
- The Second Hospital of Lanzhou University, Lanzhou University, Lanzhou, Gansu Province 730030, China.
| | - Ping Zhou
- Key Laboratory of Dental Maxillofacial Reconstruction and Biological Intelligence Manufacturing, School and Hospital of Stomatology, Lanzhou University, Lanzhou, Gansu Province 730000, China.
| |
Collapse
|
3
|
Russell G. Theoretical evaluation of the biological activity of hydrogen. Med Gas Res 2025; 15:266-275. [PMID: 39829163 PMCID: PMC11918482 DOI: 10.4103/mgr.medgasres-d-24-00083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Accepted: 10/31/2024] [Indexed: 01/22/2025] Open
Abstract
Hydrogen (H2), the simplest and most ubiquitous molecule in the universe, has garnered significant scientific interest over the past two decades because of its potential as an effective antioxidant and anti-inflammatory agent. Traditionally considered inert, H2 is now being re-evaluated for its unique bioactive properties. H2 selectively neutralizes reactive oxygen and nitrogen species, mitigating oxidative stress without disrupting essential cellular functions. This review therefore aims to provide a theoretical evaluation of the biological activity of H2, focusing on its pharmacokinetics, including absorption, distribution, and retention within biological systems. The pharmacokinetic profile of H2 is crucial for understanding its potential therapeutic applications. The interaction of H2 with protein pockets is of particular interest, as these sites may serve as reservoirs or active sites for H2, influencing its biological activity and retention time. Additionally, the impact of H2 on cellular signaling pathways, including those regulating glucose metabolism and oxidative stress responses, will be explored, offering insights into its potential as a modulator of metabolic and redox homeostasis. Finally, interactions with ferromagnetic molecules within biological environments, as well as effects on cellular signaling mechanisms, add another layer of complexity to the biological role of H2. By synthesizing the current research, this review seeks to elucidate the underlying mechanisms by which H2 may exert therapeutic effects while also identifying critical areas for further investigation. Understanding these aspects is essential for fully characterizing the pharmacodynamic profile of H2 and assessing its clinical potential in the treatment of oxidative stress-related disorders.
Collapse
Affiliation(s)
- Grace Russell
- Research Consultant, Water Fuel Engineering, Wakefield, UK
- School of Applied Sciences, University of the West of England, Frenchay Campus, Coldharbour Lane, Bristol, UK
| |
Collapse
|
4
|
Lv J, Chen F, Lv L, Zhang L, Zou H, Liu Y, Bai Y, Fang R, Qin T, Deng Z. LncRNA ABHD11-AS1 Elevates CALM2 to Promote Metastasis of Thyroid Cancer Through Sponging miR-876-5p. Biochem Genet 2025:10.1007/s10528-025-11072-9. [PMID: 40117023 DOI: 10.1007/s10528-025-11072-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Accepted: 02/21/2025] [Indexed: 03/23/2025]
Abstract
In the past decade, the treatment of thyroid cancer (TC) has been brought to a new era, but tumor metastasis still is an intractable difficulty in clinical. LncRNA ABHD11-AS1 has been confirmed to be involved in TC progression. However, its specific mechanism remains unknown. Tissues from TC patients and TC cells served as mainly experimental subjects in this study. The migration of TC cells was assessed using the scratch assay, and the ability of cell invasion was evaluated by transwell assay. RT-qPCR and western blot were conducted to determine the levels of related genes and proteins. The dual-luciferase reporter assay was used to validate the relationships among ABHD11-AS1, miR-876-5p and CALM2. ABHD11-AS1 and CALM2 are elevated in TC cancer samples and cells, while the expression of miR-876-5p is reduced. Subsequently, the ability of TC cells to migrate, invade and EMT was inhibited by both ABHD11-AS1 knockdown or miR-876-5p overexpression. Moreover, the suppression of malignant characteristics in TC cells resulting from ABHD11-AS knockdown was counteracted by the inhibition of miR-876-5p or the upregulation of CALM2. On the mechanism, ABHD11-AS1 elevated CALM2 and promoted the malignant development of TC cells by acting as a miR-876-5p sponge. ABHD11-AS1 mediated the miR-876-5p/CALM2 axis to drive the metastasis of thyroid cancer.
Collapse
Affiliation(s)
- Juan Lv
- Department of Nuclear Medicine, Yunnan Cancer Hospital, the Third Affiliated Hospital of Kunming Medical University, Peking University Cancer Hospital Yunnan, No. 519 Kunzhou Road, Xishan District, Kunming, 650118, Yunnan Province, China
| | - Fukun Chen
- Department of Nuclear Medicine, Yunnan Cancer Hospital, the Third Affiliated Hospital of Kunming Medical University, Peking University Cancer Hospital Yunnan, No. 519 Kunzhou Road, Xishan District, Kunming, 650118, Yunnan Province, China
| | - Ling Lv
- Department of Nuclear Medicine, Yunnan Cancer Hospital, the Third Affiliated Hospital of Kunming Medical University, Peking University Cancer Hospital Yunnan, No. 519 Kunzhou Road, Xishan District, Kunming, 650118, Yunnan Province, China
| | - Lu Zhang
- Department of Nuclear Medicine, Yunnan Cancer Hospital, the Third Affiliated Hospital of Kunming Medical University, Peking University Cancer Hospital Yunnan, No. 519 Kunzhou Road, Xishan District, Kunming, 650118, Yunnan Province, China
| | - Huangren Zou
- Department of Nuclear Medicine, Yunnan Cancer Hospital, the Third Affiliated Hospital of Kunming Medical University, Peking University Cancer Hospital Yunnan, No. 519 Kunzhou Road, Xishan District, Kunming, 650118, Yunnan Province, China
| | - Yanlin Liu
- Department of Nuclear Medicine, Yunnan Cancer Hospital, the Third Affiliated Hospital of Kunming Medical University, Peking University Cancer Hospital Yunnan, No. 519 Kunzhou Road, Xishan District, Kunming, 650118, Yunnan Province, China
| | - Yuke Bai
- Department of Nuclear Medicine, Yunnan Cancer Hospital, the Third Affiliated Hospital of Kunming Medical University, Peking University Cancer Hospital Yunnan, No. 519 Kunzhou Road, Xishan District, Kunming, 650118, Yunnan Province, China
| | - Ruotong Fang
- Department of Nuclear Medicine, Yunnan Cancer Hospital, the Third Affiliated Hospital of Kunming Medical University, Peking University Cancer Hospital Yunnan, No. 519 Kunzhou Road, Xishan District, Kunming, 650118, Yunnan Province, China
| | - Tiantian Qin
- Department of Nuclear Medicine, Yunnan Cancer Hospital, the Third Affiliated Hospital of Kunming Medical University, Peking University Cancer Hospital Yunnan, No. 519 Kunzhou Road, Xishan District, Kunming, 650118, Yunnan Province, China
| | - Zhiyong Deng
- Department of Nuclear Medicine, Yunnan Cancer Hospital, the Third Affiliated Hospital of Kunming Medical University, Peking University Cancer Hospital Yunnan, No. 519 Kunzhou Road, Xishan District, Kunming, 650118, Yunnan Province, China.
| |
Collapse
|
5
|
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
|
6
|
Turina P, Dal Cortivo G, Enriquez Sandoval CA, Alexov E, Ascher DB, Babbi G, Bakolitsa C, Casadio R, Fariselli P, Folkman L, Kamandula A, Katsonis P, Li D, Lichtarge O, Martelli PL, Panday SK, Pires DEV, Portelli S, Pucci F, Rodrigues CHM, Rooman M, Savojardo C, Schwersensky M, Shen Y, Strokach AV, Sun Y, Woo J, Radivojac P, Brenner SE, Dell'Orco D, Capriotti E. Assessing the predicted impact of single amino acid substitutions in calmodulin for CAGI6 challenges. Hum Genet 2025; 144:113-125. [PMID: 39714488 PMCID: PMC11975486 DOI: 10.1007/s00439-024-02720-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Accepted: 12/02/2024] [Indexed: 12/24/2024]
Abstract
Recent thermodynamic and functional studies have been conducted to evaluate the impact of amino acid substitutions on Calmodulin (CaM). The Critical Assessment of Genome Interpretation (CAGI) data provider at University of Verona (Italy) measured the melting temperature (Tm) and the percentage of unfolding (%unfold) of a set of CaM variants (CaM challenge dataset). Thermodynamic measurements for the equilibrium unfolding of CaM were obtained by monitoring far-UV Circular Dichroism as a function of temperature. These measurements were used to determine the Tm and the percentage of protein remaining unfolded at the highest temperature. The CaM challenge dataset, comprising a total of 15 single amino acid substitutions, was used to evaluate the effectiveness of computational methods in predicting the Tm and unfolding percentages associated with the variants, and categorizing them as destabilizing or not. For the sixth edition of CAGI, nine independent research groups from four continents (Asia, Australia, Europe, and North America) submitted over 52 sets of predictions, derived from various approaches. In this manuscript, we summarize the results of our assessment to highlight the potential limitations of current algorithms and provide insights into the future development of more accurate prediction tools. By evaluating the thermodynamic stability of CaM variants, this study aims to enhance our understanding of the relationship between amino acid substitutions and protein stability, ultimately contributing to more accurate predictions of the effects of genetic variants.
Collapse
Affiliation(s)
- Paola Turina
- Department of Pharmacy and Biotechnology, University of Bologna, 40126, Bologna, Italy
| | - Giuditta Dal Cortivo
- Department of Neurosciences, Biomedicine, and Movement Sciences, Section of Biological Chemistry, University of Verona, 37134, Verona, Italy
| | | | - Emil Alexov
- Department of Physics and Astronomy, Clemson University, Clemson, SC, 29634, USA
| | - David B Ascher
- Computational Biology and Clinical Informatics, Baker Heart and Diabetes Institute, Melbourne, VIC, 3004, Australia
- School of Chemistry and Molecular Biosciences, Australian Centre for Ecogenomics, University of Queensland, St Lucia, QLD, 4072, Australia
| | - Giulia Babbi
- Department of Pharmacy and Biotechnology, University of Bologna, 40126, Bologna, Italy
| | - Constantina Bakolitsa
- Department of Plant and Microbial Biology and Center for Computational Biology, University of California, Berkeley, CA, USA
| | - Rita Casadio
- Department of Pharmacy and Biotechnology, University of Bologna, 40126, Bologna, Italy
| | - Piero Fariselli
- Department of Medical Sciences, University of Torino, Turin, Italy
| | - Lukas Folkman
- Institute for Integrated and Intelligent Systems, Griffith University, Southport, QLD, Australia
| | - Akash Kamandula
- Khoury College of Computer Sciences, Northeastern University, Boston, MA, 02115, USA
| | - Panagiotis Katsonis
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Dong Li
- Computational Biology and Bioinformatics, Université Libre de Bruxelles, 50 Roosevelt Ave, 1050, Brussels, Belgium
| | - Olivier Lichtarge
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Pier Luigi Martelli
- Department of Pharmacy and Biotechnology, University of Bologna, 40126, Bologna, Italy
| | | | - Douglas E V Pires
- School of Computing and Information Systems, The University of Melbourne, Melbourne, VIC, 3053, Australia
| | - Stephanie Portelli
- Computational Biology and Clinical Informatics, Baker Heart and Diabetes Institute, Melbourne, VIC, 3004, Australia
- School of Chemistry and Molecular Biosciences, Australian Centre for Ecogenomics, University of Queensland, St Lucia, QLD, 4072, Australia
| | - Fabrizio Pucci
- Computational Biology and Bioinformatics, Université Libre de Bruxelles, 50 Roosevelt Ave, 1050, Brussels, Belgium
| | - Carlos H M Rodrigues
- Computational Biology and Clinical Informatics, Baker Heart and Diabetes Institute, Melbourne, VIC, 3004, Australia
| | - Marianne Rooman
- Computational Biology and Bioinformatics, Université Libre de Bruxelles, 50 Roosevelt Ave, 1050, Brussels, Belgium
| | - Castrense Savojardo
- Department of Pharmacy and Biotechnology, University of Bologna, 40126, Bologna, Italy
| | - Martin Schwersensky
- Computational Biology and Bioinformatics, Université Libre de Bruxelles, 50 Roosevelt Ave, 1050, Brussels, Belgium
| | - Yang Shen
- Department of Electrical and Computer Engineering Texas, A&M University, College Station, TX, USA
| | - Alexey V Strokach
- Department of Computer Science, University of Toronto, Toronto, ON, Canada
| | - Yuanfei Sun
- Department of Electrical and Computer Engineering Texas, A&M University, College Station, TX, USA
| | | | - Predrag Radivojac
- Khoury College of Computer Sciences, Northeastern University, Boston, MA, 02115, USA
| | - Steven E Brenner
- Department of Plant and Microbial Biology and Center for Computational Biology, University of California, Berkeley, CA, USA
- Biophysics Graduate Group, University of California, Berkeley, CA, 94720, USA
- Center for Computational Biology, University of California, Berkeley, CA, 94720, USA
| | - Daniele Dell'Orco
- Department of Neurosciences, Biomedicine, and Movement Sciences, Section of Biological Chemistry, University of Verona, 37134, Verona, Italy.
| | - Emidio Capriotti
- Department of Pharmacy and Biotechnology, University of Bologna, 40126, Bologna, Italy.
- Computational Genomics Platform, IRCCS University Hospital of Bologna, 40138, Bologna, Italy.
| |
Collapse
|
7
|
Gupta N, Richards EMB, Morris VS, Morris R, Wadmore K, Held M, McCormick L, Prakash O, Dart C, Helassa N. Arrhythmogenic calmodulin variants D131E and Q135P disrupt interaction with the L-type voltage-gated Ca 2+ channel (Ca v1.2) and reduce Ca 2+-dependent inactivation. Acta Physiol (Oxf) 2025; 241:e14276. [PMID: 39825574 PMCID: PMC11742489 DOI: 10.1111/apha.14276] [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: 03/16/2024] [Revised: 11/19/2024] [Accepted: 01/01/2025] [Indexed: 01/20/2025]
Abstract
AIM Long QT syndrome (LQTS) and catecholaminergic polymorphism ventricular tachycardia (CPVT) are inherited cardiac disorders often caused by mutations in ion channels. These arrhythmia syndromes have recently been associated with calmodulin (CaM) variants. Here, we investigate the impact of the arrhythmogenic variants D131E and Q135P on CaM's structure-function relationship. Our study focuses on the L-type calcium channel Cav1.2, a crucial component of the ventricular action potential and excitation-contraction coupling. METHODS We used circular dichroism (CD), 1H-15N HSQC NMR, and trypsin digestion to determine the structural and stability properties of CaM variants. The affinity of CaM for Ca2+ and interaction of Ca2+/CaM with Cav1.2 (IQ and NSCaTE domains) were investigated using intrinsic tyrosine fluorescence and isothermal titration calorimetry (ITC), respectively. The effect of CaM variants of Cav1.2 activity was determined using HEK293-Cav1.2 cells (B'SYS) and whole-cell patch-clamp electrophysiology. RESULTS Using a combination of protein biophysics and structural biology, we show that the disease-associated mutations D131E and Q135P mutations alter apo/CaM structure and stability. In the Ca2+-bound state, D131E and Q135P exhibited reduced Ca2+ binding affinity, significant structural changes, and altered interaction with Cav1.2 domains (increased affinity for Cav1.2-IQ and decreased affinity for Cav1.2-NSCaTE). We show that the mutations dramatically impair Ca2+-dependent inactivation (CDI) of Cav1.2, which would contribute to abnormal Ca2+ influx, leading to disrupted Ca2+ handling, characteristic of cardiac arrhythmia syndromes. CONCLUSIONS These findings provide insights into the molecular mechanisms behind arrhythmia caused by calmodulin mutations, contributing to our understanding of cardiac syndromes at a molecular and cellular level.
Collapse
Affiliation(s)
- Nitika Gupta
- Department of Biochemistry, Cell and Systems Biology, Institute of Systems, Molecular and Integrative Biology, Faculty of Health and Life SciencesUniversity of LiverpoolLiverpoolUK
| | - Ella M. B. Richards
- Department of Biochemistry, Cell and Systems Biology, Institute of Systems, Molecular and Integrative Biology, Faculty of Health and Life SciencesUniversity of LiverpoolLiverpoolUK
| | - Vanessa S. Morris
- Department of Biochemistry, Cell and Systems Biology, Institute of Systems, Molecular and Integrative Biology, Faculty of Health and Life SciencesUniversity of LiverpoolLiverpoolUK
| | - Rachael Morris
- Department of Biochemistry, Cell and Systems Biology, Institute of Systems, Molecular and Integrative Biology, Faculty of Health and Life SciencesUniversity of LiverpoolLiverpoolUK
| | - Kirsty Wadmore
- Department of Biochemistry, Cell and Systems Biology, Institute of Systems, Molecular and Integrative Biology, Faculty of Health and Life SciencesUniversity of LiverpoolLiverpoolUK
| | - Marie Held
- Department of Biochemistry, Cell and Systems Biology, Institute of Systems, Molecular and Integrative Biology, Faculty of Health and Life SciencesUniversity of LiverpoolLiverpoolUK
| | - Liam McCormick
- Department of Biochemistry, Cell and Systems Biology, Institute of Systems, Molecular and Integrative Biology, Faculty of Health and Life SciencesUniversity of LiverpoolLiverpoolUK
| | - Ohm Prakash
- Department of Biochemistry, Cell and Systems Biology, Institute of Systems, Molecular and Integrative Biology, Faculty of Health and Life SciencesUniversity of LiverpoolLiverpoolUK
| | - Caroline Dart
- Department of Biochemistry, Cell and Systems Biology, Institute of Systems, Molecular and Integrative Biology, Faculty of Health and Life SciencesUniversity of LiverpoolLiverpoolUK
| | - Nordine Helassa
- Department of Biochemistry, Cell and Systems Biology, Institute of Systems, Molecular and Integrative Biology, Faculty of Health and Life SciencesUniversity of LiverpoolLiverpoolUK
| |
Collapse
|
8
|
Berchtold MW, Villalobo A. Ca 2+/calmodulin signaling in organismal aging and cellular senescence: Impact on human diseases. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167583. [PMID: 39579800 DOI: 10.1016/j.bbadis.2024.167583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 11/18/2024] [Accepted: 11/18/2024] [Indexed: 11/25/2024]
Abstract
Molecular mechanisms of aging processes at the level of organisms and cells are in the focus of a large number of research laboratories. This research culminated in recent breakthroughs, which contributed to the better understanding of the natural aging process and aging associated malfunctions leading to age-related diseases. Ca2+ in connection with its master intracellular sensor protein calmodulin (CaM) regulates a plethora of crucial cellular processes orchestrating a wide range of signaling processes. This review focuses on the involvement of Ca2+/CaM in cellular mechanisms, which are associated with normal aging, as well as playing a role in the development of diseases connected with signaling processes during aging. We specifically highlight processes that involve inactivation of proteins, which take part in Ca2+/CaM regulatory systems by oxygen or nitrogen free radical species, during organismal aging and cellular senescence. As examples of organs where aging processes have recently been investigated, we chose to review the literature on molecular aging processes with involvement of Ca2+/CaM in heart and neuronal diseases, as well as in cancer and metabolic diseases, all deeply affected by aging. In addition, this article focuses on cellular senescence, a mechanism that may contribute to aging processes and therefore has been proposed as a target to interfere with the progression of age-associated diseases.
Collapse
Affiliation(s)
- Martin W Berchtold
- Department of Biology, University of Copenhagen, 13 Universitetsparken, DK-2100 Copenhagen Ø, Denmark.
| | - Antonio Villalobo
- Cancer and Human Molecular Genetics Area, Oto-Neurosurgery Research Group, University Hospital La Paz Research Institute (IdiPAZ), Paseo de la Castellana 261, E-28046 Madrid, Spain.
| |
Collapse
|
9
|
Zhang C, Shao D, Zheng X, Hao L. The mechanism of LQTS related CaM mutation E141G interfering with Ca V1.2 channels function through its C-lobe. J Physiol Biochem 2025; 81:185-197. [PMID: 39699847 DOI: 10.1007/s13105-024-01064-5] [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: 01/09/2024] [Accepted: 11/28/2024] [Indexed: 12/20/2024]
Abstract
Mutations in the CALM1-3 genes, which encode calmodulin (CaM), have been reported in clinical cases of long QT syndrome (LQTS). Specifically, the CaM mutant E141G (CaME141G) in the variant CALM1 gene has been identified as a causative factor in LQTS. This mutation disrupts the normal Ca2+-dependent inactivation (CDI) function of CaV1.2 channels. However, it is still unclear how CaME141G interferes with the regulatory role of wild-type (WT) CaM on CaV1.2 channels and leads to abnormal CDI. A CaM molecule contains two lobes with similar structure, the N-lobe and the C-lobe. In this study, a CaM-truncated C-lobe mutant E141G (C-lobeE141G) was engineered to exclude the impact of the unmutated N-lobe. Our findings revealed that at low Ca2+ concentration ([Ca2+]), the binding of C-lobeE141G to the preIQ, IQ and N-terminus (NT) of CaV1.2 channels has higher binding capacity (Bmax: 0.17, 0.22, 0.13) compared with those of WT C-lobe (Bmax: 0.04, 0.14, 0.11) in GST pull-down assay. With an increase in [Ca2+], the Ca2+-dependency for C-lobeE141G binding to CaV1.2 channels was impaired. Moreover, C-lobeE141G induced the relative channel activity to 240.58 ± 51.37% at resting [Ca2+], but it was unable to diminish the channel activity at high [Ca2+] even in the presence of WT N-lobe, which may be responsible for the abnormal CDI of CaV1.2 channels affected by the LQTS-related CaM mutation. Our research provides preliminary insights into the mechanism by which the CaM mutation interferes with CaV1.2 channels function through its C-lobe.
Collapse
Affiliation(s)
- Chenyang Zhang
- Department of Pharmaceutical Toxicology, School of Pharmacy, China Medical University, Shenyang, 110122, China
| | - Dongxue Shao
- Department of Pharmaceutical Toxicology, School of Pharmacy, China Medical University, Shenyang, 110122, China
| | - Xi Zheng
- Department of Pharmaceutical Toxicology, School of Pharmacy, China Medical University, Shenyang, 110122, China
| | - Liying Hao
- Department of Pharmaceutical Toxicology, School of Pharmacy, China Medical University, Shenyang, 110122, China.
| |
Collapse
|
10
|
Hussey JW, DeMarco E, DiSilvestre D, Brohus M, Busuioc AO, Iversen ED, Jensen HH, Nyegaard M, Overgaard MT, Ben-Johny M, Dick IE. Voltage Gated Calcium Channel Dysregulation May Contribute to Neurological Symptoms in Calmodulinopathies. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.02.626503. [PMID: 39677635 PMCID: PMC11642847 DOI: 10.1101/2024.12.02.626503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2024]
Abstract
Calmodulinopathies are caused by mutations in calmodulin (CaM), and result in debilitating cardiac arrythmias such as long-QT syndrome (LQTS) and catecholaminergic polymorphic ventricular tachycardia (CPVT). In addition, many patients exhibit neurological comorbidities, including developmental delay and autism spectrum disorder. Until now, most work into these mutations has focused on cardiac effects, identifying impairment of Ca 2+ /CaM-dependent inactivation (CDI) of Ca V 1.2 channels as a major pathogenic mechanism. However, the impact of these mutations on neurological function has yet to be fully explored. CaM regulation of voltage-gated calcium channels (VGCCs) is a critical element of neuronal function, implicating multiple VGCC subtypes in the neurological pathogenesis of calmodulinopathies. Here, we explore the potential for pathological CaM variants to impair the Ca 2+ /CaM-dependent regulation of Ca V 1.3 and Ca V 2.1, both essential for neuronal function. We find that mutations in CaM can impair the CDI of Ca V 1.3 and reduce the Ca 2+ -dependent facilitation (CDF) of Ca V 2.1 channels. We find that mutations associated with significant neurological symptoms exhibit marked effects on Ca V 1.3 CDI, with overlapping but distinct impacts on Ca V 2.1 CDF. Moreover, while the majority of CaM variants demonstrated the ability to bind the IQ region of each channel, distinct differences were noted between Ca V 1.3 and Ca V 2.1, demonstrating distinct CaM interactions across the two channel subtypes. Further, C-domain CaM variants display a reduced ability to sense Ca 2+ when in complex with the Ca V IQ domains, explaining the Ca 2+ /CaM regulation deficits. Overall, these results support the possibility that disrupted Ca 2+ /CaM regulation of VGCCs may contribute to neurological pathogenesis of calmodulinopathies.
Collapse
|
11
|
Da'as SI, Thanassoulas A, Calver BL, Saleh A, Abdelrahman D, Hasan W, Safieh-Garabedian B, Kontogianni I, Nasrallah GK, Nounesis G, Lai FA, Nomikos M. Divergent Biochemical Properties and Disparate Impact of Arrhythmogenic Calmodulin Mutations on Zebrafish Cardiac Function. J Cell Biochem 2024; 125:e30619. [PMID: 38946237 DOI: 10.1002/jcb.30619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Revised: 06/04/2024] [Accepted: 06/12/2024] [Indexed: 07/02/2024]
Abstract
Calmodulin (CaM) is a ubiquitous, small cytosolic calcium (Ca2+)-binding sensor that plays a vital role in many cellular processes by binding and regulating the activity of over 300 protein targets. In cardiac muscle, CaM modulates directly or indirectly the activity of several proteins that play a key role in excitation-contraction coupling (ECC), such as ryanodine receptor type 2 (RyR2), l-type Ca2+ (Cav1.2), sodium (NaV1.5) and potassium (KV7.1) channels. Many recent clinical and genetic studies have reported a series of CaM mutations in patients with life-threatening arrhythmogenic syndromes, such as long QT syndrome (LQTS) and catecholaminergic polymorphic ventricular tachycardia (CPVT). We recently showed that four arrhythmogenic CaM mutations (N98I, D132E, D134H, and Q136P) significantly reduce the binding of CaM to RyR2. Herein, we investigate in vivo functional effects of these CaM mutations on the normal zebrafish embryonic heart function by microinjecting complementary RNA corresponding to CaMN98I, CaMD132E, CaMD134H, and CaMQ136P mutants. Expression of CaMD132E and CaMD134H mutants results in significant reduction of the zebrafish heart rate, mimicking a severe form of human bradycardia, whereas expression of CaMQ136P results in an increased heart rate mimicking human ventricular tachycardia. Moreover, analysis of cardiac ventricular rhythm revealed that the CaMD132E and CaMN98I zebrafish groups display an irregular pattern of heart beating and increased amplitude in comparison to the control groups. Furthermore, circular dichroism spectroscopy experiments using recombinant CaM proteins reveals a decreased structural stability of the four mutants compared to the wild-type CaM protein in the presence of Ca2+. Finally, Ca2+-binding studies indicates that all CaM mutations display reduced CaM Ca2+-binding affinities, with CaMD132E exhibiting the most prominent change. Our data suggest that CaM mutations can trigger different arrhythmogenic phenotypes through multiple and complex molecular mechanisms.
Collapse
Affiliation(s)
- Sahar I Da'as
- Department of Human Genetics, Sidra Medicine, Doha, Qatar
- College of Health and Life Sciences, Hamad Bin Khalifa University, Doha, Qatar
| | | | - Brian L Calver
- Sir Geraint Evans Wales Heart Research Institute, College of Biomedical and Life Science, Cardiff University, Cardiff, UK
| | - Alaaeldin Saleh
- College of Medicine, QU Health, Qatar University, Doha, Qatar
| | | | - Waseem Hasan
- Department of Human Genetics, Sidra Medicine, Doha, Qatar
| | | | - Iris Kontogianni
- National Centre for Scientific Research "Demokritos", Agia Paraskevi, Greece
- National Technical University of Athens, Athens, Greece
| | - Gheyath K Nasrallah
- Biomedical Research Center, Qatar University, Doha, Qatar
- Department of Biological Sciences, College of Health Sciences, QU Health, Qatar University, Doha, Qatar
| | - George Nounesis
- National Centre for Scientific Research "Demokritos", Agia Paraskevi, Greece
| | - F Anthony Lai
- College of Medicine, QU Health, Qatar University, Doha, Qatar
- Sir Geraint Evans Wales Heart Research Institute, College of Biomedical and Life Science, Cardiff University, Cardiff, UK
| | - Michail Nomikos
- College of Medicine, QU Health, Qatar University, Doha, Qatar
| |
Collapse
|
12
|
Speelman-Rooms F, Vanmunster M, Coughlan A, Hinrichs M, Pontisso I, Barbeau S, Parpaite T, Bultynck G, Brohus M. 10th European Calcium Society symposium: The Ca2+-signaling toolkit in cell function, health and disease. Biol Open 2024; 13:bio060357. [PMID: 38661208 PMCID: PMC11070784 DOI: 10.1242/bio.060357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/26/2024] Open
Abstract
The 10th European Calcium Society symposium, organized in Leuven, Belgium on November 15-17, 2023, focused on the role of Ca2+ signaling in cell function, health and disease. The symposium featured six scientific sessions, 16 invited speakers - of whom two were postdoctoral researchers - and 14 short talks. The talks covered various aspects of intracellular Ca2+ signaling and its implications in pathology. Each session was opened by one or more invited speakers, followed by a series of presentations from speakers selected from submitted abstracts. Through short talks, poster presentations, awards, and sustainable travel fellowships, the symposium also fostered opportunities for the active participation of early-career researchers. At least half of the short talks were allocated to early-career researchers, thereby offering a platform for the presentation of ongoing work and unpublished results. Presentations were also broadcast in real-time for online attendees. In this Meeting Review, we aim to capture the spirit of the meeting and discuss the main take-home messages that emerged during the symposium.
Collapse
Affiliation(s)
- Femke Speelman-Rooms
- KU Leuven, Lab. Molecular & Cellular Signaling, Dep. Cellular & Molecular Medicine, Campus Gasthuisberg O/N-I bus 802, Herestraat 49, B-3000 Leuven, Belgium
- KU Leuven, Lab. Chemical Biology, Dep. Cellular & Molecular Medicine, Campus Gasthuisberg O/N-I bus 901, Herestraat 49, B-3000 Leuven, Belgium
| | - Maarten Vanmunster
- KU Leuven, Lab. Molecular & Cellular Signaling, Dep. Cellular & Molecular Medicine, Campus Gasthuisberg O/N-I bus 802, Herestraat 49, B-3000 Leuven, Belgium
| | - Aled Coughlan
- Cardiff University, Biomedicine Division, School of Biosciences, Sir Martin Evans Building, Museum Avenue, CF10 3AX, Cardiff, Wales, UK
| | - Macarena Hinrichs
- University Medical Center Hamburg-Eppendorf, The Calcium Signalling Group, Department of Biochemistry and Molecular Cell Biology, Hamburg 20251, Germany
| | - Ilaria Pontisso
- Institut de Biologie Intégrative de la Cellule (I2BC) - Université Paris-Saclay, Gif-Sur-Yvette, 91190, France
| | - Solene Barbeau
- UC Louvain, Institute of Neuroscience, Pôle Cellulaire et Moléculaire, avenue Mounier 53, 1200 Brussels, Belgium
| | - Thibaud Parpaite
- UC Louvain, Institute of Neuroscience, Pôle Cellulaire et Moléculaire, avenue Mounier 53, 1200 Brussels, Belgium
| | - Geert Bultynck
- KU Leuven, Lab. Molecular & Cellular Signaling, Dep. Cellular & Molecular Medicine, Campus Gasthuisberg O/N-I bus 802, Herestraat 49, B-3000 Leuven, Belgium
| | - Malene Brohus
- Aalborg University, Dept. Chemistry and Bioscience, Fredrik Bajers Vej 7H, 9220 Aalborg, Denmark
| |
Collapse
|
13
|
Zhang ZH, Barajas-Martinez H, Jiang H, Huang CX, Antzelevitch C, Xia H, Hu D. Gene and stem cell therapy for inherited cardiac arrhythmias. Pharmacol Ther 2024; 256:108596. [PMID: 38301770 DOI: 10.1016/j.pharmthera.2024.108596] [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: 09/26/2023] [Revised: 12/11/2023] [Accepted: 01/13/2024] [Indexed: 02/03/2024]
Abstract
Inherited cardiac arrhythmias are a group of genetic diseases predisposing to sudden cardiac arrest, mainly resulting from variants in genes encoding cardiac ion channels or proteins involved in their regulation. Currently available therapeutic options (pharmacotherapy, ablative therapy and device-based therapy) can not preclude the occurrence of arrhythmia events and/or provide complete protection. With growing understanding of the genetic background and molecular mechanisms of inherited cardiac arrhythmias, advancing insight of stem cell technology, and development of vectors and delivery strategies, gene therapy and stem cell therapy may be promising approaches for treatment of inherited cardiac arrhythmias. Recent years have witnessed impressive progress in the basic science aspects and there is a clear and urgent need to be translated into the clinical management of arrhythmic events. In this review, we present a succinct overview of gene and cell therapy strategies, and summarize the current status of gene and cell therapy. Finally, we discuss future directions for implementation of gene and cell therapy in the therapy of inherited cardiac arrhythmias.
Collapse
Affiliation(s)
- Zhong-He Zhang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, PR China; Cardiovascular Research Institute, Wuhan University, Wuhan, 430060, PR China; Hubei Key Laboratory of Cardiology, Wuhan, 430060, PR China
| | - Hector Barajas-Martinez
- Lankenau Institute for Medical Research, Lankenau Heart Institute, Wynnwood, PA, 19096, USA; Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, 19107, USA
| | - Hong Jiang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, PR China; Cardiovascular Research Institute, Wuhan University, Wuhan, 430060, PR China; Hubei Key Laboratory of Cardiology, Wuhan, 430060, PR China
| | - Cong-Xin Huang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, PR China; Cardiovascular Research Institute, Wuhan University, Wuhan, 430060, PR China; Hubei Key Laboratory of Cardiology, Wuhan, 430060, PR China
| | - Charles Antzelevitch
- Lankenau Institute for Medical Research, Lankenau Heart Institute, Wynnwood, PA, 19096, USA; Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, 19107, USA
| | - Hao Xia
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, PR China; Cardiovascular Research Institute, Wuhan University, Wuhan, 430060, PR China; Hubei Key Laboratory of Cardiology, Wuhan, 430060, PR China.
| | - Dan Hu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, PR China; Cardiovascular Research Institute, Wuhan University, Wuhan, 430060, PR China; Hubei Key Laboratory of Cardiology, Wuhan, 430060, PR China.
| |
Collapse
|
14
|
Halder R, Warshel A. Energetic and structural insights behind calcium induced conformational transition in calmodulin. Proteins 2024; 92:384-394. [PMID: 37915244 PMCID: PMC10872638 DOI: 10.1002/prot.26620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2023] [Revised: 10/01/2023] [Accepted: 10/10/2023] [Indexed: 11/03/2023]
Abstract
Calmodulin (CaM) is a key signaling protein that triggers several cellular and physiological processes inside the cell. Upon binding with calcium ion, CaM undergoes large scale conformational transition from a closed state to an open state that facilitates its interaction with various target protein and regulates their activity. This work explores the origin of the energetic and structural variation of the wild type and mutated CaM and explores the molecular origin for the structural differences between them. We first calculated the sequential calcium binding energy to CaM using the PDLD/S-LRA/β approach. This study shows a very good correlation with experimental calcium binding energies. Next we calculated the calcium binding energies to the wild type CaM and several mutated CaM systems which were reported experimentally. On the structural aspect, it has been reported experimentally that certain mutation (Q41L-K75I) in calcium bound CaM leads to complete conformational transition from an open to a closed state. By using equilibrium molecular dynamics simulation, free energy calculation and contact frequency map analysis, we have shown that the formation of a cluster of long-range hydrophobic contacts, initiated by the Q41L-K75I CaM variant is the driving force behind its closing motion. This study unravels the energetics and structural aspects behind calcium ion induced conformational changes in wild type CaM and its variant.
Collapse
Affiliation(s)
- Ritaban Halder
- Department of Chemistry, University of Southern California, Los Angeles, California, USA
| | - Arieh Warshel
- Department of Chemistry, University of Southern California, Los Angeles, California, USA
| |
Collapse
|
15
|
Yang X, Xu J, Chen X, Yao M, Pei M, Yang Y, Gao P, Zhang C, Wang Z. Co-exposure of butyl benzyl phthalate and TiO 2 nanomaterials (anatase) in Metaphire guillelmi: Gut health implications by transcriptomics. JOURNAL OF ENVIRONMENTAL MANAGEMENT 2024; 354:120429. [PMID: 38387344 DOI: 10.1016/j.jenvman.2024.120429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 01/22/2024] [Accepted: 02/17/2024] [Indexed: 02/24/2024]
Abstract
During the COVID-19 pandemic, an abundance of plastic face masks has been consumed and disposed of in the environment. In addition, substantial amounts of plastic mulch film have been used in intensive agriculture with low recovery. Butyl benzyl phthalate (BBP) and TiO2 nanomaterials (nTiO2) are widely applied in plastic products, leading to the inevitable release of BBP and nTiO2 into the soil system. However, the impact of co-exposure of BBP and nTiO2 at low concentrations on earthworms remains understudied. In the present study, transcriptomics was applied to reveal the effects of individual BBP and nTiO2 exposures at a concentration of 1 mg kg-1, along with the combined exposure of BBP and nTiO2 (1 mg kg-1 BBP + 1 mg kg-1 nTiO2 (anatase)) on Metaphire guillelmi. The result showed that BBP and nTiO2 exposures have the potential to induce neurodegeneration through glutamate accumulation, tau protein, and oxidative stress in the endoplasmic reticulum and mitochondria, as well as metabolism dysfunction. The present study contributes to our understanding of the toxic mechanisms of emerging contaminants at environmentally relevant levels and prompts consideration of the management of BBP and nTiO2 within the soil ecosystems.
Collapse
Affiliation(s)
- Xiaoqing Yang
- School of Environment & Ecology, Jiangnan University, Wuxi 214122, China
| | - Jiake Xu
- School of Environment & Ecology, Jiangnan University, Wuxi 214122, China
| | - Xiaoni Chen
- School of Environment & Ecology, Jiangnan University, Wuxi 214122, China
| | - Mengyao Yao
- School of Environment & Ecology, Jiangnan University, Wuxi 214122, China
| | - Mengyuan Pei
- School of Environment & Ecology, Jiangnan University, Wuxi 214122, China
| | - Yujian Yang
- School of Environment & Ecology, Jiangnan University, Wuxi 214122, China
| | - Peng Gao
- Department of Environmental and Occupational Health, University of Pittsburgh, Pittsburgh, PA, 15261, United States
| | - Cheng Zhang
- School of Environment & Ecology, Jiangnan University, Wuxi 214122, China.
| | - Zhenyu Wang
- School of Environment & Ecology, Jiangnan University, Wuxi 214122, China; Jiangsu Collaborative Innovation Center of Technology and Material of Water Treatment, Suzhou University of Science and Technology, 215009, China
| |
Collapse
|
16
|
Holler CV, Petersson NM, Brohus M, Niemelä MA, Iversen ED, Overgaard MT, Iwaï H, Wimmer R. Allosteric changes in protein stability and dynamics as pathogenic mechanism for calmodulin variants not affecting Ca 2+ coordinating residues. Cell Calcium 2024; 117:102831. [PMID: 37995470 DOI: 10.1016/j.ceca.2023.102831] [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: 09/11/2023] [Revised: 11/10/2023] [Accepted: 11/12/2023] [Indexed: 11/25/2023]
Abstract
Mutations in the small, calcium-sensing, protein calmodulin cause cardiac arrhythmia and can ultimately prove lethal. Here, we report the impact of the G113R variant on the structure and dynamics of the calmodulin molecule, both in the presence and in the absence of calcium. We show that the mutation introduces minor changes into the structure of calmodulin and that it changes the thermostability and thus the degree of foldedness at human body temperature. The mutation also severely impacts the intramolecular mobility of calmodulin, especially in the apo form. Glycine 113 acts as an alpha-helical C-capping residue in both apo/ - and Ca2+/calmodulin, but its exchange to arginine has very different effects on the apo and Ca2+ forms. The majority of arrhythmogenic calmodulin variants identified affects residues in the Ca2+ coordinating loops of the two C-domain EF-Hands, causing a 'direct impact on Ca2+ binding'. However, G113R lies outside a Ca2+ coordinating loop and acts differently and more similar to the previously characterized arrhythmogenic N53I. Therefore, we suggest that altered apo/CaM dynamics may be a novel general disease mechanism, defining low-calcium target affinity - or Ca2+ binding kinetics - critical for timely coordination of essential ion-channels in the excitation-contraction cycle.
Collapse
Affiliation(s)
- Christina Vallentin Holler
- Department of Chemistry and Bioscience, Aalborg University, Frederik Bajers vej 7H, 9220 Aalborg, Denmark
| | - Nina Møller Petersson
- Department of Chemistry and Bioscience, Aalborg University, Frederik Bajers vej 7H, 9220 Aalborg, Denmark
| | - Malene Brohus
- Department of Chemistry and Bioscience, Aalborg University, Frederik Bajers vej 7H, 9220 Aalborg, Denmark
| | | | - Emil Drivsholm Iversen
- Department of Chemistry and Bioscience, Aalborg University, Frederik Bajers vej 7H, 9220 Aalborg, Denmark
| | - Michael Toft Overgaard
- Department of Chemistry and Bioscience, Aalborg University, Frederik Bajers vej 7H, 9220 Aalborg, Denmark
| | - Hideo Iwaï
- Institute of Biotechnology, University of Helsinki, PO Box 65, Helsinki, FIN-00014, Finland
| | - Reinhard Wimmer
- Department of Chemistry and Bioscience, Aalborg University, Frederik Bajers vej 7H, 9220 Aalborg, Denmark.
| |
Collapse
|
17
|
Mackiewicz J, Lisek M, Boczek T. Targeting CaN/NFAT in Alzheimer's brain degeneration. Front Immunol 2023; 14:1281882. [PMID: 38077352 PMCID: PMC10701682 DOI: 10.3389/fimmu.2023.1281882] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Accepted: 11/06/2023] [Indexed: 12/18/2023] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder characterized by a progressive loss of cognitive functions. While the exact causes of this debilitating disorder remain elusive, numerous investigations have characterized its two core pathologies: the presence of β-amyloid plaques and tau tangles. Additionally, multiple studies of postmortem brain tissue, as well as results from AD preclinical models, have consistently demonstrated the presence of a sustained inflammatory response. As the persistent immune response is associated with neurodegeneration, it became clear that it may also exacerbate other AD pathologies, providing a link between the initial deposition of β-amyloid plaques and the later development of neurofibrillary tangles. Initially discovered in T cells, the nuclear factor of activated T-cells (NFAT) is one of the main transcription factors driving the expression of inflammatory genes and thus regulating immune responses. NFAT-dependent production of inflammatory mediators is controlled by Ca2+-dependent protein phosphatase calcineurin (CaN), which dephosphorylates NFAT and promotes its transcriptional activity. A substantial body of evidence has demonstrated that aberrant CaN/NFAT signaling is linked to several pathologies observed in AD, including neuronal apoptosis, synaptic deficits, and glia activation. In view of this, the role of NFAT isoforms in AD has been linked to disease progression at different stages, some of which are paralleled to diminished cognitive status. The use of classical inhibitors of CaN/NFAT signaling, such as tacrolimus or cyclosporine, or adeno-associated viruses to specifically inhibit astrocytic NFAT activation, has alleviated some symptoms of AD by diminishing β-amyloid neurotoxicity and neuroinflammation. In this article, we discuss the recent findings related to the contribution of CaN/NFAT signaling to the progression of AD and highlight the possible benefits of targeting this pathway in AD treatment.
Collapse
Affiliation(s)
| | | | - Tomasz Boczek
- Department of Molecular Neurochemistry, Medical University of Lodz, Lodz, Poland
| |
Collapse
|
18
|
Kang PW, Woodbury L, Angsutararux P, Sambare N, Shi J, Marras M, Abella C, Bedi A, Zinn D, Cui J, Silva JR. Arrhythmia-associated calmodulin variants interact with KCNQ1 to confer aberrant membrane trafficking and function. PNAS NEXUS 2023; 2:pgad335. [PMID: 37965565 PMCID: PMC10642763 DOI: 10.1093/pnasnexus/pgad335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Accepted: 10/04/2023] [Indexed: 11/16/2023]
Abstract
Missense variants in calmodulin (CaM) predispose patients to arrhythmias associated with high mortality rates ("calmodulinopathy"). As CaM regulates many key cardiac ion channels, an understanding of disease mechanism associated with CaM variant arrhythmias requires elucidating individual CaM variant effects on distinct channels. One key CaM regulatory target is the KCNQ1 (KV7.1) voltage-gated potassium channel that carries the IKs current. Yet, relatively little is known as to how CaM variants interact with KCNQ1 or affect its function. Here, we take a multipronged approach employing a live-cell fluorescence resonance energy transfer binding assay, fluorescence trafficking assay, and functional electrophysiology to characterize >10 arrhythmia-associated CaM variants for effect on KCNQ1 CaM binding, membrane trafficking, and channel function. We identify one variant (G114W) that exhibits severely weakened binding to KCNQ1 but find that most other CaM variants interact with similar binding affinity to KCNQ1 when compared with CaM wild-type over physiological Ca2+ ranges. We further identify several CaM variants that affect KCNQ1 and IKs membrane trafficking and/or baseline current activation kinetics, thereby delineating KCNQ1 dysfunction in calmodulinopathy. Lastly, we identify CaM variants with no effect on KCNQ1 function. This study provides extensive functional data that reveal how CaM variants contribute to creating a proarrhythmic substrate by causing abnormal KCNQ1 membrane trafficking and current conduction. We find that CaM variant regulation of KCNQ1 is not uniform with effects varying from benign to significant loss of function, suggesting how CaM variants predispose patients to arrhythmia via the dysregulation of multiple cardiac ion channels. Classification: Biological, Health, and Medical Sciences, Physiology.
Collapse
Affiliation(s)
- Po wei Kang
- Department of Biomedical Engineering, Washington University in St.Louis, St. Louis, MO 63130, USA
| | - Lucy Woodbury
- Department of Biomedical Engineering, Washington University in St.Louis, St. Louis, MO 63130, USA
| | - Paweorn Angsutararux
- Department of Biomedical Engineering, Washington University in St.Louis, St. Louis, MO 63130, USA
| | - Namit Sambare
- Department of Biomedical Engineering, Washington University in St.Louis, St. Louis, MO 63130, USA
| | - Jingyi Shi
- Department of Biomedical Engineering, Washington University in St.Louis, St. Louis, MO 63130, USA
| | - Martina Marras
- Department of Biomedical Engineering, Washington University in St.Louis, St. Louis, MO 63130, USA
| | - Carlota Abella
- Department of Biomedical Engineering, Washington University in St.Louis, St. Louis, MO 63130, USA
| | - Anish Bedi
- Department of Biomedical Engineering, Washington University in St.Louis, St. Louis, MO 63130, USA
| | - DeShawn Zinn
- Department of Biomedical Engineering, Washington University in St.Louis, St. Louis, MO 63130, USA
| | - Jianmin Cui
- Department of Biomedical Engineering, Washington University in St.Louis, St. Louis, MO 63130, USA
| | - Jonathan R Silva
- Department of Biomedical Engineering, Washington University in St.Louis, St. Louis, MO 63130, USA
| |
Collapse
|
19
|
Thanassoulas A, Theodoridou M, Barrak L, Riguene E, Alyaarabi T, Elrayess MA, Lai FA, Nomikos M. Arrhythmia-Associated Calmodulin E105A Mutation Alters the Binding Affinity of CaM to a Ryanodine Receptor 2 CaM-Binding Pocket. Int J Mol Sci 2023; 24:15630. [PMID: 37958614 PMCID: PMC10649572 DOI: 10.3390/ijms242115630] [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: 09/23/2023] [Revised: 10/21/2023] [Accepted: 10/24/2023] [Indexed: 11/15/2023] Open
Abstract
Calmodulin (CaM) is a small, multifunctional calcium (Ca2+)-binding sensor that binds and regulates the open probability of cardiac ryanodine receptor 2 (RyR2) at both low and high cytosolic Ca2+ concentrations. Recent isothermal titration calorimetry (ITC) studies of a number of peptides that correspond to different regions of human RyR2 showed that two regions of human RyR2 (3584-3602aa and 4255-4271aa) bind with high affinity to CaM, suggesting that these two regions might contribute to a putative RyR2 intra-subunit CaM-binding pocket. Moreover, a previously characterized de novo long QT syndrome (LQTS)-associated missense CaM mutation (E105A) which was identified in a 6-year-old boy, who experienced an aborted first episode of cardiac arrest revealed that this mutation dysregulates normal cardiac function in zebrafish by a complex mechanism that involves alterations in both CaM-Ca2+ and CaM-RyR2 interactions. Herein, to gain further insight into how the CaM E105A mutation leads to severe cardiac arrhythmia, we generated large quantities of recombinant CaMWT and CaME105A proteins. We then performed ITC experiments to investigate and compare the interactions of CaMWT and CaME105A mutant protein with two synthetic peptides that correspond to the two aforementioned human RyR2 regions, which we have proposed to contribute to the RyR2 CaM-binding pocket. Our data reveal that the E105A mutation has a significant negative effect on the interaction of CaM with both RyR2 regions in the presence and absence of Ca2+, highlighting the potential contribution of these two human RyR2 regions to an RyR2 CaM-binding pocket, which may be essential for physiological CaM/RyR2 association and thus channel regulation.
Collapse
Affiliation(s)
- Angelos Thanassoulas
- College of Medicine, QU Health, Qatar University, Doha P.O. Box 2713, Qatar; (A.T.); (L.B.); (E.R.); (T.A.); (M.A.E.); (F.A.L.)
| | - Maria Theodoridou
- Biomedical Research Center, Qatar University, Doha P.O. Box 2713, Qatar;
| | - Laila Barrak
- College of Medicine, QU Health, Qatar University, Doha P.O. Box 2713, Qatar; (A.T.); (L.B.); (E.R.); (T.A.); (M.A.E.); (F.A.L.)
| | - Emna Riguene
- College of Medicine, QU Health, Qatar University, Doha P.O. Box 2713, Qatar; (A.T.); (L.B.); (E.R.); (T.A.); (M.A.E.); (F.A.L.)
| | - Tamader Alyaarabi
- College of Medicine, QU Health, Qatar University, Doha P.O. Box 2713, Qatar; (A.T.); (L.B.); (E.R.); (T.A.); (M.A.E.); (F.A.L.)
| | - Mohamed A. Elrayess
- College of Medicine, QU Health, Qatar University, Doha P.O. Box 2713, Qatar; (A.T.); (L.B.); (E.R.); (T.A.); (M.A.E.); (F.A.L.)
- Biomedical Research Center, Qatar University, Doha P.O. Box 2713, Qatar;
| | - F. Anthony Lai
- College of Medicine, QU Health, Qatar University, Doha P.O. Box 2713, Qatar; (A.T.); (L.B.); (E.R.); (T.A.); (M.A.E.); (F.A.L.)
| | - Michail Nomikos
- College of Medicine, QU Health, Qatar University, Doha P.O. Box 2713, Qatar; (A.T.); (L.B.); (E.R.); (T.A.); (M.A.E.); (F.A.L.)
| |
Collapse
|
20
|
Kan Z, Zhang S, Liao G, Niu Z, Liu X, Sun Z, Hu X, Zhang Y, Xu S, Zhang J, Zou H, Zhang X, Song Z. Mechanism of Lactiplantibacillus plantarum regulating Ca 2+ affecting the replication of PEDV in small intestinal epithelial cells. Front Microbiol 2023; 14:1251275. [PMID: 37840713 PMCID: PMC10569473 DOI: 10.3389/fmicb.2023.1251275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Accepted: 09/05/2023] [Indexed: 10/17/2023] Open
Abstract
Porcine epidemic diarrhea virus (PEDV) mainly invades the small intestine and promotes an inflammatory response, eventually leading to severe diarrhea, vomiting, dehydration, and even death of piglets, which seriously threatens the economic development of pig farming. In recent years, researchers have found that probiotics can improve the intestinal microenvironment and reduce diarrhea. At the same time, certain probiotics have been shown to have antiviral effects; however, their mechanisms are different. Herein, we aimed to investigate the inhibitory effect of Lactiplantibacillus plantarum supernatant (LP-1S) on PEDV and its mechanism. We used IPEC-J2 cells as a model to assess the inhibitory effect of LP-1S on PEDV and to further investigate the relationship between LP-1S, Ca2+, and PEDV. The results showed that a divalent cation chelating agent (EGTA) and calcium channel inhibitors (Bepridil hydrochloride and BAPTA-acetoxymethylate) could inhibit PEDV proliferation while effectively reducing the intracellular Ca2+ concentration. Furthermore, LP-1S could reduce PEDV-induced loss of calcium channel proteins (TRPV6 and PMCA1b), alleviate intracellular Ca2+ accumulation caused by PEDV infection, and promote the balance of intra- and extracellular Ca2+ concentrations, thereby inhibiting PEDV proliferation. In summary, we found that LP-1S has potential therapeutic value against PEDV, which is realized by modulating Ca2+. This provides a potential new drug to treat PEDV infection.
Collapse
Affiliation(s)
- Zifei Kan
- College of Veterinary Medicine, Southwest University, Chongqing, China
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Shujuan Zhang
- College of Veterinary Medicine, Southwest University, Chongqing, China
| | - Guisong Liao
- College of Veterinary Medicine, Southwest University, Chongqing, China
| | - Zheng Niu
- College of Veterinary Medicine, Southwest University, Chongqing, China
- College of Veterinary Medicine, Northwest A and F University, Shanxi, China
| | - Xiangyang Liu
- College of Veterinary Medicine, Southwest University, Chongqing, China
- College of Veterinary Medicine, Xinjiang Agricultural University, Ürümqi, China
| | - Zhiwei Sun
- College of Veterinary Medicine, Southwest University, Chongqing, China
| | - Xia Hu
- College of Veterinary Medicine, Southwest University, Chongqing, China
| | - Yiling Zhang
- College of Veterinary Medicine, Southwest University, Chongqing, China
- College of Animal Scienceand Technology, Chongqing Three Gorges Vocational College, Chongqing, China
| | - Shasha Xu
- College of Veterinary Medicine, Southwest University, Chongqing, China
| | - Jingyi Zhang
- College of Veterinary Medicine, Southwest University, Chongqing, China
| | - Hong Zou
- College of Veterinary Medicine, Southwest University, Chongqing, China
| | - Xingcui Zhang
- College of Veterinary Medicine, Southwest University, Chongqing, China
| | - Zhenhui Song
- College of Veterinary Medicine, Southwest University, Chongqing, China
- Immunology Research Center, Medical Research Institute, Southwest University, Chongqing, China
| |
Collapse
|
21
|
McCormick L, Wadmore K, Milburn A, Gupta N, Morris R, Held M, Prakash O, Carr J, Barrett‐Jolley R, Dart C, Helassa N. Long QT syndrome-associated calmodulin variants disrupt the activity of the slowly activating delayed rectifier potassium channel. J Physiol 2023; 601:3739-3764. [PMID: 37428651 PMCID: PMC10952621 DOI: 10.1113/jp284994] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Accepted: 06/21/2023] [Indexed: 07/12/2023] Open
Abstract
Calmodulin (CaM) is a highly conserved mediator of calcium (Ca2+ )-dependent signalling and modulates various cardiac ion channels. Genotyping has revealed several CaM mutations associated with long QT syndrome (LQTS). LQTS patients display prolonged ventricular recovery times (QT interval), increasing their risk of incurring life-threatening arrhythmic events. Loss-of-function mutations to Kv7.1 (which drives the slow delayed rectifier potassium current, IKs, a key ventricular repolarising current) are the largest contributor to congenital LQTS (>50% of cases). CaM modulates Kv7.1 to produce a Ca2+ -sensitive IKs, but little is known about the consequences of LQTS-associated CaM mutations on Kv7.1 function. Here, we present novel data characterising the biophysical and modulatory properties of three LQTS-associated CaM variants (D95V, N97I and D131H). We showed that mutations induced structural alterations in CaM and reduced affinity for Kv7.1, when compared with wild-type (WT). Using HEK293T cells expressing Kv7.1 channel subunits (KCNQ1/KCNE1) and patch-clamp electrophysiology, we demonstrated that LQTS-associated CaM variants reduced current density at systolic Ca2+ concentrations (1 μm), revealing a direct QT-prolonging modulatory effect. Our data highlight for the first time that LQTS-associated perturbations to CaM's structure impede complex formation with Kv7.1 and subsequently result in reduced IKs. This provides a novel mechanistic insight into how the perturbed structure-function relationship of CaM variants contributes to the LQTS phenotype. KEY POINTS: Calmodulin (CaM) is a ubiquitous, highly conserved calcium (Ca2+ ) sensor playing a key role in cardiac muscle contraction. Genotyping has revealed several CaM mutations associated with long QT syndrome (LQTS), a life-threatening cardiac arrhythmia syndrome. LQTS-associated CaM variants (D95V, N97I and D131H) induced structural alterations, altered binding to Kv7.1 and reduced IKs. Our data provide a novel mechanistic insight into how the perturbed structure-function relationship of CaM variants contributes to the LQTS phenotype.
Collapse
Affiliation(s)
- Liam McCormick
- Department of Biochemistry, Cell and Systems Biology, Institute of Systems, Molecular and Integrative Biology, Faculty of Health and Life SciencesUniversity of LiverpoolLiverpoolUK
- Manchester Centre for Genomic Medicine, North West Genomic Laboratory HubSaint Mary's HospitalManchesterUK
| | - Kirsty Wadmore
- Department of Biochemistry, Cell and Systems Biology, Institute of Systems, Molecular and Integrative Biology, Faculty of Health and Life SciencesUniversity of LiverpoolLiverpoolUK
| | - Amy Milburn
- Department of Biochemistry, Cell and Systems Biology, Institute of Systems, Molecular and Integrative Biology, Faculty of Health and Life SciencesUniversity of LiverpoolLiverpoolUK
| | - Nitika Gupta
- Department of Biochemistry, Cell and Systems Biology, Institute of Systems, Molecular and Integrative Biology, Faculty of Health and Life SciencesUniversity of LiverpoolLiverpoolUK
| | - Rachael Morris
- Department of Biochemistry, Cell and Systems Biology, Institute of Systems, Molecular and Integrative Biology, Faculty of Health and Life SciencesUniversity of LiverpoolLiverpoolUK
| | - Marie Held
- Department of Biochemistry, Cell and Systems Biology, Institute of Systems, Molecular and Integrative Biology, Faculty of Health and Life SciencesUniversity of LiverpoolLiverpoolUK
| | - Ohm Prakash
- Department of Biochemistry, Cell and Systems Biology, Institute of Systems, Molecular and Integrative Biology, Faculty of Health and Life SciencesUniversity of LiverpoolLiverpoolUK
| | - Joseph Carr
- Department of Biochemistry, Cell and Systems Biology, Institute of Systems, Molecular and Integrative Biology, Faculty of Health and Life SciencesUniversity of LiverpoolLiverpoolUK
| | - Richard Barrett‐Jolley
- Department of Musculoskeletal and Ageing Science, Institute of Life Course and Medical Sciences, Faculty of Health and Life SciencesUniversity of LiverpoolLiverpoolUK
| | - Caroline Dart
- Department of Biochemistry, Cell and Systems Biology, Institute of Systems, Molecular and Integrative Biology, Faculty of Health and Life SciencesUniversity of LiverpoolLiverpoolUK
| | - Nordine Helassa
- Department of Biochemistry, Cell and Systems Biology, Institute of Systems, Molecular and Integrative Biology, Faculty of Health and Life SciencesUniversity of LiverpoolLiverpoolUK
| |
Collapse
|
22
|
Gao X, Lin J, Sun L, Hu J, Gao W, Yu J. Activation of the N-methyl-D-aspartate receptor and calcium/calmodulin-dependent protein kinase IIα signal in the rostral anterior cingulate cortex is involved in pain-related aversion in rats with peripheral nerve injury. Behav Brain Res 2023; 452:114560. [PMID: 37394125 DOI: 10.1016/j.bbr.2023.114560] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 05/28/2023] [Accepted: 06/29/2023] [Indexed: 07/04/2023]
Abstract
The rostral anterior cingulate cortex (rACC) of rat brain is associated with pain-related emotions. However, the underlying molecular mechanism remains unclear. Here, we investigated the effects of the N-methyl-D-aspartate (NMDA) receptor and Ca2+/Calmodulin-dependent protein kinase type II (CaMKII)α signal on pain-related aversion in the rACC of a rat model of neuropathic pain (NP). Mechanical and thermal hyperalgesia were examined using von Frey and hot plate tests in a rat model of NP induced by spared nerve injury (SNI) of the unilateral sciatic nerve. Bilateral rACC pretreatment with the CaMKII inhibitor tat-CN21 (derived from the cell-penetrating tat sequence and CaM-KIIN amino acids 43-63) or tat-Ctrl (the tat sequence and the scrambled sequence of CN21) was performed on postoperative days 29-35 in Sham rats or rats with SNI. Spatial memory performance was tested using an eight-arm radial maze on postoperative days 34-35. Pain-related negative emotions (aversions) were evaluated using the place escape/avoidance paradigm on postoperative day 35 following the spatial memory performance test. The percentage of time spent in the light area was used to assess pain-related negative emotions (i.e., aversion). The expression levels of the NMDA receptor GluN2B subunit, CaMKIIα, and CaMKII-Threonine at position 286 (Thr286) phosphorylation in contralateral rACC specimens were detected by Western blot or real time PCR following the aversion test. Our data showed that pretreatment of the rACC with tat-CN21 increased determinate behavior but did not alter hyperalgesia or spatial memory performance in rats with SNI. In addition, tat-CN21 reversed the enhanced CaMKII-Thr286 phosphorylation and had no effect on the upregulated expression of GluN2B, CaMKIIα protein, and mRNA. Our data suggested that activation of the NMDA receptor-CaMKIIα signal in rACC is associated with pain-related aversion in rats with NP. These data may provide a new approach for the development of drugs that modulate cognitive and emotional pain aspects.
Collapse
Affiliation(s)
- Xueqi Gao
- Experimental Center for Medical Research, School of Anesthesiology, Weifang Medical University, Weifang 261053, China
| | - Jinhai Lin
- Experimental Center for Medical Research, School of Anesthesiology, Weifang Medical University, Weifang 261053, China
| | - Lin Sun
- School of Psychology, Weifang Medical University, Weifang 261053, China
| | - Jun Hu
- Department of Anesthesiology, Weifang People's Hospital, Weifang 261044, China
| | - Wenjie Gao
- Department of Anesthesiology, Weifang People's Hospital, Weifang 261044, China
| | - Jianfeng Yu
- Experimental Center for Medical Research, School of Anesthesiology, Weifang Medical University, Weifang 261053, China.
| |
Collapse
|
23
|
Williams RB, Alam Afsar MN, Tikunova S, Kou Y, Fang X, Somarathne RP, Gyawu RF, Knotts GM, Agee TA, Garcia SA, Losordo LD, Fitzkee NC, Kekenes-Huskey PM, Davis JP, Johnson CN. Human disease-associated calmodulin mutations alter calcineurin function through multiple mechanisms. Cell Calcium 2023; 113:102752. [PMID: 37245392 PMCID: PMC10330910 DOI: 10.1016/j.ceca.2023.102752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2022] [Revised: 04/29/2023] [Accepted: 05/03/2023] [Indexed: 05/30/2023]
Abstract
Calmodulin (CaM) is a ubiquitous, calcium-sensing protein that regulates a multitude of processes throughout the body. In response to changes in [Ca2+], CaM modifies, activates, and deactivates enzymes and ion channels, as well as many other cellular processes. The importance of CaM is highlighted by the conservation of an identical amino acid sequence in all mammals. Alterations to CaM amino acid sequence were once thought to be incompatible with life. During the last decade modifications to the CaM protein sequence have been observed in patients suffering from life-threatening heart disease (calmodulinopathy). Thus far, inadequate or untimely interaction between mutant CaM and several proteins (LTCC, RyR2, and CaMKII) have been identified as mechanisms underlying calmodulinopathy. Given the extensive number of CaM interactions in the body, there are likely many consequences for altering CaM protein sequence. Here, we demonstrate that disease-associated CaM mutations alter the sensitivity and activity of the Ca2+-CaM-enhanced serine/threonine phosphatase calcineurin (CaN). Biophysical characterization by circular dichroism, solution NMR spectroscopy, stopped-flow kinetic measurements, and MD simulations provide mechanistic insight into mutation dysfunction as well as highlight important aspects of CaM Ca2+ signal transduction. We find that individual CaM point mutations (N53I, F89L, D129G, and F141L) impair CaN function, however, the mechanisms are not the same. Specifically, individual point mutations can influence or modify the following properties: CaM binding, Ca2+ binding, and/or Ca2+kinetics. Moreover, structural aspects of the CaNCaM complex can be altered in manners that indicate changes to allosteric transmission of CaM binding to the enzyme active site. Given that loss of CaN function can be fatal, as well as evidence that CaN modifies ion channels already associated with calmodulinopathy, our results raise the possibility that altered CaN function contributes to calmodulinopathy.
Collapse
Affiliation(s)
- Ryan B Williams
- Department of Chemistry, Mississippi State University, Starkville MS 39759, U.S.A
| | - Md Nure Alam Afsar
- Department of Chemistry, Mississippi State University, Starkville MS 39759, U.S.A
| | - Svetlana Tikunova
- Department of Physiology and Cell Biology, College of Medicine, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus OH 43210, U.S.A
| | - Yongjun Kou
- Department of Physiology and Cell Biology, College of Medicine, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus OH 43210, U.S.A
| | - Xuan Fang
- Department of Cell and Molecular Physiology, Loyola University of Chicago, Maywood Illinois 60153, U.S.A
| | - Radha P Somarathne
- Department of Chemistry, Mississippi State University, Starkville MS 39759, U.S.A
| | - Rita F Gyawu
- Department of Chemistry, Mississippi State University, Starkville MS 39759, U.S.A
| | - Garrett M Knotts
- Department of Chemistry, Mississippi State University, Starkville MS 39759, U.S.A
| | - Taylor A Agee
- Department of Chemistry, Mississippi State University, Starkville MS 39759, U.S.A
| | - Sara A Garcia
- Department of Chemistry, Mississippi State University, Starkville MS 39759, U.S.A
| | - Luke D Losordo
- Department of Chemistry, Mississippi State University, Starkville MS 39759, U.S.A
| | - Nicholas C Fitzkee
- Department of Chemistry, Mississippi State University, Starkville MS 39759, U.S.A
| | - Peter M Kekenes-Huskey
- Department of Cell and Molecular Physiology, Loyola University of Chicago, Maywood Illinois 60153, U.S.A
| | - Jonathan P Davis
- Department of Physiology and Cell Biology, College of Medicine, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus OH 43210, U.S.A.
| | - Christopher N Johnson
- Department of Chemistry, Mississippi State University, Starkville MS 39759, U.S.A; Vanderbilt Center for Arrhythmia Research and Therapeutics, Nashville TN 37232, U.S.A.
| |
Collapse
|
24
|
Villalobo A. Regulation of ErbB Receptors by the Ca2+ Sensor Protein Calmodulin in Cancer. Biomedicines 2023; 11:biomedicines11030661. [PMID: 36979639 PMCID: PMC10045772 DOI: 10.3390/biomedicines11030661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Revised: 02/17/2023] [Accepted: 02/19/2023] [Indexed: 02/24/2023] Open
Abstract
Overexpression and mutations of the epidermal growth factor receptor (EGFR/ErbB1/HER1) and other tyrosine kinase receptors of the ErbB family (ErbB2/HER2, ErbB3/HER3 and ErbB4/HER4) play an essential role in enhancing the proliferation, the migratory capacity and invasiveness of many tumor cells, leading to cancer progression and increased malignancy. To understand these cellular processes in detail is essential to understand at a molecular level the signaling pathways and regulatory mechanisms controlling these receptors. In this regard, calmodulin (CaM) is a Ca2+-sensor protein that directly interacts with and regulates ErbB receptors, as well as some CaM-dependent kinases that also regulate these receptors, particularly EGFR and ErbB2, adding an additional layer of CaM-dependent regulation to this system. In this short review, an update of recent advances in this area is presented, covering the direct action of Ca2+/CaM on the four ErbB family members mostly in tumor cells and the indirect action of Ca2+/CaM on the receptors via CaM-regulated kinases. It is expected that further understanding of the CaM-dependent mechanisms regulating the ErbB receptors in future studies could identify new therapeutic targets in these systems that could help to control or delay cancer progression.
Collapse
Affiliation(s)
- Antonio Villalobo
- Cancer and Human Molecular Genetics Area-Oto-Neurosurgery Research Group, University Hospital La Paz Research Institute (IdiPAZ), Paseo de la Castellana 261, E-28046 Madrid, Spain
| |
Collapse
|
25
|
Kang PW, Woodbury L, Angsutararux P, Sambare N, Shi J, Marras M, Abella C, Bedi A, Zinn D, Cui J, Silva JR. Arrhythmia-associated Calmodulin Variants Interact with KCNQ1 to Confer Aberrant Membrane Trafficking and Function. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.28.526031. [PMID: 36747728 PMCID: PMC9900995 DOI: 10.1101/2023.01.28.526031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Rationale Missense variants in calmodulin (CaM) predispose patients to arrhythmias associated with high mortality rates. As CaM regulates several key cardiac ion channels, a mechanistic understanding of CaM variant-associated arrhythmias requires elucidating individual CaM variant effect on distinct channels. One key CaM regulatory target is the KCNQ1 (K V 7.1) voltage-gated potassium channel that underlie the I Ks current. Yet, relatively little is known as to how CaM variants interact with KCNQ1 or affect its function. Objective To observe how arrhythmia-associated CaM variants affect binding to KCNQ1, channel membrane trafficking, and KCNQ1 function. Methods and Results We combine a live-cell FRET binding assay, fluorescence trafficking assay, and functional electrophysiology to characterize >10 arrhythmia-associated CaM variants effect on KCNQ1. We identify one variant (G114W) that exhibits severely weakened binding to KCNQ1 but find that most other CaM variants interact with similar binding affinity to KCNQ1 when compared to CaM wild-type over physiological Ca 2+ ranges. We further identify several CaM variants that affect KCNQ1 and I Ks membrane trafficking and/or baseline current activation kinetics, thereby contextualizing KCNQ1 dysfunction in calmodulinopathy. Lastly, we delineate CaM variants with no effect on KCNQ1 function. Conclusions This study provides comprehensive functional data that reveal how CaM variants contribute to creating a pro-arrhythmic substrate by causing abnormal KCNQ1 membrane trafficking and current conduction. We find that CaM variant regulation of KCNQ1 is not uniform with effects varying from benign to significant loss of function. This study provides a new approach to collecting details of CaM binding that are key for understanding how CaM variants predispose patients to arrhythmia via the dysregulation of multiple cardiac ion channels.
Collapse
|