Dysregulation of Calcium Signalling in Disease

A special issue of Cells (ISSN 2073-4409). This special issue belongs to the section "Cell Signaling".

Deadline for manuscript submissions: closed (15 March 2021) | Viewed by 44280

Special Issue Editor


E-Mail Website
Guest Editor
Department of Clinical and Molecular Sciences, Marche Polytechnic University, 60126 Ancona, Italy
Interests: calcium; mitochondria; Mitochondrial Ca2+ Uniporter (MCU) complex; ER-mitochondria contacts; Mitochondria-Associated Membranes (MAMs); Ca2+-mediated apoptosis

Special Issue Information

Dear Colleagues,

Calcium is the major second messenger inside the cell and acts as the most prominent signaler in a plethora of biological processes. Its homeostasis is preserved by a sophisticated system of channels, pumps, and exchangers, which ensure a low cytoplasmic Ca2+ concentration at resting conditions and prompt Ca2+ elevation upon specific stimulations.

Importantly, Ca2+ regulation is not symmetrical throughout the cell, and different subcellular districts, such as endoplasmic reticulum, mitochondria or lysosomes, are equipped with peculiar Ca2+ regulating machinery. In the last years, fantastic advances have been made in the molecular characterization of novel Ca2+ players or in the identification of unexpected Ca2+ signaling routes. It is now clear that Ca2+ alterations in a specific organelle or mutations in Ca2+ transporters and/or Ca2+-related pathways have a huge impact on the activities and functionalities of the entire cell. Therefore, it is not surprising that Ca2+ dyshomeostasis or deregulation of a single Ca2+-permeable channel have been associated with multiple pathological scenarios, including cardiovascular and neurological defects, metabolic disorders, muscular diseases, and cancers.

This Special Issue of Cells is dedicated to the role of imbalanced Ca2+ signaling in disease. Contributions in the form of original experimental articles, up-to-date reviews, or short communications are welcome. We hope that this collection of papers may help the readers to point out the contribution and the mechanistic role of Ca2+ in different pathological contexts.

Dr. Saverio Marchi
Guest Editor

Manuscript Submission Information

Manuscripts should be submitted online at www.mdpi.com by registering and logging in to this website. Once you are registered, click here to go to the submission form. Manuscripts can be submitted until the deadline. All submissions that pass pre-check are peer-reviewed. Accepted papers will be published continuously in the journal (as soon as accepted) and will be listed together on the special issue website. Research articles, review articles as well as short communications are invited. For planned papers, a title and short abstract (about 100 words) can be sent to the Editorial Office for announcement on this website.

Submitted manuscripts should not have been published previously, nor be under consideration for publication elsewhere (except conference proceedings papers). All manuscripts are thoroughly refereed through a single-blind peer-review process. A guide for authors and other relevant information for submission of manuscripts is available on the Instructions for Authors page. Cells is an international peer-reviewed open access semimonthly journal published by MDPI.

Please visit the Instructions for Authors page before submitting a manuscript. The Article Processing Charge (APC) for publication in this open access journal is 2700 CHF (Swiss Francs). Submitted papers should be well formatted and use good English. Authors may use MDPI's English editing service prior to publication or during author revisions.

Keywords

  • Calcium
  • intracellular organelles
  • plasma membrane
  • store-operated Ca2+ entry (SOCE)
  • inositol 1,4,5 trisphosphate receptors (IP3Rs)
  • sarco/endoplasmic reticulum Ca2+-ATPase (SERCA)
  • mitochondrial calcium uniporter (MCU)
  • sodium/calcium exchanger
  • ORAI and STIM proteins
  • mitochondrial permeability transition pore (mPTP)
  • TRP channels
  • cardiovascular diseases
  • neurodegenerative diseases
  • metabolic diseases
  • ageing
  • muscular diseases
  • cancer

Published Papers (11 papers)

Order results
Result details
Select all
Export citation of selected articles as:

Research

Jump to: Review

23 pages, 5509 KiB  
Article
Transient Receptor Potential C 1/4/5 Is a Determinant of MTI-101 Induced Calcium Influx and Cell Death in Multiple Myeloma
by Osama M. Elzamzamy, Brandon E. Johnson, Wei-Chih Chen, Gangqing Hu, Reinhold Penner and Lori A. Hazlehurst
Cells 2021, 10(6), 1490; https://doi.org/10.3390/cells10061490 - 13 Jun 2021
Cited by 5 | Viewed by 2659
Abstract
Multiple myeloma (MM) is a currently incurable hematologic cancer. Patients that initially respond to therapeutic intervention eventually relapse with drug resistant disease. Thus, novel treatment strategies are critically needed to improve patient outcomes. Our group has developed a novel cyclic peptide referred to [...] Read more.
Multiple myeloma (MM) is a currently incurable hematologic cancer. Patients that initially respond to therapeutic intervention eventually relapse with drug resistant disease. Thus, novel treatment strategies are critically needed to improve patient outcomes. Our group has developed a novel cyclic peptide referred to as MTI-101 for the treatment of MM. We previously reported that acquired resistance to HYD-1, the linear form of MTI-101, correlated with the repression of genes involved in store operated Ca2+ entry (SOCE): PLCβ, SERCA, ITPR3, and TRPC1 expression. In this study, we sought to determine the role of TRPC1 heteromers in mediating MTI-101 induced cationic flux. Our data indicate that, consistent with the activation of TRPC heteromers, MTI-101 treatment induced Ca2+ and Na+ influx. However, replacing extracellular Na+ with NMDG did not reduce MTI-101-induced cell death. In contrast, decreasing extracellular Ca2+ reduced both MTI-101-induced Ca2+ influx as well as cell death. The causative role of TRPC heteromers was established by suppressing STIM1, TRPC1, TRPC4, or TRPC5 function both pharmacologically and by siRNA, resulting in a reduction in MTI-101-induced Ca2+ influx. Mechanistically, MTI-101 treatment induces trafficking of TRPC1 to the membrane and co-immunoprecipitation studies indicate that MTI-101 treatment induces a TRPC1-STIM1 complex. Moreover, treatment with calpeptin inhibited MTI-101-induced Ca2+ influx and cell death, indicating a role of calpain in the mechanism of MTI-101-induced cytotoxicity. Finally, components of the SOCE pathway were found to be poor prognostic indicators among MM patients, suggesting that this pathway is attractive for the treatment of MM. Full article
(This article belongs to the Special Issue Dysregulation of Calcium Signalling in Disease)
Show Figures

Figure 1

17 pages, 9795 KiB  
Article
Repositioning Trimebutine Maleate as a Cancer Treatment Targeting Ovarian Cancer Stem Cells
by Heejin Lee, Oh-Bin Kwon, Jae-Eon Lee, Yong-Hyun Jeon, Dong-Seok Lee, Sang-Hyun Min and Jun-Woo Kim
Cells 2021, 10(4), 918; https://doi.org/10.3390/cells10040918 - 16 Apr 2021
Cited by 11 | Viewed by 2789
Abstract
The overall five-year survival rate for late-stage patients of ovarian cancer is below 29% due to disease recurrence and drug resistance. Cancer stem cells (CSCs) are known as a major contributor to drug resistance and recurrence. Accordingly, therapies targeting ovarian CSCs are needed [...] Read more.
The overall five-year survival rate for late-stage patients of ovarian cancer is below 29% due to disease recurrence and drug resistance. Cancer stem cells (CSCs) are known as a major contributor to drug resistance and recurrence. Accordingly, therapies targeting ovarian CSCs are needed to overcome the limitations of present treatments. This study evaluated the effect of trimebutine maleate (TM) targeting ovarian CSCs, using A2780-SP cells acquired by a sphere culture of A2780 epithelial ovarian cancer cells. TM is indicated as a gastrointestinal motility modulator and is known to as a peripheral opioid receptor agonist and a blocker for various channels. The GI50 of TM was approximately 0.4 µM in A2780-SP cells but over 100 µM in A2780 cells, demonstrating CSCs specific growth inhibition. TM induced G0/G1 arrest and increased the AV+/PI+ dead cell population in the A2780-SP samples. Furthermore, TM treatment significantly reduced tumor growth in A2780-SP xenograft mice. Voltage gated calcium channels (VGCC) and calcium-activated potassium channels (BKCa) were overexpressed on ovarian CSCs and targeted by TM; inhibition of both channels reduced A2780-SP cells viability. TM reduced stemness-related protein expression; this tendency was reproduced by the simultaneous inhibition of VGCC and BKCa compared to single channel inhibition. In addition, TM suppressed the Wnt/β-catenin, Notch, and Hedgehog pathways which contribute to many CSCs characteristics. Specifically, further suppression of the Wnt/β-catenin pathway by simultaneous inhibition of BKCa and VGCC is necessary for the effective and selective action of TM. Taken together, TM is a potential therapeutic drug for preventing ovarian cancer recurrence and drug resistance. Full article
(This article belongs to the Special Issue Dysregulation of Calcium Signalling in Disease)
Show Figures

Figure 1

21 pages, 4527 KiB  
Article
Lysophosphatidic Acid-Activated Calcium Signaling Is Elevated in Red Cells from Sickle Cell Disease Patients
by Jue Wang, Laura Hertz, Sandra Ruppenthal, Wassim El Nemer, Philippe Connes, Jeroen S. Goede, Anna Bogdanova, Lutz Birnbaumer and Lars Kaestner
Cells 2021, 10(2), 456; https://doi.org/10.3390/cells10020456 - 20 Feb 2021
Cited by 10 | Viewed by 3163
Abstract
(1) Background: It is known that sickle cells contain a higher amount of Ca2+ compared to healthy red blood cells (RBCs). The increased Ca2+ is associated with the most severe symptom of sickle cell disease (SCD), the vaso-occlusive crisis (VOC). The [...] Read more.
(1) Background: It is known that sickle cells contain a higher amount of Ca2+ compared to healthy red blood cells (RBCs). The increased Ca2+ is associated with the most severe symptom of sickle cell disease (SCD), the vaso-occlusive crisis (VOC). The Ca2+ entry pathway received the name of Psickle but its molecular identity remains only partly resolved. We aimed to map the involved Ca2+ signaling to provide putative pharmacological targets for treatment. (2) Methods: The main technique applied was Ca2+ imaging of RBCs from healthy donors, SCD patients and a number of transgenic mouse models in comparison to wild-type mice. Life-cell Ca2+ imaging was applied to monitor responses to pharmacological targeting of the elements of signaling cascades. Infection as a trigger of VOC was imitated by stimulation of RBCs with lysophosphatidic acid (LPA). These measurements were complemented with biochemical assays. (3) Results: Ca2+ entry into SCD RBCs in response to LPA stimulation exceeded that of healthy donors. LPA receptor 4 levels were increased in SCD RBCs. Their activation was followed by the activation of Gi protein, which in turn triggered opening of TRPC6 and CaV2.1 channels via a protein kinase Cα and a MAP kinase pathway, respectively. (4) Conclusions: We found a new Ca2+ signaling cascade that is increased in SCD patients and identified new pharmacological targets that might be promising in addressing the most severe symptom of SCD, the VOC. Full article
(This article belongs to the Special Issue Dysregulation of Calcium Signalling in Disease)
Show Figures

Graphical abstract

21 pages, 2070 KiB  
Article
The Function of Mitochondrial Calcium Uniporter at the Whole-Cell and Single Mitochondrion Levels in WT, MICU1 KO, and MICU2 KO Cells
by Syed Islamuddin Shah and Ghanim Ullah
Cells 2020, 9(6), 1520; https://doi.org/10.3390/cells9061520 - 22 Jun 2020
Cited by 6 | Viewed by 2936
Abstract
Mitochondrial Ca2+ ([Ca2+]M) uptake through its Ca2+ uniporter (MCU) is central to many cell functions such as bioenergetics, spatiotemporal organization of Ca2+ signals, and apoptosis. MCU activity is regulated by several intrinsic proteins including MICU1, MICU2, [...] Read more.
Mitochondrial Ca2+ ([Ca2+]M) uptake through its Ca2+ uniporter (MCU) is central to many cell functions such as bioenergetics, spatiotemporal organization of Ca2+ signals, and apoptosis. MCU activity is regulated by several intrinsic proteins including MICU1, MICU2, and EMRE. While significant details about the role of MICU1, MICU2, and EMRE in MCU function have emerged recently, a key challenge for the future experiments is to investigate how these regulatory proteins modulate mitochondrial Ca2+ influx through MCU in intact cells under pathophysiological conditions. This is further complicated by the fact that several variables affecting MCU function change dynamically as cell functions. To overcome this void, we develop a data-driven model that closely replicates the behavior of MCU under a wide range of cytosolic Ca2+ ([Ca2+]C), [Ca2+]M, and mitochondrial membrane potential values in WT, MICU1 knockout (KO), and MICU2 KO cells at the single mitochondrion and whole-cell levels. The model is extended to investigate how MICU1 or MICU2 KO affect mitochondrial function. Moreover, we show how Ca2+ buffering proteins, the separation between mitochondrion and Ca2+-releasing stores, and the duration of opening of Ca2+-releasing channels affect mitochondrial function under different conditions. Finally, we demonstrate an easy extension of the model to single channel function of MCU. Full article
(This article belongs to the Special Issue Dysregulation of Calcium Signalling in Disease)
Show Figures

Figure 1

16 pages, 2542 KiB  
Article
Knockout of the Mitochondrial Calcium Uniporter Strongly Suppresses Stimulus-Metabolism Coupling in Pancreatic Acinar Cells but Does Not Reduce Severity of Experimental Acute Pancreatitis
by Michael Chvanov, Svetlana Voronina, Xiaoying Zhang, Svetlana Telnova, Robert Chard, Yulin Ouyang, Jane Armstrong, Helen Tanton, Muhammad Awais, Diane Latawiec, Robert Sutton, David N. Criddle and Alexei V. Tepikin
Cells 2020, 9(6), 1407; https://doi.org/10.3390/cells9061407 - 5 Jun 2020
Cited by 11 | Viewed by 3222
Abstract
Acute pancreatitis is a frequent disease that lacks specific drug treatment. Unravelling the molecular mechanisms of acute pancreatitis is essential for the development of new therapeutics. Several inducers of acute pancreatitis trigger sustained Ca2+ increases in the cytosol and mitochondria of pancreatic [...] Read more.
Acute pancreatitis is a frequent disease that lacks specific drug treatment. Unravelling the molecular mechanisms of acute pancreatitis is essential for the development of new therapeutics. Several inducers of acute pancreatitis trigger sustained Ca2+ increases in the cytosol and mitochondria of pancreatic acinar cells. The mitochondrial calcium uniporter (MCU) mediates mitochondrial Ca2+ uptake that regulates bioenergetics and plays an important role in cell survival, damage and death. Aberrant Ca2+ signaling and mitochondrial damage in pancreatic acinar cells have been implicated in the initiation of acute pancreatitis. The primary aim of this study was to assess the involvement of the MCU in experimental acute pancreatitis. We found that pancreatic acinar cells from MCU−/− mice display dramatically reduced mitochondrial Ca2+ uptake. This is consistent with the drastic changes of stimulus-metabolism coupling, manifested by the reduction of mitochondrial NADH/FAD+ responses to cholecystokinin and in the decrease of cholecystokinin-stimulated oxygen consumption. However, in three experimental models of acute pancreatitis (induced by caerulein, taurolithocholic acid 3-sulfate or palmitoleic acid plus ethanol), MCU knockout failed to reduce the biochemical and histological changes characterizing the severity of local and systemic damage. A possible explanation of this surprising finding is the redundancy of damaging mechanisms activated by the inducers of acute pancreatitis. Full article
(This article belongs to the Special Issue Dysregulation of Calcium Signalling in Disease)
Show Figures

Figure 1

17 pages, 2124 KiB  
Article
Acute Induction of Translocon-Mediated Ca2+ Leak Protects Cardiomyocytes Against Ischemia/Reperfusion Injury
by Ribal Al-Mawla, Mallory Ducrozet, Nolwenn Tessier, Lucille Païta, Bruno Pillot, Yves Gouriou, Camille Villedieu, Zeina Harhous, Alexandre Paccalet, Claire Crola Da Silva, Michel Ovize, Gabriel Bidaux, Sylvie Ducreux and Fabien Van Coppenolle
Cells 2020, 9(5), 1319; https://doi.org/10.3390/cells9051319 - 25 May 2020
Cited by 10 | Viewed by 3287
Abstract
During myocardial infarction, dysregulation of Ca2+ homeostasis between the reticulum, mitochondria, and cytosol occurs in cardiomyocytes and leads to cell death. Ca2+ leak channels are thought to be key regulators of the reticular Ca2+ homeostasis and cell survival. The present [...] Read more.
During myocardial infarction, dysregulation of Ca2+ homeostasis between the reticulum, mitochondria, and cytosol occurs in cardiomyocytes and leads to cell death. Ca2+ leak channels are thought to be key regulators of the reticular Ca2+ homeostasis and cell survival. The present study aimed to determine whether a particular reticular Ca2+ leak channel, the translocon, also known as translocation channel, could be a relevant target against ischemia/reperfusion-mediated heart injury. To achieve this objective, we first used an intramyocardial adenoviral strategy to express biosensors in order to assess Ca2+ variations in freshly isolated adult mouse cardiomyocytes to show that translocon is a functional reticular Ca2+ leak channel. Interestingly, translocon activation by puromycin mobilized a ryanodine receptor (RyR)-independent reticular Ca2+ pool and did not affect the excitation–concentration coupling. Second, puromycin pretreatment decreased mitochondrial Ca2+ content and slowed down the mitochondrial permeability transition pore (mPTP) opening and the rate of cytosolic Ca2+ increase during hypoxia. Finally, this translocon pre-activation also protected cardiomyocytes after in vitro hypoxia reoxygenation and reduced infarct size in mice submitted to in vivo ischemia-reperfusion. Altogether, our report emphasizes the role of translocon in cardioprotection and highlights a new paradigm in cardioprotection by functionally uncoupling the RyR-dependent Ca2+ stores and translocon-dependent Ca2+ stores. Full article
(This article belongs to the Special Issue Dysregulation of Calcium Signalling in Disease)
Show Figures

Graphical abstract

22 pages, 3728 KiB  
Article
A Mathematical Model of Lysosomal Ion Homeostasis Points to Differential Effects of Cl Transport in Ca2+ Dynamics
by Rosario Astaburuaga, Orlando Daniel Quintanar Haro, Tobias Stauber and Angela Relógio
Cells 2019, 8(10), 1263; https://doi.org/10.3390/cells8101263 - 16 Oct 2019
Cited by 9 | Viewed by 5787
Abstract
The establishment and maintenance of ion gradients between the interior of lysosomes and the cytosol are crucial for numerous cellular and organismal functions. Numerous ion transport proteins ensure the required variation in luminal concentrations of the different ions along the endocytic pathway to [...] Read more.
The establishment and maintenance of ion gradients between the interior of lysosomes and the cytosol are crucial for numerous cellular and organismal functions. Numerous ion transport proteins ensure the required variation in luminal concentrations of the different ions along the endocytic pathway to fit the needs of the organelles. Failures in keeping proper ion homeostasis have pathological consequences. Accordingly, several human diseases are caused by the dysfunction of ion transporters. These include osteopetrosis, caused by the dysfunction of Cl/H+ exchange by the lysosomal transporter ClC-7. To better understand how chloride transport affects lysosomal ion homeostasis and how its disruption impinges on lysosomal function, we developed a mathematical model of lysosomal ion homeostasis including Ca2+ dynamics. The model recapitulates known biophysical properties of ClC-7 and enables the investigation of its differential activation kinetics on lysosomal ion homeostasis. We show that normal functioning of ClC-7 supports the acidification process, is associated with increased luminal concentrations of sodium, potassium, and chloride, and leads to a higher Ca2+ uptake and release. Our model highlights the role of ClC-7 in lysosomal acidification and shows the existence of differential Ca2+ dynamics upon perturbations of Cl/H+ exchange and its activation kinetics, with possible pathological consequences. Full article
(This article belongs to the Special Issue Dysregulation of Calcium Signalling in Disease)
Show Figures

Figure 1

Review

Jump to: Research

30 pages, 3869 KiB  
Review
Mitochondrial Ca2+ Signaling in Health, Disease and Therapy
by Lorenzo Modesti, Alberto Danese, Veronica Angela Maria Vitto, Daniela Ramaccini, Gianluca Aguiari, Roberta Gafà, Giovanni Lanza, Carlotta Giorgi and Paolo Pinton
Cells 2021, 10(6), 1317; https://doi.org/10.3390/cells10061317 - 25 May 2021
Cited by 58 | Viewed by 8056
Abstract
The divalent cation calcium (Ca2+) is considered one of the main second messengers inside cells and acts as the most prominent signal in a plethora of biological processes. Its homeostasis is guaranteed by an intricate and complex system of channels, pumps, [...] Read more.
The divalent cation calcium (Ca2+) is considered one of the main second messengers inside cells and acts as the most prominent signal in a plethora of biological processes. Its homeostasis is guaranteed by an intricate and complex system of channels, pumps, and exchangers. In this context, by regulating cellular Ca2+ levels, mitochondria control both the uptake and release of Ca2+. Therefore, at the mitochondrial level, Ca2+ plays a dual role, participating in both vital physiological processes (ATP production and regulation of mitochondrial metabolism) and pathophysiological processes (cell death, cancer progression and metastasis). Hence, it is not surprising that alterations in mitochondrial Ca2+ (mCa2+) pathways or mutations in Ca2+ transporters affect the activities and functions of the entire cell. Indeed, it is widely recognized that dysregulation of mCa2+ signaling leads to various pathological scenarios, including cancer, neurological defects and cardiovascular diseases (CVDs). This review summarizes the current knowledge on the regulation of mCa2+ homeostasis, the related mechanisms and the significance of this regulation in physiology and human diseases. We also highlight strategies aimed at remedying mCa2+ dysregulation as promising therapeutical approaches. Full article
(This article belongs to the Special Issue Dysregulation of Calcium Signalling in Disease)
Show Figures

Figure 1

14 pages, 2006 KiB  
Review
The Discovery of Naringenin as Endolysosomal Two-Pore Channel Inhibitor and Its Emerging Role in SARS-CoV-2 Infection
by Antonella D’Amore, Antonella Gradogna, Fioretta Palombi, Velia Minicozzi, Matteo Ceccarelli, Armando Carpaneto and Antonio Filippini
Cells 2021, 10(5), 1130; https://doi.org/10.3390/cells10051130 - 7 May 2021
Cited by 20 | Viewed by 3872
Abstract
The flavonoid naringenin (Nar), present in citrus fruits and tomatoes, has been identified as a blocker of an emerging class of human intracellular channels, namely the two-pore channel (TPC) family, whose role has been established in several diseases. Indeed, Nar was shown to [...] Read more.
The flavonoid naringenin (Nar), present in citrus fruits and tomatoes, has been identified as a blocker of an emerging class of human intracellular channels, namely the two-pore channel (TPC) family, whose role has been established in several diseases. Indeed, Nar was shown to be effective against neoangiogenesis, a process essential for solid tumor progression, by specifically impairing TPC activity. The goal of the present review is to illustrate the rationale that links TPC channels to the mechanism of coronavirus infection, and how their inhibition by Nar could be an efficient pharmacological strategy to fight the current pandemic plague COVID-19. Full article
(This article belongs to the Special Issue Dysregulation of Calcium Signalling in Disease)
Show Figures

Figure 1

15 pages, 1555 KiB  
Review
Role of the TRP Channels in Pancreatic Ductal Adenocarcinoma Development and Progression
by Gonçalo Mesquita, Natalia Prevarskaya, Albrecht Schwab and V’yacheslav Lehen’kyi
Cells 2021, 10(5), 1021; https://doi.org/10.3390/cells10051021 - 26 Apr 2021
Cited by 10 | Viewed by 2617
Abstract
The transient receptor potential channels (TRPs) have been related to several different physiologies that range from a role in sensory physiology (including thermo- and osmosensation) to a role in some pathologies like cancer. The great diversity of functions performed by these channels is [...] Read more.
The transient receptor potential channels (TRPs) have been related to several different physiologies that range from a role in sensory physiology (including thermo- and osmosensation) to a role in some pathologies like cancer. The great diversity of functions performed by these channels is represented by nine sub-families that constitute the TRP channel superfamily. From the mid-2000s, several reports have shown the potential role of the TRP channels in cancers of multiple origin. The pancreatic cancer is one of the deadliest cancers worldwide. Its prevalence is predicted to rise further. Disappointingly, the treatments currently used are ineffective. There is an urgency to find new ways to counter this disease and one of the answers may lie in the ion channels belonging to the superfamily of TRP channels. In this review, we analyse the existing knowledge on the role of TRP channels in the development and progression of pancreatic ductal adenocarcinoma (PDAC). The functions of these channels in other cancers are also considered. This might be of interest for an extrapolation to the pancreatic cancer in an attempt to identify potential therapeutic interventions. Full article
(This article belongs to the Special Issue Dysregulation of Calcium Signalling in Disease)
Show Figures

Figure 1

14 pages, 8190 KiB  
Review
Ca2+ Signaling by TRPV4 Channels in Respiratory Function and Disease
by Suhasini Rajan, Christian Schremmer, Jonas Weber, Philipp Alt, Fabienne Geiger and Alexander Dietrich
Cells 2021, 10(4), 822; https://doi.org/10.3390/cells10040822 - 6 Apr 2021
Cited by 18 | Viewed by 4584
Abstract
Members of the transient receptor potential (TRP) superfamily are broadly expressed in our body and contribute to multiple cellular functions. Most interestingly, the fourth member of the vanilloid family of TRP channels (TRPV4) serves different partially antagonistic functions in the respiratory system. This [...] Read more.
Members of the transient receptor potential (TRP) superfamily are broadly expressed in our body and contribute to multiple cellular functions. Most interestingly, the fourth member of the vanilloid family of TRP channels (TRPV4) serves different partially antagonistic functions in the respiratory system. This review highlights the role of TRPV4 channels in lung fibroblasts, the lung endothelium, as well as the alveolar and bronchial epithelium, during physiological and pathophysiological mechanisms. Data available from animal models and human tissues confirm the importance of this ion channel in cellular signal transduction complexes with Ca2+ ions as a second messenger. Moreover, TRPV4 is an excellent therapeutic target with numerous specific compounds regulating its activity in diseases, like asthma, lung fibrosis, edema, and infections. Full article
(This article belongs to the Special Issue Dysregulation of Calcium Signalling in Disease)
Show Figures

Figure 1

Back to TopTop