1
|
Zeng X, Sun A, Cheng W, Hou X, Zhu M, Liao Y. Inhibition of STIM1 alleviates high glucose-induced proliferation and fibrosis by inducing autophagy in mesangial cells. Mol Cell Biochem 2024; 479:2365-2379. [PMID: 37736800 DOI: 10.1007/s11010-023-04844-7] [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: 06/15/2023] [Accepted: 08/24/2023] [Indexed: 09/23/2023]
Abstract
Diabetic nephropathy (DN) is a renal microvascular complication caused by diabetes mellitus. One of the most typical characteristics of DN is glomerular mesangial cells (GMCs) proliferation. Stromal interaction molecule 1 (STIM1), a Ca2+ channel, is involved in many diseases. In this study, we investigated the role of STIM1 in the proliferation and fibrosis in high glucose (HG)-induced HBZY-1 cells. We found that the expression of STIM1 was increased in renal tissues of diabetic rat and HBZY-1 cells stimulated by HG. Downregulation of STIM1-mediated SOCE suppressed hyperglycemic cell proliferation and fibrosis by activating autophagy. In addition, the inhibitory effect of downregulating STIM1 on cells was blocked by autophagy inhibitor Bafilomycin A1 (BafA1). Moreover, this experiment also showed that STIM1 regulated autophagy, cell proliferation and fibrosis via PI3K/AKT/mTOR signal pathway. These results clarify the role of STIM1 in HBZY-1 cells and its mechanism, and provide a new target for the treatment of DN.
Collapse
Affiliation(s)
- Xixi Zeng
- Department of Anatomy, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, 430030, Hubei, People's Republic of China
| | - Anbang Sun
- Department of Anatomy, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, 430030, Hubei, People's Republic of China
| | - Weiyi Cheng
- Department of Emergency Surgery, Union Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, 430030, Hubei, People's Republic of China
| | - Xin Hou
- Medical College, Affiliated Hospital, Hebei University of Engineering, Handan, People's Republic of China
| | - Min Zhu
- Department of Thoracic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China.
| | - Yanhong Liao
- Department of Anatomy, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, 430030, Hubei, People's Republic of China.
| |
Collapse
|
2
|
Ma R, Tao Y, Wade ML, Mallet RT. Non-voltage-gated Ca 2+ channel signaling in glomerular cells in kidney health and disease. Am J Physiol Renal Physiol 2024; 327:F249-F264. [PMID: 38867675 PMCID: PMC11460346 DOI: 10.1152/ajprenal.00130.2024] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 06/10/2024] [Accepted: 06/10/2024] [Indexed: 06/14/2024] Open
Abstract
Positioned at the head of the nephron, the renal corpuscle generates a plasma ultrafiltrate to initiate urine formation. Three major cell types within the renal corpuscle, the glomerular mesangial cells, podocytes, and glomerular capillary endothelial cells, communicate via endocrine- and paracrine-signaling mechanisms to maintain the structure and function of the glomerular capillary network and filtration barrier. Ca2+ signaling mediated by several distinct plasma membrane Ca2+ channels impacts the functions of all three cell types. The past two decades have witnessed pivotal advances in understanding of non-voltage-gated Ca2+ channel function and regulation in the renal corpuscle in health and renal disease. This review summarizes the current knowledge of the physiological and pathological impact of non-voltage-gated Ca2+ channel signaling in mesangial cells, podocytes and glomerular capillary endothelium. The main focus is on transient receptor potential and store-operated Ca2+ channels, but ionotropic N-methyl-d-aspartate receptors and purinergic receptors also are discussed. This update of Ca2+ channel functions and their cellular signaling cascades in the renal corpuscle is intended to inform the development of therapeutic strategies targeting these channels to treat kidney diseases, particularly diabetic nephropathy.
Collapse
Affiliation(s)
- Rong Ma
- Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, Texas, United States
| | - Yu Tao
- Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, Texas, United States
| | - Michael L Wade
- Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, Texas, United States
| | - Robert T Mallet
- Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, Texas, United States
| |
Collapse
|
3
|
Staruschenko A, Alexander RT, Caplan MJ, Ilatovskaya DV. Calcium signalling and transport in the kidney. Nat Rev Nephrol 2024; 20:541-555. [PMID: 38641658 PMCID: PMC12036682 DOI: 10.1038/s41581-024-00835-z] [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] [Accepted: 03/27/2024] [Indexed: 04/21/2024]
Abstract
The kidney plays a pivotal role in regulating calcium levels within the body. Approximately 98% of the filtered calcium is reabsorbed in the nephron, and this process is tightly controlled to maintain calcium homeostasis, which is required to facilitate optimal bone mineralization, preserve serum calcium levels within a narrow range, and support intracellular signalling mechanisms. The maintenance of these functions is attributed to a delicate balance achieved by various calcium channels, transporters, and calcium-binding proteins in renal cells. Perturbation of this balance due to deficiency or dysfunction of calcium channels and calcium-binding proteins can lead to severe complications. For example, polycystic kidney disease is linked to aberrant calcium transport and signalling. Furthermore, dysregulation of calcium levels can promote the formation of kidney stones. This Review provides an updated description of the key aspects of calcium handling in the kidney, focusing on the function of various calcium channels and the physiological stimuli that control these channels or are communicated through them. A discussion of the role of calcium as an intracellular second messenger and the pathophysiology of renal calcium dysregulation, as well as a summary of gaps in knowledge and future prospects, are also included.
Collapse
Affiliation(s)
- Alexander Staruschenko
- Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, FL, USA.
- Hypertension and Kidney Research Center, University of South Florida, Tampa, FL, USA.
- James A. Haley Veterans Hospital, Tampa, FL, USA.
| | - R Todd Alexander
- Department of Paediatrics, University of Alberta, Edmonton, AB, Canada
- Women's and Children's Health Institute, Edmonton, AB, Canada
| | - Michael J Caplan
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT, USA
| | - Daria V Ilatovskaya
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, GA, USA
| |
Collapse
|
4
|
Staruschenko A, Ma R, Palygin O, Dryer SE. Ion channels and channelopathies in glomeruli. Physiol Rev 2023; 103:787-854. [PMID: 36007181 PMCID: PMC9662803 DOI: 10.1152/physrev.00013.2022] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 08/15/2022] [Accepted: 08/21/2022] [Indexed: 11/22/2022] Open
Abstract
An essential step in renal function entails the formation of an ultrafiltrate that is delivered to the renal tubules for subsequent processing. This process, known as glomerular filtration, is controlled by intrinsic regulatory systems and by paracrine, neuronal, and endocrine signals that converge onto glomerular cells. In addition, the characteristics of glomerular fluid flow, such as the glomerular filtration rate and the glomerular filtration fraction, play an important role in determining blood flow to the rest of the kidney. Consequently, disease processes that initially affect glomeruli are the most likely to lead to end-stage kidney failure. The cells that comprise the glomerular filter, especially podocytes and mesangial cells, express many different types of ion channels that regulate intrinsic aspects of cell function and cellular responses to the local environment, such as changes in glomerular capillary pressure. Dysregulation of glomerular ion channels, such as changes in TRPC6, can lead to devastating glomerular diseases, and a number of channels, including TRPC6, TRPC5, and various ionotropic receptors, are promising targets for drug development. This review discusses glomerular structure and glomerular disease processes. It also describes the types of plasma membrane ion channels that have been identified in glomerular cells, the physiological and pathophysiological contexts in which they operate, and the pathways by which they are regulated and dysregulated. The contributions of these channels to glomerular disease processes, such as focal segmental glomerulosclerosis (FSGS) and diabetic nephropathy, as well as the development of drugs that target these channels are also discussed.
Collapse
Affiliation(s)
- Alexander Staruschenko
- Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, Florida
- Hypertension and Kidney Research Center, University of South Florida, Tampa, Florida
- James A. Haley Veterans Hospital, Tampa, Florida
| | - Rong Ma
- Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, Texas
| | - Oleg Palygin
- Division of Nephrology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina
| | - Stuart E Dryer
- Department of Biology and Biochemistry, University of Houston, Houston, Texas
- Department of Biomedical Sciences, Tilman J. Fertitta Family College of Medicine, University of Houston, Houston, Texas
| |
Collapse
|
5
|
Yu Y, Li W, Jiang J. TRPC channels as emerging targets for seizure disorders. Trends Pharmacol Sci 2022; 43:787-798. [PMID: 35840362 PMCID: PMC9378536 DOI: 10.1016/j.tips.2022.06.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 06/09/2022] [Accepted: 06/17/2022] [Indexed: 10/17/2022]
Abstract
Epilepsy is characterized by seizures of diverse types that affect about 1-2% of the population worldwide. Current antiseizure medications are unsatisfactory, as they merely provide symptomatic relief, are ineffective in about one-third of patients, and cause unbearable adverse effects. Transient receptor potential canonical (TRPC) channels are a group of nonselective cation channels involved in many physiological functions. In this review, we provide an overview of recent preclinical studies using both genetic and pharmacological strategies that reveal these receptor-operated calcium-permeable channels may also play fundamental roles in many aspects of epileptic seizures. We also propose that TRPC channels represent appealing targets for epilepsy treatment, with a goal of helping to advance the discovery and development of new antiseizure and/or antiepileptogenic therapies.
Collapse
Affiliation(s)
- Ying Yu
- Department of Pharmaceutical Sciences and Drug Discovery Center, College of Pharmacy, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Wei Li
- Department of Pharmaceutical Sciences and Drug Discovery Center, College of Pharmacy, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Jianxiong Jiang
- Department of Pharmaceutical Sciences and Drug Discovery Center, College of Pharmacy, University of Tennessee Health Science Center, Memphis, TN 38163, USA.
| |
Collapse
|
6
|
Tao Y, Chaudhari S, Shotorbani PY, Ding Y, Chen Z, Kasetti R, Zode G, Ma R. Enhanced Orai1-mediated store-operated Ca 2+ channel/calpain signaling contributes to high glucose-induced podocyte injury. J Biol Chem 2022; 298:101990. [PMID: 35490782 PMCID: PMC9136128 DOI: 10.1016/j.jbc.2022.101990] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 04/19/2022] [Accepted: 04/21/2022] [Indexed: 02/09/2023] Open
Abstract
Podocyte injury induced by hyperglycemia is the main cause of kidney dysfunction in diabetic nephropathy. However, the underlying mechanism is unclear. Store-operated Ca2+ entry (SOCE) regulates a diversity of cellular processes in a variety of cell types. Calpain, a Ca2+-dependent cysteine protease, was recently shown to be involved in podocyte injury. In the present study, we sought to determine whether increased SOCE contributed to high glucose (HG)-induced podocyte injury through activation of the calpain pathway. In cultured human podocytes, whole-cell patch clamp indicated the presence of functional store-operated Ca2+ channels, which are composed of Orai1 proteins and mediate SOCE. Western blots showed that HG treatment increased the protein abundance of Orai1 in a dose-dependent manner. Consistently, calcium imaging experiments revealed that SOCE was significantly enhanced in podocytes following HG treatment. Furthermore, HG treatment caused overt podocyte F-actin disorganization as well as a significant decrease in nephrin protein abundance, both of which are indications of podocyte injury. These podocyte injury responses were significantly blunted by both pharmacological inhibition of Orai1 using the small molecule inhibitor BTP2 or by genetic deletion of Orai1 using CRISPR-Cas9 lentivirus. Moreover, activation of SOCE by thapsigargin, an inhibitor of Ca2+ pump on the endoplasmic/sarcoplasmic reticulum membrane, significantly increased the activity of calpain, which was inhibited by BTP2. Finally, the calpain-1/calpain-2 inhibitor calpeptin significantly blunted the nephrin protein reduction induced by HG treatment. Taken together, our results suggest that enhanced signaling via an Orai1/SOCE/Calpain axis contributes to HG-induced podocyte injury.
Collapse
Affiliation(s)
- Yu Tao
- Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, Texas, USA
| | - Sarika Chaudhari
- Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, Texas, USA
| | | | - Yanfeng Ding
- Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, Texas, USA
| | - Zhenglan Chen
- Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, Texas, USA
| | - Ramesh Kasetti
- The North Texas Eye Research Institute and Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, Texas, USA
| | - Gulab Zode
- The North Texas Eye Research Institute and Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, Texas, USA
| | - Rong Ma
- Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, Texas, USA.
| |
Collapse
|
7
|
Liu Y, Lyu Y, Wang H. TRP Channels as Molecular Targets to Relieve Endocrine-Related Diseases. Front Mol Biosci 2022; 9:895814. [PMID: 35573736 PMCID: PMC9095829 DOI: 10.3389/fmolb.2022.895814] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 03/28/2022] [Indexed: 12/03/2022] Open
Abstract
Transient receptor potential (TRP) channels are polymodal channels capable of sensing environmental stimuli, which are widely expressed on the plasma membrane of cells and play an essential role in the physiological or pathological processes of cells as sensors. TRPs often form functional homo- or heterotetramers that act as cation channels to flow Na+ and Ca2+, change membrane potential and [Ca2+]i (cytosolic [Ca2+]), and change protein expression levels, channel attributes, and regulatory factors. Under normal circumstances, various TRP channels respond to intracellular and extracellular stimuli such as temperature, pH, osmotic pressure, chemicals, cytokines, and cell damage and depletion of Ca2+ reserves. As cation transport channels and physical and chemical stimulation receptors, TRPs play an important role in regulating secretion, interfering with cell proliferation, and affecting neural activity in these glands and their adenocarcinoma cells. Many studies have proved that TRPs are widely distributed in the pancreas, adrenal gland, and other glands. This article reviews the specific regulatory mechanisms of various TRP channels in some common glands (pancreas, salivary gland, lacrimal gland, adrenal gland, mammary gland, gallbladder, and sweat gland).
Collapse
|
8
|
Agarwal S, Sudhini YR, Polat OK, Reiser J, Altintas MM. Renal cell markers: lighthouses for managing renal diseases. Am J Physiol Renal Physiol 2021; 321:F715-F739. [PMID: 34632812 DOI: 10.1152/ajprenal.00182.2021] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Kidneys, one of the vital organs in our body, are responsible for maintaining whole body homeostasis. The complexity of renal function (e.g., filtration, reabsorption, fluid and electrolyte regulation, and urine production) demands diversity not only at the level of cell types but also in their overall distribution and structural framework within the kidney. To gain an in depth molecular-level understanding of the renal system, it is imperative to discern the components of kidney and the types of cells residing in each of the subregions. Recent developments in labeling, tracing, and imaging techniques have enabled us to mark, monitor, and identify these cells in vivo with high efficiency in a minimally invasive manner. In this review, we summarize different cell types, specific markers that are uniquely associated with those cell types, and their distribution in the kidney, which altogether make kidneys so special and different. Cellular sorting based on the presence of certain proteins on the cell surface allowed for the assignment of multiple markers for each cell type. However, different studies using different techniques have found contradictions in cell type-specific markers. Thus, the term "cell marker" might be imprecise and suboptimal, leading to uncertainty when interpreting the data. Therefore, we strongly believe that there is an unmet need to define the best cell markers for a cell type. Although the compendium of renal-selective marker proteins presented in this review is a resource that may be useful to researchers, we acknowledge that the list may not be necessarily exhaustive.
Collapse
Affiliation(s)
- Shivangi Agarwal
- Department of Internal Medicine, Rush University, Chicago, Illinois
| | | | - Onur K Polat
- Department of Internal Medicine, Rush University, Chicago, Illinois
| | - Jochen Reiser
- Department of Internal Medicine, Rush University, Chicago, Illinois
| | | |
Collapse
|
9
|
Lefranc F. Transient Receptor Potential (TRP) Ion Channels Involved in Malignant Glioma Cell Death and Therapeutic Perspectives. Front Cell Dev Biol 2021; 9:618961. [PMID: 34458247 PMCID: PMC8388852 DOI: 10.3389/fcell.2021.618961] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Accepted: 04/29/2021] [Indexed: 01/22/2023] Open
Abstract
Among the most biologically, thus clinically, aggressive primary brain tumors are found malignant gliomas. Despite recent advances in adjuvant therapies, which include targeted and immunotherapies, after surgery and radio/chemotherapy, the tumor is recurrent and always lethal. Malignant gliomas also contain a pool of initiating stem cells that are highly invasive and resistant to conventional treatment. Ion channels and transporters are markedly involved in cancer cell biology, including glioma cell biology. Transient receptor potential (TRP) ion channels are calcium-permeable channels implicated in Ca2+ changes in multiple cellular compartments by modulating the driving force for Ca2+ entry. Recent scientific reports have shown that these channels contribute to the increase in glioblastoma aggressiveness, with glioblastoma representing the ultimate level of glioma malignancy. The current review focuses on each type of TRP ion channel potentially involved in malignant glioma cell death, with the ultimate goal of identifying new therapeutic targets to clinically combat malignant gliomas. It thus appears that cannabidiol targeting the TRPV2 type could be such a potential target.
Collapse
Affiliation(s)
- Florence Lefranc
- Department of Neurosurgery, Hôpital Erasme, Université Libre de Bruxelles, Brussels, Belgium
| |
Collapse
|
10
|
Zhang Y, Maitikuerban B, Chen Y, Li Y, Cao Y, Xu X. Correlation between classical transient receptor potential channel 1 gene polymorphism and microalbuminuria in patients with primary hypertension. Clin Exp Hypertens 2021; 43:443-449. [PMID: 33877007 DOI: 10.1080/10641963.2021.1901107] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
OBJECTIVE To investigate the correlation between transient receptor potential channel 1 (TRPC1) gene polymorphism and microalbuminuria in patients with primary hypertension. Methods: A total of 468 patients with primary hypertension were admitted to the Department of Hypertension of the First Affiliated Hospital of Xinjiang Medical University from April 2015 to November 2017. According to microalbuminuria, the patients were divided into two groups: high urinary albumin group (EH+mALB group, n = 71) and normal urinary microalbuminuria group (EH group, n = 397). The Sequenom detection technology was used for genotyping the single nucleotide polymorphism (SNP) sites of the TRPC1 gene, such as rs1382688, rs3821647, rs7638459, rs953239, and rs7621642. RESULTS (1) No significant differences were detected in gender, smoking history, drinking history, family history, course of hypertension, fasting blood glucose, urea, creatinine, triglyceride, total cholesterol, high-density lipoprotein cholesterol, low-density lipoprotein cholesterol, glycosylated hemoglobin, vitamin D, homocysteine, and cystatin C between the two groups (P > .05). However, age, body mass index (BMI), 24-h mean systolic and diastolic blood pressure, and 24-h average pulse pressure were statistically significant (P < .05). (2) No significant difference was detected in the distribution frequency of the polymorphisms of the TRPC1 gene between the two groups (P > .05), while the genotype, allele, and recessive model of rs7638459 differed significantly difference (P < .05). (3) Logistic regression analysis showed that BMI and rs7638459 CC genotype were the risk factors of increased microalbuminuria in patients with primary hypertension. CONCLUSION TRPC1 gene polymorphism is associated with increased microalbuminuria in patients with primary hypertension. The CC genotype of rs7638459 may increase the risk of microalbuminuria in patients with essential hypertension, while BMI and rs7638459 CC genotype may be the risk factors of increased microalbuminuria in patients with primary hypertension.
Collapse
Affiliation(s)
- Yu Zhang
- Department of Hypertension, The First Affiliated Hospital of Xinjiang Medical University, Urumqi,Xinjiang, China
| | | | - Yulan Chen
- Department of Hypertension, The First Affiliated Hospital of Xinjiang Medical University, Urumqi,Xinjiang, China
| | - Yu Li
- Second Department of General Internal Medicine, The First Affiliated Hospital of Xinjiang Medical University, Urumqi,Xinjiang, China
| | - Yaping Cao
- Department of Hypertension, The First Affiliated Hospital of Xinjiang Medical University, Urumqi,Xinjiang, China
| | - Xinjuan Xu
- Department of Hypertension, The First Affiliated Hospital of Xinjiang Medical University, Urumqi,Xinjiang, China
| |
Collapse
|
11
|
Song N, Yang M, Zhang H, Yang SK. Intracellular Calcium Homeostasis and Kidney Disease. Curr Med Chem 2021; 28:3647-3665. [PMID: 33138745 DOI: 10.2174/0929867327666201102114257] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 09/30/2020] [Accepted: 09/30/2020] [Indexed: 11/22/2022]
Abstract
Kidney disease is a serious health problem that burdens our healthcare system. It is crucial to find the accurate pathogenesis of various types of kidney disease to provide guidance for precise therapies for patients suffering from these diseases. However, the exact molecular mechanisms underlying these diseases have not been fully understood. Disturbance of calcium homeostasis in renal cells plays a fundamental role in the development of various types of kidney disease, such as primary glomerular disease, diabetic nephropathy, acute kidney injury and polycystic kidney disease, through promoting cell proliferation, stimulating extracellular matrix accumulation, aggravating podocyte injury, disrupting cellular energetics as well as dysregulating cell survival and death dynamics. As a result, preventing the disturbance of calcium homeostasis in specific renal cells (such as tubular cells, podocytes and mesangial cells) is becoming one of the most promising therapeutic strategies in the treatment of kidney disease. The endoplasmic reticulum and mitochondria are two vital organelles in this process. Calcium ions cycle between the endoplasmic reticulum and mitochondria at the conjugation of these two organelles known as the mitochondria-associated endoplasmic reticulum membrane, maintaining calcium homeostasis. The pharmacologic modulation of cellular calcium homeostasis can be viewed as a novel therapeutic method for renal diseases. Here, we will introduce calcium homeostasis under physiological conditions and the disturbance of calcium homeostasis in kidney diseases. We will focus on the calcium homeostasis regulation in renal cells (including tubular cells, podocytes and mesangial cells), especially in the mitochondria- associated endoplasmic reticulum membranes of these renal cells.
Collapse
Affiliation(s)
- Na Song
- Department of Nephrology, The Third Xiangya Hospital of Central South University, Changsha 410013, Hunan Province, China
| | - Ming Yang
- Department of Nephrology, The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan Province, China
| | - Hao Zhang
- Department of Nephrology, The Third Xiangya Hospital of Central South University, Changsha 410013, Hunan Province, China
| | - Shi-Kun Yang
- Department of Nephrology, The Third Xiangya Hospital of Central South University, Changsha 410013, Hunan Province, China
| |
Collapse
|
12
|
Shalygin A, Kolesnikov D, Glushankova L, Gusev K, Skopin A, Skobeleva K, Kaznacheyeva EV. Role of STIM2 and Orai proteins in regulating TRPC1 channel activity upon calcium store depletion. Cell Calcium 2021; 97:102432. [PMID: 34157631 DOI: 10.1016/j.ceca.2021.102432] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2020] [Revised: 05/08/2021] [Accepted: 06/04/2021] [Indexed: 11/17/2022]
Abstract
Store-operated calcium channels are the major player in calcium signaling in non-excitable cells. Store-operated calcium entry is associated with the Orai, stromal interaction molecule (STIM), and transient receptor potential canonical (TRPC) protein families. Researchers have provided conflicting data about TRPC1 channel regulation by Orai and STIM. To determine how Orai and STIM influence endogenous TRPC1 pore properties and regulation, we used single channel patch-clamp recordings. Here we showed that knockout or knockdown of Orai1 or Orai3 or overexpression of the dominant-negative mutant Orai1 E106Q did not change the conductance or selectivity of single TRPC1 channels. In addition, these TRPC1 channel properties did not depend on the amount of STIM1 and STIM2 proteins. To study STIM2-mediated regulation of TRPC1 channels, we utilized partial calcium store depletion induced by application of 10 nM thapsigargin (Tg). TRPC1 activation by endogenous STIM2 was greatly decreased in acute extracellular calcium-free experiments. STIM2 overexpression increased both the basal activity and number of silent TRPC1 channels in the plasma membrane. After calcium store depletion, overexpressed STIM2 directly activated TRPC1 in the plasma membrane even without calcium entry in acute experiments. However, this effect was abrogated by co-expression with the non-permeable Orai1 E106Q mutant protein. Taken together, our single-channel patch clamp experiments clearly demonstrated that endogenous TRPC1 forms a channel pore without involving Orai proteins. Calcium entry through Orai triggered TRPC1 channel activation in the plasma membrane, while subsequent STIM2-mediated TRPC1 activity regulation was not dependent on calcium entry.
Collapse
Affiliation(s)
- A Shalygin
- Institute of Cytology, Russian Academy of Sciences, 4 Tikhoretsky Avenue, St. Petersburg 194064, Russia.
| | - D Kolesnikov
- Institute of Cytology, Russian Academy of Sciences, 4 Tikhoretsky Avenue, St. Petersburg 194064, Russia
| | - L Glushankova
- Institute of Cytology, Russian Academy of Sciences, 4 Tikhoretsky Avenue, St. Petersburg 194064, Russia
| | - K Gusev
- Institute of Cytology, Russian Academy of Sciences, 4 Tikhoretsky Avenue, St. Petersburg 194064, Russia
| | - A Skopin
- Institute of Cytology, Russian Academy of Sciences, 4 Tikhoretsky Avenue, St. Petersburg 194064, Russia
| | - K Skobeleva
- Institute of Cytology, Russian Academy of Sciences, 4 Tikhoretsky Avenue, St. Petersburg 194064, Russia
| | - E V Kaznacheyeva
- Institute of Cytology, Russian Academy of Sciences, 4 Tikhoretsky Avenue, St. Petersburg 194064, Russia.
| |
Collapse
|
13
|
Wang WA, Demaurex N. Proteins Interacting with STIM1 and Store-Operated Ca 2+ Entry. PROGRESS IN MOLECULAR AND SUBCELLULAR BIOLOGY 2021; 59:51-97. [PMID: 34050862 DOI: 10.1007/978-3-030-67696-4_4] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
The endoplasmic reticulum (ER) Ca2+ sensor stromal interaction molecule 1 (STIM1) interacts with ORAI Ca2+ channels at the plasma membrane to regulate immune and muscle cell function. The conformational changes underlying STIM1 activation, translocation, and ORAI1 trapping and gating, are stringently regulated by post-translational modifications and accessory proteins. Here, we review the recent progress in the identification and characterization of ER and cytosolic proteins interacting with STIM1 to control its activation and deactivation during store-operated Ca2+ entry (SOCE).
Collapse
Affiliation(s)
- Wen-An Wang
- Department of Cellular Physiology and Metabolism, University of Geneva, Geneva, Switzerland
| | - Nicolas Demaurex
- Department of Cellular Physiology and Metabolism, University of Geneva, Geneva, Switzerland.
| |
Collapse
|
14
|
Chaudhari S, Mallet RT, Shotorbani PY, Tao Y, Ma R. Store-operated calcium entry: Pivotal roles in renal physiology and pathophysiology. Exp Biol Med (Maywood) 2020; 246:305-316. [PMID: 33249888 DOI: 10.1177/1535370220975207] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Research conducted over the last two decades has dramatically advanced the understanding of store-operated calcium channels (SOCC) and their impact on renal function. Kidneys contain many types of cells, including those specialized for glomerular filtration (fenestrated capillary endothelium, podocytes), water and solute transport (tubular epithelium), and regulation of glomerular filtration and renal blood flow (vascular smooth muscle cells, mesangial cells). The highly integrated function of these myriad cells effects renal control of blood pressure, extracellular fluid volume and osmolality, electrolyte balance, and acid-base homeostasis. Many of these cells are regulated by Ca2+ signaling. Recent evidence demonstrates that SOCCs are major Ca2+ entry portals in several renal cell types. SOCC is activated by depletion of Ca2+ stores in the sarco/endoplasmic reticulum, which communicates with plasma membrane SOCC via the Ca2+ sensor Stromal Interaction Molecule 1 (STIM1). Orai1 is recognized as the main pore-forming subunit of SOCC in the plasma membrane. Orai proteins alone can form highly Ca2+ selective SOCC channels. Also, members of the Transient Receptor Potential Canonical (TRPC) channel family are proposed to form heteromeric complexes with Orai1 subunits, forming SOCC with low Ca2+ selectivity. Recently, Ca2+ entry through SOCC, known as store-operated Ca2+ entry (SOCE), was identified in glomerular mesangial cells, tubular epithelium, and renovascular smooth muscle cells. The physiological and pathological relevance and the characterization of SOCC complexes in those cells are still unclear. In this review, we summarize the current knowledge of SOCC and their roles in renal glomerular, tubular and vascular cells, including studies from our laboratory, emphasizing SOCE regulation of fibrotic protein deposition. Understanding the diverse roles of SOCE in different renal cell types is essential, as SOCC and its signaling pathways are emerging targets for treatment of SOCE-related diseases.
Collapse
Affiliation(s)
- Sarika Chaudhari
- Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| | - Robert T Mallet
- Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| | - Parisa Y Shotorbani
- Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| | - Yu Tao
- Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| | - Rong Ma
- Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| |
Collapse
|
15
|
Chen X, Yin J, Xu Y, Qiu Z, Liu J, Chen X. Effect of selective inhibition or activation of PGE2 EP1 receptor on glomerulosclerosis. Mol Med Rep 2020; 22:2887-2895. [PMID: 32700746 PMCID: PMC7453572 DOI: 10.3892/mmr.2020.11353] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Accepted: 06/19/2020] [Indexed: 11/16/2022] Open
Abstract
Prostaglandin E2 (PGE2) is involved in numerous physiological and pathological processes of the kidney via its four receptors. A previous study has suggested that a defect in the PGE2 receptor 1 (EP1) gene markedly suppressed the transforming growth factor‑β1 (TGF‑β1)‑induced mesangial cell (MC) proliferation and extracellular matrix aggregation. Therefore, the present study aimed to adopt a pharmacological method of specifically suppressing or activating the EP1 receptor to further verify and demonstrate these results. The EP1 receptor antagonist SC‑19220 and EP1 receptor agonist 17‑phenyl‑trinor‑PGE2 ethyl amide (17‑pt‑PGE2) were selectively used to treat five‑sixths nephrectomy renal fibrosis model mice and TGF‑β1‑stimulated MCs. An Alpha screen PGE2 assay kit, flow cytometry, western blotting and immunohistochemical techniques were adopted to perform in vivo and in vitro experiments. The present results suggested that compared with the control group, the selective EP1 receptor antagonist SC‑19220 improved renal function, markedly reduced the plasma blood urea nitrogen and creatinine levels (P<0.05) and alleviated glomerulosclerosis (P<0.05). By contrast, the EP1 receptor agonist 17‑pt‑PGE2 aggravated renal dysfunction and glomerulosclerosis (P<0.05). To verify the renal protection mechanisms mediated by suppression of the EP1 receptor, the expression levels of endoplasmic reticulum stress (ERS)‑related proteins, including chaperone glucose‑regulated protein 78 (GRP78), transient receptor potential channel 1 (TRPC1) and protein kinase R‑like endoplasmic reticulum kinase (PERK), were further evaluated histologically. The expression of GRP78, TRPC1 and PERK in the antagonist treatment group were markedly downregulated (P<0.05), whereas those in the agonist treatment group were upregulated (P<0.05). The present in vitro experiments demonstrated that, compared with the control group, the EP1 receptor antagonist suppressed the expression of GRP78, TRPC1 and PERK (P<0.05), reduced the production of PGE2 (P<0.05) and decreased the MC apoptosis rate (P<0.05), thus alleviating TGF‑β1‑stimulated MC injury. Consequently, consistent with previous results, selectively antagonizing the EP1 receptor improved renal function and mitigated glomerulosclerosis, and its potential mechanism might be associated with the suppression of ERS.
Collapse
MESH Headings
- Animals
- Apoptosis/drug effects
- Cells, Cultured
- Dibenz(b,f)(1,4)oxazepine-10(11H)-carboxylic acid, 8-chloro-, 2-acetylhydrazide/pharmacology
- Dinoprostone/analogs & derivatives
- Dinoprostone/metabolism
- Dinoprostone/pharmacology
- Disease Models, Animal
- Endoplasmic Reticulum Chaperone BiP
- Endoplasmic Reticulum Stress/drug effects
- Glomerulonephritis/drug therapy
- Glomerulonephritis/etiology
- Glomerulonephritis/physiopathology
- Heat-Shock Proteins/drug effects
- Heat-Shock Proteins/metabolism
- Kidney/drug effects
- Kidney/metabolism
- Kidney/pathology
- Kidney/physiopathology
- Male
- Mesangial Cells/drug effects
- Mesangial Cells/metabolism
- Mice
- Mice, Inbred C57BL
- Nephrectomy/adverse effects
- Prostaglandin Antagonists/pharmacology
- Receptors, Prostaglandin E, EP1 Subtype/agonists
- Receptors, Prostaglandin E, EP1 Subtype/antagonists & inhibitors
- TRPC Cation Channels/drug effects
- TRPC Cation Channels/metabolism
- Transforming Growth Factor beta1/toxicity
- eIF-2 Kinase/drug effects
- eIF-2 Kinase/metabolism
Collapse
Affiliation(s)
- Xu Chen
- Department of Nephrology, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Jun Yin
- Department of Nephrology, Wuxi No. 2 People's Hospital, Wuxi, Jiangsu 214000, P.R. China
| | - Yuyin Xu
- Department of Nephrology, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Zhi Qiu
- Department of Emergency Medicine, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Jing Liu
- Department of Nephrology, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Xiaolan Chen
- Department of Nephrology, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| |
Collapse
|
16
|
Formoso K, Susperreguy S, Freichel M, Birnbaumer L. RNA-seq analysis reveals TRPC genes to impact an unexpected number of metabolic and regulatory pathways. Sci Rep 2020; 10:7227. [PMID: 32350291 PMCID: PMC7190874 DOI: 10.1038/s41598-020-61177-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Accepted: 02/21/2020] [Indexed: 12/12/2022] Open
Abstract
The seven-member transient receptor potential canonical genes (TRPC1-7) encode cation channels linked to several human diseases. There is little understanding of the participation of each TRPC in each pathology, considering functional redundancy. Also, most of the inhibitors available are not specific. Thus, we developed mice that lack all of the TRPCs and performed a transcriptome analysis in eight tissues. The aim of this research was to address the impact of the absence of all TRPC channels on gene expression. We obtained a total of 4305 differentially expressed genes (DEGs) in at least one tissue where spleen showed the highest number of DEGs (1371). Just 21 genes were modified in all the tissues. Performing a pathway enrichment analysis, we found that many important signaling pathways were modified in more than one tissue, including PI3K (phosphatidylinositol 3-kinase/protein kinase-B) signaling pathway, cytokine-cytokine receptor interaction, extracellular matrix (ECM)-receptor interaction and circadian rhythms. We describe for the first time the changes at the transcriptome level due to the lack of all TRPC proteins in a mouse model and provide a starting point to understand the function of TRPC channels and their possible roles in pathologies.
Collapse
Affiliation(s)
- Karina Formoso
- Institute for Biomedical Research (BIOMED UCA-CONICET). School of Medical Sciences, Catholic University of Argentina (UCA), Buenos Aires, C1107AFF, Argentina
| | - Sebastian Susperreguy
- Institute for Biomedical Research (BIOMED UCA-CONICET). School of Medical Sciences, Catholic University of Argentina (UCA), Buenos Aires, C1107AFF, Argentina
| | - Marc Freichel
- Institute of Physiology and Pathophysiology, Heidelberg University, 69120, Heidelberg, Germany
| | - Lutz Birnbaumer
- Institute for Biomedical Research (BIOMED UCA-CONICET). School of Medical Sciences, Catholic University of Argentina (UCA), Buenos Aires, C1107AFF, Argentina. .,Neurobiology Laboratory, National Institute of Environmental Health Sciences (NIEHS), Research Triangle Park, North Carolina, 27709, USA.
| |
Collapse
|
17
|
Dyrda A, Koenig S, Frieden M. STIM1 long and STIM1 gate differently TRPC1 during store-operated calcium entry. Cell Calcium 2020; 86:102134. [DOI: 10.1016/j.ceca.2019.102134] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Revised: 11/13/2019] [Accepted: 11/25/2019] [Indexed: 11/30/2022]
|
18
|
Shotorbani PY, Chaudhari S, Tao Y, Tsiokas L, Ma R. Inhibitor of myogenic differentiation family isoform a, a new positive regulator of fibronectin production by glomerular mesangial cells. Am J Physiol Renal Physiol 2020; 318:F673-F682. [PMID: 31984795 PMCID: PMC7099507 DOI: 10.1152/ajprenal.00508.2019] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Revised: 01/14/2020] [Accepted: 01/16/2020] [Indexed: 12/12/2022] Open
Abstract
Overproduction of extracellular matrix proteins, including fibronectin by mesangial cells (MCs), contributes to diabetic nephropathy. Inhibitor of myogenic differentiation family isoform a (I-mfa) is a multifunctional cytosolic protein functioning as a transcriptional modulator or plasma channel protein regulator. However, its renal effects are unknown. The present study was conducted to determine whether I-mfa regulated fibronectin production by glomerular MCs. In human MCs, overexpression of I-mfa significantly increased fibronectin abundance. Silencing I-mfa significantly reduced the level of fibronectin mRNA and blunted transforming growth factor-β1-stimulated production of fibronectin. We further found that high glucose increased I-mfa protein content in a time course (≥48 h) and concentration (≥25 mM)-dependent manner. Although high glucose exposure increased I-mfa at the protein level, it did not significantly alter transcripts of I-mfa in MCs. Furthermore, the abundance of I-mfa protein was significantly increased in the renal cortex of rats with diabetic nephropathy. The I-mfa protein level was also elevated in the glomerulus of mice with diabetic kidney disease. However, there was no significant difference in glomerular I-mfa mRNA levels between mice with and without diabetic nephropathy. Moreover, H2O2 significantly increased I-mfa protein abundance in a dose-dependent manner in cultured human MCs. The antioxidants polyethylene glycol-catalase, ammonium pyrrolidithiocarbamate, and N-acetylcysteine significantly blocked the high glucose-induced increase of I-mfa protein. Taken together, our results suggest that I-mfa, increased by high glucose/diabetes through the production of reactive oxygen species, stimulates fibronectin production by MCs.
Collapse
Affiliation(s)
| | - Sarika Chaudhari
- Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, Texas
| | - Yu Tao
- Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, Texas
| | - Leonidas Tsiokas
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Rong Ma
- Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, Texas
| |
Collapse
|
19
|
Wang H, Cheng X, Tian J, Xiao Y, Tian T, Xu F, Hong X, Zhu MX. TRPC channels: Structure, function, regulation and recent advances in small molecular probes. Pharmacol Ther 2020; 209:107497. [PMID: 32004513 DOI: 10.1016/j.pharmthera.2020.107497] [Citation(s) in RCA: 141] [Impact Index Per Article: 28.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Accepted: 01/14/2020] [Indexed: 02/08/2023]
Abstract
Transient receptor potential canonical (TRPC) channels constitute a group of receptor-operated calcium-permeable nonselective cation channels of the TRP superfamily. The seven mammalian TRPC members, which can be further divided into four subgroups (TRPC1, TRPC2, TRPC4/5, and TRPC3/6/7) based on their amino acid sequences and functional similarities, contribute to a broad spectrum of cellular functions and physiological roles. Studies have revealed complexity of their regulation involving several components of the phospholipase C pathway, Gi and Go proteins, and internal Ca2+ stores. Recent advances in cryogenic electron microscopy have provided several high-resolution structures of TRPC channels. Growing evidence demonstrates the involvement of TRPC channels in diseases, particularly the link between genetic mutations of TRPC6 and familial focal segmental glomerulosclerosis. Because TRPCs were discovered by the molecular identity first, their pharmacology had lagged behind. This is rapidly changing in recent years owning to great efforts from both academia and industry. A number of potent tool compounds from both synthetic and natural products that selective target different subtypes of TRPC channels have been discovered, including some preclinical drug candidates. This review will cover recent advancements in the understanding of TRPC channel regulation, structure, and discovery of novel TRPC small molecular probes over the past few years, with the goal of facilitating drug discovery for the study of TRPCs and therapeutic development.
Collapse
Affiliation(s)
- Hongbo Wang
- Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education; Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai 264005, China.
| | - Xiaoding Cheng
- State Key Laboratory of Virology, Key Laboratory of Combinatorial Biosynthesis and Drug Discovery (MOE) and Hubei Province Engineering and Technology Research Center for Fluorinated Pharmaceuticals, Wuhan University School of Pharmaceutical Sciences, Wuhan 430071, China
| | - Jinbin Tian
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Yuling Xiao
- State Key Laboratory of Virology, Key Laboratory of Combinatorial Biosynthesis and Drug Discovery (MOE) and Hubei Province Engineering and Technology Research Center for Fluorinated Pharmaceuticals, Wuhan University School of Pharmaceutical Sciences, Wuhan 430071, China
| | - Tian Tian
- Innovation Center for Traditional Tibetan Medicine Modernization and Quality Control, Medical College, Department of Chemistry and Environmental Science, School of Science, Tibet University, Lhasa 850000, China
| | - Fuchun Xu
- Innovation Center for Traditional Tibetan Medicine Modernization and Quality Control, Medical College, Department of Chemistry and Environmental Science, School of Science, Tibet University, Lhasa 850000, China
| | - Xuechuan Hong
- State Key Laboratory of Virology, Key Laboratory of Combinatorial Biosynthesis and Drug Discovery (MOE) and Hubei Province Engineering and Technology Research Center for Fluorinated Pharmaceuticals, Wuhan University School of Pharmaceutical Sciences, Wuhan 430071, China; Innovation Center for Traditional Tibetan Medicine Modernization and Quality Control, Medical College, Department of Chemistry and Environmental Science, School of Science, Tibet University, Lhasa 850000, China.
| | - Michael X Zhu
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA.
| |
Collapse
|
20
|
TRPP2 associates with STIM1 to regulate cerebral vasoconstriction and enhance high salt intake-induced hypertensive cerebrovascular spasm. Hypertens Res 2019; 42:1894-1904. [PMID: 31541223 DOI: 10.1038/s41440-019-0324-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Revised: 08/05/2019] [Accepted: 08/14/2019] [Indexed: 11/08/2022]
Abstract
Cerebrovascular spasm is a life-threatening event in salt-sensitive hypertension. The relationship between store-operated calcium entry (SOCE) and vasoconstriction in hypertension has not been fully clarified. This study investigated the changes in cerebrovascular contractile responses in high salt intake-induced hypertension and the functional roles of the main components of SOCE, namely, polycystin-2 (TRPP2), stromal interaction molecule 1 (STIM1), and Orai3. Polycystic kidney disease 2 (which encodes TRPP2) knockout mice displayed decreased cerebrovascular SOCE-induced contraction. The blood pressure of age-matched rats fed a normal or high-salt diet for 4 weeks was monitored weekly using noninvasive tail-cuff plethysmography. The systolic blood pressure of the rats fed a high-salt diet was significantly higher than that of controls. Western blotting and immunohistochemical results showed that these hypertensive rats expressed higher levels of cerebrovascular TRPP2, STIM1, and Orai3 than controls. Cerebrovascular tension measurements of the basilar artery indicated that SOCE-mediated contraction was significantly increased in hypertensive rats compared with control rats. In addition, SOCE-mediated contraction was decreased in the basilar arteries of rats pretreated with the SOCE inhibitor BTP-2 (10 μM) or transfected with TRPP2-specific or STIM1-specific small interfering RNA. Staining with 2,3,5-triphenyltetrazolium chloride (TTC) was used to quantify the infarcted brain area 24 h after middle cerebral artery occlusion, a model of ischemic stroke, in rodents. The infarcted brain area was significantly greater in hypertensive rats and significantly lower in BTP-2-treated rats than in controls. Taken together, these findings indicate that SOCE-induced contraction may be overactive in the basilar arteries of salt-sensitive hypertensive rats, suggesting the dysregulation of TRPP2 and SOCE and its other components.
Collapse
|
21
|
Reboreda A, Theissen FM, Valero-Aracama MJ, Arboit A, Corbu MA, Yoshida M. Do TRPC channels support working memory? Comparing modulations of TRPC channels and working memory through G-protein coupled receptors and neuromodulators. Behav Brain Res 2018; 354:64-83. [PMID: 29501506 DOI: 10.1016/j.bbr.2018.02.042] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Revised: 02/27/2018] [Accepted: 02/27/2018] [Indexed: 12/11/2022]
Abstract
Working memory is a crucial ability we use in daily life. However, the cellular mechanisms supporting working memory still remain largely unclear. A key component of working memory is persistent neural firing which is believed to serve short-term (hundreds of milliseconds up to tens of seconds) maintenance of necessary information. In this review, we will focus on the role of transient receptor potential canonical (TRPC) channels as a mechanism underlying persistent firing. Many years of in vitro work have been suggesting a crucial role of TRPC channels in working memory and temporal association tasks. If TRPC channels are indeed a central mechanism for working memory, manipulations which impair or facilitate working memory should have a similar effect on TRPC channel modulation. However, modulations of working memory and TRPC channels were never systematically compared, and it remains unanswered whether TRPC channels indeed contribute to working memory in vivo or not. In this article, we review the effects of G-protein coupled receptors (GPCR) and neuromodulators, including acetylcholine, noradrenalin, serotonin and dopamine, on working memory and TRPC channels. Based on comparisons, we argue that GPCR and downstream signaling pathways that activate TRPC, generally support working memory, while those that suppress TRPC channels impair it. However, depending on the channel types, areas, and systems tested, this is not the case in all studies. Further work to clarify involvement of specific TRPC channels in working memory tasks and how they are affected by neuromodulators is still necessary in the future.
Collapse
Affiliation(s)
- Antonio Reboreda
- Leibniz Institute for Neurobiology (LIN) Magdeburg, Brenneckestraße 6, 39118 Magdeburg, Germany; German Center for Neurodegenerative Diseases (DZNE) Magdeburg, Leipziger Str. 44/Haus 64, 39120, Magdeburg, Germany.
| | - Frederik M Theissen
- German Center for Neurodegenerative Diseases (DZNE) Magdeburg, Leipziger Str. 44/Haus 64, 39120, Magdeburg, Germany
| | - Maria J Valero-Aracama
- Institute of Physiology and Pathophysiology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Universitätsstraße 17, 91054 Erlangen, Germany
| | - Alberto Arboit
- German Center for Neurodegenerative Diseases (DZNE) Magdeburg, Leipziger Str. 44/Haus 64, 39120, Magdeburg, Germany
| | - Mihaela A Corbu
- Ruhr University Bochum (RUB), Universitätsstraße 150, 44801, Bochum, Germany
| | - Motoharu Yoshida
- Leibniz Institute for Neurobiology (LIN) Magdeburg, Brenneckestraße 6, 39118 Magdeburg, Germany; German Center for Neurodegenerative Diseases (DZNE) Magdeburg, Leipziger Str. 44/Haus 64, 39120, Magdeburg, Germany; Center for Behavioral Brain Sciences, 39106, Magdeburg, Germany.
| |
Collapse
|
22
|
Urotensin II-induced store-operated Ca 2+ entry contributes to glomerular mesangial cell proliferation and extracellular matrix protein production under high glucose conditions. Sci Rep 2017; 7:18049. [PMID: 29273760 PMCID: PMC5741753 DOI: 10.1038/s41598-017-18143-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Accepted: 12/06/2017] [Indexed: 12/19/2022] Open
Abstract
Glomerular mesangial cell (GMC) proliferation and matrix expansion are pathological hallmarks of a wide range of kidney diseases, including diabetic nephropathy. Although the circulating level of peptide hormone urotensin II (UII) and kidney tissue expression of UII and UII receptors (UTR) are increased in diabetic nephropathy, it remains unclear whether UII regulates GMC growth and extracellular matrix (ECM) accumulation. In this study, we tested the hypothesis that UII-induced Ca2+ signaling controls GMC proliferation and ECM production under normal and high glucose conditions. Mouse GMCs cultured under normal glucose conditions proliferated and synthesized ECM proteins in response to stimulation by mouse UII. UII-induced GMC proliferation and ECM protein synthesis were dependent on TRPC4 channel-mediated store-operated Ca2+ entry (SOCE) and sequential activation of Ca2+/calmodulin-dependent protein kinase II (CaMKII) and Ca2+/cAMP response element-binding protein (CREB) transcription factor. Under high glucose conditions, GMCs synthesized UII. Moreover, proliferation and ECM production in high glucose-challenged GMCs were attenuated by selective UTR antagonist, TRPC4 channel blocker, and CaMKII and CREB-binding protein/p300 inhibitors. These findings indicate that UII-induced SOCE via TRPC4 channels stimulates CaMKII/CREB-dependent GMC proliferation and ECM protein production. Our data also suggest that UII synthesis contributes to GMC proliferation and ECM accumulation under high glucose conditions.
Collapse
|
23
|
Zeng B, Chen GL, Garcia-Vaz E, Bhandari S, Daskoulidou N, Berglund LM, Jiang H, Hallett T, Zhou LP, Huang L, Xu ZH, Nair V, Nelson RG, Ju W, Kretzler M, Atkin SL, Gomez MF, Xu SZ. ORAI channels are critical for receptor-mediated endocytosis of albumin. Nat Commun 2017; 8:1920. [PMID: 29203863 PMCID: PMC5714946 DOI: 10.1038/s41467-017-02094-y] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Accepted: 11/06/2017] [Indexed: 01/15/2023] Open
Abstract
Impaired albumin reabsorption by proximal tubular epithelial cells (PTECs) has been highlighted in diabetic nephropathy (DN), but little is known about the underlying molecular mechanisms. Here we find that ORAI1-3, are preferentially expressed in PTECs and downregulated in patients with DN. Hyperglycemia or blockade of insulin signaling reduces the expression of ORAI1-3. Inhibition of ORAI channels by BTP2 and diethylstilbestrol or silencing of ORAI expression impairs albumin uptake. Transgenic mice expressing a dominant-negative Orai1 mutant (E108Q) increases albuminuria, and in vivo injection of BTP2 exacerbates albuminuria in streptozotocin-induced and Akita diabetic mice. The albumin endocytosis is Ca2+-dependent and accompanied by ORAI1 internalization. Amnionless (AMN) associates with ORAIs and forms STIM/ORAI/AMN complexes after Ca2+ store depletion. STIM1/ORAI1 colocalizes with clathrin, but not with caveolin, at the apical membrane of PTECs, which determines clathrin-mediated endocytosis. These findings provide insights into the mechanisms of protein reabsorption and potential targets for treating diabetic proteinuria. Patients with diabetic nephropathy suffer from impaired albumin reabsorption by proximal tubular epithelial cells. Here authors use diabetic and transgenic mouse models and in vitro models to show the cause for this lies in the down regulation and internalization of the ion channels, ORAI1-3.
Collapse
Affiliation(s)
- Bo Zeng
- Centre for Cardiovascular and Metabolic Research, Hull York Medical School, University of Hull, Hull, HU6 7RX, UK. .,Key Laboratory of Medical Electrophysiology, Ministry of Education, and Institute of Cardiovascular Research, Southwest Medical University, Luzhou, 646000, China.
| | - Gui-Lan Chen
- Centre for Cardiovascular and Metabolic Research, Hull York Medical School, University of Hull, Hull, HU6 7RX, UK.,Key Laboratory of Medical Electrophysiology, Ministry of Education, and Institute of Cardiovascular Research, Southwest Medical University, Luzhou, 646000, China
| | - Eliana Garcia-Vaz
- Department of Clinical Sciences in Malmö, Lund University Diabetes Centre, Lund University, Malmö, 214 28 Malmö, Sweden
| | - Sunil Bhandari
- Department of Renal Medicine and Hull York Medical School, Hull Royal Infirmary, Hull and East Yorkshire Hospitals NHS Trust, Hull, HU3 2JZ, UK
| | - Nikoleta Daskoulidou
- Centre for Cardiovascular and Metabolic Research, Hull York Medical School, University of Hull, Hull, HU6 7RX, UK
| | - Lisa M Berglund
- Department of Clinical Sciences in Malmö, Lund University Diabetes Centre, Lund University, Malmö, 214 28 Malmö, Sweden
| | - Hongni Jiang
- Centre for Cardiovascular and Metabolic Research, Hull York Medical School, University of Hull, Hull, HU6 7RX, UK
| | - Thomas Hallett
- Centre for Cardiovascular and Metabolic Research, Hull York Medical School, University of Hull, Hull, HU6 7RX, UK
| | - Lu-Ping Zhou
- Key Laboratory of Medical Electrophysiology, Ministry of Education, and Institute of Cardiovascular Research, Southwest Medical University, Luzhou, 646000, China
| | - Li Huang
- Key Laboratory of Medical Electrophysiology, Ministry of Education, and Institute of Cardiovascular Research, Southwest Medical University, Luzhou, 646000, China
| | - Zi-Hao Xu
- Key Laboratory of Medical Electrophysiology, Ministry of Education, and Institute of Cardiovascular Research, Southwest Medical University, Luzhou, 646000, China
| | - Viji Nair
- Department of Internal Medicine & Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Robert G Nelson
- Chronic Kidney Disease Section, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Phoenix, AZ, 85014, USA
| | - Wenjun Ju
- Department of Internal Medicine & Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Matthias Kretzler
- Department of Internal Medicine & Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Stephen L Atkin
- Centre for Cardiovascular and Metabolic Research, Hull York Medical School, University of Hull, Hull, HU6 7RX, UK.,Weill Cornell Medical College Qatar, PO Box, 24144, Doha, Qatar
| | - Maria F Gomez
- Department of Clinical Sciences in Malmö, Lund University Diabetes Centre, Lund University, Malmö, 214 28 Malmö, Sweden
| | - Shang-Zhong Xu
- Centre for Cardiovascular and Metabolic Research, Hull York Medical School, University of Hull, Hull, HU6 7RX, UK.
| |
Collapse
|
24
|
Abstract
This chapter offers a brief introduction of the functions of TRPC channels in non-neuronal systems. We focus on three major organs of which the research on TRPC channels have been most focused on: kidney, heart, and lung. The chapter highlights on cellular functions and signaling pathways mediated by TRPC channels. It also summarizes several inherited diseases in humans that are related to or caused by TRPC channel mutations and malfunction. A better understanding of TRPC channels functions and the importance of TRPC channels in health and disease should lead to new insights and discovery of new therapeutic approaches for intractable disease.
Collapse
|
25
|
Antigny F, Sabourin J, Saüc S, Bernheim L, Koenig S, Frieden M. TRPC1 and TRPC4 channels functionally interact with STIM1L to promote myogenesis and maintain fast repetitive Ca 2+ release in human myotubes. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2017; 1864:806-813. [PMID: 28185894 DOI: 10.1016/j.bbamcr.2017.02.003] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/10/2016] [Revised: 01/25/2017] [Accepted: 02/03/2017] [Indexed: 12/29/2022]
Abstract
STIM1 and Orai1 are essential players of store-operated Ca2+ entry (SOCE) in human skeletal muscle cells and are required for adult muscle differentiation. Besides these two proteins, TRPC (transient receptor potential canonical) channels and STIM1L (a longer STIM1 isoform) are also present on muscle cells. In the present study, we assessed the role of TRPC1, TRPC4 and STIM1L in SOCE, in the maintenance of repetitive Ca2+ transients and in muscle differentiation. Knockdown of TRPC1 and TRPC4 reduced SOCE by about 50% and significantly delayed the onset of Ca2+ entry, both effects similar to STIM1L invalidation. Upon store depletion, TRPC1 and TRPC4 appeared to interact preferentially with STIM1L compared to STIM1. STIM1L invalidation affected myoblast differentiation, with the formation of smaller myotubes, an effect similar to what we reported for TRPC1 and TRPC4 knockdown. On the contrary, the overexpression of STIM1L leads to the formation of larger myotubes. All together, these data strongly suggest that STIM1L and TRPC1/4 are working together in myotubes to ensure efficient store refilling and a proper differentiation program.
Collapse
Affiliation(s)
- Fabrice Antigny
- Department of Basic Neurosciences, Geneva Medical Center, 1, Rue Michel Servet, 1211 Geneva 4, Switzerland
| | - Jessica Sabourin
- Inserm UMR S1180, Faculté de Pharmacie, Université Paris Sud, Université Paris-Saclay, Châtenay-Malabry, France
| | - Sophie Saüc
- Department of Basic Neurosciences, Geneva Medical Center, 1, Rue Michel Servet, 1211 Geneva 4, Switzerland; Department of Cell Physiology and Metabolism, Geneva Medical Center, 1, Rue Michel Servet, 1211 Geneva 4, Switzerland
| | - Laurent Bernheim
- Department of Basic Neurosciences, Geneva Medical Center, 1, Rue Michel Servet, 1211 Geneva 4, Switzerland
| | - Stéphane Koenig
- Department of Basic Neurosciences, Geneva Medical Center, 1, Rue Michel Servet, 1211 Geneva 4, Switzerland
| | - Maud Frieden
- Department of Cell Physiology and Metabolism, Geneva Medical Center, 1, Rue Michel Servet, 1211 Geneva 4, Switzerland.
| |
Collapse
|
26
|
Lee JM, Noguchi S. Calcium Dyshomeostasis in Tubular Aggregate Myopathy. Int J Mol Sci 2016; 17:ijms17111952. [PMID: 27879676 PMCID: PMC5133946 DOI: 10.3390/ijms17111952] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Revised: 11/15/2016] [Accepted: 11/15/2016] [Indexed: 11/16/2022] Open
Abstract
Calcium is a crucial mediator of cell signaling in skeletal muscles for basic cellular functions and specific functions, including contraction, fiber-type differentiation and energy production. The sarcoplasmic reticulum (SR) is an organelle that provides a large supply of intracellular Ca2+ in myofibers. Upon excitation, it releases Ca2+ into the cytosol, inducing contraction of myofibrils. During relaxation, it takes up cytosolic Ca2+ to terminate the contraction. During exercise, Ca2+ is cycled between the cytosol and the SR through a system by which the Ca2+ pool in the SR is restored by uptake of extracellular Ca2+ via a specific channel on the plasma membrane. This channel is called the store-operated Ca2+ channel or the Ca2+ release-activated Ca2+ channel. It is activated by depletion of the Ca2+ store in the SR by coordination of two main molecules: stromal interaction molecule 1 (STIM1) and calcium release-activated calcium channel protein 1 (ORAI1). Recently, myopathies with a dominant mutation in these genes have been reported and the pathogenic mechanism of such diseases have been proposed. This review overviews the calcium signaling in skeletal muscles and role of store-operated Ca2+ entry in calcium homeostasis. Finally, we discuss the phenotypes and the pathomechanism of myopathies caused by mutations in the STIM1 and ORAI1 genes.
Collapse
Affiliation(s)
- Jong-Mok Lee
- Department of Genome Medicine Development, Medical Genome Center, National Center of Neurology and Neuropsychiatry, Kodaira, Tokyo 187-8551, Japan.
- Department of Neuromuscular Research, National Institute of Neuroscience, National Center of Neurology and Neuropsychiatry, Kodaira, Tokyo 187-8502, Japan.
| | - Satoru Noguchi
- Department of Genome Medicine Development, Medical Genome Center, National Center of Neurology and Neuropsychiatry, Kodaira, Tokyo 187-8551, Japan.
- Department of Neuromuscular Research, National Institute of Neuroscience, National Center of Neurology and Neuropsychiatry, Kodaira, Tokyo 187-8502, Japan.
| |
Collapse
|
27
|
Tao J, Lan Z, Wang Y, Hei H, Tian L, Pan W, Zhang X, Peng W. Large-Conductance Calcium-Activated Potassium Channels in Glomerulus: From Cell Signal Integration to Disease. Front Physiol 2016; 7:248. [PMID: 27445840 PMCID: PMC4915313 DOI: 10.3389/fphys.2016.00248] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2016] [Accepted: 06/06/2016] [Indexed: 12/21/2022] Open
Abstract
Large-conductance calcium-activated potassium (BK) channels are currently considered as vital players in a variety of renal physiological processes. In podocytes, BK channels become active in response to stimuli that increase local cytosolic Ca2+, possibly secondary to activation of slit diaphragm TRPC6 channels by chemical or mechanical stimuli. Insulin increases filtration barrier permeability through mobilization of BK channels. In mesangial cells, BK channels co-expressed with β1 subunits act as a major component of the counteractive response to contraction in order to regulate glomerular filtration. This review aims to highlight recent discoveries on the localization, physiological and pathological roles of BK channels in glomerulus.
Collapse
Affiliation(s)
- Jie Tao
- Department of Nephrology and Central Laboratory, Putuo Hospital, Shanghai University of Traditional Chinese Medicine Shanghai, China
| | - Zhen Lan
- Department of Nephrology and Central Laboratory, Putuo Hospital, Shanghai University of Traditional Chinese Medicine Shanghai, China
| | - Yunman Wang
- Department of Nephrology and Central Laboratory, Putuo Hospital, Shanghai University of Traditional Chinese Medicine Shanghai, China
| | - Hongya Hei
- Department of Pharmacology, School of Pharmacy, Fudan University Shanghai, China
| | - Lulu Tian
- Department of Pharmacology, School of Pharmacy, Fudan University Shanghai, China
| | - Wanma Pan
- Department of Pharmacology, School of Pharmacy, Fudan University Shanghai, China
| | - Xuemei Zhang
- Department of Pharmacology, School of Pharmacy, Fudan University Shanghai, China
| | - Wen Peng
- Department of Nephrology and Central Laboratory, Putuo Hospital, Shanghai University of Traditional Chinese Medicine Shanghai, China
| |
Collapse
|
28
|
Zhou Y, Greka A. Calcium-permeable ion channels in the kidney. Am J Physiol Renal Physiol 2016; 310:F1157-67. [PMID: 27029425 DOI: 10.1152/ajprenal.00117.2016] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2016] [Accepted: 03/29/2016] [Indexed: 02/07/2023] Open
Abstract
Calcium ions (Ca(2+)) are crucial for a variety of cellular functions. The extracellular and intracellular Ca(2+) concentrations are thus tightly regulated to maintain Ca(2+) homeostasis. The kidney, one of the major organs of the excretory system, regulates Ca(2+) homeostasis by filtration and reabsorption. Approximately 60% of the Ca(2+) in plasma is filtered, and 99% of that is reabsorbed by the kidney tubules. Ca(2+) is also a critical signaling molecule in kidney development, in all kidney cellular functions, and in the emergence of kidney diseases. Recently, studies using genetic and molecular biological approaches have identified several Ca(2+)-permeable ion channel families as important regulators of Ca(2+) homeostasis in kidney. These ion channel families include transient receptor potential channels (TRP), voltage-gated calcium channels, and others. In this review, we provide a brief and systematic summary of the expression, function, and pathological contribution for each of these Ca(2+)-permeable ion channels. Moreover, we discuss their potential as future therapeutic targets.
Collapse
Affiliation(s)
- Yiming Zhou
- Department of Medicine and Glom-NExT Center for Glomerular Kidney Disease and Novel Experimental Therapeutics, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts; and
| | - Anna Greka
- Department of Medicine and Glom-NExT Center for Glomerular Kidney Disease and Novel Experimental Therapeutics, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts; and The Broad Institute of MIT and Harvard, Cambridge, Massachusetts
| |
Collapse
|
29
|
Abstract
Store-operated Ca(2+) entry (SOCE) is mediated by the store-operated Ca(2+) channel (SOC) that opens upon depletion of internal Ca(2+) stores following activation of G protein-coupled receptors or receptor tyrosine kinases. Over the past two decades, the physiological and pathological relevance of SOCE has been extensively studied. Recently, accumulating evidence suggests associations of altered SOCE with diabetic complications. This review focuses on the implication of SOCE as it pertains to various complications resulting from diabetes. We summarize recent findings by us and others on the involvement of abnormal SOCE in the development of diabetic complications, such as diabetic nephropathy and diabetic vasculopathy. The underlying mechanisms that mediate the diabetes-associated alterations of SOCE are also discussed. The SOCE pathway may be considered as a potential therapeutic target for diabetes-associated diseases.
Collapse
Affiliation(s)
- Sarika Chaudhari
- Department of Integrative Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth 76107, TX, USA
| | - Rong Ma
- Department of Integrative Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth 76107, TX, USA
| |
Collapse
|
30
|
Ong HL, de Souza LB, Ambudkar IS. Role of TRPC Channels in Store-Operated Calcium Entry. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 898:87-109. [DOI: 10.1007/978-3-319-26974-0_5] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
|
31
|
Albarran L, Lopez JJ, Salido GM, Rosado JA. Historical Overview of Store-Operated Ca(2+) Entry. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 898:3-24. [PMID: 27161222 DOI: 10.1007/978-3-319-26974-0_1] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Calcium influx is an essential mechanism for the activation of cellular functions both in excitable and non-excitable cells. In non-excitable cells, activation of phospholipase C by occupation of G protein-coupled receptors leads to the generation of inositol 1,4,5-trisphosphate (IP3) and diacylglycerol (DAG), which, in turn, initiate two Ca(2+) entry pathways: Ca(2+) release from intracellular Ca(2+) stores, signaled by IP3, leads to the activation of store-operated Ca(2+) entry (SOCE); on the other hand, DAG activates a distinct second messenger-operated pathway. SOCE is regulated by the filling state of the intracellular calcium stores. The search for the molecular components of SOCE has identified the stromal interaction molecule 1 (STIM1) as the Ca(2+) sensor in the endoplasmic reticulum and Orai1 as a store-operated channel (SOC) subunit. Furthermore, a number of reports have revealed that several members of the TRPC family of channels also take part of the SOC macromolecular complex. This introductory chapter summarizes the early pieces of evidence that led to the concept of SOCE and the components of the store-operated signaling pathway.
Collapse
Affiliation(s)
- Letizia Albarran
- Department of Physiology (Cell Physiology Research Group), University of Extremadura, Av. Universidad s/n, 10003, Cáceres, Spain
| | - Jose J Lopez
- Department of Physiology (Cell Physiology Research Group), University of Extremadura, Av. Universidad s/n, 10003, Cáceres, Spain
| | - Ginés M Salido
- Department of Physiology (Cell Physiology Research Group), University of Extremadura, Av. Universidad s/n, 10003, Cáceres, Spain
| | - Juan A Rosado
- Departamento de Fisiología, University of Extremadura, Cáceres, Spain.
| |
Collapse
|
32
|
Zhong X, Fu J, Song K, Xue N, Gong R, Sun D, Luo H, He W, Pan X, Shen B, Du J. The role of TRPP2 in agonist-induced gallbladder smooth muscle contraction. SCIENCE CHINA-LIFE SCIENCES 2015; 59:409-16. [DOI: 10.1007/s11427-015-4958-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/26/2015] [Accepted: 09/01/2015] [Indexed: 11/30/2022]
|
33
|
Wang Y, Chaudhari S, Ren Y, Ma R. Impairment of hepatic nuclear factor-4α binding to the Stim1 promoter contributes to high glucose-induced upregulation of STIM1 expression in glomerular mesangial cells. Am J Physiol Renal Physiol 2015; 308:F1135-45. [PMID: 25786776 PMCID: PMC4437002 DOI: 10.1152/ajprenal.00563.2014] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2014] [Accepted: 03/16/2015] [Indexed: 11/22/2022] Open
Abstract
The present study was carried out to investigate if hepatic nuclear factor (HNF)4α contributed to the high glucose-induced increase in stromal interacting molecule (STIM)1 protein abundance in glomerular mesangial cells (MCs). Western blot and immunofluorescence experiments showed HNF4α expression in MCs. Knockdown of HNF4α using a small interfering RNA approach significantly increased mRNA expression levels of both STIM1 and Orai1 and protein expression levels of STIM1 in cultured human MCs. Consistently, overexpression of HNF4α reduced expressed STIM1 protein expression in human embryonic kidney-293 cells. Furthermore, high glucose treatment did not significantly change the abundance of HNF4α protein in MCs but significantly attenuated HNF4α binding activity to the Stim1 promoter. Moreover, knockdown of HNF4α significantly augmented store-operated Ca(2+) entry, which is known to be gated by STIM1 and has recently been found to be antifibrotic in MCs. In agreement with those results, knockdown of HNF4α significantly attenuated the fibrotic response of high glucose. These results suggest that HNF4α negatively regulates STIM1 transcription in MCs. High glucose increases STIM1 expression levels by impairing HNF4α binding activity to the Stim1 promoter, which subsequently releases Stim1 transcription from HNF4α repression. Since the STIM1-gated store-operated Ca(2+) entry pathway in MCs has an antifibrotic effect, inhibition of HNF4α in MCs might be a potential therapeutic option for diabetic kidney disease.
Collapse
Affiliation(s)
- Yanxia Wang
- Department of Integrative Physiology and Cardiovascular Research Institute, University of North Texas Health Science Center, Fort Worth, Texas; and
| | - Sarika Chaudhari
- Department of Integrative Physiology and Cardiovascular Research Institute, University of North Texas Health Science Center, Fort Worth, Texas; and
| | - Yuezhong Ren
- Department of Endocrinology, The Second Affiliated Hospital of Zhejiang University College of Medicine, Hangzhou, Zhejiang, China
| | - Rong Ma
- Department of Integrative Physiology and Cardiovascular Research Institute, University of North Texas Health Science Center, Fort Worth, Texas; and
| |
Collapse
|
34
|
Wu P, Wang Y, Davis ME, Zuckerman JE, Chaudhari S, Begg M, Ma R. Store-Operated Ca2+ Channels in Mesangial Cells Inhibit Matrix Protein Expression. J Am Soc Nephrol 2015; 26:2691-702. [PMID: 25788524 DOI: 10.1681/asn.2014090853] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2014] [Accepted: 12/22/2014] [Indexed: 11/03/2022] Open
Abstract
Accumulation of extracellular matrix derived from glomerular mesangial cells is an early feature of diabetic nephropathy. Ca(2+) signals mediated by store-operated Ca(2+) channels regulate protein production in a variety of cell types. The aim of this study was to determine the effect of store-operated Ca(2+) channels in mesangial cells on extracellular matrix protein expression. In cultured human mesangial cells, activation of store-operated Ca(2+) channels by thapsigargin significantly decreased fibronectin protein expression and collagen IV mRNA expression in a dose-dependent manner. Conversely, inhibition of the channels by 2-aminoethyl diphenylborinate significantly increased the expression of fibronectin and collagen IV. Similarly, overexpression of stromal interacting molecule 1 reduced, but knockdown of calcium release-activated calcium channel protein 1 (Orai1) increased fibronectin protein expression. Furthermore, 2-aminoethyl diphenylborinate significantly augmented angiotensin II-induced fibronectin protein expression, whereas thapsigargin abrogated high glucose- and TGF-β1-stimulated matrix protein expression. In vivo knockdown of Orai1 in mesangial cells of mice using a targeted nanoparticle siRNA delivery system resulted in increased expression of glomerular fibronectin and collagen IV, and mice showed significant mesangial expansion compared with controls. Similarly, in vivo knockdown of stromal interacting molecule 1 in mesangial cells by recombinant adeno-associated virus-encoded shRNA markedly increased collagen IV protein expression in renal cortex and caused mesangial expansion in rats. These results suggest that store-operated Ca(2+) channels in mesangial cells negatively regulate extracellular matrix protein expression in the kidney, which may serve as an endogenous renoprotective mechanism in diabetes.
Collapse
Affiliation(s)
- Peiwen Wu
- Department of Integrative Physiology and Anatomy and Cardiovascular Research Institute, University of North Texas Health Science Center, Fort Worth, Texas; Department of Endocrinology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, People's Republic of China
| | - Yanxia Wang
- Department of Integrative Physiology and Anatomy and Cardiovascular Research Institute, University of North Texas Health Science Center, Fort Worth, Texas
| | - Mark E Davis
- Chemical Engineering, California Institute of Technology, Pasadena, California; and
| | - Jonathan E Zuckerman
- Chemical Engineering, California Institute of Technology, Pasadena, California; and
| | - Sarika Chaudhari
- Department of Integrative Physiology and Anatomy and Cardiovascular Research Institute, University of North Texas Health Science Center, Fort Worth, Texas
| | - Malcolm Begg
- Respiratory Therapy Area Unit, Medicines Research Center, GlaxoSmithKline, Stevenage, United Kingdom
| | - Rong Ma
- Department of Integrative Physiology and Anatomy and Cardiovascular Research Institute, University of North Texas Health Science Center, Fort Worth, Texas;
| |
Collapse
|
35
|
Redondo PC, Rosado JA. Store-operated calcium entry: unveiling the calcium handling signalplex. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2015; 316:183-226. [PMID: 25805125 DOI: 10.1016/bs.ircmb.2015.01.007] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Store-operated Ca(2+) entry (SOCE) is an important mechanism for Ca(2+) influx in non-excitable cells, also present in excitable cells. The activation of store-operated channels (SOCs) is finely regulated by the filling state of the intracellular agonist-sensitive Ca(2+) compartments, and both, the mechanism of sensing the Ca(2+) stores and the nature and functional properties of the SOCs, have been a matter of intense investigation and debate. The identification of STIM1 as the endoplasmic reticulum Ca(2+) sensor and both Orai1, as the pore-forming subunit of the channels mediating the Ca(2+)-selective store-operated current, and the members of the TRPC subfamily of proteins, as the channels mediating the cation-permeable SOCs, has shed new light on the underlying events. This review summarizes the initial hypothesis and the current advances on the mechanism of activation of SOCE.
Collapse
Affiliation(s)
- Pedro C Redondo
- Department of Physiology, University of Extremadura, Cáceres, Spain
| | - Juan A Rosado
- Department of Physiology, University of Extremadura, Cáceres, Spain
| |
Collapse
|
36
|
Fu J, Gao Z, Shen B, Zhu MX. Canonical transient receptor potential 4 and its small molecule modulators. SCIENCE CHINA-LIFE SCIENCES 2014; 58:39-47. [PMID: 25480324 DOI: 10.1007/s11427-014-4772-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/16/2014] [Accepted: 09/06/2014] [Indexed: 02/04/2023]
Abstract
Canonical transient receptor potential 4 (TRPC4) forms non-selective cation channels that contribute to phospholipase C-dependent Ca(2+) entry into cells following stimulation of G protein coupled receptors and receptor tyrosine kinases. Moreover, the channels are regulated by pertussis toxin-sensitive Gi/o proteins, lipids, and various other signaling mechanisms. TRPC4-containing channels participate in the regulation of a variety of physiological functions, including excitability of both gastrointestinal smooth muscles and brain neurons. This review is to present recent advances in the understanding of physiology and development of small molecular modulators of TRPC4 channels.
Collapse
Affiliation(s)
- Jie Fu
- Department of Physiology, Anhui Medical University, Hefei, 230032, China
| | | | | | | |
Collapse
|
37
|
Nielsen N, Lindemann O, Schwab A. TRP channels and STIM/ORAI proteins: sensors and effectors of cancer and stroma cell migration. Br J Pharmacol 2014; 171:5524-40. [PMID: 24724725 DOI: 10.1111/bph.12721] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2013] [Revised: 03/24/2014] [Accepted: 04/03/2014] [Indexed: 01/05/2023] Open
Abstract
UNLABELLED Cancer cells are strongly influenced by host cells within the tumour stroma and vice versa. This leads to the development of a tumour microenvironment with distinct physical and chemical properties that are permissive for tumour progression. The ability to migrate plays a central role in this mutual interaction. Migration of cancer cells is considered as a prerequisite for tumour metastasis and the migration of host stromal cells is required for reaching the tumour site. Increasing evidence suggests that transient receptor potential (TRP) channels and STIM/ORAI proteins affect key calcium-dependent mechanisms implicated in both cancer and stroma cell migration. These include, among others, cytoskeletal remodelling, growth factor/cytokine signalling and production, and adaptation to tumour microenvironmental properties such as hypoxia and oxidative stress. In this review, we will summarize the current knowledge regarding TRP channels and STIM/ORAI proteins in cancer and stroma cell migration. We focus on how TRP channel or STIM/ORAI-mediated Ca(2+) signalling directly or indirectly influences cancer and stroma cell migration by affecting the above listed mechanisms. LINKED ARTICLES This article is part of a themed section on Cytoskeleton, Extracellular Matrix, Cell Migration, Wound Healing and Related Topics. To view the other articles in this section visit http://dx.doi.org/10.1111/bph.2014.171.issue-24.
Collapse
Affiliation(s)
- N Nielsen
- Institute of Physiology II, University of Münster, Münster, Germany
| | | | | |
Collapse
|
38
|
Meng K, Xu J, Zhang C, Zhang R, Yang H, Liao C, Jiao J. Calcium sensing receptor modulates extracellular calcium entry and proliferation via TRPC3/6 channels in cultured human mesangial cells. PLoS One 2014; 9:e98777. [PMID: 24905090 PMCID: PMC4048219 DOI: 10.1371/journal.pone.0098777] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2014] [Accepted: 05/07/2014] [Indexed: 12/31/2022] Open
Abstract
Calcium-sensing receptor (CaSR) has been demonstrated to be present in several tissues and cells unrelated to systemic calcium homeostasis, where it regulates a series of diverse cellular functions. A previous study indicated that CaSR is expressed in mouse glomerular mesangial cells (MCs), and stimulation of CaSR induces cell proliferation. However, the signaling cascades initiated by CaSR activation in MCs are currently unknown. In this study, our data demonstrate that CaSR mRNA and protein are expressed in a human mesangial cell line. Activating CaSR with high extracellular Ca2+ concentration ([Ca2+]o) or spermine induces a phospholipase C (PLC)-dependent increase in intracellular Ca2+ concentration ([Ca2+]i). Interestingly, the CaSR activation-induced increase in [Ca2+]i results not only from intracellular Ca2+ release from internal stores but also from canonical transient receptor potential (TRPC)-dependent Ca2+ influx. This increase in Ca2+ was attenuated by treatment with a nonselective TRPC channel blocker but not by treatment with a voltage-gated calcium blocker or Na+/Ca2+ exchanger inhibitor. Furthermore, stimulation of CaSR by high [Ca2+]o enhanced the expression of TRPC3 and TRPC6 but not TRPC1 and TRPC4, and siRNA targeting TRPC3 and TRPC6 attenuated the CaSR activation-induced [Ca2+]i increase. Further experiments indicate that 1-oleoyl-2-acetyl-sn-glycerol (OAG), a known activator of receptor-operated calcium channels, significantly enhances the CaSR activation-induced [Ca2+]i increase. Moreover, under conditions in which intracellular stores were already depleted with thapsigargin (TG), CaSR agonists also induced an increase in [Ca2+]i, suggesting that calcium influx stimulated by CaSR agonists does not require the release of calcium stores. Finally, our data indicate that pharmacological inhibition and knock down of TRPC3 and TRPC6 attenuates the CaSR activation-induced cell proliferation in human MCs. With these data, we conclude that CaSR activation mediates Ca2+ influx and cell proliferation via TRPC3 and TRPC6 in human MCs.
Collapse
Affiliation(s)
- Kexin Meng
- Department of Nephrology, The Second Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Jia Xu
- Department of Nephrology, The Fourth Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Chengwei Zhang
- Department of Nephrology, The Second Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Rui Zhang
- Department of Nephrology, The Second Affiliated Hospital, Harbin Medical University, Harbin, China
| | - He Yang
- Department of Nephrology, The Second Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Chang Liao
- Department of Nephrology, The Second Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Jundong Jiao
- Department of Nephrology, The Second Affiliated Hospital, Harbin Medical University, Harbin, China
- Institute of Nephrology, Harbin Medical University, Harbin, China
- * E-mail:
| |
Collapse
|
39
|
ADEBIYI ADEBOWALE. RGS2 regulates urotensin II-induced intracellular Ca2+ elevation and contraction in glomerular mesangial cells. J Cell Physiol 2014; 229:502-11. [PMID: 24105430 PMCID: PMC11250777 DOI: 10.1002/jcp.24470] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2012] [Accepted: 09/06/2013] [Indexed: 01/01/2023]
Abstract
Urotensin II (UII), a vasoactive peptide modulates renal hemodynamics. However, the physiological functions of UII in glomerular cells are unclear. In particular, whether UII alters mesangial tone remains largely unknown. The present study investigates the physiological effects of UII on glomerular mesangial cells (GMCs). This study also tested the hypothesis that the regulator of G-protein signaling (RGS) controls UII receptor (UTR) activity in GMCs. RT-PCR, Western immunoblotting, and immunofluorescence revealed UTR expression in cultured murine GMCs. Mouse UII (mUII) stimulated Ca(2+) release from intracellular stores and activated store-operated Ca(2+) entry (SOCE) in the cells. mUII also caused a reduction in planar GMC surface area. mUII-induced [Ca(2+)]i elevation and contraction were attenuated by SB 657510, a UTR antagonist, araguspongin B, an inositol 1,4,5-trisphosphate receptor antagonist, thapsigargin, a sarco/endoplasmic reticulum Ca(2+)-ATPase inhibitor, and La(3+), a store-operated Ca(2+) channel blocker, but not nimodipine, an L-type Ca(2+) channel blocker. In situ proximity ligation assay indicated molecular proximity between endogenous RGS2 and UTR in the cells. Treatment of GMCs with mUII elevated plasma membrane expression of RGS2 by ∼2-fold. mUII also increased the interaction between RGS2 and UTR in the cells. siRNA-mediated knockdown of RGS2 in murine GMCs increased mUII-induced [Ca(2+)]i elevation and contraction by ∼35 and 31%, respectively. These findings indicate that mUII-induced SOCE results in murine GMC contraction. These data also suggest that UTR activation stimulates RGS2 recruitment to GMC plasma membrane as a negative feedback mechanism to regulate UTR signaling.
Collapse
Affiliation(s)
- ADEBOWALE ADEBIYI
- Department of Physiology, University of Tennessee Health Science Center, Memphis, Tennessee
| |
Collapse
|
40
|
Chaudhari S, Wu P, Wang Y, Ding Y, Yuan J, Begg M, Ma R. High glucose and diabetes enhanced store-operated Ca(2+) entry and increased expression of its signaling proteins in mesangial cells. Am J Physiol Renal Physiol 2014; 306:F1069-80. [PMID: 24623143 DOI: 10.1152/ajprenal.00463.2013] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The present study was conducted to determine whether and how store-operated Ca(2+) entry (SOCE) in glomerular mesangial cells (MCs) was altered by high glucose (HG) and diabetes. Human MCs were treated with either normal glucose or HG for different time periods. Cyclopiazonic acid-induced SOCE was significantly greater in the MCs with 7-day HG treatment and the response was completely abolished by GSK-7975A, a selective inhibitor of store-operated Ca(2+) channels. Similarly, the inositol 1,4,5-trisphosphate-induced store-operated Ca(2+) currents were significantly enhanced in the MCs treated with HG for 7 days, and the enhanced response was abolished by both GSK-7975A and La(3+). In contrast, receptor-operated Ca(2+) entry in MCs was significantly reduced by HG treatment. Western blotting showed that HG increased the expression levels of STIM1 and Orai1 in cultured MCs. A significant HG effect occurred at a concentration as low as 10 mM, but required a minimum of 7 days. The HG effect in cultured MCs was recapitulated in renal glomeruli/cortex of both type I and II diabetic rats. Furthermore, quantitative real-time RT-PCR revealed that a 6-day HG treatment significantly increased the mRNA expression level of STIM1. However, the expressions of STIM2 and Orai1 transcripts were not affected by HG. Taken together, these results suggest that HG/diabetes enhanced SOCE in MCs by increasing STIM1/Orai1 protein expressions. HG upregulates STIM1 by promoting its transcription but increases Orai1 protein through a posttranscriptional mechanism.
Collapse
Affiliation(s)
- Sarika Chaudhari
- 3500 Camp Bowie Blvd., Dept. of Integrative Physiology, Univ. of North Texas Health Science Center, Fort Worth, TX 76107.
| | | | | | | | | | | | | |
Collapse
|
41
|
Ong HL, de Souza LB, Cheng KT, Ambudkar IS. Physiological functions and regulation of TRPC channels. Handb Exp Pharmacol 2014; 223:1005-34. [PMID: 24961978 DOI: 10.1007/978-3-319-05161-1_12] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The TRP-canonical (TRPC) subfamily, which consists of seven members (TRPC1-TRPC7), are Ca(2+)-permeable cation channels that are activated in response to receptor-mediated PIP2 hydrolysis via store-dependent and store-independent mechanisms. These channels are involved in a variety of physiological functions in different cell types and tissues. Of these, TRPC6 has been linked to a channelopathy resulting in human disease. Two key players of the store-dependent regulatory pathway, STIM1 and Orai1, interact with some TRPC channels to gate and regulate channel activity. The Ca(2+) influx mediated by TRPC channels generates distinct intracellular Ca(2+) signals that regulate downstream signaling events and consequent cell functions. This requires localization of TRPC channels in specific plasma membrane microdomains and precise regulation of channel function which is coordinated by various scaffolding, trafficking, and regulatory proteins.
Collapse
Affiliation(s)
- Hwei Ling Ong
- Secretory Physiology Section, Molecular Physiology and Therapeutics Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, 20892, USA
| | | | | | | |
Collapse
|
42
|
Abstract
TRPC4 proteins comprise six transmembrane domains, a putative pore-forming region, and an intracellularly located amino- and carboxy-terminus. Among eleven splice variants identified so far, TRPC4α and TRPC4β are the most abundantly expressed and functionally characterized. TRPC4 is expressed in various organs and cell types including the soma and dendrites of numerous types of neurons; the cardiovascular system including endothelial, smooth muscle, and cardiac cells; myometrial and skeletal muscle cells; kidney; and immune cells such as mast cells. Both recombinant and native TRPC4-containing channels differ tremendously in their permeability and other biophysical properties, pharmacological modulation, and mode of activation depending on the cellular environment. They vary from inwardly rectifying store-operated channels with a high Ca(2+) selectivity to non-store-operated channels predominantly carrying Na(+) and activated by Gαq- and/or Gαi-coupled receptors with a complex U-shaped current-voltage relationship. Thus, individual TRPC4-containing channels contribute to agonist-induced Ca(2+) entry directly or indirectly via depolarization and activation of voltage-gated Ca(2+) channels. The differences in channel properties may arise from variations in the composition of the channel complexes, in the specific regulatory pathways in the corresponding cell system, and/or in the expression pattern of interaction partners which comprise other TRPC proteins to form heteromultimeric channels. Additional interaction partners of TRPC4 that can mediate the activity of TRPC4-containing channels include (1) scaffolding proteins (e.g., NHERF) that may mediate interactions with signaling molecules in or in close vicinity to the plasma membrane such as Gα proteins or phospholipase C and with the cytoskeleton, (2) proteins in specific membrane microdomains (e.g., caveolin-1), or (3) proteins on cellular organelles (e.g., Stim1). The diversity of TRPC4-containing channels hampers the development of specific agonists or antagonists, but recently, ML204 was identified as a blocker of both recombinant and endogenous TRPC4-containing channels with an IC50 in the lower micromolar range that lacks activity on most voltage-gated channels and other TRPs except TRPC5 and TRPC3. Lanthanides are specific activators of heterologously expressed TRPC4- and TRPC5-containing channels but can block individual native TRPC4-containing channels. The biological relevance of TRPC4-containing channels was demonstrated by knockdown of TRPC4 expression in numerous native systems including gene expression, cell differentiation and proliferation, formation of myotubes, and axonal regeneration. Studies of TRPC4 single and TRPC compound knockout mice uncovered their role for the regulation of vascular tone, endothelial permeability, gastrointestinal contractility and motility, neurotransmitter release, and social exploratory behavior as well as for excitotoxicity and epileptogenesis. Recently, a single-nucleotide polymorphism (SNP) in the Trpc4 gene was associated with a reduced risk for experience of myocardial infarction.
Collapse
Affiliation(s)
- Marc Freichel
- Pharmakologisches Institut, Ruprecht-Karls-Universität Heidelberg, Im Neuenheimer Feld 366, 69120, Heidelberg, Germany,
| | | | | |
Collapse
|
43
|
Shen B, Zhu J, Zhang J, Jiang F, Wang Z, Zhang Y, Li J, Huang D, Ke D, Ma R, Du J. Attenuated mesangial cell proliferation related to store-operated Ca2+ entry in aged rat: the role of STIM 1 and Orai 1. AGE (DORDRECHT, NETHERLANDS) 2013; 35:2193-2202. [PMID: 23334602 PMCID: PMC3824990 DOI: 10.1007/s11357-013-9511-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/25/2012] [Accepted: 01/09/2013] [Indexed: 06/01/2023]
Abstract
Store-operated Ca(2+) entry (SOCE) is a common and ubiquitous mechanism regulating Ca(2+) influx into cells and participates in numerous biological processes including cell proliferation. Glomerular mesangial cells (GMCs) play a role in the regulation of the glomerular filtration rate. From a clinical point of view, many physiological functions alter with age. In the present study, we used angiotensin II, glucagon, and the sarco/endoplasmic reticulum membrane Ca(2+) pump inhibitor thapsigargin to deplete the internal Ca(2+) stores for the activation of SOCE. We found that SOCE was significantly attenuated in GMCs from aged (22-month-old) rats. The expression of SOCE-related components, stromal interaction molecule 1 (STIM 1) and Orai 1, in freshly isolated glomeruli notably decreased, and STIM 1 and Orai 1 puncta formation significantly reduced in primary-cultured GMCs in aged rats. Moreover, specific knockdown of STIM 1 and Orai 1 by small interfering RNA markedly suppressed SOCE and cell proliferation of GMCs isolated from young (3-month-old) rats. We conclude that the attenuation of GMCs proliferation can be attributed to the decreased SOCE partially caused by reduced expression of STIM 1 and Orai 1.
Collapse
Affiliation(s)
- Bing Shen
- />Department of Physiology, Anhui Medical University, Hefei, China
| | - Jinhang Zhu
- />Department of Physiology, Anhui Medical University, Hefei, China
| | - Jin Zhang
- />Department of Physiology, Anhui Medical University, Hefei, China
| | - Feifei Jiang
- />Department of Physiology, Anhui Medical University, Hefei, China
| | - Zhaoyi Wang
- />Department of Physiology, Anhui Medical University, Hefei, China
| | - Yang Zhang
- />Comprehensive Surgery, Anhui Provincial Hospital, Hefei, 230032 China
| | - Jie Li
- />Department of Physiology, Anhui Medical University, Hefei, China
| | - Dake Huang
- />Comprehensive Laboratory of Basic Medical School, Anhui Medical University, Hefei, China
| | - Daoping Ke
- />Department of Physiology, Anhui Medical University, Hefei, China
| | - Rong Ma
- />Department of Integrative Physiology, University of North Texas Health Science Center at Fort Worth, Fort Worth, TX USA
| | - Juan Du
- />Department of Physiology, Anhui Medical University, Hefei, China
| |
Collapse
|
44
|
Saul S, Stanisz H, Backes CS, Schwarz EC, Hoth M. How ORAI and TRP channels interfere with each other: interaction models and examples from the immune system and the skin. Eur J Pharmacol 2013; 739:49-59. [PMID: 24291108 DOI: 10.1016/j.ejphar.2013.10.071] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2013] [Revised: 10/08/2013] [Accepted: 10/17/2013] [Indexed: 11/16/2022]
Abstract
Four types of Ca(2+) selective ion channels are known, ten voltage gated Ca(2+) (CaV) channels, four CatSper channels, three store operated CRAC channels (ORAI channels) and at least two members of the TRPV subfamily (TRPV5, TRPV6). Some of the other TRP channels also show some Ca(2+) selectivity like certain splice variants of TRPM3. In addition to Ca(2+) selective channels, various cation channels play an important role for Ca(2+) entry and furthermore, they may also regulate Ca(2+) entry through other channels by modulating the membrane potential or other means as outlined in this review. Of the different types of cation channels, TRP channels form one of the most prominent families of non-selective cation channels with functional relevance in electrically non-excitable and electrically excitable cell types. Among these, the seven channels of the TRPC subfamily are rather non-selective with very modest Ca(2+) selectivity, whereas in the other subfamilies, cation selectivity ranges from monovalent selectivity (i.e. TRPM4, TRPM5) to divalent selectivity (i.e. TRPM6, TRPM7) or Ca(2+) selectivity (i.e. TRPV5, TRPV6). Rather than discussing the heavily reviewed individual functions of ORAI or TRP channels, we summarize data and present models how TRP and ORAI may functionally interact to guide cellular functions. We focus on T lymphocytes representing a more ORAI-dominated tissue and skin as model system in which both ORAI and TRP channel have been reported to control relevant functions. We present several interaction models how ORAI and TRP may interfere with each other's function.
Collapse
Affiliation(s)
- Stephanie Saul
- Department of Biophysics, School of Medicine, Saarland University, Homburg, Germany
| | - Hedwig Stanisz
- Department of Dermatology, School of Medicine, Saarland University, Homburg, Germany
| | - Christian S Backes
- Department of Biophysics, School of Medicine, Saarland University, Homburg, Germany
| | - Eva C Schwarz
- Department of Biophysics, School of Medicine, Saarland University, Homburg, Germany
| | - Markus Hoth
- Department of Biophysics, School of Medicine, Saarland University, Homburg, Germany.
| |
Collapse
|
45
|
Wang Y, Ding M, Chaudhari S, Ding Y, Yuan J, Stankowska D, He S, Krishnamoorthy R, Cunningham JT, Ma R. Nuclear factor κB mediates suppression of canonical transient receptor potential 6 expression by reactive oxygen species and protein kinase C in kidney cells. J Biol Chem 2013; 288:12852-65. [PMID: 23525112 PMCID: PMC3642329 DOI: 10.1074/jbc.m112.410357] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2012] [Revised: 03/21/2013] [Indexed: 02/05/2023] Open
Abstract
This study was carried out to explore the molecular mechanism for down-regulation of TRPC6 expression in the reactive oxygen species (ROS)/PKC signaling in kidney cells. In cultured human mesangial cells, H2O2 and TNF-α inhibited TRPC6 mRNA expression in a time-dependent manner. Inhibition of NF-κB reversed both H2O2- and phorbol 12-myristate 13-acetate (PMA)-induced decrease in TRPC6 protein expression. Activation of NF-κB by knocking down IκBα using siRNA could mimic the suppressive effect of ROS/PKC on TRPC6. a Ca(2+) imaging study showed that activation and inhibition of NF-κB significantly decreased and increased the TRPC6-mediated Ca(2+) entry, respectively. Further experiments showed that PMA, but not its inactive analog 4α-phorbol 12, 13-didecanoate (4α-PDD), caused phosphorylation of IκBα and stimulated the nuclear translocation of NF-κB p50 and p65 subunits. The PMA-dependent IκBα phosphorylation was significantly inhibited by Gö6976. Electrophoretic mobility shift assay revealed that PMA stimulated DNA binding activity of NF-κB. Furthermore, specific knockdown of p65, but not p50, prevented an H2O2 inhibitory effect on TRPC6 protein expression, suggesting p65 as a predominant NF-κB subunit repressing TRPC6. In agreement with a major role of p65, chromatin immunoprecipitation assays showed that PMA treatment induced p65 binding to the TRPC6 promoter. Moreover, PMA treatment increased the association of p65 with histone deacetylase (HDAC) and decreased histone acetylation at the TRPC6 promoter. Consistently, knockdown of HDAC2 by siRNA or inhibition of HDAC with trichostatin A prevented a H2O2-induced decrease in TRPC6 mRNA and protein expressions, respectively. Taken together, our findings imply an important role of NF-κB in a negative regulation of TRPC6 expression at the gene transcription level in kidney cells.
Collapse
Affiliation(s)
- Yanxia Wang
- From the Department of Integrative Physiology and Cardiovascular Research Institute and
| | - Min Ding
- From the Department of Integrative Physiology and Cardiovascular Research Institute and
| | - Sarika Chaudhari
- From the Department of Integrative Physiology and Cardiovascular Research Institute and
| | - Yanfeng Ding
- From the Department of Integrative Physiology and Cardiovascular Research Institute and
| | - Joseph Yuan
- From the Department of Integrative Physiology and Cardiovascular Research Institute and
| | - Dorota Stankowska
- the Department of Cell Biology, University of North Texas Health Science Center, Fort Worth, Texas 76107
| | - Shaoqing He
- the Department of Cell Biology, University of North Texas Health Science Center, Fort Worth, Texas 76107
| | - Raghu Krishnamoorthy
- the Department of Cell Biology, University of North Texas Health Science Center, Fort Worth, Texas 76107
| | - Joseph T. Cunningham
- From the Department of Integrative Physiology and Cardiovascular Research Institute and
| | - Rong Ma
- From the Department of Integrative Physiology and Cardiovascular Research Institute and
| |
Collapse
|
46
|
Jardin I, López JJ, Berna-Erro A, Salido GM, Rosado JA. Homer Proteins in Ca2+Entry. IUBMB Life 2013; 65:497-504. [DOI: 10.1002/iub.1162] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2012] [Accepted: 02/07/2013] [Indexed: 11/08/2022]
|
47
|
Contribution and regulation of TRPC channels in store-operated Ca2+ entry. CURRENT TOPICS IN MEMBRANES 2013; 71:149-79. [PMID: 23890115 DOI: 10.1016/b978-0-12-407870-3.00007-x] [Citation(s) in RCA: 152] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/05/2022]
Abstract
Store-operated calcium entry (SOCE) is activated in response to depletion of the endoplasmic reticulum-Ca(2+) stores following stimulation of plasma membrane receptors that couple to PIP2 hydrolysis and IP3 generation. Search for the molecular components of SOCE channels led to the identification of mammalian transient receptor potential canonical (TRPC) family of calcium-permeable channels (TRPC1-TRPC7), which are all activated in response to stimuli that result in PIP2 hydrolysis. While several TRPCs, including TRPC1, TRPC3, and TRPC4, have been implicated in SOCE, the data are most consistent for TRPC1. Extensive studies in cell lines and knockout mouse models have established the contribution of TRPC1 to SOCE. Furthermore, there is a critical functional interaction between TRPC1 and the key components of SOCE, STIM1, and Orai1, which determines the activation of TRPC1. Orai1-mediated Ca(2+) entry is required for recruitment of TRPC1 and its insertion into surface membranes while STIM1 gates the channel. Notably, TRPC1 and Orai1 generate distinct patterns of Ca(2+) signals in cells that are decoded for the regulation of specific cellular functions. Thus, SOCE appears to be a complex process that depends on temporal and spatial coordination of several distinct steps mediated by proteins in different cellular compartments. Emerging data suggest that, in many cell types, the net Ca(2+) entry measured in response to store depletion is the result of the coordinated regulation of different calcium-permeable ion channels. Orai1 and STIM1 are central players in this process, and by mediating recruitment or activation of other Ca(2+) channels, Orai1-CRAC function can elicit rapid changes in global and local [Ca(2+)]i signals in cells. It is most likely that the type of channels and the [Ca(2+)]i signature that are generated by this process reflect the physiological function of the cell that is regulated by Ca(2+).
Collapse
|
48
|
Soboloff J, Rothberg BS, Madesh M, Gill DL. STIM proteins: dynamic calcium signal transducers. Nat Rev Mol Cell Biol 2012; 13:549-65. [PMID: 22914293 DOI: 10.1038/nrm3414] [Citation(s) in RCA: 537] [Impact Index Per Article: 41.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Stromal interaction molecule (STIM) proteins function in cells as dynamic coordinators of cellular calcium (Ca(2+)) signals. Spanning the endoplasmic reticulum (ER) membrane, they sense tiny changes in the levels of Ca(2+) stored within the ER lumen. As ER Ca(2+) is released to generate primary Ca(2+) signals, STIM proteins undergo an intricate activation reaction and rapidly translocate into junctions formed between the ER and the plasma membrane. There, STIM proteins tether and activate the highly Ca(2+)-selective Orai channels to mediate finely controlled Ca(2+) signals and to homeostatically balance cellular Ca(2+). Details are emerging on the remarkable organization within these STIM-induced junctional microdomains and the identification of new regulators and alternative target proteins for STIM.
Collapse
Affiliation(s)
- Jonathan Soboloff
- Department of Biochemistry, Temple University School of Medicine, 3400 North Broad Street, Philadelphia, Pennsylvania 19140, USA
| | | | | | | |
Collapse
|
49
|
Toll-like receptor 4 is involved in bacterial endotoxin-induced endothelial cell injury and SOC-mediated calcium regulation. Cell Biol Int 2012; 36:475-81. [PMID: 22288713 DOI: 10.1042/cbi20110535] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Bacterial endotoxins may lead to vascular endothelial cell injury. Our study explored the role of TLR4 (Toll-like receptor 4) and STIM1 (stromal interaction molecule 1) in bacterial endotoxin-induced calcium overload and inflammatory reactions in HUVECs (human umbilical vein endothelial cells). It showed that under LPS (lipopolysaccharide) stimulation, LBP (LPS-binding protein) mRNA levels peaked at 24 h, TLR4 levels at 12 h and NF-κB (nuclear factor κB) levels at 6 h (all P<0.01). LBP levels increased gradually and peaked at 24 h of LPS treatment. TLR4 protein levels increased significantly at 1 h and peaked at 12 h. NF-κB protein levels markedly increased at 1 h and peaked at 6 h. Knockdown of STIM1 alone, TLR4 alone or both STIM1 and TLR4 together, markedly abolished LPS-induced increase in calcium influx into cells (P<0.05, P<0.01 and P<0.01 respectively). LBP-TLR4 and STIM-NF-κB interactions were detected without LPS treatment, enhanced by LPS stimulation, and markedly reduced by knocking down TLR4 and STIM respectively. Both the NF-κB inhibitor, PDTC (pyrrolidine dithiocarbamate) and TLR4 knockdown could block LPS induction of NF-κB, STIM, TNFα (tumour necrosis factor α) and IL-6 (interleukin 6). The data indicate LPS-LBP may activate TLR4 signalling and downstream transcription factor NF-κB, which further can activate STIM1 and eventually lead to calcium influx and injury of HUVECs. Inhibition of TLR4 effectively reverses LPS induction of inflammatory mediator generation and extracellular calcium influx mediated by STIM1.
Collapse
|
50
|
Cioffi DL, Wu S, Chen H, Alexeyev M, St Croix CM, Pitt BR, Uhlig S, Stevens T. Orai1 determines calcium selectivity of an endogenous TRPC heterotetramer channel. Circ Res 2012; 110:1435-44. [PMID: 22534489 DOI: 10.1161/circresaha.112.269506] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
RATIONALE Canonical transient receptor potential 4 (TRPC4) contributes to the molecular composition of a channel encoding for a calcium selective store-operated current, I(SOC), whereas Orai1 critically comprises a channel encoding for the highly selective calcium release activated calcium current, I(CRAC). However, Orai1 may interact with TRPC proteins and influence their activation and permeation characteristics. Endothelium expresses both TRPC4 and Orai1, and it remains unclear as to whether Orai1 interacts with TRPC4 and contributes to calcium permeation through the TPRC4 channel. OBJECTIVE We tested the hypothesis that Orai1 interacts with TRPC4 and contributes to the channel's selective calcium permeation important for endothelial barrier function. METHODS AND RESULTS A novel method to purify the endogenous TRPC4 channel and probe for functional interactions was developed, using TRPC4 binding to protein 4.1 as bait. Isolated channel complexes were conjugated to anti-TRPC protein antibodies labeled with cy3-cy5 pairs. Förster Resonance Energy Transfer among labeled subunits revealed the endogenous protein alignment. One TRPC1 and at least 2 TRPC4 subunits constituted the endogenous channel (TRPC1/4). Orai1 interacted with TRPC4. Conditional Orai1 knockdown reduced the probability for TRPC1/4 channel activation and converted it from a calcium-selective to a nonselective channel, an effect that was rescued on Orai1 reexpression. Loss of Orai1 improved endothelial cell barrier function. CONCLUSION Orai1 interacts with TRPC4 in the endogenous channel complex, where it controls TRPC1/4 activation and channel permeation characteristics, including calcium selectivity, important for control of endothelial cell barrier function.
Collapse
Affiliation(s)
- Donna L Cioffi
- Department of Biochemistry and Molecular Biology, University of South Alabama College of Medicine, Mobile, AL 36688, USA. dlcioffi@ usouthal.edu
| | | | | | | | | | | | | | | |
Collapse
|