1
|
Ran Q, Zhang J, Zhong J, Lin J, Zhang S, Li G, You B. Organ preservation: current limitations and optimization approaches. Front Med (Lausanne) 2025; 12:1566080. [PMID: 40206471 PMCID: PMC11980443 DOI: 10.3389/fmed.2025.1566080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Accepted: 02/28/2025] [Indexed: 04/11/2025] Open
Abstract
Despite the annual rise in patients with end-stage diseases necessitating organ transplantation, the scarcity of high-quality grafts constrains the further development of transplantation. The primary causes of the graft shortage are the scarcity of standard criteria donors, unsatisfactory organ preservation strategies, and mismatching issues. Organ preservation strategies are intimately related to pre-transplant graft viability and the incidence of adverse clinical outcomes. Static cold storage (SCS) is the current standard practice of organ preservation, characterized by its cost-effectiveness, ease of transport, and excellent clinical outcomes. However, cold-induced injury during static cold preservation, toxicity of organ preservation solution components, and post-transplantation reperfusion injury could further exacerbate graft damage. Long-term ex vivo dynamic machine perfusion (MP) preserves grafts in a near-physiological condition, evaluates graft viability, and cures damage to grafts, hence enhancing the usage and survival rates of marginal organs. With the increased use of extended criteria donors (ECD) and advancements in machine perfusion technology, static cold storage is being gradually replaced by machine perfusion. This review encapsulates the latest developments in cryopreservation, subzero non-freezing storage, static cold storage, and machine perfusion. The emphasis is on the injury mechanisms linked to static cold storage and optimization strategies, which may serve as references for the optimization of machine perfusion techniques.
Collapse
Affiliation(s)
- Qiulin Ran
- Department of Cardiovascular Surgery, Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Jiayi Zhang
- Translational Medicine Center, Beijing Chest Hospital, Capital Medical University, Beijing, China
| | - Jisheng Zhong
- Department of Cardiovascular Surgery, Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Ji Lin
- Department of Cardiovascular Surgery, Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Shuai Zhang
- Department of Cardiovascular Surgery, Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Guang Li
- Department of Cardiovascular Surgery, Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Bin You
- Department of Cardiovascular Surgery, Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
2
|
Li X, Du YX, Yu CL, Niu N. Ion channels in macrophages: Implications for disease progression. Int Immunopharmacol 2025; 144:113628. [PMID: 39566388 DOI: 10.1016/j.intimp.2024.113628] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 11/09/2024] [Accepted: 11/11/2024] [Indexed: 11/22/2024]
Abstract
RATIONALE Macrophages are immune cells found throughout the body and exhibit morphological and functional diversity. Macrophages have been implicated in a wide range of diseases, including autoimmune diseases, acute liver injury, cardiovascular diseases, lung diseases and tumours. Ion channels are transmembrane glycoproteins with important functions in maintaining homeostasis in the intra- and extracellular environment and mediating signal transduction. Many studies have shown that different types of ion channels influence the role of macrophages in the development of various diseases. In recent years, studies on the role of ion channels in macrophages in immune regulation and inflammatory responses have attracted much attention. OBJECTIVE AND FINDINGS In order to gain a deeper understanding of the role of macrophage ion channels, this paper reviews the recent research progress on the role of macrophage ion channels in recent years. The aim is to explore the role of different ion channels in the regulation of macrophage function and their impact on a variety of disease processes. The most studied channels are calcium, sodium and potassium channels, most of which are located in the cell membrane. Among these, TRP channels have a more complex role in M1 and M2 macrophage types. CONCLUSION Ion channels are critical for the functional regulation of macrophages. Targeting ion channels provides new avenues for disease prevention and treatment. This review provides researchers with new ideas and introduces readers to the current state of research on ion channels in macrophages.
Collapse
Affiliation(s)
- Xu Li
- School of Pharmacy, North Sichuan Medical College, Nanchong 637000, China
| | - Yan-Xi Du
- School of Clinical Medicine, North Sichuan Medical College, Nanchong 637000, China
| | - Chun-Lei Yu
- School of Pharmacy, North Sichuan Medical College, Nanchong 637000, China
| | - Na Niu
- School of Pharmacy, North Sichuan Medical College, Nanchong 637000, China.
| |
Collapse
|
3
|
Zheng JH, Zhu YH, Yang J, Ji PX, Zhao RK, Duan ZH, Yao HF, Jia QY, Yin YF, Hu LP, Li Q, Jiang SH, Huo YM, Liu W, Sun YW, Liu DJ. A CLIC1 network coordinates matrix stiffness and the Warburg effect to promote tumor growth in pancreatic cancer. Cell Rep 2024; 43:114633. [PMID: 39154343 DOI: 10.1016/j.celrep.2024.114633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Revised: 06/19/2024] [Accepted: 07/30/2024] [Indexed: 08/20/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) features substantial matrix stiffening and reprogrammed glucose metabolism, particularly the Warburg effect. However, the complex interplay between these traits and their impact on tumor advancement remains inadequately explored. Here, we integrated clinical, cellular, and bioinformatics approaches to explore the connection between matrix stiffness and the Warburg effect in PDAC, identifying CLIC1 as a key mediator. Elevated CLIC1 expression, induced by matrix stiffness through Wnt/β-catenin/TCF4 signaling, signifies poorer prognostic outcomes in PDAC. Functionally, CLIC1 serves as a catalyst for glycolytic metabolism, propelling tumor proliferation. Mechanistically, CLIC1 fortifies HIF1α stability by curbing hydroxylation via reactive oxygen species (ROS). Collectively, PDAC cells elevate CLIC1 levels in a matrix-stiffness-responsive manner, bolstering the Warburg effect to drive tumor growth via ROS/HIF1α signaling. Our insights highlight opportunities for targeted therapies that concurrently address matrix properties and metabolic rewiring, with CLIC1 emerging as a promising intervention point.
Collapse
Affiliation(s)
- Jia-Hao Zheng
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, P.R. China; Department of Biliary-Pancreatic Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, P.R. China
| | - Yu-Heng Zhu
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, P.R. China; Department of Biliary-Pancreatic Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, P.R. China
| | - Jian Yang
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, P.R. China; Department of Biliary-Pancreatic Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, P.R. China
| | - Pei-Xuan Ji
- Shanghai Institute of Digestive Disease, Division of Gastroenterology and Hepatology, NHC Key Laboratory of Digestive Diseases, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200001, P.R. China
| | - Rui-Kang Zhao
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai 200438, P.R. China
| | - Zong-Hao Duan
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, P.R. China; Department of Biliary-Pancreatic Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, P.R. China
| | - Hong-Fei Yao
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, P.R. China; Department of Biliary-Pancreatic Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, P.R. China
| | - Qin-Yuan Jia
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, P.R. China; Department of Biliary-Pancreatic Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, P.R. China
| | - Yi-Fan Yin
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, P.R. China; Department of Biliary-Pancreatic Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, P.R. China
| | - Li-Peng Hu
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, P.R. China
| | - Qing Li
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, P.R. China
| | - Shu-Heng Jiang
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, P.R. China
| | - Yan-Miao Huo
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, P.R. China; Department of Biliary-Pancreatic Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, P.R. China.
| | - Wei Liu
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, P.R. China; Department of Biliary-Pancreatic Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, P.R. China.
| | - Yong-Wei Sun
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, P.R. China; Department of Biliary-Pancreatic Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, P.R. China.
| | - De-Jun Liu
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, P.R. China; Department of Biliary-Pancreatic Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, P.R. China.
| |
Collapse
|
4
|
Tapia M, Levay K, Tsoulfas P, Park KK. Retrograde AAV-mediated gene modulation reveals chloride intracellular channel proteins as potent regulators of retinal ganglion cell death. Exp Neurol 2024; 377:114810. [PMID: 38714284 PMCID: PMC11660818 DOI: 10.1016/j.expneurol.2024.114810] [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: 03/01/2024] [Revised: 04/20/2024] [Accepted: 05/03/2024] [Indexed: 05/09/2024]
Abstract
Most projection neurons, including retinal ganglion cells (RGCs), undergo cell death after axotomy proximal to the cell body. Specific RGC subtypes, such as ON-OFF direction selective RGCs (ooDSGCs) are particularly vulnerable, whereas intrinsically photosensitive RGCs (ipRGCs) exhibit resilience to axonal injury. Through the application of RNA sequencing and fluorescent in situ hybridization, we show that the expression of chloride intracellular channel protein 1 and 4 (Clic1 and Clic4) are highly increased in the ooDSGCs after axonal injury. Toward determining a gene's role in RGCs, we optimized the utility and efficacy of adenovirus associated virus (AAV)-retro expressing short hairpin RNA (shRNA). Injection of AAV2-retro into the superior colliculus results in efficient shRNA expression in RGCs. Incorporating histone H2B gene fused with mGreenLantern results in bright nuclear reporter expression, thereby enhancing single RGC identification and cell quantitation in live retinas. Lastly, we demonstrate that AAV2-retro mediated knockdown of both Clic1 and Clic4 promotes RGC survival after injury. Our findings establish an integrated use of AAV2-retro-shRNA and real-time fundus imaging and reveal CLICs' contribution to RGC death.
Collapse
Affiliation(s)
- Mary Tapia
- Department of Neurological Surgery, The Miami Project to Cure Paralysis, The University of Miami Miller School of Medicine, 1501 NW 10th Avenue, Miami, FL 33136, United States of America
| | - Konstantin Levay
- Department of Neurological Surgery, The Miami Project to Cure Paralysis, The University of Miami Miller School of Medicine, 1501 NW 10th Avenue, Miami, FL 33136, United States of America
| | - Pantelis Tsoulfas
- Department of Neurological Surgery, The Miami Project to Cure Paralysis, The University of Miami Miller School of Medicine, 1501 NW 10th Avenue, Miami, FL 33136, United States of America
| | - Kevin K Park
- Department of Ophthalmology, Department of Neuroscience, Peter O'Donnell Jr. Brain Institute, The University of Texas Southwestern Medical Center, 5901 Forest Park Rd, Dallas, TX 75235, United States of America.
| |
Collapse
|
5
|
Raut S, Singh K, Sanghvi S, Loyo-Celis V, Varghese L, Singh E, Gururaja Rao S, Singh H. Chloride ions in health and disease. Biosci Rep 2024; 44:BSR20240029. [PMID: 38573803 PMCID: PMC11065649 DOI: 10.1042/bsr20240029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 03/26/2024] [Accepted: 04/04/2024] [Indexed: 04/06/2024] Open
Abstract
Chloride is a key anion involved in cellular physiology by regulating its homeostasis and rheostatic processes. Changes in cellular Cl- concentration result in differential regulation of cellular functions such as transcription and translation, post-translation modifications, cell cycle and proliferation, cell volume, and pH levels. In intracellular compartments, Cl- modulates the function of lysosomes, mitochondria, endosomes, phagosomes, the nucleus, and the endoplasmic reticulum. In extracellular fluid (ECF), Cl- is present in blood/plasma and interstitial fluid compartments. A reduction in Cl- levels in ECF can result in cell volume contraction. Cl- is the key physiological anion and is a principal compensatory ion for the movement of the major cations such as Na+, K+, and Ca2+. Over the past 25 years, we have increased our understanding of cellular signaling mediated by Cl-, which has helped in understanding the molecular and metabolic changes observed in pathologies with altered Cl- levels. Here, we review the concentration of Cl- in various organs and cellular compartments, ion channels responsible for its transportation, and recent information on its physiological roles.
Collapse
Affiliation(s)
- Satish K. Raut
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, OH, U.S.A
| | - Kulwinder Singh
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, OH, U.S.A
| | - Shridhar Sanghvi
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, OH, U.S.A
- Department of Molecular Cellular and Developmental Biology, The Ohio State University, Columbus, OH, U.S.A
| | - Veronica Loyo-Celis
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, OH, U.S.A
| | - Liyah Varghese
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, OH, U.S.A
| | - Ekam R. Singh
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, OH, U.S.A
| | | | - Harpreet Singh
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, OH, U.S.A
- Department of Molecular Cellular and Developmental Biology, The Ohio State University, Columbus, OH, U.S.A
| |
Collapse
|
6
|
Loyo-Celis V, Patel D, Sanghvi S, Kaur K, Ponnalagu D, Zheng Y, Bindra S, Bhachu HR, Deschenes I, Gururaja Rao S, Singh H. Biophysical characterization of chloride intracellular channel 6 (CLIC6). J Biol Chem 2023; 299:105349. [PMID: 37838179 PMCID: PMC10641671 DOI: 10.1016/j.jbc.2023.105349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 09/27/2023] [Accepted: 09/30/2023] [Indexed: 10/16/2023] Open
Abstract
Chloride intracellular channels (CLICs) are a family of proteins that exist in soluble and transmembrane forms. The newest discovered member of the family CLIC6 is implicated in breast, ovarian, lung gastric, and pancreatic cancers and is also known to interact with dopamine-(D(2)-like) receptors. The soluble structure of the channel has been resolved, but the exact physiological role of CLIC6, biophysical characterization, and the membrane structure remain unknown. Here, we aimed to characterize the biophysical properties of this channel using a patch-clamp approach. To determine the biophysical properties of CLIC6, we expressed CLIC6 in HEK-293 cells. On ectopic expression, CLIC6 localizes to the plasma membrane of HEK-293 cells. We established the biophysical properties of CLIC6 by using electrophysiological approaches. Using various anions and potassium (K+) solutions, we determined that CLIC6 is more permeable to chloride-(Cl-) as compared to bromide-(Br-), fluoride-(F-), and K+ ions. In the whole-cell configuration, the CLIC6 currents were inhibited after the addition of 10 μM of IAA-94 (CLIC-specific blocker). CLIC6 was also found to be regulated by pH and redox potential. We demonstrate that the histidine residue at 648 (H648) in the C terminus and cysteine residue in the N terminus (C487) are directly involved in the pH-induced conformational change and redox regulation of CLIC6, respectively. Using qRT-PCR, we identified that CLIC6 is most abundant in the lung and brain, and we recorded the CLIC6 current in mouse lung epithelial cells. Overall, we have determined the biophysical properties of CLIC6 and established it as a Cl- channel.
Collapse
Affiliation(s)
- Veronica Loyo-Celis
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Devendra Patel
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Shridhar Sanghvi
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, Ohio, USA; Department of Molecular Cellular and Developmental Biology, The Ohio State University, Columbus, Ohio, USA
| | - Kamalpreet Kaur
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Devasena Ponnalagu
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, Ohio, USA; Department of Pharmacology, The University of Washington, Seattle, Washington, USA
| | - Yang Zheng
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Sahej Bindra
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Harmeet Rireika Bhachu
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Isabelle Deschenes
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, Ohio, USA
| | | | - Harpreet Singh
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, Ohio, USA; Department of Molecular Cellular and Developmental Biology, The Ohio State University, Columbus, Ohio, USA.
| |
Collapse
|
7
|
Zapata RC, Zhang D, Yoon D, Nasamran CA, Chilin-Fuentes DR, Libster A, Chaudry BS, Lopez-Valencia M, Ponnalagu D, Singh H, Petrascheck M, Osborn O. Targeting Clic1 for the treatment of obesity: A novel therapeutic strategy to reduce food intake and body weight. Mol Metab 2023; 76:101794. [PMID: 37604246 PMCID: PMC10480059 DOI: 10.1016/j.molmet.2023.101794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Revised: 08/13/2023] [Accepted: 08/17/2023] [Indexed: 08/23/2023] Open
Abstract
OBJECTIVE Despite great advances in obesity therapeutics in recent years, there is still a need to identify additional therapeutic targets for the treatment of this disease. We previously discovered a signature of genes, including Chloride intracellular channel 1 (Clic1), whose expression was associated with drug-induced weight gain, and in these studies, we assess the effect of Clic1 inhibition on food intake and body weight in mice. METHODS We studied the impact of Clic1 inhibition in mouse models of binge-eating, diet-induced obese mice and genetic models of obesity (Magel2 KO mice). RESULTS Clic1 knockout (KO) mice ate significantly less and had a lower body weight than WT littermates when either fed chow or high fat diet. Furthermore, pharmacological inhibition of Clic1 in diet-induced obese mice resulted in suppression of food intake and promoted highly efficacious weight loss. Clic1 inhibition also reduced food intake in binge-eating models and hyperphagic Magel2 KO mice. We observed that chronic obesity resulted in a significant change in subcellular localization of Clic1 with an increased ratio of Clic1 in the membrane in the obese state. These observations provide a novel therapeutic strategy to block Clic1 translocation as a potential mechanism to reduce food intake and lower body weight. CONCLUSIONS These studies attribute a novel role of Clic1 as a driver of food intake and overconsumption. In summary, we have identified hypothalamic expression of Clic1 plays a key role in food intake, providing a novel therapeutic target to treat overconsumption that is the root cause of modern obesity.
Collapse
Affiliation(s)
- Rizaldy C Zapata
- Division of Endocrinology and Metabolism, School of Medicine, University of California San Diego, La Jolla, CA, 92093, USA
| | - Dinghong Zhang
- Division of Endocrinology and Metabolism, School of Medicine, University of California San Diego, La Jolla, CA, 92093, USA
| | - Dongmin Yoon
- Division of Endocrinology and Metabolism, School of Medicine, University of California San Diego, La Jolla, CA, 92093, USA
| | - Chanond A Nasamran
- Center for Computational Biology & Bioinformatics, Department of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Daisy R Chilin-Fuentes
- Center for Computational Biology & Bioinformatics, Department of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Avraham Libster
- Division of Endocrinology and Metabolism, School of Medicine, University of California San Diego, La Jolla, CA, 92093, USA
| | - Besma S Chaudry
- Division of Endocrinology and Metabolism, School of Medicine, University of California San Diego, La Jolla, CA, 92093, USA
| | - Mariela Lopez-Valencia
- Division of Endocrinology and Metabolism, School of Medicine, University of California San Diego, La Jolla, CA, 92093, USA
| | - Devasena Ponnalagu
- Department of Physiology and Cell Biology, The Ohio State University, Columbus, OH, USA; Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, USA
| | - Harpreet Singh
- Department of Physiology and Cell Biology, The Ohio State University, Columbus, OH, USA; Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, USA
| | - Michael Petrascheck
- Department of Molecular Medicine, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA, 92037, USA; Department of Neuroscience, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA, 92037, USA
| | - Olivia Osborn
- Division of Endocrinology and Metabolism, School of Medicine, University of California San Diego, La Jolla, CA, 92093, USA.
| |
Collapse
|
8
|
Randhawa K, Jahani-Asl A. CLIC1 regulation of cancer stem cells in glioblastoma. CURRENT TOPICS IN MEMBRANES 2023; 92:99-123. [PMID: 38007271 DOI: 10.1016/bs.ctm.2023.09.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/27/2023]
Abstract
Chloride intracellular channel 1 (CLIC1) has emerged as a therapeutic target in various cancers. CLIC1 promotes cell cycle progression and cancer stem cell (CSC) self-renewal. Furthermore, CLIC1 is shown to play diverse roles in proliferation, cell volume regulation, tumour invasion, migration, and angiogenesis. In glioblastoma (GB), CLIC1 facilitates the G1/S phase transition and tightly regulates glioma stem-like cells (GSCs), a rare population of self-renewing CSCs with central roles in tumour resistance to therapy and tumour recurrence. CLIC1 is found as either a monomeric soluble protein or as a non-covalent dimeric protein that can form an ion channel. The ratio of dimeric to monomeric protein is altered in GSCs and depends on the cell redox state. Elucidating the mechanisms underlying the alterations in CLIC1 expression and structural transitions will further our understanding of its role in GSC biology. This review will highlight the role of CLIC1 in GSCs and its significance in facilitating different hallmarks of cancer.
Collapse
Affiliation(s)
- Kamaldeep Randhawa
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, Canada; Brain and Mind Research Institute, University of Ottawa, Ottawa, ON, Canada
| | - Arezu Jahani-Asl
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, Canada; Brain and Mind Research Institute, University of Ottawa, Ottawa, ON, Canada; Regenerative Medicine Program and Cancer Therapeutics Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada.
| |
Collapse
|
9
|
Alghalayini A, Hossain KR, Moghaddasi S, Turkewitz DR, D’Amario C, Wallach M, Valenzuela SM. In Vitro Enzymatic Studies Reveal pH and Temperature Sensitive Properties of the CLIC Proteins. Biomolecules 2023; 13:1394. [PMID: 37759794 PMCID: PMC10526857 DOI: 10.3390/biom13091394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 09/09/2023] [Accepted: 09/10/2023] [Indexed: 09/29/2023] Open
Abstract
Chloride intracellular ion channel (CLIC) proteins exist as both soluble and integral membrane proteins, with CLIC1 capable of shifting between two distinct structural conformations. New evidence has emerged indicating that members of the CLIC family act as moonlighting proteins, referring to the ability of a single protein to carry out multiple functions. In addition to their ion channel activity, CLIC family members possess oxidoreductase enzymatic activity and share significant structural and sequence homology, along with varying overlaps in their tissue distribution and cellular localization. In this study, the 2-hydroxyethyl disulfide (HEDS) assay system was used to characterize kinetic properties, as well as the temperature and pH profiles of three CLIC protein family members (CLIC1, CLIC3, CLIC4). We also assessed the effects of the drugs rapamycin and amphotericin B, on the three CLIC proteins' enzymatic activity in the HEDS assay. Our results demonstrate CLIC1 to be highly heat-sensitive, with optimal enzymatic activity observed at neutral pH7 and at a temperature of 37 °C, while CLIC3 had higher oxidoreductase activity in more acidic pH5 and was found to be relatively heat stable. CLIC4, like CLIC1, was temperature sensitive with optimal enzymatic activity observed at 37 °C; however, it showed optimal activity in more alkaline conditions of pH8. Our current study demonstrates individual differences in the enzymatic activity between the three CLIC proteins, suggesting each CLIC protein is likely regulated in discrete ways, involving changes in the subcellular milieu and microenvironment.
Collapse
Affiliation(s)
- Amani Alghalayini
- School of Life Sciences, University of Technology Sydney, Sydney, NSW 2007, Australia; (A.A.); (K.R.H.); (S.M.); (D.R.T.); (C.D.); (M.W.)
- ARC Research Hub for Integrated Device for End-User Analysis at Low-Levels (IDEAL), Faculty of Science, University of Technology Sydney, Sydney, NSW 2007, Australia
| | - Khondker Rufaka Hossain
- School of Life Sciences, University of Technology Sydney, Sydney, NSW 2007, Australia; (A.A.); (K.R.H.); (S.M.); (D.R.T.); (C.D.); (M.W.)
- ARC Research Hub for Integrated Device for End-User Analysis at Low-Levels (IDEAL), Faculty of Science, University of Technology Sydney, Sydney, NSW 2007, Australia
| | - Saba Moghaddasi
- School of Life Sciences, University of Technology Sydney, Sydney, NSW 2007, Australia; (A.A.); (K.R.H.); (S.M.); (D.R.T.); (C.D.); (M.W.)
| | - Daniel R. Turkewitz
- School of Life Sciences, University of Technology Sydney, Sydney, NSW 2007, Australia; (A.A.); (K.R.H.); (S.M.); (D.R.T.); (C.D.); (M.W.)
| | - Claudia D’Amario
- School of Life Sciences, University of Technology Sydney, Sydney, NSW 2007, Australia; (A.A.); (K.R.H.); (S.M.); (D.R.T.); (C.D.); (M.W.)
| | - Michael Wallach
- School of Life Sciences, University of Technology Sydney, Sydney, NSW 2007, Australia; (A.A.); (K.R.H.); (S.M.); (D.R.T.); (C.D.); (M.W.)
| | - Stella M. Valenzuela
- School of Life Sciences, University of Technology Sydney, Sydney, NSW 2007, Australia; (A.A.); (K.R.H.); (S.M.); (D.R.T.); (C.D.); (M.W.)
- ARC Research Hub for Integrated Device for End-User Analysis at Low-Levels (IDEAL), Faculty of Science, University of Technology Sydney, Sydney, NSW 2007, Australia
| |
Collapse
|
10
|
Huang Q, Lv Q, Tang W, Pan Y, Xing Y, He M, Wu H, Huang J, Huang C, Lan H, Chen J, Xiao G. A comprehensively prognostic and immunological analysis of chloride intracellular channel protein 5 (CLIC5) in pan-cancer and identification in ovarian cancer. J Cancer Res Clin Oncol 2023; 149:10561-10583. [PMID: 37286734 DOI: 10.1007/s00432-023-04927-4] [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: 03/21/2023] [Accepted: 05/23/2023] [Indexed: 06/09/2023]
Abstract
CLIC5 encoded protein associates with actin-based cytoskeletal and is increasingly thought to play significant roles in human cancers. We use TCGA and GEO to explore CLIC5 expression differences, mutation and DNA methylation, TMB, MSI, and immune cell infiltration. We verified the mRNA expression of CLIC5 in human ovarian cancer cells by real-time PCR and detected the expression of CLIC5 as well as immune marker genes in ovarian cancer by immunohistochemistry. The pan-cancer analysis showed that CLIC5 is highly expressed in several malignant tumors. In some cancers, CLIC5 expression in tumor samples is associated with poorer overall survival. For example, patients with ovarian cancer with high expression of CLIC5 have a poor prognosis. CLIC5 mutation frequency increased in all tumor types. The CLIC5 promoter is hypomethylated in most tumors. CLIC5 was associated with tumor immunity and different immune cells of different tumor types, such as CD8 + T cells, tumor-associated fibroblasts, macrophages, etc. CLIC5 was positively correlated with various immune checkpoints, and TMB and MSI were correlated with dysregulation of CLIC5 in tumors. The expression of CLIC5 in ovarian cancer was detected by qPCR and IHC, and the results were consistent with the bioinformatics results. There were a strong positive correlation between CLIC5 expression and M2 macrophage (CD163) infiltration and a negative correlation with CD8 + T-cell infiltration. In conclusions, our first pan-cancer analysis offered a detailed grasp of the cancerogenic functions of CLIC5 in a variety of malignancies. CLIC5 participated in immunomodulation and performed a crucial function in the tumor microenvironment.
Collapse
Affiliation(s)
- Qiaoling Huang
- Translational Medicine Center, Guangzhou Key Laboratory for Research and Development of Nano-Biomedical Technology for Diagnosis and Therapy and Guangdong Provincial Education Department Key Laboratory of Nano-Immunoregulation Tumour Microenvironment, Department of Gynecology, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510260, Guangdong, People's Republic of China
- Central Laboratory, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510260, Guangdong, People's Republic of China
| | - Quankun Lv
- Emergency Department, the Sixth Affiliated Hospital, South China University of Technology, Foshan, 528000, Guangdong, People's Republic of China
| | - Waner Tang
- Translational Medicine Center, Guangzhou Key Laboratory for Research and Development of Nano-Biomedical Technology for Diagnosis and Therapy and Guangdong Provincial Education Department Key Laboratory of Nano-Immunoregulation Tumour Microenvironment, Department of Gynecology, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510260, Guangdong, People's Republic of China
- Central Laboratory, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510260, Guangdong, People's Republic of China
| | - Yuhua Pan
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China
| | - Yue Xing
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, 151 Yanjiang Road, Guangzhou, 510120, Guangdong, People's Republic of China
| | - Min He
- Translational Medicine Center, Guangzhou Key Laboratory for Research and Development of Nano-Biomedical Technology for Diagnosis and Therapy and Guangdong Provincial Education Department Key Laboratory of Nano-Immunoregulation Tumour Microenvironment, Department of Gynecology, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510260, Guangdong, People's Republic of China
| | - Huiyi Wu
- Translational Medicine Center, Guangzhou Key Laboratory for Research and Development of Nano-Biomedical Technology for Diagnosis and Therapy and Guangdong Provincial Education Department Key Laboratory of Nano-Immunoregulation Tumour Microenvironment, Department of Gynecology, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510260, Guangdong, People's Republic of China
| | - Jiamin Huang
- Translational Medicine Center, Guangzhou Key Laboratory for Research and Development of Nano-Biomedical Technology for Diagnosis and Therapy and Guangdong Provincial Education Department Key Laboratory of Nano-Immunoregulation Tumour Microenvironment, Department of Gynecology, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510260, Guangdong, People's Republic of China
| | - Che Huang
- Hubei University of Medicine, Shiyan, 442000, China
| | - Haifeng Lan
- Department of Orthopaedic Surgery, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, Guangdong, People's Republic of China
| | - Jingqi Chen
- Translational Medicine Center, Guangzhou Key Laboratory for Research and Development of Nano-Biomedical Technology for Diagnosis and Therapy and Guangdong Provincial Education Department Key Laboratory of Nano-Immunoregulation Tumour Microenvironment, Department of Gynecology, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510260, Guangdong, People's Republic of China.
- Oncology Department, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510260, Guangdong, People's Republic of China.
| | - Guohong Xiao
- Translational Medicine Center, Guangzhou Key Laboratory for Research and Development of Nano-Biomedical Technology for Diagnosis and Therapy and Guangdong Provincial Education Department Key Laboratory of Nano-Immunoregulation Tumour Microenvironment, Department of Gynecology, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510260, Guangdong, People's Republic of China.
| |
Collapse
|
11
|
Hosler J, Hoang N, Edwards KS. The cyclic lipopeptide micafungin induces rupture of isolated mitochondria by reprograming the mitochondrial inner membrane anion channel. Mitochondrion 2023; 71:50-62. [PMID: 37201620 PMCID: PMC10524837 DOI: 10.1016/j.mito.2023.05.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 04/17/2023] [Accepted: 05/07/2023] [Indexed: 05/20/2023]
Abstract
The antifungal activity of the drug micafungin, a cyclic lipopeptide that interacts with membrane proteins, may involve inhibition of fungal mitochondria. In humans, mitochondria are spared by the inability of micafungin to cross the cytoplasmic membrane. Using isolated mitochondria, we find that micafungin initiates the uptake of salts, causing rapid swelling and rupture of mitochondria with release of cytochrome c. The inner membrane anion channel (IMAC) is altered by micafungin to transfer both cations and anions. We propose that binding of anionic micafungin to IMAC attracts cations into the ion pore for the rapid transfer of ion pairs.
Collapse
Affiliation(s)
- Jonathan Hosler
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, 2500 N. State St., Jackson, MS 39216, United States
| | - Ngoc Hoang
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, 2500 N. State St., Jackson, MS 39216, United States
| | - Kristin Shirey Edwards
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, 2500 N. State St., Jackson, MS 39216, United States.
| |
Collapse
|
12
|
Hossain KR, Turkewitz DR, Holt SA, Le Brun AP, Valenzuela SM. Sterol Structural Features' Impact on the Spontaneous Membrane Insertion of CLIC1 into Artificial Lipid Membranes. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2023; 39:3286-3300. [PMID: 36821411 DOI: 10.1021/acs.langmuir.2c03129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Background: A membrane protein interaction with lipids shows distinct specificity in terms of the sterol structure. The structure of the sterol's polar headgroup, steroidal rings, and aliphatic side chains have all been shown to influence protein membrane interactions, including the initial binding and subsequent oligomerization to form functional channels. Previous studies have provided some insights into the regulatory role that cholesterol plays in the spontaneous membrane insertion of the chloride intracellular ion channel protein, CLIC1. However, the manner in which cholesterol interacts with CLIC1 is yet largely unknown. Method: In this study, the CLIC1 interaction with different lipid:sterol monolayers was studied using the Langmuir trough and neutron reflectometry in order to investigate the structural features of cholesterol essential for the spontaneous membrane insertion of the CLIC1 protein. Molecular docking simulations were also performed to study the binding affinities between CLIC1 and the different sterol molecules. Results: This study, for the first time, highlights the vital role of the free sterol 3β-OH group as an essential structural requirement for the interaction of CLIC1 with cholesterol. Furthermore, the presence of additional hydroxyl groups, methylation of the sterol skeleton, and the structure of the sterol alkyl side chain have also been shown to modulate the magnitude of CLIC1 interaction with sterols and hence their spontaneous membrane insertion. This study also reports the ability of CLIC1 to interact with other naturally existing sterol molecules. General Significance: Like the sterol molecules, CLIC proteins are evolutionarily conserved with almost all vertebrates expressing six CLIC proteins (CLIC1-6), and CLIC-like proteins are also present in invertebrates and have also been reported in plants. This discovery of CLIC1 protein interaction with other natural sterols and the sterol structural requirements for CLIC membrane insertion provide key information to explore the feasibility of exploiting these properties for therapeutic and prophylactic purposes.
Collapse
Affiliation(s)
- Khondker R Hossain
- School of Life Sciences, University of Technology Sydney, Sydney, New South Wales 2007, Australia
- Australian Centre for Neutron Scattering, Australian Nuclear Science and Technology Organisation (ANSTO), Lucas Heights, New South Wales 2234, Australia
| | - Daniel R Turkewitz
- School of Life Sciences, University of Technology Sydney, Sydney, New South Wales 2007, Australia
| | - Stephen A Holt
- Australian Centre for Neutron Scattering, Australian Nuclear Science and Technology Organisation (ANSTO), Lucas Heights, New South Wales 2234, Australia
| | - Anton P Le Brun
- Australian Centre for Neutron Scattering, Australian Nuclear Science and Technology Organisation (ANSTO), Lucas Heights, New South Wales 2234, Australia
| | - Stella M Valenzuela
- School of Life Sciences, University of Technology Sydney, Sydney, New South Wales 2007, Australia
- Institute for Biomedical Materials and Devices (IBMD), University of Technology Sydney, Sydney, New South Wales 2007, Australia
- ARC Research Hub for Integrated Device for End-User Analysis at Low-Levels (IDEAL Hub), Faculty of Science, University of Technology Sydney, , Sydney, New South Wales 2007, Australia
| |
Collapse
|
13
|
Faerch O, Worth R, Achilonu I, Dirr H. Nuclear localisation sequences of chloride intracellular channels 1 and 4 facilitate nuclear import via interactions with import mediator importin-α: An empirical and theoretical perspective. J Mol Recognit 2023; 36:e2996. [PMID: 36175369 PMCID: PMC10078197 DOI: 10.1002/jmr.2996] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 09/15/2022] [Accepted: 09/26/2022] [Indexed: 01/14/2023]
Abstract
Chloride intracellular channel proteins (CLICs) display ubiquitous expression, with each member exhibiting specific subcellular localisation. While all CLICs, except CLIC3, exhibit a highly conserved putative nuclear localisation sequence (NLS), only CLIC1, CLIC3 and CLIC4 exist within the nucleus. The CLIC4 NLS, 199-KVVAKKYR-206, appears crucial for nuclear entry and interacts with mouse nuclear import mediator Impα isoform 1, omitting the IBB domain (mImpα1ΔIBB). The essential nature of the basic residues in the CLIC4 NLS has been established by the fact that mutating out these residues inhibits nuclear import, which in turn is linked to cutaneous squamous cell cancer. Given the conservation of the CLIC NLS, CLIC1 likely follows a similar import pathway to CLIC4. Peptides of the CLIC1 (Pep1; Pep1_S C/S mutant) and CLIC4 (Pep4) NLSs were designed to examine binding to human Impα isoform 1, omitting the IBB domain (hImpα1ΔIBB). Molecular docking indicated that the core CLIC NLS region (KKYR) forms a similar binding pattern to both mImpα1ΔIBB and hImpα1ΔIBB. Fluorescence quenching demonstrated that Pep1_S (Kd ≈ 237 μM) and Pep4 (Kd ≈ 317 μM) bind hImpα1ΔIBB weakly. Isothermal titration calorimetry confirmed the weak binding interaction between Pep4 and hImpα1ΔIBB (Kd ≈ 130 μM) and the presence of a proton-linked effect. This weak interaction may be due to regions distal from the CLIC NLS needed to stabilise and strengthen hImpα1ΔIBB binding. Additionally, this NLS may preferentially bind another hImpα isoform with different flexibility properties.
Collapse
Affiliation(s)
- Olga Faerch
- Protein Structure-Function and Research Unit, School of Molecular and Cell Biology, Faculty of Science, University of the Witwatersrand, Johannesburg, South Africa
| | - Roland Worth
- Protein Structure-Function and Research Unit, School of Molecular and Cell Biology, Faculty of Science, University of the Witwatersrand, Johannesburg, South Africa
| | - Ikechukwu Achilonu
- Protein Structure-Function and Research Unit, School of Molecular and Cell Biology, Faculty of Science, University of the Witwatersrand, Johannesburg, South Africa
| | - Heini Dirr
- Protein Structure-Function and Research Unit, School of Molecular and Cell Biology, Faculty of Science, University of the Witwatersrand, Johannesburg, South Africa
| |
Collapse
|
14
|
Structure-based discovery and in vitro validation of inhibitors of chloride intracellular channel 4 protein. Comput Struct Biotechnol J 2022; 21:688-701. [PMID: 36659928 PMCID: PMC9826898 DOI: 10.1016/j.csbj.2022.12.040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 12/22/2022] [Accepted: 12/23/2022] [Indexed: 12/25/2022] Open
Abstract
The use of computer-aided methods have continued to propel accelerated drug discovery across various disease models, interestingly allowing the specific inhibition of pathogenic targets. Chloride Intracellular Channel Protein 4 (CLIC4) is a novel class of intracellular ion channel highly implicated in tumor and vascular biology. It regulates cell proliferation, apoptosis and angiogenesis; and is involved in multiple pathologic signaling pathways. Absence of specific inhibitors however impedes its advancement to translational research. Here, we integrate structural bioinformatics and experimental research approaches for the discovery and validation of small-molecule inhibitors of CLIC4. High-affinity allosteric binders were identified from a library of 1615 Food and Drug Administration (FDA)-approved drugs via a high-performance computing-powered blind-docking approach, resulting in the selection of amphotericin B and rapamycin. NMR assays confirmed the binding and conformational disruptive effects of both drugs while they also reversed stress-induced membrane translocation of CLIC4 and inhibited endothelial cell migration. Structural and dynamics simulation studies further revealed that the inhibitory mechanisms of these compounds were hinged on the allosteric modulation of the catalytic glutathione (GSH)-like site loop and the extended catalytic β loop which may elicit interference with the catalytic activities of CLIC4. Structure-based insights from this study provide the basis for the selective targeting of CLIC4 to treat the associated pathologies.
Collapse
Key Words
- A9C, 9-Anthracenecarboxylic acid
- AMPhB, Amphotericin B
- Ad, Adenovirus
- Allosteric inhibition
- Bad, BCL2 associated agonist of cell death
- Bcl-2, B-cell lymphoma 2
- Bcl-xL, B-cell lymphoma-extra large
- CDK, Cyclin-dependent kinases
- CLIC, Chloride intracellular channel protein
- Chloride intracellular channel protein 4
- Computational high-throughput screening
- DAPI, 4′,6-diamidino-2-phenylindole
- DIDS, 4,4′-Diisothiocyano-2,2′-stilbenedisulfonic acid
- DMSO, Dimethyl sulfoxide
- DOPE, Discrete optimized protein energy
- GPU, Graphics Processing Unit
- GSH-like catalytic site
- GST, glutathione S-transferases
- GUI, Graphical User Interface
- HEPES, (4-(2-hydroxyethyl)− 1-piperazineethanesulfonic acid;
- HIF, Hypoxia-inducible factor
- HSQC, Heteronuclear single quantum coherence spectroscopy
- HUVEC, Human umbilical vein endothelial cells
- IKKβ, Inhibitor of nuclear kappa-B-kinase subunit beta
- JNK, c-Jun N-terminal kinase
- MKK6, Mitogen-activated protein kinase kinase-6
- MOI, Multiplicity of infection
- NF-κB, Nuclear factor kappa-light-chain-enhancer of activated B cells
- NMR, Nuclear magnetic resonance
- NPT, The constant-temperature, constant-pressure ensemble
- NaCL, Sodium chloride
- Nuclear magnetic resonance
- PAH, Pulmonary arterial hypertension
- RAPA, Rapamycin
- SASA, Solvent accessible surface area
- SEK1, Dual specificity mitogen-activated protein kinase kinase 4
- Smad, Suppressor of Mothers against Decapentaplegic
- Structure-based drug discovery
- TEV, Tobacco etch virus
- TIP3P, Transferable intermolecular potential 3 P
- TROSY, Transverse relaxation optimized spectroscopy
- UCSF, University of California, San Francisco
- VEGF, Vascular endothelial growth factor
- p38, Mitogen activated protein kinases
Collapse
|
15
|
Ozaki S, Mikami K, Kunieda T, Tanaka J. Chloride Intracellular Channel Proteins (CLICs) and Malignant Tumor Progression: A Focus on the Preventive Role of CLIC2 in Invasion and Metastasis. Cancers (Basel) 2022; 14:cancers14194890. [PMID: 36230813 PMCID: PMC9562003 DOI: 10.3390/cancers14194890] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 10/03/2022] [Accepted: 10/03/2022] [Indexed: 11/27/2022] Open
Abstract
Simple Summary Although chloride intracellular channel proteins (CLICs) have been identified as ion channel proteins, their true functions are still elusive. Recent in silico analyses show that CLICs may be prognostic markers in cancer. This review focuses on CLIC2 that plays preventive roles in malignant cell invasion and metastasis. CLIC2 is secreted extracellularly and binds to matrix metalloproteinase 14 (MMP14), while inhibiting its activity. As a result, CLIC2 may contribute to the development/maintenance of junctions between blood vessel endothelial cells and the inhibition of invasion and metastasis of tumor cells. CLIC2 may be a novel therapeutic target for malignancies. Abstract CLICs are the dimorphic protein present in both soluble and membrane fractions. As an integral membrane protein, CLICs potentially possess ion channel activity. However, it is not fully clarified what kinds of roles CLICs play in physiological and pathological conditions. In vertebrates, CLICs are classified into six classes: CLIC1, 2, 3, 4, 5, and 6. Recently, in silico analyses have revealed that the expression level of CLICs may have prognostic significance in cancer. In this review, we focus on CLIC2, which has received less attention than other CLICs, and discuss its role in the metastasis and invasion of malignant tumor cells. CLIC2 is expressed at higher levels in benign tumors than in malignant ones, most likely preventing tumor cell invasion into surrounding tissues. CLIC2 is also expressed in the vascular endothelial cells of normal tissues and maintains their intercellular adhesive junctions, presumably suppressing the hematogenous metastasis of malignant tumor cells. Surprisingly, CLIC2 is localized in secretory granules and secreted into the extracellular milieu. Secreted CLIC2 binds to MMP14 and inhibits its activity, leading to suppressed MMP2 activity. CLIC4, on the other hand, promotes MMP14 activity. These findings challenge the assumption that CLICs are ion channels, implying that they could be potential new targets for the treatment of malignant tumors.
Collapse
Affiliation(s)
- Saya Ozaki
- Department of Neurosurgery, Graduate School of Medicine, Ehime University, Toon 791-0295, Japan
- Department of Neurosurgery, National Cerebral and Cardiovascular Center Hospital, Suita 564-8565, Japan
- Correspondence: (S.O.); (J.T.)
| | - Kanta Mikami
- Department of Molecular and Cellular Physiology, Graduate School of Medicine, Ehime University, Toon 791-0295, Japan
| | - Takeharu Kunieda
- Department of Neurosurgery, Graduate School of Medicine, Ehime University, Toon 791-0295, Japan
| | - Junya Tanaka
- Department of Molecular and Cellular Physiology, Graduate School of Medicine, Ehime University, Toon 791-0295, Japan
- Correspondence: (S.O.); (J.T.)
| |
Collapse
|
16
|
Sanchez VC, Yang HH, Craig-Lucas A, Dubois W, Carofino BL, Lack J, Dwyer JE, Simpson RM, Cataisson C, Lee MP, Luo J, Hunter KW, Yuspa SH. Host CLIC4 expression in the tumor microenvironment is essential for breast cancer metastatic competence. PLoS Genet 2022; 18:e1010271. [PMID: 35727842 PMCID: PMC9249210 DOI: 10.1371/journal.pgen.1010271] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 07/01/2022] [Accepted: 05/22/2022] [Indexed: 11/18/2022] Open
Abstract
The TGF-β-regulated Chloride Intracellular Channel 4 (CLIC4) is an essential participant in the formation of breast cancer stroma. Here, we used data available from the TCGA and METABRIC datasets to show that CLIC4 expression was higher in breast cancers from younger women and those with early-stage metastatic disease. Elevated CLIC4 predicted poor outcome in breast cancer patients and was linked to the TGF-β pathway. However, these associations did not reveal the underlying biological contribution of CLIC4 to breast cancer progression. Constitutive ablation of host Clic4 in two murine metastatic breast cancer models nearly eliminated lung metastases without reducing primary tumor weight, while tumor cells ablated of Clic4 retained metastatic capability in wildtype hosts. Thus, CLIC4 was required for host metastatic competence. Pre- and post-metastatic proteomic analysis identified circulating pro-metastatic soluble factors that differed in tumor-bearing CLIC4-deficient and wildtype hosts. Vascular abnormalities and necrosis increased in primary tumors from CLIC4-deficient hosts. Transcriptional profiles of both primary tumors and pre-metastatic lungs of tumor-bearing CLIC4-deficient hosts were consistent with a microenvironment where inflammatory pathways were elevated. Altogether, CLIC4 expression in human breast cancers may serve as a prognostic biomarker; therapeutic targeting of CLIC4 could reduce primary tumor viability and host metastatic competence.
Collapse
Affiliation(s)
- Vanesa C. Sanchez
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Center, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Howard H. Yang
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Center, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Alayna Craig-Lucas
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Center, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Wendy Dubois
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Center, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Brandi L. Carofino
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Center, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Justin Lack
- NIAID Collaborative Bioinformatics Resource (NCBR), National Institute of Allergy and Infectious Disease, National Institutes of Health, Bethesda, Maryland, United States of America
- Advanced Biomedical Computational Science, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Inc., Frederick, Maryland, United States of America
| | - Jennifer E. Dwyer
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Center, National Institutes of Health, Bethesda, Maryland, United States of America
| | - R. Mark Simpson
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Center, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Christophe Cataisson
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Center, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Max P. Lee
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Center, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Ji Luo
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Center, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Kent W. Hunter
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Center, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Stuart H. Yuspa
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Center, National Institutes of Health, Bethesda, Maryland, United States of America
| |
Collapse
|
17
|
Jaramillo Ponce JR, Kapps D, Paulus C, Chicher J, Frugier M. Discovery of two distinct aminoacyl-tRNA synthetase complexes anchored to the Plasmodium surface tRNA import protein. J Biol Chem 2022; 298:101987. [PMID: 35487244 PMCID: PMC9136112 DOI: 10.1016/j.jbc.2022.101987] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 04/19/2022] [Accepted: 04/20/2022] [Indexed: 11/21/2022] Open
Abstract
Aminoacyl-tRNA synthetases (aaRSs) attach amino acids to their cognate transfer RNAs. In eukaryotes, a subset of cytosolic aaRSs is organized into a multisynthetase complex (MSC), along with specialized scaffolding proteins referred to as aaRS-interacting multifunctional proteins (AIMPs). In Plasmodium, the causative agent of malaria, the tRNA import protein (tRip), is a membrane protein that participates in tRNA trafficking; we show that tRip also functions as an AIMP. We identified three aaRSs, the glutamyl-tRNA synthetase (ERS), glutaminyl-tRNA synthetase (QRS), and methionyl-tRNA synthetase (MRS), which were specifically coimmunoprecipitated with tRip in Plasmodium berghei blood stage parasites. All four proteins contain an N-terminal glutathione-S-transferase (GST)-like domain that was demonstrated to be involved in MSC assembly. In contrast to previous studies, further dissection of GST-like interactions identified two exclusive heterotrimeric complexes: the Q-complex (tRip-ERS-QRS) and the M-complex (tRip-ERS-MRS). Gel filtration and light scattering suggest a 2:2:2 stoichiometry for both complexes but with distinct biophysical properties and mutational analysis further revealed that the GST-like domains of QRS and MRS use different strategies to bind ERS. Taken together, our results demonstrate that neither the singular homodimerization of tRip nor its localization in the parasite plasma membrane prevents the formation of MSCs in Plasmodium. Besides, the extracellular localization of the tRNA-binding module of tRip is compensated by the presence of additional tRNA-binding modules fused to MRS and QRS, providing each MSC with two spatially distinct functions: aminoacylation of intraparasitic tRNAs and binding of extracellular tRNAs. This unique host-pathogen interaction is discussed.
Collapse
Affiliation(s)
- José R Jaramillo Ponce
- Université de Strasbourg, CNRS, Architecture et Réactivité de l'ARN, UPR 9002, Strasbourg, France
| | - Delphine Kapps
- Université de Strasbourg, CNRS, Architecture et Réactivité de l'ARN, UPR 9002, Strasbourg, France
| | - Caroline Paulus
- Université de Strasbourg, CNRS, Architecture et Réactivité de l'ARN, UPR 9002, Strasbourg, France
| | - Johana Chicher
- Strasbourg-Esplanade Proteomics Facility, Université de Strasbourg, Strasbourg, France
| | - Magali Frugier
- Université de Strasbourg, CNRS, Architecture et Réactivité de l'ARN, UPR 9002, Strasbourg, France.
| |
Collapse
|
18
|
Abstract
Mitochondria have been recognized as key organelles in cardiac physiology and are potential targets for clinical interventions to improve cardiac function. Mitochondrial dysfunction has been accepted as a major contributor to the development of heart failure. The main function of mitochondria is to meet the high energy demands of the heart by oxidative metabolism. Ionic homeostasis in mitochondria directly regulates oxidative metabolism, and any disruption in ionic homeostasis causes mitochondrial dysfunction and eventually contractile failure. The mitochondrial ionic homeostasis is closely coupled with inner mitochondrial membrane potential. To regulate and maintain ionic homeostasis, mitochondrial membranes are equipped with ion transporting proteins. Ion transport mechanisms involving several different ion channels and transporters are highly efficient and dynamic, thus helping to maintain the ionic homeostasis of ions as well as their salts present in the mitochondrial matrix. In recent years, several novel proteins have been identified on the mitochondrial membranes and these proteins are actively being pursued in research for roles in the organ as well as organelle physiology. In this article, the role of mitochondrial ion channels in cardiac function is reviewed. In recent times, the major focus of the mitochondrial ion channel field is to establish molecular identities as well as assigning specific functions to them. Given the diversity of mitochondrial ion channels and their unique roles in cardiac function, they present novel and viable therapeutic targets for cardiac diseases.
Collapse
Affiliation(s)
- Harpreet Singh
- Department of Physiology and Cell Biology, The Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University College of Medicine, Columbus, Ohio
| |
Collapse
|
19
|
Lukasiak A, Zajac M. The Distribution and Role of the CFTR Protein in the Intracellular Compartments. MEMBRANES 2021; 11:membranes11110804. [PMID: 34832033 PMCID: PMC8618639 DOI: 10.3390/membranes11110804] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 10/18/2021] [Accepted: 10/21/2021] [Indexed: 12/11/2022]
Abstract
Cystic fibrosis is a hereditary disease that mainly affects secretory organs in humans. It is caused by mutations in the gene encoding CFTR with the most common phenylalanine deletion at position 508. CFTR is an anion channel mainly conducting Cl− across the apical membranes of many different epithelial cells, the impairment of which causes dysregulation of epithelial fluid secretion and thickening of the mucus. This, in turn, leads to the dysfunction of organs such as the lungs, pancreas, kidney and liver. The CFTR protein is mainly localized in the plasma membrane; however, there is a growing body of evidence that it is also present in the intracellular organelles such as the endosomes, lysosomes, phagosomes and mitochondria. Dysfunction of the CFTR protein affects not only the ion transport across the epithelial tissues, but also has an impact on the proper functioning of the intracellular compartments. The review aims to provide a summary of the present state of knowledge regarding CFTR localization and function in intracellular compartments, the physiological role of this localization and the consequences of protein dysfunction at cellular, epithelial and organ levels. An in-depth understanding of intracellular processes involved in CFTR impairment may reveal novel opportunities in pharmacological agents of cystic fibrosis.
Collapse
|
20
|
Laurent Q, Martinent R, Lim B, Pham AT, Kato T, López-Andarias J, Sakai N, Matile S. Thiol-Mediated Uptake. JACS AU 2021; 1:710-728. [PMID: 34467328 PMCID: PMC8395643 DOI: 10.1021/jacsau.1c00128] [Citation(s) in RCA: 83] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Indexed: 05/19/2023]
Abstract
This Perspective focuses on thiol-mediated uptake, that is, the entry of substrates into cells enabled by oligochalcogenides or mimics, often disulfides, and inhibited by thiol-reactive agents. A short chronology from the initial observations in 1990 until today is followed by a summary of cell-penetrating poly(disulfide)s (CPDs) and cyclic oligochalcogenides (COCs) as privileged scaffolds in thiol-mediated uptake and inhibitors of thiol-mediated uptake as potential antivirals. In the spirit of a Perspective, the main part brings together topics that possibly could help to explain how thiol-mediated uptake really works. Extreme sulfur chemistry mostly related to COCs and their mimics, cyclic disulfides, thiosulfinates/-onates, diselenolanes, benzopolysulfanes, but also arsenics and Michael acceptors, is viewed in the context of acidity, ring tension, exchange cascades, adaptive networks, exchange affinity columns, molecular walkers, ring-opening polymerizations, and templated polymerizations. Micellar pores (or lipid ion channels) are considered, from cell-penetrating peptides and natural antibiotics to voltage sensors, and a concise gallery of membrane proteins, as possible targets of thiol-mediated uptake, is provided, including CLIC1, a thiol-reactive chloride channel; TMEM16F, a Ca-activated scramblase; EGFR, the epithelial growth factor receptor; and protein-disulfide isomerase, known from HIV entry or the transferrin receptor, a top hit in proteomics and recently identified in the cellular entry of SARS-CoV-2.
Collapse
Affiliation(s)
- Quentin Laurent
- Department of Organic Chemistry, University of Geneva, 1211 Geneva, Switzerland
| | - Rémi Martinent
- Department of Organic Chemistry, University of Geneva, 1211 Geneva, Switzerland
| | - Bumhee Lim
- Department of Organic Chemistry, University of Geneva, 1211 Geneva, Switzerland
| | - Anh-Tuan Pham
- Department of Organic Chemistry, University of Geneva, 1211 Geneva, Switzerland
| | - Takehiro Kato
- Department of Organic Chemistry, University of Geneva, 1211 Geneva, Switzerland
| | | | - Naomi Sakai
- Department of Organic Chemistry, University of Geneva, 1211 Geneva, Switzerland
| | - Stefan Matile
- Department of Organic Chemistry, University of Geneva, 1211 Geneva, Switzerland
| |
Collapse
|
21
|
Mitochondrial osmoregulation in evolution, cation transport and metabolism. BIOCHIMICA ET BIOPHYSICA ACTA-BIOENERGETICS 2021; 1862:148368. [PMID: 33422486 DOI: 10.1016/j.bbabio.2021.148368] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 12/22/2020] [Accepted: 01/04/2021] [Indexed: 11/24/2022]
Abstract
This review provides a retrospective on the role of osmotic regulation in the process of eukaryogenesis. Specifically, it focuses on the adjustments which must have been made by the original colonizing α-proteobacteria that led to the evolution of modern mitochondria. We focus on the cations that are fundamentally involved in volume determination and cellular metabolism and define the transporter landscape in relation to these ions in mitochondria as we know today. We provide analysis on how the cations interplay and together maintain osmotic balance that allows for effective ATP synthesis in the organelle.
Collapse
|
22
|
Novel biomarkers of bronchopulmonary dysplasia and bronchopulmonary dysplasia-associated pulmonary hypertension. J Perinatol 2020; 40:1634-1643. [PMID: 32811975 PMCID: PMC7664991 DOI: 10.1038/s41372-020-00788-8] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2019] [Revised: 07/23/2020] [Accepted: 08/07/2020] [Indexed: 11/29/2022]
Abstract
OBJECTIVE To quantify and compare levels of potential biomarkers in neonates with (i) Bronchopulmonary dysplasia (BPD); (ii) BPD-associated pulmonary hypertension (BPD-PH); (iii) PH without BPD; and (iv) neonates without lung disease at ~36 weeks postmenstrual age. STUDY DESIGN Multiple potential biomarkers were measured in plasma samples of 90 patients using a multi-spot enzyme-linked immunosorbent assay. Statistical tests done included one-way ANOVA to compare levels of biomarkers between different groups. RESULTS Higher levels of ICAM-1 were present in infants with BPD and correlated with its severity. Infants with BPD have significantly higher levels of ANG-2 and lower levels of ANG-1. Infants with PH have higher levels of: IL-6, IL-8, IL-10, and TNF-α. Infants with BPD-PH have significantly lower levels of MCP-1 and higher levels of IL-1β than infants with PH without BPD. CONCLUSION ICAM-1 may be used as a specific biomarker for diagnosis of BPD and its severity.
Collapse
|
23
|
Zheng S, Jiang J, Lee A, Barboiu M. A Voltage‐Responsive Synthetic Cl−‐Channel Regulated by pH. Angew Chem Int Ed Engl 2020. [DOI: 10.1002/ange.202008393] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Shao‐Ping Zheng
- Lehn Institute of Functional Materials School of Chemistry Sun Yat-Sen University Guangzhou 510275 China
- Institut Europeen des Membranes Adaptive Supramolecular Nanosystems Group University of Montpellier ENSCM-CNRS Place E. Bataillon CC047 34095 Montpellier France
| | - Ji‐Jun Jiang
- Lehn Institute of Functional Materials School of Chemistry Sun Yat-Sen University Guangzhou 510275 China
| | - Arie Lee
- Institut Europeen des Membranes Adaptive Supramolecular Nanosystems Group University of Montpellier ENSCM-CNRS Place E. Bataillon CC047 34095 Montpellier France
| | - Mihail Barboiu
- Lehn Institute of Functional Materials School of Chemistry Sun Yat-Sen University Guangzhou 510275 China
- Institut Europeen des Membranes Adaptive Supramolecular Nanosystems Group University of Montpellier ENSCM-CNRS Place E. Bataillon CC047 34095 Montpellier France
| |
Collapse
|
24
|
Zheng S, Jiang J, Lee A, Barboiu M. A Voltage‐Responsive Synthetic Cl−‐Channel Regulated by pH. Angew Chem Int Ed Engl 2020; 59:18920-18926. [DOI: 10.1002/anie.202008393] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2020] [Revised: 08/10/2020] [Indexed: 12/11/2022]
Affiliation(s)
- Shao‐Ping Zheng
- Lehn Institute of Functional Materials School of Chemistry Sun Yat-Sen University Guangzhou 510275 China
- Institut Europeen des Membranes Adaptive Supramolecular Nanosystems Group University of Montpellier ENSCM-CNRS Place E. Bataillon CC047 34095 Montpellier France
| | - Ji‐Jun Jiang
- Lehn Institute of Functional Materials School of Chemistry Sun Yat-Sen University Guangzhou 510275 China
| | - Arie Lee
- Institut Europeen des Membranes Adaptive Supramolecular Nanosystems Group University of Montpellier ENSCM-CNRS Place E. Bataillon CC047 34095 Montpellier France
| | - Mihail Barboiu
- Lehn Institute of Functional Materials School of Chemistry Sun Yat-Sen University Guangzhou 510275 China
- Institut Europeen des Membranes Adaptive Supramolecular Nanosystems Group University of Montpellier ENSCM-CNRS Place E. Bataillon CC047 34095 Montpellier France
| |
Collapse
|
25
|
Ling CK, Santos LL, Zhou W, Dimitriadis E. Chloride intracellular channel 4 is dysregulated in endometrium of women with infertility and alters receptivity. Biochem Biophys Res Commun 2020; 531:490-496. [PMID: 32807494 DOI: 10.1016/j.bbrc.2020.07.046] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Accepted: 07/10/2020] [Indexed: 02/07/2023]
Abstract
The endometrium remodels in each menstrual cycle to become receptive in preparation for embryo implantation which occurs in the mid-secretory phase of the cycle. Failure of blastocyst adhesion and implantation cause infertility. We compared chloride intracellular channel 4 (CLIC4) expression in human endometrium from women with normal fertility and primary unexplained infertility in the mid-secretory/receptive phase of the menstrual cycle. CLIC4 localised to both the epithelial and stromal regions of the endometrium of fertile tissues across the cycle. CLIC4 expression was significantly reduced in the luminal and glandular epithelium and remained unchanged in the stromal region of mid-secretory infertile endometrium compared to fertile endometrium. siRNA knockdown of CLIC4 significantly compromised adhesive capacity of Ishikawa cells (endometrial epithelial cell line). This reduced adhesion and CLIC4 expression was associated with elevated SGK1, p53, SIRT1, BCL2 and MCL1 gene expression in the Ishikawa cells. CLIC4 expression was increased in primary human endometrial stromal cells during decidualization, however, siRNA knockdown of CLIC4 did not affect decidualization. Our data provide evidence that CLIC4 may regulate receptivity and facilitate blastocyst attachment initiating implantation. Reduced CLIC4 levels may be causative of implantation failure in women.
Collapse
Affiliation(s)
- Cheuk Kwan Ling
- Department of Obstetrics and Gynaecology, University of Melbourne, Parkville, Victoria, 3010, Australia; Gynaecology Research Centre, Royal Women's Hospital, Parkville, Victoria, 3052, Australia
| | - Leilani L Santos
- Department of Obstetrics and Gynaecology, University of Melbourne, Parkville, Victoria, 3010, Australia; Gynaecology Research Centre, Royal Women's Hospital, Parkville, Victoria, 3052, Australia
| | - Wei Zhou
- Department of Obstetrics and Gynaecology, University of Melbourne, Parkville, Victoria, 3010, Australia; Gynaecology Research Centre, Royal Women's Hospital, Parkville, Victoria, 3052, Australia
| | - Evdokia Dimitriadis
- Department of Obstetrics and Gynaecology, University of Melbourne, Parkville, Victoria, 3010, Australia; Gynaecology Research Centre, Royal Women's Hospital, Parkville, Victoria, 3052, Australia.
| |
Collapse
|
26
|
Measurement of Oxidative Stress Markers In Vitro Using Commercially Available Kits. ACTA ACUST UNITED AC 2020. [DOI: 10.1007/978-3-030-47318-1_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/24/2023]
|
27
|
Nguyen LTS, Robinson DN. The Unusual Suspects in Cytokinesis: Fitting the Pieces Together. Front Cell Dev Biol 2020; 8:441. [PMID: 32626704 PMCID: PMC7314909 DOI: 10.3389/fcell.2020.00441] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Accepted: 05/11/2020] [Indexed: 01/24/2023] Open
Abstract
Cytokinesis is the step of the cell cycle in which the cell must faithfully separate the chromosomes and cytoplasm, yielding two daughter cells. The assembly and contraction of the contractile network is spatially and temporally coupled with the formation of the mitotic spindle to ensure the successful completion of cytokinesis. While decades of studies have elucidated the components of this machinery, the so-called usual suspects, and their functions, many lines of evidence are pointing to other unexpected proteins and sub-cellular systems as also being involved in cytokinesis. These we term the unusual suspects. In this review, we introduce recent discoveries on some of these new unusual suspects and begin to consider how these subcellular systems snap together to help complete the puzzle of cytokinesis.
Collapse
Affiliation(s)
- Ly T. S. Nguyen
- Department of Cell Biology, Johns Hopkins School of Medicine, Baltimore, MD, United States
| | - Douglas N. Robinson
- Department of Cell Biology, Johns Hopkins School of Medicine, Baltimore, MD, United States
- Department of Pharmacology and Molecular Sciences, Johns Hopkins School of Medicine, Baltimore, MD, United States
- Department of Medicine, Johns Hopkins School of Medicine, Baltimore, MD, United States
- Department of Oncology, Johns Hopkins School of Medicine, Baltimore, MD, United States
- Chemical and Biomolecular Engineering, Johns Hopkins University Whiting School of Engineering, Baltimore, MD, United States
| |
Collapse
|
28
|
Ponnalagu D, Singh H. Insights Into the Role of Mitochondrial Ion Channels in Inflammatory Response. Front Physiol 2020; 11:258. [PMID: 32327997 PMCID: PMC7160495 DOI: 10.3389/fphys.2020.00258] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Accepted: 03/05/2020] [Indexed: 12/14/2022] Open
Abstract
Mitochondria are the source of many pro-inflammatory signals that cause the activation of the immune system and generate inflammatory responses. They are also potential targets of pro-inflammatory mediators, thus triggering a severe inflammatory response cycle. As mitochondria are a central hub for immune system activation, their dysfunction leads to many inflammatory disorders. Thus, strategies aiming at regulating mitochondrial dysfunction can be utilized as a therapeutic tool to cure inflammatory disorders. Two key factors that determine the structural and functional integrity of mitochondria are mitochondrial ion channels and transporters. They are not only important for maintaining the ionic homeostasis of the cell, but also play a role in regulating reactive oxygen species generation, ATP production, calcium homeostasis and apoptosis, which are common pro-inflammatory signals. The significance of the mitochondrial ion channels in inflammatory response is still not clearly understood and will need further investigation. In this article, we review the different mechanisms by which mitochondria can generate the inflammatory response as well as highlight how mitochondrial ion channels modulate these mechanisms and impact the inflammatory processes.
Collapse
Affiliation(s)
- Devasena Ponnalagu
- Department of Physiology and Cell Biology, Davis Heart and Lung Research Institute, The Ohio State University, Wexner Medical Center, Columbus, OH, United States
| | - Harpreet Singh
- Department of Physiology and Cell Biology, Davis Heart and Lung Research Institute, The Ohio State University, Wexner Medical Center, Columbus, OH, United States
| |
Collapse
|
29
|
Gururaja Rao S, Patel NJ, Singh H. Intracellular Chloride Channels: Novel Biomarkers in Diseases. Front Physiol 2020; 11:96. [PMID: 32116799 PMCID: PMC7034325 DOI: 10.3389/fphys.2020.00096] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Accepted: 01/27/2020] [Indexed: 12/27/2022] Open
Abstract
Ion channels are integral membrane proteins present on the plasma membrane as well as intracellular membranes. In the human genome, there are more than 400 known genes encoding ion channel proteins. Ion channels are known to regulate several cellular, organellar, and physiological processes. Any mutation or disruption in their function can result in pathological disorders, both common or rare. Ion channels present on the plasma membrane are widely acknowledged for their role in various biological processes, but in recent years, several studies have pointed out the importance of ion channels located in intracellular organelles. However, ion channels located in intracellular organelles are not well-understood in the context of physiological conditions, such as the generation of cellular excitability and ionic homeostasis. Due to the lack of information regarding their molecular identity and technical limitations of studying them, intracellular organelle ion channels have thus far been overlooked as potential therapeutic targets. In this review, we focus on a novel class of intracellular organelle ion channels, Chloride Intracellular Ion Channels (CLICs), mainly documented for their role in cardiovascular, neurophysiology, and tumor biology. CLICs have a single transmembrane domain, and in cells, they exist in cytosolic as well as membranous forms. They are predominantly present in intracellular organelles and have recently been shown to be localized to cardiomyocyte mitochondria as well as exosomes. In fact, a member of this family, CLIC5, is the first mitochondrial chloride channel to be identified on the molecular level in the inner mitochondrial membrane, while another member, CLIC4, is located predominantly in the outer mitochondrial membrane. In this review, we discuss this unique class of intracellular chloride channels, their role in pathologies, such as cardiovascular, cancer, and neurodegenerative diseases, and the recent developments concerning their usage as theraputic targets.
Collapse
Affiliation(s)
- Shubha Gururaja Rao
- Department of Physiology and Cell Biology, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| | - Neel J Patel
- Department of Cardiology, Hospital of the University of Pennsylvania, Philadelphia, PA, United States
| | - Harpreet Singh
- Department of Physiology and Cell Biology, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| |
Collapse
|
30
|
The influence of monovalent and divalent metal cations on the stability of the DNA-protein interaction in the nucleosome core particle. ADVANCES IN QUANTUM CHEMISTRY 2020. [DOI: 10.1016/bs.aiq.2020.05.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
31
|
Uretmen Kagiali ZC, Saner N, Akdag M, Sanal E, Degirmenci BS, Mollaoglu G, Ozlu N. CLIC4 and CLIC1 bridge plasma membrane and cortical actin network for a successful cytokinesis. Life Sci Alliance 2019; 3:3/2/e201900558. [PMID: 31879279 PMCID: PMC6933522 DOI: 10.26508/lsa.201900558] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Revised: 12/20/2019] [Accepted: 12/20/2019] [Indexed: 11/24/2022] Open
Abstract
CLIC members are required for the progression of cytokinesis by coupling the plasma membrane and cortical actin network at the cleavage furrow and polar cortex. CLIC4 and CLIC1 are members of the well-conserved chloride intracellular channel proteins (CLICs) structurally related to glutathione-S-transferases. Here, we report new roles of CLICs in cytokinesis. At the onset of cytokinesis, CLIC4 accumulates at the cleavage furrow and later localizes to the midbody in a RhoA-dependent manner. The cell cycle–dependent localization of CLIC4 is abolished when its glutathione S-transferase activity–related residues (C35A and F37D) are mutated. Ezrin, anillin, and ALIX are identified as interaction partners of CLIC4 at the cleavage furrow and midbody. Strikingly, CLIC4 facilitates the activation of ezrin at the cleavage furrow and reciprocally inhibition of ezrin activation diminishes the translocation of CLIC4 to the cleavage furrow. Furthermore, knockouts of CLIC4and CLIC1 cause abnormal blebbing at the polar cortex and regression of the cleavage furrow at late cytokinesis leading to multinucleated cells. We conclude that CLIC4 and CLIC1 function together with ezrin where they bridge plasma membrane and actin cytoskeleton at the polar cortex and cleavage furrow to promote cortical stability and successful completion of cytokinesis in mammalian cells.
Collapse
Affiliation(s)
| | - Nazan Saner
- Department of Molecular Biology and Genetics, Koç University, Istanbul, Turkey
| | - Mehmet Akdag
- Department of Molecular Biology and Genetics, Koç University, Istanbul, Turkey
| | - Erdem Sanal
- Department of Molecular Biology and Genetics, Koç University, Istanbul, Turkey
| | | | - Gurkan Mollaoglu
- Department of Molecular Biology and Genetics, Koç University, Istanbul, Turkey
| | - Nurhan Ozlu
- Department of Molecular Biology and Genetics, Koç University, Istanbul, Turkey .,Koç University Research Center for Translational Medicine (KUTTAM), Istanbul, Turkey
| |
Collapse
|
32
|
Ponnalagu D, Hussain AT, Thanawala R, Meka J, Bednarczyk P, Feng Y, Szewczyk A, GururajaRao S, Bopassa JC, Khan M, Singh H. Chloride channel blocker IAA-94 increases myocardial infarction by reducing calcium retention capacity of the cardiac mitochondria. Life Sci 2019; 235:116841. [PMID: 31494173 PMCID: PMC7664129 DOI: 10.1016/j.lfs.2019.116841] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Revised: 08/28/2019] [Accepted: 09/04/2019] [Indexed: 01/14/2023]
Abstract
Indanyloxyacetic acid-94 (IAA-94), an intracellular chloride channel blocker, is shown to ablate cardioprotection rendered by ischemic preconditioning (IPC), N (6)-2-(4-aminophenyl) ethyladenosine or the PKC activator phorbol 12-myristate 13-acetate and cyclosporin A (CsA) in both ex-vivo and in-vivo ischemia-reperfusion (IR) injury. Thus signifying the role of the IAA-94 sensitive chloride channels in mediating cardio-protection upon IR injury. Although IAA-94 sensitive chloride currents are recorded in cardiac mitoplast, there is still a lack of understanding of the mechanism by which IAA-94 increases myocardial infarction (MI) by IR injury. Mitochondria are the key arbitrators of cell life and death pathways. Both oxidative stress and calcium overload in the mitochondria, elicit pathways resulting in the opening of mitochondrial permeability transition pore (mPTP) leading to cell death. Therefore, in this study we explored the role of IAA-94 in MI and in maintaining calcium retention capacity (CRC) of cardiac mitochondria after IR. IAA-94 inhibited the CRC of the isolated cardiac mitochondria in a concentration-dependent manner as measured spectrofluorimetrically using calcium green-5 N. Interestingly, IAA-94 did not change the mitochondrial membrane potential. Further, CsA a blocker of mPTP opening could not override the effect of IAA-94. We also showed for the first time that IAA-94 perfusion after ischemic event augments MI by reducing the CRC of mitochondria. To conclude, our results demonstrate that the mechanism of IAA-94 mediated cardio-deleterious effects is via modulating the mitochondria CRC, thereby playing a role in mPTP opening. These findings highlight new pharmacological targets, which can mediate cardioprotection from IR injury.
Collapse
Affiliation(s)
- Devasena Ponnalagu
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA 19102, United States of America; Department of Physiology and Cell Biology, The Ohio State University, Columbus, OH 43210, United States of America.
| | - Ahmed Tafsirul Hussain
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA 19102, United States of America
| | - Rushi Thanawala
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA 19102, United States of America
| | - Jahnavi Meka
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA 19102, United States of America
| | - Piotr Bednarczyk
- Department of Biophysics, Warsaw University of Life Sciences - SGGW, Poland
| | - Yansheng Feng
- Department of Cellular and Integrative Physiology, University of Texas Health Science Center, San Antonio, TX 78229, United States of America
| | - Adam Szewczyk
- Department of Biochemistry, Nencki Institute of Experimental Biology, Poland
| | - Shubha GururajaRao
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA 19102, United States of America; Department of Physiology and Cell Biology, The Ohio State University, Columbus, OH 43210, United States of America
| | - Jean C Bopassa
- Department of Cellular and Integrative Physiology, University of Texas Health Science Center, San Antonio, TX 78229, United States of America
| | - Mahmood Khan
- Department of Physiology and Cell Biology, The Ohio State University, Columbus, OH 43210, United States of America; Department of Emergency Medicine, The Ohio State University, Columbus, OH 43210, United States of America
| | - Harpreet Singh
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA 19102, United States of America; Department of Physiology and Cell Biology, The Ohio State University, Columbus, OH 43210, United States of America.
| |
Collapse
|
33
|
Friedrich SR, Lovell PV, Kaser TM, Mello CV. Exploring the molecular basis of neuronal excitability in a vocal learner. BMC Genomics 2019; 20:629. [PMID: 31375088 PMCID: PMC6679542 DOI: 10.1186/s12864-019-5871-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Accepted: 05/31/2019] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND Vocal learning, the ability to learn to produce vocalizations through imitation, relies on specialized brain circuitry known in songbirds as the song system. While the connectivity and various physiological properties of this system have been characterized, the molecular genetic basis of neuronal excitability in song nuclei remains understudied. We have focused our efforts on examining voltage-gated ion channels to gain insight into electrophysiological and functional features of vocal nuclei. A previous investigation of potassium channel genes in zebra finches (Taeniopygia guttata) revealed evolutionary modifications unique to songbirds, as well as transcriptional specializations in the song system [Lovell PV, Carleton JB, Mello CV. BMC Genomics 14:470 2013]. Here, we expand this approach to sodium, calcium, and chloride channels along with their modulatory subunits using comparative genomics and gene expression analysis encompassing microarrays and in situ hybridization. RESULTS We found 23 sodium, 38 calcium, and 33 chloride channel genes (HGNC-based classification) in the zebra finch genome, several of which were previously unannotated. We determined 15 genes are missing relative to mammals, including several genes (CLCAs, BEST2) linked to olfactory transduction. The majority of sodium and calcium but few chloride channels showed differential expression in the song system, among them SCN8A and CACNA1E in the direct motor pathway, and CACNG4 and RYR2 in the anterior forebrain pathway. In several cases, we noted a seemingly coordinated pattern across multiple nuclei (SCN1B, SCN3B, SCN4B, CACNB4) or sparse expression (SCN1A, CACNG5, CACNA1B). CONCLUSION The gene families examined are highly conserved between avian and mammalian lineages. Several cases of differential expression likely support high-frequency and burst firing in specific song nuclei, whereas cases of sparse patterns of expression may contribute to the unique electrophysiological signatures of distinct cell populations. These observations lay the groundwork for manipulations to determine how ion channels contribute to the neuronal excitability properties of vocal learning systems.
Collapse
Affiliation(s)
- Samantha R. Friedrich
- Department of Behavioral Neuroscience, Oregon Health and Science University, 3181 Sam Jackson Park Rd L470, Portland, OR USA
| | - Peter V. Lovell
- Department of Behavioral Neuroscience, Oregon Health and Science University, 3181 Sam Jackson Park Rd L470, Portland, OR USA
| | - Taylor M. Kaser
- Department of Behavioral Neuroscience, Oregon Health and Science University, 3181 Sam Jackson Park Rd L470, Portland, OR USA
| | - Claudio V. Mello
- Department of Behavioral Neuroscience, Oregon Health and Science University, 3181 Sam Jackson Park Rd L470, Portland, OR USA
| |
Collapse
|
34
|
Hossain KR, Turkewitz DR, Holt SA, Herson L, Brown LJ, Cornell BA, Curmi PMG, Valenzuela SM. A conserved GXXXG motif in the transmembrane domain of CLIC proteins is essential for their cholesterol-dependant membrane interaction. Biochim Biophys Acta Gen Subj 2019; 1863:1243-1253. [PMID: 31075359 DOI: 10.1016/j.bbagen.2019.04.020] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Revised: 04/25/2019] [Accepted: 04/29/2019] [Indexed: 12/21/2022]
Abstract
BACKGROUND Sterols have been reported to modulate conformation and hence the function of several membrane proteins. One such group is the Chloride Intracellular Ion Channel (CLIC) family of proteins. The CLIC protein family consists of six evolutionarily conserved protein members in vertebrates. These proteins exist as both monomeric soluble proteins and as membrane bound proteins. To date, the structure of their membrane-bound form remains unknown. In addition to several studies indicating cellular redox environment and pH as facilitators of CLIC1 insertion into membranes, we have also demonstrated that the spontaneous membrane insertion of CLIC1 is regulated by membrane cholesterol. METHOD We have performed Langmuir-film, Impedance Spectroscopy and Molecular Docking Simulations to study the role of this GXXXG motif in CLIC1 interaction with cholesterol. RESULTS Unlike CLIC1-wild-type protein, the G18A and G22A mutants, that form part of the GXXXG motif, showed much slower initial kinetics and lower ion channel activity compared to the native protein. This difference can be attributed to the significantly reduced membrane interaction and insertion rate of the mutant proteins and/or slower formation of the final membrane configuration of the mutant proteins once in the membrane. CONCLUSION In this study, our findings uncover the identification of a GXXXG motif in CLIC1, which likely serves as the cholesterol-binding domain, that facilitates the protein's membrane interaction and insertion. Furthermore, we were able to postulate a model by which CLIC1 can autonomously insert into membranes to form functional ion channels. GENERAL SIGNIFICANCE Members of the CLIC family of proteins demonstrate unusual structural and dual functional properties - as ion channels and enzymes. Elucidating how the CLIC proteins' interact with membranes, thus allowing them to switch between their soluble and membrane form, will provide key information as to a mechanism of moonlighting activity and a novel regulatory role for cholesterol in such a process.
Collapse
Affiliation(s)
- Khondker Rufaka Hossain
- School of Life Sciences, University of Technology Sydney, Sydney, New South Wales 2007, Australia
| | - Daniel R Turkewitz
- School of Life Sciences, University of Technology Sydney, Sydney, New South Wales 2007, Australia
| | - Stephen A Holt
- Australian Nuclear Science and Technology Organisation (ANSTO), Lucas Height, Australian Centre for Neutron Scattering, New South Wales 2234, Australia
| | - Leonie Herson
- School of Life Sciences, University of Technology Sydney, Sydney, New South Wales 2007, Australia
| | - Louise J Brown
- Department of Molecular Sciences, Macquarie University, New South Wales 2109, Australia
| | - Bruce A Cornell
- Surgical Diagnostics Pty Ltd., Roseville, Sydney 2069, Australia; Institute for Biomedical Materials and Devices, University of Technology Sydney, Sydney, New South Wales 2007, Australia
| | - Paul M G Curmi
- School of Physics, The University of New South Wales, Sydney, New South Wales 2052, Australia
| | - Stella M Valenzuela
- School of Life Sciences, University of Technology Sydney, Sydney, New South Wales 2007, Australia; Institute for Biomedical Materials and Devices, University of Technology Sydney, Sydney, New South Wales 2007, Australia.
| |
Collapse
|
35
|
Lam A, Karekar P, Shah K, Hariharan G, Fleyshman M, Kaur H, Singh H, Gururaja Rao S. Drosophila Voltage-Gated Calcium Channel α1-Subunits Regulate Cardiac Function in the Aging Heart. Sci Rep 2018; 8:6910. [PMID: 29720608 PMCID: PMC5932002 DOI: 10.1038/s41598-018-25195-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Accepted: 04/17/2018] [Indexed: 12/20/2022] Open
Abstract
Ion channels maintain numerous physiological functions and regulate signaling pathways. They are the key targets for cellular reactive oxygen species (ROS), acting as signaling switches between ROS and ionic homeostasis. We have carried out a paraquat (PQ) screen in Drosophila to identify ion channels regulating the ROS handling and survival in Drosophila melanogaster. Our screen has revealed that α1-subunits (D-type, T-type, and cacophony) of voltage-gated calcium channels (VGCCs) handle PQ-mediated ROS stress differentially in a gender-based manner. Since ROS are also involved in determining the lifespan, we discovered that the absence of T-type and cacophony decreased the lifespan while the absence of D-type maintained a similar lifespan to that of the wild-type strain. VGCCs are also responsible for electrical signaling in cardiac cells. The cardiac function of each mutant was evaluated through optical coherence tomography (OCT), which revealed that α1-subunits of VGCCs are essential in maintaining cardiac rhythmicity and cardiac function in an age-dependent manner. Our results establish specific roles of α1-subunits of VGCCs in the functioning of the aging heart.
Collapse
Affiliation(s)
- Alexander Lam
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA, 19102, USA
| | - Priyanka Karekar
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA, 19102, USA
| | - Kajol Shah
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA, 19102, USA
| | - Girija Hariharan
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA, 19102, USA
| | - Michelle Fleyshman
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA, 19102, USA
| | - Harmehak Kaur
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA, 19102, USA
| | - Harpreet Singh
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA, 19102, USA. .,Division of Cardiology, Department of Medicine, Drexel University College of Medicine, Philadelphia, PA, 19102, USA.
| | - Shubha Gururaja Rao
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA, 19102, USA.
| |
Collapse
|
36
|
Gururaja Rao S, Ponnalagu D, Patel NJ, Singh H. Three Decades of Chloride Intracellular Channel Proteins: From Organelle to Organ Physiology. CURRENT PROTOCOLS IN PHARMACOLOGY 2018; 80:11.21.1-11.21.17. [PMID: 30040212 PMCID: PMC6060641 DOI: 10.1002/cpph.36] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Intracellular organelles are membranous structures central for maintaining cellular physiology and the overall health of the cell. To maintain cellular function, intracellular organelles are required to tightly regulate their ionic homeostasis. Any imbalance in ionic concentrations can disrupt energy production (mitochondria), protein degradation (lysosomes), DNA replication (nucleus), or cellular signaling (endoplasmic reticulum). Ionic homeostasis is also important for volume regulation of intracellular organelles and is maintained by cation and anion channels as well as transporters. One of the major classes of ion channels predominantly localized to intracellular membranes is chloride intracellular channel proteins (CLICs). They are non-canonical ion channels with six homologs in mammals, existing as either soluble or integral membrane protein forms, with dual functions as enzymes and channels. Provided in this overview is a brief introduction to CLICs, and a summary of recent information on their localization, biophysical properties, and physiological roles. © 2018 by John Wiley & Sons, Inc.
Collapse
Affiliation(s)
- Shubha Gururaja Rao
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, Pennsylvania
| | - Devasena Ponnalagu
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, Pennsylvania
| | - Neel J Patel
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, Pennsylvania
| | - Harpreet Singh
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, Pennsylvania
| |
Collapse
|
37
|
Zeng J, Li Z, Lui EY, Lam SH, Swaminathan K. Tilapia and human CLIC2 structures are highly conserved. Biochem Biophys Res Commun 2017; 495:1752-1757. [PMID: 29198705 DOI: 10.1016/j.bbrc.2017.11.189] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Accepted: 11/28/2017] [Indexed: 01/11/2023]
Abstract
Chloride intracellular channels (CLICs) exist in soluble and membrane bound forms. We have determined the crystal structure of soluble Clic2 from the euryhaline teleost fish Oreochromis mossambicus. Structural comparison of tilapia and human CLIC2 with other CLICs shows that these proteins are highly conserved. We have also compared the expression levels of clic2 in selected osmoregulatory organs of tilapia, acclimated to freshwater, seawater and hypersaline water. Structural conservation of vertebrate CLICs implies that they might play conserved roles. Also, tissue-specific responsiveness of clic2 suggests that it might be involved in iono-osmoregulation under extreme conditions in tilapia.
Collapse
Affiliation(s)
- Jiao Zeng
- Department of Biological Sciences, National University of Singapore 117543, Singapore
| | - Zhengjun Li
- NUS Environmental Research Institute, National University of Singapore 117411, Singapore
| | - Eei Yin Lui
- NUS Environmental Research Institute, National University of Singapore 117411, Singapore
| | - Siew Hong Lam
- Department of Biological Sciences, National University of Singapore 117543, Singapore; NUS Environmental Research Institute, National University of Singapore 117411, Singapore
| | | |
Collapse
|
38
|
Choi H, Gim J, Won S, Kim YJ, Kwon S, Park C. Network analysis for count data with excess zeros. BMC Genet 2017; 18:93. [PMID: 29110633 PMCID: PMC5674822 DOI: 10.1186/s12863-017-0561-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2017] [Accepted: 10/25/2017] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Undirected graphical models or Markov random fields have been a popular class of models for representing conditional dependence relationships between nodes. In particular, Markov networks help us to understand complex interactions between genes in biological processes of a cell. Local Poisson models seem to be promising in modeling positive as well as negative dependencies for count data. Furthermore, when zero counts are more frequent than are expected, excess zeros should be considered in the model. METHODS We present a penalized Poisson graphical model for zero inflated count data and derive an expectation-maximization (EM) algorithm built on coordinate descent. Our method is shown to be effective through simulated and real data analysis. RESULTS Results from the simulated data indicate that our method outperforms the local Poisson graphical model in the presence of excess zeros. In an application to a RNA sequencing data, we also investigate the gender effect by comparing the estimated networks according to different genders. Our method may help us in identifying biological pathways linked to sex hormone regulation and thus understanding underlying mechanisms of the gender differences. CONCLUSIONS We have presented a penalized version of zero inflated spatial Poisson regression and derive an efficient EM algorithm built on coordinate descent. We discuss possible improvements of our method as well as potential research directions associated with our findings from the RNA sequencing data.
Collapse
Affiliation(s)
- Hosik Choi
- Department of Applied Statistics, Kyonggi University, Suwon, 16227 Korea
| | - Jungsoo Gim
- Institute of Health and Environment, Seoul National University, Seoul, 08826 Korea
| | - Sungho Won
- Graduate School of Public Health, Seoul National University, 08826Seoul, Korea
| | - You Jin Kim
- Department of Nutritional Science and Food Management, Ewha Womans University, Seoul, 03760 Korea
| | - Sunghoon Kwon
- Department of Applied Statistics, Konkuk University, Seoul, 05029 Korea
| | - Changyi Park
- Department of Statistics, University of Seoul, Seoul, 02504 Korea
| |
Collapse
|
39
|
Hossain KR, Holt SA, Le Brun AP, Al Khamici H, Valenzuela SM. X-ray and Neutron Reflectivity Study Shows That CLIC1 Undergoes Cholesterol-Dependent Structural Reorganization in Lipid Monolayers. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2017; 33:12497-12509. [PMID: 29016141 DOI: 10.1021/acs.langmuir.7b02872] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
CLIC1 belongs to the ubiquitous family of chloride intracellular ion channel proteins that are evolutionarily conserved across species. The CLICs are unusual in that they exist mainly as soluble proteins but possess the intriguing property of spontaneous conversion from the soluble to an integral membrane-bound form. This conversion is regulated by the membrane lipid composition, especially by cholesterol, together with external factors such as oxidation and pH. However, the precise physiological mechanism regulating CLIC1 membrane insertion is currently unknown. In this study, X-ray and neutron reflectivity experiments were performed to study the interaction of CLIC1 with different phospholipid monolayers prepared using POPC, POPE, or POPS with and without cholesterol in order to better understand the regulatory role of cholesterol in CLIC1 membrane insertion. Our findings demonstrate for the first time two different structural orientations of CLIC1 within phospholipid monolayers, dependent upon the absence or presence of cholesterol. In phospholipid monolayers devoid of cholesterol, CLIC1 was unable to insert into the lipid acyl chain region. However, in the presence of cholesterol, CLIC1 showed significant insertion within the phospholipid acyl chains occupying an area per protein molecule of 6-7 nm2 with a total CLIC1 thickness ranging from ∼50 to 56 Å across the entire monolayer. Our data strongly suggests that cholesterol not only facilitates the initial docking or binding of CLIC1 to the membrane but also promotes deeper penetration of CLIC1 into the hydrophobic tails of the lipid monolayer.
Collapse
Affiliation(s)
- Khondker R Hossain
- Australian Centre for Neutron Scattering, Australian Nuclear Science and Technology Organisation (ANSTO), Lucas Heights, New South Wales 2234, Australia
| | - Stephen A Holt
- Australian Centre for Neutron Scattering, Australian Nuclear Science and Technology Organisation (ANSTO), Lucas Heights, New South Wales 2234, Australia
| | - Anton P Le Brun
- Australian Centre for Neutron Scattering, Australian Nuclear Science and Technology Organisation (ANSTO), Lucas Heights, New South Wales 2234, Australia
| | | | | |
Collapse
|
40
|
Identification and Characterization of a Bacterial Homolog of Chloride Intracellular Channel (CLIC) Protein. Sci Rep 2017; 7:8500. [PMID: 28819106 PMCID: PMC5561075 DOI: 10.1038/s41598-017-08742-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Accepted: 07/17/2017] [Indexed: 11/25/2022] Open
Abstract
Chloride intracellular channels (CLIC) are non-classical ion channels lacking a signal sequence for membrane targeting. In eukaryotes, they are implicated in cell volume regulation, acidification, and cell cycle. CLICs resemble the omega class of Glutathione S-transferases (GST), yet differ from them in their ability to form ion channels. They are ubiquitously found in eukaryotes but no prokaryotic homolog has been characterized. We found that indanyloxyacetic acid-94 (IAA-94), a blocker of CLICs, delays the growth of Escherichia coli. In silico analysis showed that the E. coli stringent starvation protein A (SspA) shares sequence and structural homology with CLICs. Similar to CLICs, SspA lacks a signal sequence but contains an omega GST fold. Electrophysiological analysis revealed that SspA auto-inserts into lipid bilayers and forms IAA-94-sensitive ion channels. Substituting the ubiquitously conserved residue leucine 29 to alanine in the pore-forming region increased its single-channel conductance. SspA is essential for cell survival during acid-induced stress, and we found that acidic pH increases the open probability of SspA. Further, IAA-94 delayed the growth of wild-type but not sspA null mutant E. coli. Our results for the first time show that CLIC-like proteins exist in bacteria in the form of SspA, forming functional ion channels.
Collapse
|
41
|
Argenzio E, Moolenaar WH. Emerging biological roles of Cl- intracellular channel proteins. J Cell Sci 2017; 129:4165-4174. [PMID: 27852828 DOI: 10.1242/jcs.189795] [Citation(s) in RCA: 74] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Cl- intracellular channels (CLICs) are a family of six evolutionary conserved cytosolic proteins that exist in both soluble and membrane-associated forms; however, their functions have long been elusive. Soluble CLICs adopt a glutathione S-transferase (GST)-fold, can induce ion currents in artificial membranes and show oxidoreductase activity in vitro, but there is no convincing evidence of CLICs having such activities in vivo. Recent studies have revealed a role for CLIC proteins in Rho-regulated cortical actin dynamics as well as vesicular trafficking and integrin recycling, the latter of which are under the control of Rab GTPases. In this Commentary, we discuss the emerging roles of CLIC proteins in these processes and the lessons learned from gene-targeting studies. We also highlight outstanding questions regarding the molecular function(s) of these important but still poorly understood proteins.
Collapse
Affiliation(s)
- Elisabetta Argenzio
- Division of Cell Biology, The Netherlands Cancer Institute, Amsterdam 1066CX, The Netherlands
| | - Wouter H Moolenaar
- Division of Cell Biology, The Netherlands Cancer Institute, Amsterdam 1066CX, The Netherlands
| |
Collapse
|
42
|
Tomasek M, Misak A, Grman M, Tomaskova Z. Subconductance states of mitochondrial chloride channels: implication for functionally-coupled tetramers. FEBS Lett 2017. [PMID: 28640976 DOI: 10.1002/1873-3468.12721] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Recently, it has been discovered that isoforms of intracellular chloride channels (CLIC) are present in cardiac mitochondria. By reconstituting rat cardiac mitochondrial chloride channels into bilayer lipid membranes, we detected three equally separated subconductance states with conductance increment of 45 pS and < 2% occupancy. The observed rare events of channel decomposition into substates, accompanied by disrupted gating, provide an insight into channel quaternary structure. Our findings suggest that the observed channels work as four functionally coupled subunits with synchronized gating. We discuss the putative connection of channel activity from native mitochondria with the recombinant CLIC channels. However, conclusive evidence is needed to prove this connection.
Collapse
Affiliation(s)
| | - Anton Misak
- Institute of Clinical and Translational Research, Biomedical Research Center, Slovak Academy of Sciences, Bratislava, Slovak
| | - Marian Grman
- Institute of Clinical and Translational Research, Biomedical Research Center, Slovak Academy of Sciences, Bratislava, Slovak
| | - Zuzana Tomaskova
- Institute of Molecular Physiology and Genetics, Centre of Biosciences, Slovak Academy of Sciences, Bratislava, Slovak
| |
Collapse
|
43
|
Brum AM, van der Leije CS, Schreuders-Koedam M, Verhoeven J, Janssen M, Dekkers DH, Demmers JA, Eijken M, van de Peppel J, van Leeuwen JP, van der Eerden BC. Identification of Chloride Intracellular Channel Protein 3 as a Novel Gene Affecting Human Bone Formation. JBMR Plus 2017; 1:16-26. [PMID: 30283877 PMCID: PMC6124162 DOI: 10.1002/jbm4.10003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Accepted: 03/09/2017] [Indexed: 12/14/2022] Open
Abstract
Osteoporosis is a common skeletal disorder characterized by low bone mass leading to increased bone fragility and fracture susceptibility. The bone building cells, osteoblasts, are derived from mesenchymal stromal cells (MSCs); however, with increasing age osteogenic differentiation is diminished and more adipocytes are seen in the bone marrow, suggesting a shift in MSC lineage commitment. Identification of specific factors that stimulate osteoblast differentiation from human MSCs may deliver therapeutic targets to treat osteoporosis. The aim of this study was to identify novel genes involved in osteoblast differentiation of human bone marrow–derived MSCs (hMSCs). We identified the gene chloride intracellular channel protein 3 (CLIC3) to be strongly upregulated during MSC‐derived osteoblast differentiation. Lentiviral overexpression of CLIC3 in hMSCs caused a 60% increase of matrix mineralization. Conversely, knockdown of CLIC3 in hMSCs using two short‐hairpin RNAs (shRNAs) against CLIC3 resulted in a 69% to 76% reduction in CLIC3 mRNA expression, 53% to 37% less alkaline phosphatase (ALP) activity, and 78% to 88% less matrix mineralization compared to scrambled control. Next, we used an in vivo human bone formation model in which hMSCs lentivirally transduced with the CLIC3 overexpression construct were loaded onto a scaffold (hydroxyapatite‐tricalcium‐phosphate), implanted under the skin of NOD‐SCID mice, and analyzed for bone formation 8 weeks later. CLIC3 overexpression led to a 15‐fold increase in bone formation (0.33% versus 5.05% bone area relative to scaffold). Using a Clic3‐His‐tagged pull‐down assay and liquid chromatography–mass spectrometry (LS/MS)‐based proteomics analysis in lysates of osteogenically differentiated hMSCs, we showed that CLIC3 interacts with NIMA‐related kinase 9 (NEK9) and phosphatidylserine synthase 1 (PTDSS1) in vitro, and this finding was supported by immunofluorescent analysis. In addition, inhibition of NEK9 or PTDSS1 gene expression by shRNAs inhibited osteoblast differentiation and mineralization. In conclusion, we successfully identified CLIC3 to be a lineage‐specific gene regulating osteoblast differentiation and bone formation through its interaction with NEK9 and PTDSS1. © The Authors. JBMR Plus is published by Wiley Periodicals, Inc. on behalf of the American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Andrea M Brum
- Department of Internal Medicine School of Molecular Medicine Erasmus University Medical Center Rotterdam the Netherlands
| | - Cindy S van der Leije
- Department of Internal Medicine School of Molecular Medicine Erasmus University Medical Center Rotterdam the Netherlands
| | - Marijke Schreuders-Koedam
- Department of Internal Medicine School of Molecular Medicine Erasmus University Medical Center Rotterdam the Netherlands
| | - Jeroen Verhoeven
- Department of Internal Medicine School of Molecular Medicine Erasmus University Medical Center Rotterdam the Netherlands
| | | | - Dick Hw Dekkers
- Proteomics Center Erasmus University Medical Center Rotterdam The Netherlands
| | - Jeroen Aa Demmers
- Proteomics Center Erasmus University Medical Center Rotterdam The Netherlands
| | | | - Jeroen van de Peppel
- Department of Internal Medicine School of Molecular Medicine Erasmus University Medical Center Rotterdam the Netherlands
| | - Johannes Ptm van Leeuwen
- Department of Internal Medicine School of Molecular Medicine Erasmus University Medical Center Rotterdam the Netherlands
| | - Bram Cj van der Eerden
- Department of Internal Medicine School of Molecular Medicine Erasmus University Medical Center Rotterdam the Netherlands
| |
Collapse
|
44
|
Abstract
Mitochondria are the "power house" of a cell continuously generating ATP to ensure its proper functioning. The constant production of ATP via oxidative phosphorylation demands a large electrochemical force that drives protons across the highly selective and low-permeable mitochondrial inner membrane. Besides the conventional role of generating ATP, mitochondria also play an active role in calcium signaling, generation of reactive oxygen species (ROS), stress responses, and regulation of cell-death pathways. Deficiencies in these functions result in several pathological disorders like aging, cancer, diabetes, neurodegenerative and cardiovascular diseases. A plethora of ion channels and transporters are present in the mitochondrial inner and outer membranes which work in concert to preserve the ionic equilibrium of a cell for the maintenance of cell integrity, in physiological as well as pathophysiological conditions. For, e.g., mitochondrial cation channels KATP and BKCa play a significant role in cardioprotection from ischemia-reperfusion injury. In addition to the cation channels, mitochondrial anion channels are equally essential, as they aid in maintaining electro-neutrality by regulating the cell volume and pH. This chapter focusses on the information on molecular identity, structure, function, and physiological relevance of mitochondrial chloride channels such as voltage dependent anion channels (VDACs), uncharacterized mitochondrial inner membrane anion channels (IMACs), chloride intracellular channels (CLIC) and the aspects of forthcoming chloride channels.
Collapse
Affiliation(s)
- Devasena Ponnalagu
- Department of Pharmacology and Physiology, Drexel University College of Medicine, 245 N. 15th Street, Room 8154, Mail Stop 488, Philadelphia, PA, 19102-1192, USA
| | - Harpreet Singh
- Department of Pharmacology and Physiology, Drexel University College of Medicine, 245 N. 15th Street, Room 8154, Mail Stop 488, Philadelphia, PA, 19102-1192, USA.
| |
Collapse
|
45
|
Hare JE, Goodchild SC, Breit SN, Curmi PMG, Brown LJ. Interaction of Human Chloride Intracellular Channel Protein 1 (CLIC1) with Lipid Bilayers: A Fluorescence Study. Biochemistry 2016; 55:3825-33. [PMID: 27299171 DOI: 10.1021/acs.biochem.6b00080] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Chloride intracellular channel protein 1 (CLIC1) is very unusual as it adopts a soluble glutathione S-transferase-like canonical fold but can also autoinsert into lipid bilayers to form an ion channel. The conversion between these forms involves a large, but reversible, structural rearrangement of the CLIC1 module. The only identified environmental triggers controlling the metamorphic transition of CLIC1 are pH and oxidation. Until now, there have been no high-resolution structural data available for the CLIC1 integral membrane state, and consequently, a limited understanding of how CLIC1 unfolds and refolds across the bilayer to form a membrane protein with ion channel activity exists. Here we show that fluorescence spectroscopy can be used to establish the interaction and position of CLIC1 in a lipid bilayer. Our method employs a fluorescence energy transfer (FRET) approach between CLIC1 and a dansyl-labeled lipid analogue to probe the CLIC1-lipid interface. Under oxidizing conditions, a strong FRET signal between the single tryptophan residue of CLIC1 (Trp35) and the dansyl-lipid analogue was detected. When considering the proportion of CLIC1 interacting with the lipid bilayer, as estimated by fluorescence quenching experiments, the FRET distance between Trp35 and the dansyl moiety on the membrane surface was determined to be ∼15 Å. This FRET-detected interaction provides direct structural evidence that CLIC1 associates with membranes. The results presented support the current model of an oxidation-driven interaction of CLIC1 with lipid bilayers and also propose a membrane anchoring role for Trp35.
Collapse
Affiliation(s)
- Joanna E Hare
- Department of Chemistry and Biomolecular Sciences, Macquarie University , Sydney, New South Wales 2109, Australia
| | - Sophia C Goodchild
- Astbury Centre for Structural Molecular Biology and School of Molecular and Cellular Biology, University of Leeds , Leeds LS29JT, United Kingdom
| | - Samuel N Breit
- St Vincent's Centre for Applied Medical Research, St Vincent's Hospital , Sydney, New South Wales 2010, Australia
| | - Paul M G Curmi
- School of Physics, University of New South Wales , Sydney, New South Wales 2052, Australia
| | - Louise J Brown
- Department of Chemistry and Biomolecular Sciences, Macquarie University , Sydney, New South Wales 2109, Australia
| |
Collapse
|
46
|
Ponnalagu D, Gururaja Rao S, Farber J, Xin W, Hussain AT, Shah K, Tanda S, Berryman M, Edwards JC, Singh H. Molecular identity of cardiac mitochondrial chloride intracellular channel proteins. Mitochondrion 2016; 27:6-14. [PMID: 26777142 DOI: 10.1016/j.mito.2016.01.001] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2015] [Revised: 12/08/2015] [Accepted: 01/07/2016] [Indexed: 01/08/2023]
Abstract
Emerging evidences demonstrate significance of chloride channels in cardiac function and cardioprotection from ischemia-reperfusion (IR) injury. Unlike mitochondrial potassium channels sensitive to calcium (BKCa) and ATP (KATP), molecular identity of majority of cardiac mitochondrial chloride channels located at the inner membrane is not known. In this study, we report the presence of unique dimorphic chloride intracellular channel (CLIC) proteins namely CLIC1, CLIC4 and CLIC5 as abundant CLICs in the rodent heart. Further, CLIC4, CLIC5, and an ortholog present in Drosophila (DmCLIC) localize to adult cardiac mitochondria. We found that CLIC4 is enriched in the outer mitochondrial membrane, whereas CLIC5 is present in the inner mitochondrial membrane. Also, CLIC5 plays a direct role in regulating mitochondrial reactive oxygen species (ROS) generation. Our study highlights that CLIC5 is localized to the cardiac mitochondria and directly modulates mitochondrial function.
Collapse
Affiliation(s)
- Devasena Ponnalagu
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA 19102, United States
| | - Shubha Gururaja Rao
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA 19102, United States
| | - Jason Farber
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA 19102, United States
| | - Wenyu Xin
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA 19102, United States
| | - Ahmed Tafsirul Hussain
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA 19102, United States
| | - Kajol Shah
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA 19102, United States
| | - Soichi Tanda
- Department of Biological Sciences, Ohio University, Athens, OH 45701, United States
| | - Mark Berryman
- Department of Biomedical Sciences, Ohio University, Athens, OH 45701, United States
| | - John C Edwards
- Division of Nephrology, St. Louis University, St. Louis, MO 63110, United States
| | - Harpreet Singh
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA 19102, United States.
| |
Collapse
|
47
|
Kamano S, Kume S, Iida K, Lei KJ, Nakano M, Nakayama Y, Iida H. Transmembrane Topologies of Ca2+-permeable Mechanosensitive Channels MCA1 and MCA2 in Arabidopsis thaliana. J Biol Chem 2015; 290:30901-9. [PMID: 26555262 DOI: 10.1074/jbc.m115.692574] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Indexed: 11/06/2022] Open
Abstract
Sensing mechanical stresses, including touch, stretch, compression, and gravity, is crucial for growth and development in plants. A good mechanosensor candidate is the Ca(2+)-permeable mechanosensitive (MS) channel, the pore of which opens to permeate Ca(2+) in response to mechanical stresses. However, the structure-function relationships of plant MS channels are poorly understood. Arabidopsis MCA1 and MCA2 form a homotetramer and exhibit Ca(2+)-permeable MS channel activity; however, their structures have only been partially elucidated. The transmembrane topologies of these ion channels need to be determined in more detail to elucidate the underlying regulatory mechanisms. We herein determined the topologies of MCA1 and MCA2 using two independent methods, the Suc2C reporter and split-ubiquitin yeast two-hybrid methods, and found that both proteins are single-pass type I integral membrane proteins with extracellular N termini and intracellular C termini. These results imply that an EF hand-like motif, coiled-coil motif, and plac8 motif are all present in the cytoplasm. Thus, the activities of both channels can be regulated by intracellular Ca(2+) and protein interactions.
Collapse
Affiliation(s)
- Shumpei Kamano
- From the Department of Biology, Tokyo Gakugei University, 4-1-1 Nukui kita-machi, Koganei, Tokyo 184-8501, Japan and
| | - Shinichiro Kume
- From the Department of Biology, Tokyo Gakugei University, 4-1-1 Nukui kita-machi, Koganei, Tokyo 184-8501, Japan and
| | - Kazuko Iida
- Laboratory of Biomembrane, Tokyo Metropolitan Institute of Medical Science, 2-1-6 Kamikitazawa, Setagaya, Tokyo 156-8506, Japan
| | - Kai-Jian Lei
- From the Department of Biology, Tokyo Gakugei University, 4-1-1 Nukui kita-machi, Koganei, Tokyo 184-8501, Japan and
| | - Masataka Nakano
- From the Department of Biology, Tokyo Gakugei University, 4-1-1 Nukui kita-machi, Koganei, Tokyo 184-8501, Japan and
| | - Yoshitaka Nakayama
- From the Department of Biology, Tokyo Gakugei University, 4-1-1 Nukui kita-machi, Koganei, Tokyo 184-8501, Japan and
| | - Hidetoshi Iida
- From the Department of Biology, Tokyo Gakugei University, 4-1-1 Nukui kita-machi, Koganei, Tokyo 184-8501, Japan and
| |
Collapse
|
48
|
Al Khamici H, Brown LJ, Hossain KR, Hudson AL, Sinclair-Burton AA, Ng JPM, Daniel EL, Hare JE, Cornell BA, Curmi PMG, Davey MW, Valenzuela SM. Members of the chloride intracellular ion channel protein family demonstrate glutaredoxin-like enzymatic activity. PLoS One 2015; 10:e115699. [PMID: 25581026 PMCID: PMC4291220 DOI: 10.1371/journal.pone.0115699] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2014] [Accepted: 11/26/2014] [Indexed: 01/07/2023] Open
Abstract
The Chloride Intracellular Ion Channel (CLIC) family consists of six evolutionarily conserved proteins in humans. Members of this family are unusual, existing as both monomeric soluble proteins and as integral membrane proteins where they function as chloride selective ion channels, however no function has previously been assigned to their soluble form. Structural studies have shown that in the soluble form, CLIC proteins adopt a glutathione S-transferase (GST) fold, however, they have an active site with a conserved glutaredoxin monothiol motif, similar to the omega class GSTs. We demonstrate that CLIC proteins have glutaredoxin-like glutathione-dependent oxidoreductase enzymatic activity. CLICs 1, 2 and 4 demonstrate typical glutaredoxin-like activity using 2-hydroxyethyl disulfide as a substrate. Mutagenesis experiments identify cysteine 24 as the catalytic cysteine residue in CLIC1, which is consistent with its structure. CLIC1 was shown to reduce sodium selenite and dehydroascorbate in a glutathione-dependent manner. Previous electrophysiological studies have shown that the drugs IAA-94 and A9C specifically block CLIC channel activity. These same compounds inhibit CLIC1 oxidoreductase activity. This work for the first time assigns a functional activity to the soluble form of the CLIC proteins. Our results demonstrate that the soluble form of the CLIC proteins has an enzymatic activity that is distinct from the channel activity of their integral membrane form. This CLIC enzymatic activity may be important for protecting the intracellular environment against oxidation. It is also likely that this enzymatic activity regulates the CLIC ion channel function.
Collapse
Affiliation(s)
- Heba Al Khamici
- School of Medical and Molecular Biosciences, University of Technology Sydney, Sydney, New South Wales 2007, Australia
| | - Louise J. Brown
- Department of Chemistry and Bimolecular Sciences, Macquarie University, Sydney, New South Wales 2109, Australia
| | - Khondker R. Hossain
- School of Medical and Molecular Biosciences, University of Technology Sydney, Sydney, New South Wales 2007, Australia
- Bragg Institute, Australian Nuclear Science and Technology Organisation, Sydney, New South Wales 2234, Australia
| | - Amanda L. Hudson
- School of Medical and Molecular Biosciences, University of Technology Sydney, Sydney, New South Wales 2007, Australia
| | - Alxcia A. Sinclair-Burton
- School of Medical and Molecular Biosciences, University of Technology Sydney, Sydney, New South Wales 2007, Australia
| | - Jane Phui Mun Ng
- School of Medical and Molecular Biosciences, University of Technology Sydney, Sydney, New South Wales 2007, Australia
| | - Elizabeth L. Daniel
- Department of Chemistry and Bimolecular Sciences, Macquarie University, Sydney, New South Wales 2109, Australia
| | - Joanna E. Hare
- Department of Chemistry and Bimolecular Sciences, Macquarie University, Sydney, New South Wales 2109, Australia
| | - Bruce A. Cornell
- Surgical Diagnostics, Roseville, Sydney, New South Wales 2069, Australia
| | - Paul M. G. Curmi
- School of Physics, University of New South Wales, Sydney, New South Wales 2052, Australia
- Centre for Applied Medical Research, St Vincent's Hospital, Sydney, New South Wales 2010, Australia
| | - Mary W. Davey
- School of Medical and Molecular Biosciences, University of Technology Sydney, Sydney, New South Wales 2007, Australia
| | - Stella M. Valenzuela
- School of Medical and Molecular Biosciences, University of Technology Sydney, Sydney, New South Wales 2007, Australia
- Centre for Health Technologies, University of Technology Sydney, Sydney, New South Wales 2007, Australia
| |
Collapse
|
49
|
Correani V, Francesco LD, Cera I, Mignogna G, Giorgi A, Mazzanti M, Fumagalli L, Fabrizi C, Maras B, Schininà ME. Reversible redox modifications in the microglial proteome challenged by beta amyloid. MOLECULAR BIOSYSTEMS 2015; 11:1584-93. [DOI: 10.1039/c4mb00703d] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Reversible redox modifications of the microglial proteome contribute to switching of these neuronal sentinel cells toward a neuroinflammatory phenotype.
Collapse
Affiliation(s)
- Virginia Correani
- Dipartimento di Scienze Biochimiche
- Sapienza University of Rome
- 00185 Rome
- Italy
| | - Laura Di Francesco
- Dipartimento di Scienze Biochimiche
- Sapienza University of Rome
- 00185 Rome
- Italy
| | - Isabella Cera
- Dipartimento di Scienze Biochimiche
- Sapienza University of Rome
- 00185 Rome
- Italy
| | - Giuseppina Mignogna
- Dipartimento di Scienze Biochimiche
- Sapienza University of Rome
- 00185 Rome
- Italy
| | - Alessandra Giorgi
- Dipartimento di Scienze Biochimiche
- Sapienza University of Rome
- 00185 Rome
- Italy
| | - Michele Mazzanti
- Dipartimento di Bioscienze
- Università degli Studi di Milano
- Milan
- Italy
| | - Lorenzo Fumagalli
- Dipartimento di Scienze Anatomiche
- Istologiche
- Medico-Legali e dell'Apparato Locomotore
- Sapienza University of Rome
- Rome
| | - Cinzia Fabrizi
- Dipartimento di Scienze Anatomiche
- Istologiche
- Medico-Legali e dell'Apparato Locomotore
- Sapienza University of Rome
- Rome
| | - Bruno Maras
- Dipartimento di Scienze Biochimiche
- Sapienza University of Rome
- 00185 Rome
- Italy
| | - M. Eugenia Schininà
- Dipartimento di Scienze Biochimiche
- Sapienza University of Rome
- 00185 Rome
- Italy
| |
Collapse
|
50
|
Haltaufderhyde KD, Oancea E. Genome-wide transcriptome analysis of human epidermal melanocytes. Genomics 2014; 104:482-9. [PMID: 25451175 DOI: 10.1016/j.ygeno.2014.09.010] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2014] [Revised: 09/19/2014] [Accepted: 09/22/2014] [Indexed: 01/09/2023]
Abstract
Because human epidermal melanocytes (HEMs) provide critical protection against skin cancer, sunburn, and photoaging, a genome-wide perspective of gene expression in these cells is vital to understanding human skin physiology. In this study we performed high throughput sequencing of HEMs to obtain a complete data set of transcript sizes, abundances, and splicing. As expected, we found that melanocyte specific genes that function in pigmentation were among the highest expressed genes. We analyzed receptor, ion channel and transcription factor gene families to get a better understanding of the cell signaling pathways used by melanocytes. We also performed a comparative transcriptomic analysis of lightly versus darkly pigmented HEMs and found 16 genes differentially expressed in the two pigmentation phenotypes; of those, only one putative melanosomal transporter (SLC45A2) has known function in pigmentation. In addition, we found 166 transcript isoforms expressed exclusively in one pigmentation phenotype, 17 of which are genes involved in signal transduction. Our melanocyte transcriptome study provides a comprehensive view and may help identify novel pigmentation genes and potential pharmacological targets.
Collapse
Affiliation(s)
- Kirk D Haltaufderhyde
- Department of Molecular Pharmacology, Physiology, and Biotechnology, Brown University, Providence, RI 02192, USA
| | - Elena Oancea
- Department of Molecular Pharmacology, Physiology, and Biotechnology, Brown University, Providence, RI 02192, USA.
| |
Collapse
|