1
|
Kulkarni GC, Saha R, Peters CJ. Ion channel expression and function in glioblastoma multiforme (GBM): pathophysiological mechanisms and therapeutic potential. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2025; 1872:119982. [PMID: 40328081 DOI: 10.1016/j.bbamcr.2025.119982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 03/29/2025] [Accepted: 04/10/2025] [Indexed: 05/08/2025]
Abstract
Glioblastoma Multiforme (GBM) is a highly malignant and diffusely invasive WHO Grade IV brain tumor arising from glial and neural stem cells. GBM is characterized by rapid proliferation and migration, aggressive invasion of local brain parenchyma, a hypoxic microenvironment, resistance to apoptosis and high vascular remodeling and angiogenesis. These hallmarks contribute to a near universal tumor recurrence after treatment or resection and poor patient prognosis. Ion channels, a superfamily of proteins responsible for permitting ion flux across otherwise impermeant membranes, show extensive remodeling in GBM with aberrant function mechanistically linked to manipulation of each of these hallmarks. In this review, we will discuss the known links between ion channel expression and activity and cellular processes that are enhanced or perturbed during GBM formation or progression. We will also discuss the extent to which basic or translational findings on ion channels in GBM samples or cell lines have shown preclinical promise towards the development of improved therapeutics against GBMs.
Collapse
Affiliation(s)
- Gauri C Kulkarni
- Department of Anatomy and Cell Biology, University of Illinois Chicago, Chicago, IL, USA
| | - Rayna Saha
- Department of Anatomy and Cell Biology, University of Illinois Chicago, Chicago, IL, USA
| | - Christian J Peters
- Department of Anatomy and Cell Biology, University of Illinois Chicago, Chicago, IL, USA.
| |
Collapse
|
2
|
Chen R, Pan C, Mao X, Zhang Y, Chen G, Xu M, Nivar J, Tao Y, Cao H, Li J. Chloride intracellular channel 4 blockade improves cognition in mice with Alzheimer's disease: CLIC4 protein expression and tau protein hyperphosphorylation. Int J Biol Macromol 2024; 278:134972. [PMID: 39181373 DOI: 10.1016/j.ijbiomac.2024.134972] [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: 06/28/2024] [Revised: 08/12/2024] [Accepted: 08/20/2024] [Indexed: 08/27/2024]
Abstract
Numerous academic literature suggests that amyloid-β (Aβ) deposition, tau protein phosphorylation, and irreversible neuronal death are the three major causes of AD. The chloride intracellular channel (CLIC) protein family not only regulates the polarisation of neurons, but also has important implications for neuronal survival. Chloride intracellular channel 4 (CLIC4) can be pathologically activated by cyclin-dependent kinase 5 (Cdk5), which causes a significant increase in the expression of CLIC4 and mediates neuronal apoptosis. CLIC4 knockdown inhibits H2O2-induced neuronal apoptosis; however, the relationship between CLIC4 and AD remains unknown. In the present study, we showed that CLIC4 expression was elevated in the hippocampus of AD mice; knockdown of hippocampal CLIC4 alleviated Aβ25-35-induced cognitive impairment in mice; overexpression of hippocampal CLIC4 accelerated Aβ deposition and tau protein hyperphosphorylation in young AD mice (APP/PS1 mice at three months of age). CLIC4 overexpressing mice had a longer escape latency compared to controls in behavioural testing (Morris water maze and T-maze tests). By Co-immunoprecipitation/mass spectrometry (Co-IP/MS) of HT22 cells to identify proteins that specifically bind to CLIC4, we found interactions with CCAAT enhancer binding protein (C/EBPβ); a critical pathway involved in the development of various neurodegenerative diseases. In addition, the knockdown of hippocampal CLIC4 alleviated AD-like pathology by inhibiting the C/EBPβ/AEP signaling pathway. These data suggest an essential role for high CLIC4 expression in the pathophysiology of AD and reveal that inhibition of CLIC4 expression may provide an opportunity for treatment.
Collapse
Affiliation(s)
- Rui Chen
- The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Department of Anesthesiology, Wenzhou 325000, Zhejiang Province, China; The Second Affiliated Hospital Zhejiang University School of Medicine, Department of Anesthesiology, Hangzhou 310000, Zhejiang Province, China
| | - Chi Pan
- The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Department of Anesthesiology, Wenzhou 325000, Zhejiang Province, China
| | - Xinyu Mao
- The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Department of Anesthesiology, Wenzhou 325000, Zhejiang Province, China
| | - Yantong Zhang
- The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Department of Anesthesiology, Wenzhou 325000, Zhejiang Province, China
| | - Gang Chen
- The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Department of Anesthesiology, Wenzhou 325000, Zhejiang Province, China
| | - Mengting Xu
- The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Department of Anesthesiology, Wenzhou 325000, Zhejiang Province, China
| | - John Nivar
- Rutgers New Jersey Medical School, Department of Anesthesiology, Newark, NJ, USA
| | - Yuanxiang Tao
- Rutgers New Jersey Medical School, Department of Anesthesiology, Newark, NJ, USA
| | - Hong Cao
- The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Department of Anesthesiology, Wenzhou 325000, Zhejiang Province, China
| | - Jun Li
- The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Department of Anesthesiology, Wenzhou 325000, Zhejiang Province, China.
| |
Collapse
|
3
|
Yang L, Ying J, Tao Q, Zhang Q. RNA N 6-methyladenosine modifications in urological cancers: from mechanism to application. Nat Rev Urol 2024; 21:460-476. [PMID: 38347160 DOI: 10.1038/s41585-023-00851-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/16/2023] [Indexed: 08/04/2024]
Abstract
The N6-methyladenosine (m6A) modification is the most common modification of messenger RNAs in eukaryotes and has crucial roles in multiple cancers, including in urological malignancies such as renal cell carcinoma, bladder cancer and prostate cancer. The m6A RNA modification is controlled by three types of regulators, including methyltransferases (writers), demethylases (erasers) and RNA-binding proteins (readers), which are responsible for gene regulation at the post-transcriptional level. This Review summarizes the current evidence indicating that aberrant or dysregulated m6A modification is associated with urological cancer development, progression and prognosis. The complex and context-dependent effects of dysregulated m6A modifications in urological cancers are described, along with the potential for aberrantly expressed m6A regulators to provide valuable diagnostic and prognostic biomarkers as well as new therapeutic targets.
Collapse
Affiliation(s)
- Lei Yang
- Department of Urology, Peking University First Hospital, Institute of Urology, National Research Center for Genitourinary Oncology, Peking University, Beijing, China
| | - Jianming Ying
- Department of Pathology, Cancer Institute and Cancer Hospital, Peking Union Medical College (PUMC), Chinese Academy of Medical Sciences, Beijing, China
| | - Qian Tao
- Cancer Epigenetics Laboratory, Department of Clinical Oncology, State Key Laboratory of Translational Oncology, Sir Y.K. Pao Center for Cancer and Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Qian Zhang
- Department of Urology, Peking University First Hospital, Institute of Urology, National Research Center for Genitourinary Oncology, Peking University, Beijing, China.
- Department of Urology, Peking University Binhai Hospital, Tianjin, 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
|
Blondeau-Bidet E, Tine M, Gonzalez AA, Guinand B, Lorin-Nebel C. Coping with salinity extremes: Gill transcriptome profiling in the black-chinned tilapia (Sarotherodon melanotheron). THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 929:172620. [PMID: 38642748 DOI: 10.1016/j.scitotenv.2024.172620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 03/21/2024] [Accepted: 04/17/2024] [Indexed: 04/22/2024]
Abstract
Steeper and sometimes extreme salinity gradients increasingly affect aquatic organisms because of climate change. Hypersalinity habitats demand powerful physiological adaptive strategies. Few teleost species have the capacity to spend their whole life cycle in salinities way over seawater levels. Focusing on the multifunctional gill, we unraveled the tilapia S. melanotheron key strategies to cope with different environmental conditions, ranging from freshwater up to hypersaline habitats. De novo transcriptome assembly based on RNAseq allowed for the analysis of 40,967 annotated transcripts among samples collected in three wild populations at 0, 40 and 80 ‰. A trend analysis of the expression patterns revealed responses across the salinity gradient with different gene pathways involved. Genes linked to ion transport, pH regulation and cell surface receptor signaling were mainly upregulated in the high salinity habitat. We identified tight junction proteins that were critical in high salinity habitats and that were different from the well-known tightening junctional proteins identified and expressed in fresh water. Expression profiles also suggest a change in the vascular tone that could be linked to an osmorespiratory compromise not only in fresh water, but also in high salinity environments. A striking downregulation of genes linked to the immune system and to the heat shock response was observed suggesting an energetic trade-off between immunity and acclimation/adaptation in the hypersaline habitat. The high expression of transcripts coding for immune and heat shock response in the freshwater habitat suggests the establishment of powerful mechanisms to protect gills from environmental threats and to maintain protein integrity. Non-directional expression trends were also detected with an upregulation of genes only in the hypersaline habitat (80 ‰) or only in the marine habitat (40 ‰). Unravel physiological strategies in S. melanotheron populations will help to better understand the molecular basis of fish euryhalinity in salinity-contrasted environments.
Collapse
Affiliation(s)
| | - Mbaye Tine
- UFR of Agricultural Sciences, Aquaculture and Food Technologies (UFR S2ATA), Gaston Berger University, Saint-Louis, Senegal
| | | | - Bruno Guinand
- ISEM, Univ Montpellier, CNRS, IRD, Montpellier, France
| | | |
Collapse
|
6
|
Jin T, Wang H, Liu Y, Wang H. Circular RNAs: Regulators of endothelial cell dysfunction in atherosclerosis. J Mol Med (Berl) 2024; 102:313-335. [PMID: 38265445 DOI: 10.1007/s00109-023-02413-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 12/09/2023] [Accepted: 12/21/2023] [Indexed: 01/25/2024]
Abstract
Endothelial cell (EC) dysfunction is associated with atherosclerosis. Circular RNAs (circRNAs) are covalently closed loops formed by back-splicing, are highly expressed in a tissue-specific or cell-specific manner, and regulate ECs mainly through miRNAs (mircoRNAs) or protein sponges. This review describes the regulatory mechanisms and physiological functions of circRNAs, as well as the differential expression of circRNAs in aberrant ECs. This review focuses on their roles in inflammation, proliferation, migration, angiogenesis, apoptosis, senescence, and autophagy in ECs from the perspective of signaling pathways, such as nuclear factor κB (NF-κB), nucleotide-binding domain, leucine-rich-repeat family, pyrin-domain-containing 3 (NLRP3)/caspase-1, Janus kinase/signal transducer and activator of transcription (JAK/STAT), and phosphoinositide-3 kinase/protein kinase B (PI3K/Akt). Finally, we address the issues and recent advances in circRNAs as well as circRNA-mediated regulation of ECs to improve our understanding of the molecular mechanisms underlying the progression of atherosclerosis and provide a reference for studies on circRNAs that regulate EC dysfunction and thus affect atherosclerosis.
Collapse
Affiliation(s)
- Tengyu Jin
- Hebei Medical University, Shijiazhuang 050011, Hebei, China
- Hebei General Hospital, Affiliated to Hebei Medical University, Shijiazhuang 050051, Hebei, China
| | - Haoyuan Wang
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China
| | - Yuelin Liu
- Hebei Medical University, Shijiazhuang 050011, Hebei, China
| | - Hebo Wang
- Hebei Medical University, Shijiazhuang 050011, Hebei, China.
- Hebei General Hospital, Affiliated to Hebei Medical University, Shijiazhuang 050051, Hebei, China.
- Hebei Provincial Key Laboratory of Cerebral Networks and Cognitive Disorders, Shijiazhuang 050051, Hebei, China.
| |
Collapse
|
7
|
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
|
8
|
Zhang L, Chen X, Yao S, Zheng L, Yang X, Wang Y, Li X, Wu E, Tuo B. Intracellular chloride channel 1 and tumor. Am J Cancer Res 2023; 13:3300-3314. [PMID: 37693147 PMCID: PMC10492100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2023] [Accepted: 07/08/2023] [Indexed: 09/12/2023] Open
Abstract
As the major intracellular anion, chloride plays an important role in maintaining intracellular and extracellular ion homeostasis, osmotic pressure, and cell volume. Intracellular chloride channel 1, which has the physiological role of forming membrane proteins in the lipid bilayer and playing ion channels, is a hot research topic in recent years. It has been found that CLIC1 does not only act as an ion channel but also participates in cell cycle regulation, apoptosis, and intracellular oxidation; thus, it participates in the proliferation, invasion, and migration of various tumor cells in various systems throughout the body. At the same time, CLIC1 is highly expressed in tumor cells and is associated with malignancy and a poor prognosis. This paper reviews the pathological mechanisms of CLIC1 in systemic diseases, which is important for the early diagnosis, treatment, and prognosis of systemic diseases associated with CLIC1 expression.
Collapse
Affiliation(s)
- Li Zhang
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical UniversityZunyi, Guizhou, China
| | - Xingyue Chen
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical UniversityZunyi, Guizhou, China
| | - Shun Yao
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical UniversityZunyi, Guizhou, China
| | - Liming Zheng
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical UniversityZunyi, Guizhou, China
| | - Xingyue Yang
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical UniversityZunyi, Guizhou, China
| | - Yongfeng Wang
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical UniversityZunyi, Guizhou, China
| | - Xin Li
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical UniversityZunyi, Guizhou, China
| | - Enqin Wu
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical UniversityZunyi, Guizhou, China
| | - Biguang Tuo
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical UniversityZunyi, Guizhou, China
- The Collaborative Innovation Center of Tissue Damage Repair and Regenerative Medicine of Zunyi Medical UniversityZunyi, Guizhou, China
| |
Collapse
|
9
|
Kadam A, Jadiya P, Tomar D. Post-translational modifications and protein quality control of mitochondrial channels and transporters. Front Cell Dev Biol 2023; 11:1196466. [PMID: 37601094 PMCID: PMC10434574 DOI: 10.3389/fcell.2023.1196466] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Accepted: 07/24/2023] [Indexed: 08/22/2023] Open
Abstract
Mitochondria play a critical role in energy metabolism and signal transduction, which is tightly regulated by proteins, metabolites, and ion fluxes. Metabolites and ion homeostasis are mainly mediated by channels and transporters present on mitochondrial membranes. Mitochondria comprise two distinct compartments, the outer mitochondrial membrane (OMM) and the inner mitochondrial membrane (IMM), which have differing permeabilities to ions and metabolites. The OMM is semipermeable due to the presence of non-selective molecular pores, while the IMM is highly selective and impermeable due to the presence of specialized channels and transporters which regulate ion and metabolite fluxes. These channels and transporters are modulated by various post-translational modifications (PTMs), including phosphorylation, oxidative modifications, ions, and metabolites binding, glycosylation, acetylation, and others. Additionally, the mitochondrial protein quality control (MPQC) system plays a crucial role in ensuring efficient molecular flux through the mitochondrial membranes by selectively removing mistargeted or defective proteins. Inefficient functioning of the transporters and channels in mitochondria can disrupt cellular homeostasis, leading to the onset of various pathological conditions. In this review, we provide a comprehensive overview of the current understanding of mitochondrial channels and transporters in terms of their functions, PTMs, and quality control mechanisms.
Collapse
Affiliation(s)
- Ashlesha Kadam
- Department of Internal Medicine, Section of Cardiovascular Medicine, Section of Molecular Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, United States
| | - Pooja Jadiya
- Department of Internal Medicine, Section of Gerontology and Geriatric Medicine, Section of Molecular Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, United States
| | - Dhanendra Tomar
- Department of Internal Medicine, Section of Cardiovascular Medicine, Section of Molecular Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, United States
| |
Collapse
|
10
|
Chiliquinga AJ, Acosta B, Ogonaga-Borja I, Villarruel-Melquiades F, de la Garza J, Gariglio P, Ocádiz-Delgado R, Ramírez A, Sánchez-Pérez Y, García-Cuellar CM, Bañuelos C, Camacho J. Ion Channels as Potential Tools for the Diagnosis, Prognosis, and Treatment of HPV-Associated Cancers. Cells 2023; 12:1376. [PMID: 37408210 PMCID: PMC10217072 DOI: 10.3390/cells12101376] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 04/19/2023] [Accepted: 05/05/2023] [Indexed: 07/07/2023] Open
Abstract
The human papilloma virus (HPV) group comprises approximately 200 genetic types that have a special affinity for epithelial tissues and can vary from producing benign symptoms to developing into complicated pathologies, such as cancer. The HPV replicative cycle affects various cellular and molecular processes, including DNA insertions and methylation and relevant pathways related to pRb and p53, as well as ion channel expression or function. Ion channels are responsible for the flow of ions across cell membranes and play very important roles in human physiology, including the regulation of ion homeostasis, electrical excitability, and cell signaling. However, when ion channel function or expression is altered, the channels can trigger a wide range of channelopathies, including cancer. In consequence, the up- or down-regulation of ion channels in cancer makes them attractive molecular markers for the diagnosis, prognosis, and treatment of the disease. Interestingly, the activity or expression of several ion channels is dysregulated in HPV-associated cancers. Here, we review the status of ion channels and their regulation in HPV-associated cancers and discuss the potential molecular mechanisms involved. Understanding the dynamics of ion channels in these cancers should help to improve early diagnosis, prognosis, and treatment in the benefit of HPV-associated cancer patients.
Collapse
Affiliation(s)
| | - Brenda Acosta
- Grupo de Investigación de Ciencias en Red, Universidad Técnica del Norte, Ibarra 100105, Ecuador
- Departamento de Farmacología, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Ciudad de Mexico CP 07360, Mexico
| | - Ingrid Ogonaga-Borja
- Grupo de Investigación de Ciencias en Red, Universidad Técnica del Norte, Ibarra 100105, Ecuador
- Departamento de Farmacología, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Ciudad de Mexico CP 07360, Mexico
| | - Fernanda Villarruel-Melquiades
- Departamento de Farmacología, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Ciudad de Mexico CP 07360, Mexico
| | - Jaime de la Garza
- Unidad de Oncología Torácica y Laboratorio de Medicina Personalizada, Instituto Nacional de Cancerología (INCan), Tlalpan, Ciudad de Mexico CP 14080, Mexico
| | - Patricio Gariglio
- Departamento de Genética y Biología Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Ciudad de Mexico CP 07360, Mexico
| | - Rodolfo Ocádiz-Delgado
- Departamento de Genética y Biología Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Ciudad de Mexico CP 07360, Mexico
| | - Ana Ramírez
- Facultad de Ciencias Químicas e Ingeniería, Universidad Autónoma de Baja California, Calzada Universidad 14418, Tijuana 22390, Mexico
| | - Yesennia Sánchez-Pérez
- Subdirección de Investigación Básica, Instituto Nacional de Cancerología (INCan), Tlalpan, Ciudad de Mexico CP 14080, Mexico
| | - Claudia M. García-Cuellar
- Subdirección de Investigación Básica, Instituto Nacional de Cancerología (INCan), Tlalpan, Ciudad de Mexico CP 14080, Mexico
| | - Cecilia Bañuelos
- Programa Transdisciplinario en Desarrollo Científico y Tecnológico para la Sociedad, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Ciudad de Mexico CP 07360, Mexico
| | - Javier Camacho
- Departamento de Farmacología, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Ciudad de Mexico CP 07360, Mexico
| |
Collapse
|
11
|
Wang Y, Geng H, Li X, Chen P, Xu S, Zhang S, Weng P, Guo J, Huang M, Wu Y, Chen Y. A novel nomogram for predicting overall survival in peripheral T cell lymphoma patients.. [DOI: 10.21203/rs.3.rs-2823604/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/28/2023]
Abstract
Abstract
Background The prognosis of peripheral T cell lymphomas (PTCLs) varies greatly. This study aimed at generating a prognostic nomogram based on differentially expressed genes (DEGs).Methods Firstly, we collected RNA transcripts from Gene Expression Omnibus and identified DEGs. Secondly we used univariate Cox regression, Least absolute shrinkage and selection operator (LASSO) to screen the independent risk factors to construct nomogram in the training cohort. Thirdly, we evaluate its prediction accuracy via decision curves analysis (DCA), receiver operating characteristic (ROC) and calibration rate to confirm its performance on survival in training and validation cohort. Then we carried out subgroup analysis in training and validation to eliminate the effects of age, gender, and pathological subtype. Lastly, to verify feasibility of nomogram in practice, we applied immunohistochemistry to clinical samples and analyzed the relationship between IHC scores and prognosis.Results The 702 DEGs between 40 PTCLs and 20 non-tumor patients were identified. Then ANGPTL2, CPSF4, CLIC4 and OTUD6B were screened out as independent risk factors via univariate Cox regression and LASSO. The DCA, ROC, Harrell’s concordance index (c-index) and calibration rate showed nomogram predicting more accurately than any single specific transcript. The results showed PTCLs with higher nomogram-score had a longer survival, regardless of age, gender and pathological subtype. Finally, the high expression level of ANGPTL2, CPSF4 and OTUD6B related to poor prognosis. Higher expression of CLIC4 related to longer survival.Conclusion This nomogram showed the favorable clinical applicability, regardless of age, gender and pathological subtype.
Collapse
Affiliation(s)
- Yi-Ting Wang
- Fujian Institute of Hematology, Fujian Medical University Union Hospital
| | - Hai-Li Geng
- Fujian Institute of Hematology, Fujian Medical University Union Hospital
| | - Xiao-Fan Li
- Fujian Institute of Hematology, Fujian Medical University Union Hospital
| | - Ping Chen
- Fujian Institute of Hematology, Fujian Medical University Union Hospital
| | - Shu-Juan Xu
- Fujian Institute of Hematology, Fujian Medical University Union Hospital
| | - Shu-Xia Zhang
- Fujian Institute of Hematology, Fujian Medical University Union Hospital
| | - Ping Weng
- Fujian Institute of Hematology, Fujian Medical University Union Hospital
| | - Jiang-Rui Guo
- Fujian Institute of Hematology, Fujian Medical University Union Hospital
| | - Mei-Juan Huang
- Fujian Institute of Hematology, Fujian Medical University Union Hospital
| | - Yong Wu
- Fujian Institute of Hematology, Fujian Medical University Union Hospital
| | - Yuan-Zhong Chen
- Fujian Institute of Hematology, Fujian Medical University Union Hospital
| |
Collapse
|
12
|
Schwab K, Coronel L, Riege K, Sacramento EK, Rahnis N, Häckes D, Cirri E, Groth M, Hoffmann S, Fischer M. Multi-omics analysis identifies RFX7 targets involved in tumor suppression and neuronal processes. Cell Death Discov 2023; 9:80. [PMID: 36864036 PMCID: PMC9981735 DOI: 10.1038/s41420-023-01378-1] [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: 01/23/2023] [Revised: 02/19/2023] [Accepted: 02/22/2023] [Indexed: 03/04/2023] Open
Abstract
Recurrently mutated in lymphoid neoplasms, the transcription factor RFX7 is emerging as a tumor suppressor. Previous reports suggested that RFX7 may also have a role in neurological and metabolic disorders. We recently reported that RFX7 responds to p53 signaling and cellular stress. Furthermore, we found RFX7 target genes to be dysregulated in numerous cancer types also beyond the hematological system. However, our understanding of RFX7's target gene network and its role in health and disease remains limited. Here, we generated RFX7 knock-out cells and employed a multi-omics approach integrating transcriptome, cistrome, and proteome data to obtain a more comprehensive picture of RFX7 targets. We identify novel target genes linked to RFX7's tumor suppressor function and underscoring its potential role in neurological disorders. Importantly, our data reveal RFX7 as a mechanistic link that enables the activation of these genes in response to p53 signaling.
Collapse
Affiliation(s)
- Katjana Schwab
- grid.418245.e0000 0000 9999 5706Computational Biology Group, Leibniz Institute on Aging—Fritz Lipmann Institute (FLI), Beutenbergstraße 11, 07745 Jena, Germany
| | - Luis Coronel
- grid.418245.e0000 0000 9999 5706Computational Biology Group, Leibniz Institute on Aging—Fritz Lipmann Institute (FLI), Beutenbergstraße 11, 07745 Jena, Germany
| | - Konstantin Riege
- grid.418245.e0000 0000 9999 5706Computational Biology Group, Leibniz Institute on Aging—Fritz Lipmann Institute (FLI), Beutenbergstraße 11, 07745 Jena, Germany
| | - Erika K. Sacramento
- grid.418245.e0000 0000 9999 5706Core Facility for Proteomics, Leibniz Institute on Aging—Fritz Lipmann Institute (FLI), Beutenbergstraße 11, 07745 Jena, Germany
| | - Norman Rahnis
- grid.418245.e0000 0000 9999 5706Core Facility for Proteomics, Leibniz Institute on Aging—Fritz Lipmann Institute (FLI), Beutenbergstraße 11, 07745 Jena, Germany
| | - David Häckes
- grid.418245.e0000 0000 9999 5706Computational Biology Group, Leibniz Institute on Aging—Fritz Lipmann Institute (FLI), Beutenbergstraße 11, 07745 Jena, Germany
| | - Emilio Cirri
- grid.418245.e0000 0000 9999 5706Core Facility for Proteomics, Leibniz Institute on Aging—Fritz Lipmann Institute (FLI), Beutenbergstraße 11, 07745 Jena, Germany
| | - Marco Groth
- grid.418245.e0000 0000 9999 5706Core Facility for Next-Generation Sequencing, Leibniz Institute on Aging—Fritz Lipmann Institute (FLI), Beutenbergstraße 11, 07745 Jena, Germany
| | - Steve Hoffmann
- grid.418245.e0000 0000 9999 5706Computational Biology Group, Leibniz Institute on Aging—Fritz Lipmann Institute (FLI), Beutenbergstraße 11, 07745 Jena, Germany
| | - Martin Fischer
- Computational Biology Group, Leibniz Institute on Aging-Fritz Lipmann Institute (FLI), Beutenbergstraße 11, 07745, Jena, Germany.
| |
Collapse
|
13
|
Alzaydi MM, Abdul-Salam VB, Whitwell HJ, Russomanno G, Glynos A, Capece D, Szabadkai G, Wilkins MR, Wojciak-Stothard B. Intracellular Chloride Channels Regulate Endothelial Metabolic Reprogramming in Pulmonary Arterial Hypertension. Am J Respir Cell Mol Biol 2023; 68:103-115. [PMID: 36264759 PMCID: PMC9817916 DOI: 10.1165/rcmb.2022-0111oc] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Mitochondrial fission and a metabolic switch from oxidative phosphorylation to glycolysis are key features of vascular pathology in pulmonary arterial hypertension (PAH) and are associated with exuberant endothelial proliferation and apoptosis. The underlying mechanisms are poorly understood. We describe the contribution of two intracellular chloride channel proteins, CLIC1 and CLIC4, both highly expressed in PAH and cancer, to mitochondrial dysfunction and energy metabolism in PAH endothelium. Pathological overexpression of CLIC proteins induces mitochondrial fragmentation, inhibits mitochondrial cristae formation, and induces metabolic shift toward glycolysis in human pulmonary artery endothelial cells, consistent with changes observed in patient-derived cells. Interactions of CLIC proteins with structural components of the inner mitochondrial membrane offer mechanistic insights. Endothelial CLIC4 excision and mitofusin 2 supplementation have protective effects in human PAH cells and preclinical PAH. This study is the first to demonstrate the key role of endothelial intracellular chloride channels in the regulation of mitochondrial structure, biogenesis, and metabolic reprogramming in expression of the PAH phenotype.
Collapse
Affiliation(s)
- Mai M. Alzaydi
- National Heart and Lung Institute,,National Center for Biotechnology, King Abdulaziz City for Science and Technology (KACST), Riyadh, Saudi Arabia
| | - Vahitha B. Abdul-Salam
- National Heart and Lung Institute,,Centre for Cardiovascular Medicine and Device Innovation, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Harry J. Whitwell
- National Phenome Centre and Imperial Clinical Phenotyping Centre, and,Section of Bioanalytical Chemistry, Division of Systems Medicine, Department of Metabolism, Digestion, and Reproduction, and
| | - Giusy Russomanno
- National Heart and Lung Institute,,Medical Research Council (MRC) Centre for Drug Safety Science, Department of Pharmacology and Therapeutics, Institute of Systems, Molecular, and Integrative Biology, University of Liverpool, Liverpool, United Kingdom
| | - Angelos Glynos
- Mitochondrial Biology Unit, Medical Research Council, University of Cambridge, Cambridge, United Kingdom; and
| | - Daria Capece
- Centre for Cell Signalling and Inflammation, Department of Immunology and Inflammation, Imperial College London, London, United Kingdom
| | - Gyorgy Szabadkai
- Cell and Developmental Biology, University College London, London, United Kingdom
| | | | | |
Collapse
|
14
|
Bousseau S, Lahm T. Hungry for Chloride: Reprogramming Endothelial Cell Metabolism in Pulmonary Arterial Hypertension. Am J Respir Cell Mol Biol 2023; 68:11-12. [PMID: 36269721 PMCID: PMC9817906 DOI: 10.1165/rcmb.2022-0386ed] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Affiliation(s)
- Simon Bousseau
- Division of Pulmonary, Sleep, and Critical Care Medicine National Jewish Health Denver, Colorado
| | - Tim Lahm
- Division of Pulmonary, Sleep, and Critical Care Medicine National Jewish Health Denver, Colorado
- Division of Pulmonary Sciences and Critical Care Medicine University of Colorado Anschutz Medical Campus Aurora, Colorado
- Rocky Mountain Regional Veteran Affairs Medical Center Aurora, Colorado
| |
Collapse
|
15
|
Ponnalagu D, Hamilton S, Sanghvi S, Antelo D, Schwieterman N, Hansra I, Xu X, Gao E, Edwards JC, Bansal SS, Wold LE, Terentyev D, Janssen PML, Hund TJ, Khan M, Kohut AR, Koch WJ, Singh H. CLIC4 localizes to mitochondrial-associated membranes and mediates cardioprotection. SCIENCE ADVANCES 2022; 8:eabo1244. [PMID: 36269835 PMCID: PMC9586484 DOI: 10.1126/sciadv.abo1244] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 08/25/2022] [Indexed: 06/12/2023]
Abstract
Mitochondrial-associated membranes (MAMs) are known to modulate organellar and cellular functions and can subsequently affect pathophysiology including myocardial ischemia-reperfusion (IR) injury. Thus, identifying molecular targets in MAMs that regulate the outcome of IR injury will hold a key to efficient therapeutics. Here, we found chloride intracellular channel protein (CLIC4) presence in MAMs of cardiomyocytes and demonstrate its role in modulating ER and mitochondrial calcium homeostasis under physiological and pathological conditions. In a murine model, loss of CLIC4 increased myocardial infarction and substantially reduced cardiac function after IR injury. CLIC4 null cardiomyocytes showed increased apoptosis and mitochondrial dysfunction upon hypoxia-reoxygenation injury in comparison to wild-type cardiomyocytes. Overall, our results indicate that MAM-CLIC4 is a key mediator of cellular response to IR injury and therefore may have a potential implication on other pathophysiological processes.
Collapse
Affiliation(s)
- 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
| | - Shanna Hamilton
- 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
| | - Shridhar Sanghvi
- 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
| | - Diego Antelo
- 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
| | - Neill Schwieterman
- 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
- College of Nursing, The Ohio State University, Columbus, OH, USA
| | - Inderjot Hansra
- Department of Physiology and Cell Biology, The Ohio State University, Columbus, OH, USA
| | - Xianyao Xu
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, USA
- Departments of Biomedical Engineering and Internal Medicine, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, USA
| | - Erhe Gao
- Center for Translational Medicine, Department of Pharmacology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - John C. Edwards
- Nephrology Division, Department of Internal Medicine, St. Louis University, St. Louis, MO, USA
| | - Shyam S. Bansal
- 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
| | - Loren E. Wold
- 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
- College of Nursing, The Ohio State University, Columbus, OH, USA
| | - Dmitry Terentyev
- 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
| | - Paul M. L. Janssen
- 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
| | - Thomas J. Hund
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, USA
- Departments of Biomedical Engineering and Internal Medicine, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, USA
| | - Mahmood Khan
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, USA
- Department of Emergency Medicine, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Andrew R. Kohut
- Penn Heart and Vascular Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Walter J. Koch
- Center for Translational Medicine, Department of Pharmacology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, 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
| |
Collapse
|
16
|
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
|
17
|
Cdk5-p25 as a key element linking amyloid and tau pathologies in Alzheimer's disease: Mechanisms and possible therapeutic interventions. Life Sci 2022; 308:120986. [PMID: 36152679 DOI: 10.1016/j.lfs.2022.120986] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 09/15/2022] [Accepted: 09/19/2022] [Indexed: 11/24/2022]
Abstract
Despite the fact that the small atypical serine/threonine cyclin-dependent kinase 5 (Cdk5) is expressed in a number of tissues, its activity is restricted to the central nervous system due to the neuron-only localization of its activators p35 and p39. Although its importance for the proper development and function of the brain and its role as a switch between neuronal survival and death are unmistakable and unquestionable, Cdk5 is nevertheless increasingly emerging, as supported by a large number of publications on the subject, as a therapeutic target of choice in the fight against Alzheimer's disease. Thus, its aberrant over activation via the calpain-dependent conversion of p35 into p25 is observed during the pathogenesis of the disease where it leads to the hyperphosphorylation of the β-amyloid precursor protein and tau. The present review highlights the pivotal roles of the hyperactive Cdk5-p25 complex activity in contributing to the development of Alzheimer's disease pathogenesis, with a particular emphasis on the linking function between Aβ and tau that this kinase fulfils and on the fact that Cdk5-p25 is part of a deleterious feed forward loop giving rise to a molecular machinery runaway leading to AD pathogenesis. Additionally, we discuss the advances and challenges related to the possible strategies aimed at specifically inhibiting Cdk5-p25 activity and which could lead to promising anti-AD therapeutics.
Collapse
|
18
|
Al Khamici H, Sanchez VC, Yan H, Cataisson C, Michalowski AM, Yang HH, Li L, Lee MP, Huang J, Yuspa SH. The oxidoreductase CLIC4 is required to maintain mitochondrial function and resistance to exogenous oxidants in breast cancer cells. J Biol Chem 2022; 298:102275. [PMID: 35863434 PMCID: PMC9418444 DOI: 10.1016/j.jbc.2022.102275] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 06/01/2022] [Accepted: 06/02/2022] [Indexed: 02/07/2023] Open
Abstract
The chloride intracellular channel-4 (CLIC4) is one of the six highly conserved proteins in the CLIC family that share high structural homology with GST-omega in the GST superfamily. While CLIC4 is a multifunctional protein that resides in multiple cellular compartments, the discovery of its enzymatic glutaredoxin-like activity in vitro suggested that it could function as an antioxidant. Here, we found that deleting CLIC4 from murine 6DT1 breast tumor cells using CRISPR enhanced the accumulation of reactive oxygen species (ROS) and sensitized cells to apoptosis in response to H2O2 as a ROS-inducing agent. In intact cells, H2O2 increased the expression of both CLIC4 mRNA and protein. In addition, increased superoxide production in 6DT1 cells lacking CLIC4 was associated with mitochondrial hyperactivity including increased mitochondrial membrane potential and mitochondrial organelle enlargement. In the absence of CLIC4, however, H2O2-induced apoptosis was associated with low expression and degradation of the antiapoptotic mitochondrial protein Bcl2 and the negative regulator of mitochondrial ROS, UCP2. Furthermore, transcriptomic profiling of H2O2-treated control and CLIC4-null cells revealed upregulation of genes associated with ROS-induced apoptosis and downregulation of genes that sustain mitochondrial functions. Accordingly, tumors that formed from transplantation of CLIC4-deficient 6DT1 cells were highly necrotic. These results highlight a critical role for CLIC4 in maintaining redox-homeostasis and mitochondrial functions in 6DT1 cells. Our findings also raise the possibility of targeting CLIC4 to increase cancer cell sensitivity to chemotherapeutic drugs that are based on elevating ROS in cancer cells.
Collapse
Affiliation(s)
- Heba Al Khamici
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Center, National Institutes of Health. Bethesda, Maryland, USA
| | - Vanesa C Sanchez
- Office of Science, Division of Nonclinical Science, Center for Tobacco Products, U.S. Food and Drug Administration, Silver Spring, Maryland, USA
| | - Hualong Yan
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Center, National Institutes of Health. Bethesda, Maryland, USA
| | - Christophe Cataisson
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Center, National Institutes of Health. Bethesda, Maryland, USA
| | - Aleksandra M Michalowski
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Center, National Institutes of Health. Bethesda, Maryland, USA
| | - Howard H Yang
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Center, National Institutes of Health. Bethesda, Maryland, USA
| | - Luowei Li
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Center, National Institutes of Health. Bethesda, Maryland, USA
| | - Maxwell P Lee
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Center, National Institutes of Health. Bethesda, Maryland, USA
| | - Jing Huang
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Center, National Institutes of Health. Bethesda, Maryland, USA
| | - Stuart H Yuspa
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Center, National Institutes of Health. Bethesda, Maryland, USA.
| |
Collapse
|
19
|
Abrigo J, Olguín H, Gutierrez D, Tacchi F, Arrese M, Cabrera D, Valero-Breton M, Elorza AA, Simon F, Cabello-Verrugio C. Bile Acids Induce Alterations in Mitochondrial Function in Skeletal Muscle Fibers. Antioxidants (Basel) 2022; 11:antiox11091706. [PMID: 36139784 PMCID: PMC9495846 DOI: 10.3390/antiox11091706] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 08/25/2022] [Accepted: 08/27/2022] [Indexed: 11/16/2022] Open
Abstract
Cholestatic chronic liver disease is characterized by developing sarcopenia and elevated serum levels of bile acids. Sarcopenia is a skeletal muscle disorder with the hallmarks of muscle weakness, muscle mass loss, and muscle strength decline. Our previous report demonstrated that deoxycholic acid (DCA) and cholic acid (CA), through the membrane receptor TGR5, induce a sarcopenia-like phenotype in myotubes and muscle fibers. The present study aimed to evaluate the impact of DCA and CA on mitochondrial mass and function in muscle fibers and the role of the TGR5 receptor. To this end, muscle fibers obtained from wild-type and TGR5−/− mice were incubated with DCA and CA. Our results indicated that DCA and CA decreased mitochondrial mass, DNA, and potential in a TGR5-dependent fashion. Furthermore, with TGR5 participation, DCA and CA also reduced the oxygen consumption rate and complexes I and II from the mitochondrial electron transport chain. In addition, DCA and CA generated more mitochondrial reactive oxygen species than the control, which were abolished in TGR5−/− mice muscle fibers. Our results indicate that DCA and CA induce mitochondrial dysfunction in muscle fibers through a TGR5-dependent mechanism.
Collapse
Affiliation(s)
- Johanna Abrigo
- Laboratory of Muscle Pathology, Fragility and Aging, Faculty of Life Sciences, Universidad Andres Bello, Santiago 8370146, Chile
- Millennium Institute on Immunology and Immunotherapy, Faculty of Life Sciences, Universidad Andres Bello, Santiago 8370146, Chile
- Center for the Development of Nanoscience and Nanotechnology (CEDENNA), Universidad de Santiago de Chile, Santiago 8350709, Chile
| | - Hugo Olguín
- Laboratory of Tissue Repair and Adult Stem Cells, Department of Cellular and Molecular Biology, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago 8330077, Chile
| | - Danae Gutierrez
- Laboratory of Muscle Pathology, Fragility and Aging, Faculty of Life Sciences, Universidad Andres Bello, Santiago 8370146, Chile
- Millennium Institute on Immunology and Immunotherapy, Faculty of Life Sciences, Universidad Andres Bello, Santiago 8370146, Chile
- Center for the Development of Nanoscience and Nanotechnology (CEDENNA), Universidad de Santiago de Chile, Santiago 8350709, Chile
| | - Franco Tacchi
- Laboratory of Muscle Pathology, Fragility and Aging, Faculty of Life Sciences, Universidad Andres Bello, Santiago 8370146, Chile
- Millennium Institute on Immunology and Immunotherapy, Faculty of Life Sciences, Universidad Andres Bello, Santiago 8370146, Chile
- Center for the Development of Nanoscience and Nanotechnology (CEDENNA), Universidad de Santiago de Chile, Santiago 8350709, Chile
| | - Marco Arrese
- Departamento de Gastroenterología, Escuela de Medicina. Centro de Envejecimiento y Regeneración (CARE), Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago 8330077, Chile
| | - Daniel Cabrera
- Departamento de Gastroenterología, Escuela de Medicina. Centro de Envejecimiento y Regeneración (CARE), Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago 8330077, Chile
- Facultad de Ciencias Médicas, Universidad Bernardo O Higgins, Santiago 8370993, Chile
| | - Mayalen Valero-Breton
- Laboratory of Muscle Pathology, Fragility and Aging, Faculty of Life Sciences, Universidad Andres Bello, Santiago 8370146, Chile
- Millennium Institute on Immunology and Immunotherapy, Faculty of Life Sciences, Universidad Andres Bello, Santiago 8370146, Chile
- Center for the Development of Nanoscience and Nanotechnology (CEDENNA), Universidad de Santiago de Chile, Santiago 8350709, Chile
| | - Alvaro A. Elorza
- Millennium Institute on Immunology and Immunotherapy, Faculty of Life Sciences, Universidad Andres Bello, Santiago 8370146, Chile
- Institute of Biomedical Sciences, Faculty of Medicine and Life Sciences, Universidad Andres Bello, Santiago 8370146, Chile
| | - Felipe Simon
- Millennium Institute on Immunology and Immunotherapy, Faculty of Life Sciences, Universidad Andres Bello, Santiago 8370146, Chile
- Millennium Nucleus of Ion Channel-Associated Diseases (MiNICAD), Universidad de Chile, Santiago 8370146, Chile
- Laboratory of Integrative Physiopathology, Faculty of Life Sciences, Universidad Andres Bello, Santiago 8370146, Chile
- Correspondence: (F.S.); (C.C.-V.); Tel.: +56-227-703-665 (F.S. & C.C.-V.)
| | - Claudio Cabello-Verrugio
- Laboratory of Muscle Pathology, Fragility and Aging, Faculty of Life Sciences, Universidad Andres Bello, Santiago 8370146, Chile
- Millennium Institute on Immunology and Immunotherapy, Faculty of Life Sciences, Universidad Andres Bello, Santiago 8370146, Chile
- Center for the Development of Nanoscience and Nanotechnology (CEDENNA), Universidad de Santiago de Chile, Santiago 8350709, Chile
- Correspondence: (F.S.); (C.C.-V.); Tel.: +56-227-703-665 (F.S. & C.C.-V.)
| |
Collapse
|
20
|
N6-methyladenosine demethylase FTO suppressed prostate cancer progression by maintaining CLIC4 mRNA stability. Cell Death Dis 2022; 8:184. [PMID: 35397614 PMCID: PMC8994758 DOI: 10.1038/s41420-022-01003-7] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 01/29/2022] [Accepted: 02/09/2022] [Indexed: 12/13/2022]
Abstract
The fat mass and obesity-associated protein (FTO) is an N6-Methyladenosine (m6A) demethylase, which has been revealed to play critical roles in tumorigenesis. However, its role in the development and progression of prostate cancer (PCa) remains poorly understood. Here, we aimed to investigate the function and clinical relevance of FTO in PCa. Our results demonstrated that FTO was notably downregulated in PCa tissues compared with the paired normal tissues. In addition, the decreased expression of FTO was correlated with poor prognosis of PCa. Functional experiments showed that depletion of FTO promoted the proliferation and metastasis of PCa both in vitro and in vivo. Conversely, ectopic expression of FTO exhibited the opposite effects. Combined with RNA-sequencing, MeRIP-RT-qPCR, and mRNA stability assays indicated chloride intracellular channel 4(CLIC4) was a functional target of FTO-mediated m6A modification. FTO depletion significantly increased the m6A level of CLIC4 mRNA and then reduced the mRNA stability. In conclusion, our findings suggest that FTO suppresses PCa proliferation and metastasis through reducing the degradation of CLIC4 mRNA in an m6A dependent manner. FTO may be used as a promising novel therapeutic target and prognostic evaluation biomarker for PCa.
Collapse
|
21
|
Wasson CW, Caballero-Ruiz B, Gillespie J, Derrett-Smith E, Mankouri J, Denton CP, Canettieri G, Riobo-Del Galdo NA, Del Galdo F. Induction of Pro-Fibrotic CLIC4 in Dermal Fibroblasts by TGF-β/Wnt3a Is Mediated by GLI2 Upregulation. Cells 2022; 11:cells11030530. [PMID: 35159339 PMCID: PMC8834396 DOI: 10.3390/cells11030530] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2022] [Revised: 01/21/2022] [Accepted: 02/01/2022] [Indexed: 11/16/2022] Open
Abstract
Chloride intracellular channel 4 (CLIC4) is a recently discovered driver of fibroblast activation in Scleroderma (SSc) and cancer-associated fibroblasts (CAF). CLIC4 expression and activity are regulated by TGF-β signalling through the SMAD3 transcription factor. In view of the aberrant activation of canonical Wnt-3a and Hedgehog (Hh) signalling in fibrosis, we investigated their role in CLIC4 upregulation. Here, we show that TGF-β/SMAD3 co-operates with Wnt3a/β-catenin and Smoothened/GLI signalling to drive CLIC4 expression in normal dermal fibroblasts, and that the inhibition of β-catenin and GLI expression or activity abolishes TGF-β/SMAD3-dependent CLIC4 induction. We further show that the expression of the pro-fibrotic marker α-smooth muscle actin strongly correlates with CLIC4 expression in dermal fibroblasts. Further investigations revealed that the inhibition of CLIC4 reverses morphogen-dependent fibroblast activation. Our data highlights that CLIC4 is a common downstream target of TGF-β, Hh, and Wnt-3a through signalling crosstalk and we propose a potential therapeutic avenue using CLIC4 inhibitors
Collapse
Affiliation(s)
- Christopher W. Wasson
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, Faculty of Medicine and Health, University of Leeds, Leeds LS29JT, UK; (J.G.); (F.D.G.)
- Correspondence:
| | - Begoña Caballero-Ruiz
- School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds LS29JT, UK; (B.C.-R.); (J.M.); (N.A.R.-D.G.)
- Department of Molecular Medicine, Sapienza University of Rome, 00196 Rome, Italy;
| | - Justin Gillespie
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, Faculty of Medicine and Health, University of Leeds, Leeds LS29JT, UK; (J.G.); (F.D.G.)
| | - Emma Derrett-Smith
- Centre for Rheumatology and Connective Tissue Diseases, UCL Division of Medicine, London NW32PF, UK; (E.D.-S.); (C.P.D.)
| | - Jamel Mankouri
- School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds LS29JT, UK; (B.C.-R.); (J.M.); (N.A.R.-D.G.)
| | - Christopher P. Denton
- Centre for Rheumatology and Connective Tissue Diseases, UCL Division of Medicine, London NW32PF, UK; (E.D.-S.); (C.P.D.)
| | - Gianluca Canettieri
- Department of Molecular Medicine, Sapienza University of Rome, 00196 Rome, Italy;
| | - Natalia A. Riobo-Del Galdo
- School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds LS29JT, UK; (B.C.-R.); (J.M.); (N.A.R.-D.G.)
- Leeds Institute of Medical Research, Faculty of Medicine and Health, University of Leeds, Leeds LS29JT, UK
| | - Francesco Del Galdo
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, Faculty of Medicine and Health, University of Leeds, Leeds LS29JT, UK; (J.G.); (F.D.G.)
- Scleroderma Programme, NIHR Leeds Musculoskeletal Biomedical Research Centre, Leeds LS29JT, UK
| |
Collapse
|
22
|
Kouyoumdzian NM, Kim G, Rudi MJ, Rukavina Mikusic NL, Fernández BE, Choi MR. Clues and new evidences in arterial hypertension: unmasking the role of the chloride anion. Pflugers Arch 2022; 474:155-176. [PMID: 34966955 DOI: 10.1007/s00424-021-02649-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2021] [Revised: 12/02/2021] [Accepted: 12/03/2021] [Indexed: 02/06/2023]
Abstract
The present review will focus on the role of chloride anion in cardiovascular disease, with special emphasis in the development of hypertensive disease and vascular inflammation. It is known that acute and chronic overload of sodium chloride increase blood pressure and have pro-inflammatory and pro-fibrotic effects on different target organs, but it is unknown how chloride may influence these processes. Chloride anion is the predominant anion in the extracellular fluid and its intracellular concentration is dynamically regulated. As the queen of the electrolytes, it is of crucial importance to understand the physiological mechanisms that regulate the cellular handling of this anion including the different transporters and cellular chloride channels, which exert a variety of functions, such as regulation of cellular proliferation, differentiation, migration, apoptosis, intracellular pH and cellular redox state. In this article, we will also review the relationship between dietary, serum and intracellular chloride and how these different sources of chloride in the organism are affected in hypertension and their impact on cardiovascular disease. Additionally, we will discuss the approach of potential strategies that affect chloride handling and its potential effect on cardiovascular system, including pharmacological blockade of chloride channels and non-pharmacological interventions by replacing chloride by another anion.
Collapse
Affiliation(s)
- Nicolás Martín Kouyoumdzian
- Universidad de Buenos Aires, CONICET, Instituto Alberto C. Taquini de Investigaciones en Medicina Traslacional (IATIMET), Buenos Aires, Argentina.
| | - Gabriel Kim
- Facultad de Farmacia Y Bioquímica, Departamento de Ciencias Biológicas, Cátedra de Anatomía e Histología, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - María Julieta Rudi
- Facultad de Farmacia Y Bioquímica, Departamento de Ciencias Biológicas, Cátedra de Anatomía e Histología, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Natalia Lucía Rukavina Mikusic
- Facultad de Farmacia Y Bioquímica, Departamento de Ciencias Biológicas, Cátedra de Anatomía e Histología, Universidad de Buenos Aires, Buenos Aires, Argentina
| | | | - Marcelo Roberto Choi
- Universidad de Buenos Aires, CONICET, Instituto Alberto C. Taquini de Investigaciones en Medicina Traslacional (IATIMET), Buenos Aires, Argentina
- Facultad de Farmacia Y Bioquímica, Departamento de Ciencias Biológicas, Cátedra de Anatomía e Histología, Universidad de Buenos Aires, Buenos Aires, Argentina
- Instituto Universitario de Ciencias de La Salud, Fundación H.A. Barceló, Buenos Aires, Argentina
| |
Collapse
|
23
|
Bogenpohl JW, Weston RM, Foreman TN, Kitchen KE, Miles MF. Chloride intracellular channel 4 (CLIC4) expression profile in the mouse medial prefrontal cortex and its regulation by ethanol. Alcohol Clin Exp Res 2022; 46:29-39. [PMID: 34839533 PMCID: PMC8799520 DOI: 10.1111/acer.14754] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 11/12/2021] [Accepted: 11/22/2021] [Indexed: 01/03/2023]
Abstract
BACKGROUND Chloride intracellular channel 4 (CLIC4) is a multifunctional metamorphic protein for which a growing body of evidence supports a major role in the brain's molecular and behavioral responses to ethanol (EtOH). Although key to understanding the functional biology underlying this role, little is known about the cellular and subcellular expression patterns of CLIC4 in brain and how they are affected by EtOH. METHODS We used qRT-PCR to assess Clic4 mRNA expression in the medial prefrontal cortex (mPFC) of C57BL/6J mice in the absence and presence of acute EtOH exposure. Two complementary immunohistochemical techniques were employed to assess the subcellular localization of the CLIC4 protein and its pattern of expression across brain cell types in the mPFC in the absence and presence of acute EtOH. RESULTS Through immunohistochemical and stereological techniques, we show that CLIC4 protein is robustly expressed by oligodendrocytes (most abundant), microglia, and astrocytes, with minimal expression in neurons. Following acute EtOH exposure, we observed a rapid increase in Clic4 mRNA expression in female but not male mice and an overall increase in the number of oligodendrocytes and astrocytes expressing the CLIC4 protein. CONCLUSIONS These findings suggest that Clic4 functions as an early response gene for acute EtOH in brain, which likely underlies its ability to modulate EtOH behavior. Our results also suggest that the role of CLIC4 in the brain's response to EtOH is mediated through oligodendrocytes.
Collapse
Affiliation(s)
- James W. Bogenpohl
- Department of Molecular Biology and Chemistry, Christopher
Newport University, Newport News, VA, USA
| | - Rory M. Weston
- Department of Pharmacology and Toxicology, Virginia
Commonwealth University, Richmond, VA, USA
| | - Taylor N. Foreman
- Department of Molecular Biology and Chemistry, Christopher
Newport University, Newport News, VA, USA
| | - Kaitlyn E. Kitchen
- Department of Molecular Biology and Chemistry, Christopher
Newport University, Newport News, VA, USA
| | - Michael F. Miles
- Department of Pharmacology and Toxicology, Virginia
Commonwealth University, Richmond, VA, USA
- VCU Alcohol Research Center, Virginia Commonwealth
University, Richmond, VA, USA
| |
Collapse
|
24
|
Carotenuto P, Amato F, Lampis A, Rae C, Hedayat S, Previdi MC, Zito D, Raj M, Guzzardo V, Sclafani F, Lanese A, Parisi C, Vicentini C, Said-Huntingford I, Hahne JC, Hallsworth A, Kirkin V, Young K, Begum R, Wotherspoon A, Kouvelakis K, Azevedo SX, Michalarea V, Upstill-Goddard R, Rao S, Watkins D, Starling N, Sadanandam A, Chang DK, Biankin AV, Jamieson NB, Scarpa A, Cunningham D, Chau I, Workman P, Fassan M, Valeri N, Braconi C. Modulation of pancreatic cancer cell sensitivity to FOLFIRINOX through microRNA-mediated regulation of DNA damage. Nat Commun 2021; 12:6738. [PMID: 34795259 PMCID: PMC8602334 DOI: 10.1038/s41467-021-27099-6] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Accepted: 10/29/2021] [Indexed: 01/17/2023] Open
Abstract
FOLFIRINOX, a combination of chemotherapy drugs (Fluorouracil, Oxaliplatin, Irinotecan -FOI), provides the best clinical benefit in pancreatic ductal adenocarcinoma (PDAC) patients. In this study we explore the role of miRNAs (MIR) as modulators of chemosensitivity to identify potential biomarkers of response. We find that 41 and 84 microRNA inhibitors enhance the sensitivity of Capan1 and MiaPaCa2 PDAC cells respectively. These include a MIR1307-inhibitor that we validate in further PDAC cell lines. Chemotherapy-induced apoptosis and DNA damage accumulation are higher in MIR1307 knock-out (MIR1307KO) versus control PDAC cells, while re-expression of MIR1307 in MIR1307KO cells rescues these effects. We identify binding of MIR1307 to CLIC5 mRNA through covalent ligation of endogenous Argonaute-bound RNAs cross-linking immunoprecipitation assay. We validate these findings in an in vivo model with MIR1307 disruption. In a pilot cohort of PDAC patients undergoing FOLFIRONX chemotherapy, circulating MIR1307 correlates with clinical outcome.
Collapse
Affiliation(s)
- Pietro Carotenuto
- Division of Cancer Therapeutics, The Institute of Cancer Research, London, UK
- TIGEM - Telethon Institute of Genetics and Medicine, Naples, Italy
| | - Francesco Amato
- Institute of Cancer Sciences, University of Glasgow, Glasgow, UK
| | - Andrea Lampis
- Division of Molecular Pathology, The Institute of Cancer Research, London, UK
| | - Colin Rae
- Institute of Cancer Sciences, University of Glasgow, Glasgow, UK
| | - Somaieh Hedayat
- Division of Molecular Pathology, The Institute of Cancer Research, London, UK
| | - Maria C Previdi
- Division of Cancer Therapeutics, The Institute of Cancer Research, London, UK
| | - Domenico Zito
- Division of Molecular Pathology, The Institute of Cancer Research, London, UK
| | - Maya Raj
- Division of Cancer Therapeutics, The Institute of Cancer Research, London, UK
| | | | | | - Andrea Lanese
- The Royal Marsden NHS Trust, London and Surrey, London, UK
| | - Claudia Parisi
- The Royal Marsden NHS Trust, London and Surrey, London, UK
| | - Caterina Vicentini
- ARC-Net Research Centre and Department of Diagnostics and Public Health, Section of Pathology, , University of Verona, Verona, Italy
| | | | - Jens C Hahne
- Division of Molecular Pathology, The Institute of Cancer Research, London, UK
| | - Albert Hallsworth
- Division of Cancer Therapeutics, The Institute of Cancer Research, London, UK
| | - Vladimir Kirkin
- Division of Cancer Therapeutics, The Institute of Cancer Research, London, UK
| | - Kate Young
- The Royal Marsden NHS Trust, London and Surrey, London, UK
| | - Ruwaida Begum
- The Royal Marsden NHS Trust, London and Surrey, London, UK
| | | | | | | | | | | | - Sheela Rao
- The Royal Marsden NHS Trust, London and Surrey, London, UK
| | - David Watkins
- The Royal Marsden NHS Trust, London and Surrey, London, UK
| | | | - Anguraj Sadanandam
- Division of Molecular Pathology, The Institute of Cancer Research, London, UK
| | - David K Chang
- Institute of Cancer Sciences, University of Glasgow, Glasgow, UK
- West of Scotland Pancreatic Unit, Glasgow Royal Infirmary, Glasgow, UK
| | - Andrew V Biankin
- Institute of Cancer Sciences, University of Glasgow, Glasgow, UK
- West of Scotland Pancreatic Unit, Glasgow Royal Infirmary, Glasgow, UK
- South Western Sydney Clinical School, Faculty of Medicine, University of NSW, Sydney, NSW, Australia
| | - Nigel B Jamieson
- Institute of Cancer Sciences, University of Glasgow, Glasgow, UK
- West of Scotland Pancreatic Unit, Glasgow Royal Infirmary, Glasgow, UK
| | - Aldo Scarpa
- ARC-Net Research Centre and Department of Diagnostics and Public Health, Section of Pathology, , University of Verona, Verona, Italy
| | | | - Ian Chau
- The Royal Marsden NHS Trust, London and Surrey, London, UK
| | - Paul Workman
- Division of Cancer Therapeutics, The Institute of Cancer Research, London, UK
| | - Matteo Fassan
- Department of Medicine, University of Padua, Padua, Italy
- Veneto Institute of Oncology (IOV-IRCCS), Padua, Italy
| | - Nicola Valeri
- Division of Molecular Pathology, The Institute of Cancer Research, London, UK
- The Royal Marsden NHS Trust, London and Surrey, London, UK
| | - Chiara Braconi
- Division of Cancer Therapeutics, The Institute of Cancer Research, London, UK.
- Institute of Cancer Sciences, University of Glasgow, Glasgow, UK.
- The Royal Marsden NHS Trust, London and Surrey, London, UK.
- Beatson West of Scotland Cancer Centre, Glasgow, UK.
| |
Collapse
|
25
|
Chen Y, Li L, Zhang J, Cui H, Wang J, Wang C, Shi M, Fan H. Dexmedetomidine Alleviates Lipopolysaccharide-Induced Hippocampal Neuronal Apoptosis via Inhibiting the p38 MAPK/c-Myc/CLIC4 Signaling Pathway in Rats. Mol Neurobiol 2021; 58:5533-5547. [PMID: 34363182 DOI: 10.1007/s12035-021-02512-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Accepted: 08/02/2021] [Indexed: 12/11/2022]
Abstract
Dexmedetomidine (DEX) has multiple biological effects. Here, we investigated the neuroprotective role and molecular mechanism of DEX against lipopolysaccharide (LPS)-induced hippocampal neuronal apoptosis. Sprague Dawley rats were intraperitoneally injected with LPS (10 mg/kg) and/or DEX (30 µg/kg). We found that DEX improved LPS-induced alterations of hippocampal microstructure (necrosis and neuronal loss in the CA1 and CA3 regions) and ultrastructure (mitochondrial damage). DEX also attenuated LPS-induced inflammation and hippocampal apoptosis by inhibiting the increase of interleukin-1β, interleukin-6, interleukin-18, and tumor necrosis factor-α levels and downregulating the expression of mitochondrial apoptosis pathway-related proteins. Moreover, DEX prevented the LPS-induced activation of the c-Myc/chloride intracellular channel 4 (CLIC4) pathway. DEX inhibited the p38 MAPK pathway, but not JNK and ERK. To further clarify whether DEX alleviated LPS-induced neuronal apoptosis through the p38 MAPK/c-Myc/CLIC4 pathway, we treated PC12 cells with p38 MAPK inhibitor SB203582 (10 µM). DEX had the same effect as SB203582 in reducing the protein and mRNA expression of c-Myc and CLIC4. Furthermore, DEX and SB203582 diminished LPS-induced apoptosis, indicated by decreased Bax and Tom20 fluorescent double-stained cells, reduced annexin V-FITC/PI apoptosis rate, and reduced protein expression levels of Bax, cytochrome C, cleaved caspase-9, and cleaved caspase-3. Taken together, the findings indicate that DEX attenuates LPS-induced hippocampal neuronal apoptosis by regulating the p38 MAPK/c-Myc/CLIC4 signaling pathway. These findings provide new insights into the mechanism of Alzheimer's disease and depression and may help aid in drug development for these diseases.
Collapse
Affiliation(s)
- Yongping Chen
- College of Veterinary Medicine, Northeast Agricultural University, Heilongjiang Province, Harbin, 150030, People's Republic of China
| | - Lin Li
- College of Veterinary Medicine, Northeast Agricultural University, Heilongjiang Province, Harbin, 150030, People's Republic of China
| | - Jiuyan Zhang
- College of Veterinary Medicine, Northeast Agricultural University, Heilongjiang Province, Harbin, 150030, People's Republic of China
| | - Hailin Cui
- College of Veterinary Medicine, Northeast Agricultural University, Heilongjiang Province, Harbin, 150030, People's Republic of China
| | - Jiucheng Wang
- College of Veterinary Medicine, Northeast Agricultural University, Heilongjiang Province, Harbin, 150030, People's Republic of China
| | - Chuqiao Wang
- College of Veterinary Medicine, Northeast Agricultural University, Heilongjiang Province, Harbin, 150030, People's Republic of China
| | - Mingxian Shi
- College of Veterinary Medicine, Northeast Agricultural University, Heilongjiang Province, Harbin, 150030, People's Republic of China
| | - Honggang Fan
- College of Veterinary Medicine, Northeast Agricultural University, Heilongjiang Province, Harbin, 150030, People's Republic of China.
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, Northeast Agricultural University, Harbin, 150030, People's Republic of China.
| |
Collapse
|
26
|
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
|
27
|
Wang H, An J, He S, Liao C, Wang J, Tuo B. Chloride intracellular channels as novel biomarkers for digestive system tumors (Review). Mol Med Rep 2021; 24:630. [PMID: 34278487 DOI: 10.3892/mmr.2021.12269] [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: 01/17/2021] [Accepted: 05/19/2021] [Indexed: 11/06/2022] Open
Abstract
Digestive system malignant tumors are common tumors, and the traditional treatment methods for these tumors include surgical resection, radiotherapy, chemotherapy, and molecularly targeted drugs. However, diagnosis remains challenging, and the early detection of postoperative recurrence is complicated. Therefore, it is necessary to explore novel biomarkers to facilitate clinical diagnosis and treatment. Accumulating evidence supports the crucial role of chloride channels in the development of multiple types of cancers. Given that chloride channels are widely expressed and involved in cell proliferation, apoptosis and cell cycle, among other processes, they may serve as a promising diagnostic and therapeutic target. Chloride intracellular channels (CLICs) are a class of chloride channels that are upregulated or downregulated in certain types of cancer. Furthermore, in certain cases, during cell cycle progression, the localization and function of the cytosolic form of the transmembrane proteins of CLICs are also altered, which may provide a key target for cancer therapy. The aim of the present review was to focus on CLICs as biomarkers for digestive system tumors.
Collapse
Affiliation(s)
- Hui Wang
- Department of Gastroenterology, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, P.R. China
| | - Jiaxing An
- Department of Gastroenterology, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, P.R. China
| | - Suyu He
- The Fourth Department of the Digestive Disease Center, Suining Central Hospital, Suining, Sichuan 629000, P.R. China
| | - Chengcheng Liao
- Special Key Laboratory of Oral Disease Research, Higher Education Institution in Guizhou Province, School of Stomatology, Zunyi Medical University, Zunyi, Guizhou 563006, P.R. China
| | - Juan Wang
- Department of Gastroenterology, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, P.R. China
| | - Biguang Tuo
- Department of Gastroenterology, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, P.R. China
| |
Collapse
|
28
|
Weston RM, Schmitt RE, Grotewiel M, Miles MF. Transcriptome analysis of chloride intracellular channel knockdown in Drosophila identifies oxidation-reduction function as possible mechanism of altered sensitivity to ethanol sedation. PLoS One 2021; 16:e0246224. [PMID: 34228751 PMCID: PMC8259981 DOI: 10.1371/journal.pone.0246224] [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: 01/15/2021] [Accepted: 06/18/2021] [Indexed: 01/22/2023] Open
Abstract
Chloride intracellular channels (CLICs) are a unique family of evolutionarily conserved metamorphic proteins, switching between stable conformations based on redox conditions. CLICs have been implicated in a wide variety biological processes including ion channel activity, apoptosis, membrane trafficking, and enzymatic oxidoreductase activity. Understanding the molecular mechanisms by which CLICs engage in these activities is an area of active research. Here, the sole Drosophila melanogaster ortholog, Clic, was targeted for RNAi knockdown to identify genes and biological processes associated with Clic expression. Clic knockdown had a substantial impact on global transcription, altering expression of over 7% of transcribed Drosophila genes. Overrepresentation analysis of differentially expressed genes identified enrichment of Gene Ontology terms including Cytoplasmic Translation, Oxidation-Reduction Process, Heme Binding, Membrane, Cell Junction, and Nucleolus. The top term, Cytoplasmic Translation, was enriched almost exclusively with downregulated genes. Drosophila Clic and vertebrate ortholog Clic4 have previously been tied to ethanol sensitivity and ethanol-regulated expression. Clic knockdown-responsive genes from the present study were found to overlap significantly with gene sets from 4 independently published studies related to ethanol exposure and sensitivity in Drosophila. Bioinformatic analysis of genes shared between these studies revealed an enrichment of genes related to amino acid metabolism, protein processing, oxidation-reduction processes, and lipid particles among others. To determine whether the modulation of ethanol sensitivity by Clic may be related to co-regulated oxidation-reduction processes, we evaluated the effect of hyperoxia on ethanol sedation in Clic knockdown flies. Consistent with previous findings, Clic knockdown reduced acute ethanol sedation sensitivity in flies housed under normoxia. However, this effect was reversed by exposure to hyperoxia, suggesting a common set of molecular-genetic mechanism may modulate each of these processes. This study suggests that Drosophila Clic has a major influence on regulation of oxidative stress signaling and that this function overlaps with the molecular mechanisms of acute ethanol sensitivity in the fly.
Collapse
Affiliation(s)
- Rory M. Weston
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, Virginia, United States of America
- VCU Alcohol Research Center, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Rebecca E. Schmitt
- VCU Alcohol Research Center, Virginia Commonwealth University, Richmond, Virginia, United States of America
- Department of Human and Molecular Genetics, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Mike Grotewiel
- VCU Alcohol Research Center, Virginia Commonwealth University, Richmond, Virginia, United States of America
- Department of Human and Molecular Genetics, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Michael F. Miles
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, Virginia, United States of America
- VCU Alcohol Research Center, Virginia Commonwealth University, Richmond, Virginia, United States of America
| |
Collapse
|
29
|
Marín-Garzón NA, Magalhães AFB, Schmidt PI, Serna M, Fonseca LFS, Salatta BM, Frezarim GB, Fernandes-Júnior GA, Bresolin T, Carvalheiro R, Albuquerque LG. Genome-wide scan reveals genomic regions and candidate genes underlying direct and maternal effects of preweaning calf mortality in Nellore cattle. Genomics 2021; 113:1386-1395. [PMID: 33716185 DOI: 10.1016/j.ygeno.2021.02.021] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 01/25/2021] [Accepted: 02/23/2021] [Indexed: 11/26/2022]
Abstract
We conducted analysis to estimate genetic parameters and to identify genomic regions and candidate genes affecting direct and maternal effects of preweaning calf mortality (PWM) in Nellore cattle. Phenotypic records of 67,196 animals, and 8443 genotypes for 410,936 SNPs were used. Analysis were performed through the weighted single-step GBLUP approach and considering a threshold animal model via Bayesian Inference. Direct and maternal heritability estimates were of 0.2143 ± 0.0348 and 0.0137 ± 0.0066, respectively. The top 10 genomic regions accounted for 13.61 and 14.23% of the direct and maternal additive genetic variances and harbored a total of 63 and 91 positional candidate genes, respectively. Two overlapping regions on BTA2 were identified for both direct and maternal effects. Candidate genes are involved in biological mechanisms i.e. embryogenesis, immune response, feto-maternal communication, circadian rhythm, hormone alterations, myometrium adaptation, and milk secretion, which are critical for the successful calf growth and survival during preweaning period.
Collapse
Affiliation(s)
- N A Marín-Garzón
- São Paulo State University (Unesp), College of Agricultural and Veterinarian Sciences, Via de Acesso Professor Paulo Donato Castelane Castellane S/N - Vila Industrial, 14884-900 Jaboticabal, SP, Brazil.
| | - A F B Magalhães
- São Paulo State University (Unesp), College of Agricultural and Veterinarian Sciences, Via de Acesso Professor Paulo Donato Castelane Castellane S/N - Vila Industrial, 14884-900 Jaboticabal, SP, Brazil
| | - P I Schmidt
- São Paulo State University (Unesp), College of Agricultural and Veterinarian Sciences, Via de Acesso Professor Paulo Donato Castelane Castellane S/N - Vila Industrial, 14884-900 Jaboticabal, SP, Brazil
| | - M Serna
- São Paulo State University (Unesp), College of Agricultural and Veterinarian Sciences, Via de Acesso Professor Paulo Donato Castelane Castellane S/N - Vila Industrial, 14884-900 Jaboticabal, SP, Brazil
| | - L F S Fonseca
- São Paulo State University (Unesp), College of Agricultural and Veterinarian Sciences, Via de Acesso Professor Paulo Donato Castelane Castellane S/N - Vila Industrial, 14884-900 Jaboticabal, SP, Brazil
| | - B M Salatta
- São Paulo State University (Unesp), College of Agricultural and Veterinarian Sciences, Via de Acesso Professor Paulo Donato Castelane Castellane S/N - Vila Industrial, 14884-900 Jaboticabal, SP, Brazil
| | - G B Frezarim
- São Paulo State University (Unesp), College of Agricultural and Veterinarian Sciences, Via de Acesso Professor Paulo Donato Castelane Castellane S/N - Vila Industrial, 14884-900 Jaboticabal, SP, Brazil
| | - G A Fernandes-Júnior
- São Paulo State University (Unesp), College of Agricultural and Veterinarian Sciences, Via de Acesso Professor Paulo Donato Castelane Castellane S/N - Vila Industrial, 14884-900 Jaboticabal, SP, Brazil
| | - T Bresolin
- São Paulo State University (Unesp), College of Agricultural and Veterinarian Sciences, Via de Acesso Professor Paulo Donato Castelane Castellane S/N - Vila Industrial, 14884-900 Jaboticabal, SP, Brazil
| | - R Carvalheiro
- São Paulo State University (Unesp), College of Agricultural and Veterinarian Sciences, Via de Acesso Professor Paulo Donato Castelane Castellane S/N - Vila Industrial, 14884-900 Jaboticabal, SP, Brazil
| | - L G Albuquerque
- São Paulo State University (Unesp), College of Agricultural and Veterinarian Sciences, Via de Acesso Professor Paulo Donato Castelane Castellane S/N - Vila Industrial, 14884-900 Jaboticabal, SP, Brazil; National Council for Science and Technological Development (CNPq), Brasília, Brazil
| |
Collapse
|
30
|
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
|
31
|
Capatina AL, Lagos D, Brackenbury WJ. Targeting Ion Channels for Cancer Treatment: Current Progress and Future Challenges. Rev Physiol Biochem Pharmacol 2020; 183:1-43. [PMID: 32865696 DOI: 10.1007/112_2020_46] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Ion channels are key regulators of cancer cell pathophysiology. They contribute to a variety of processes such as maintenance of cellular osmolarity and membrane potential, motility (via interactions with the cytoskeleton), invasion, signal transduction, transcriptional activity and cell cycle progression, leading to tumour progression and metastasis. Ion channels thus represent promising targets for cancer therapy. Ion channels are attractive targets because many of them are expressed at the plasma membrane and a broad range of existing inhibitors are already in clinical use for other indications. However, many of the ion channels identified in cancer cells are also active in healthy normal cells, so there is a risk that certain blockers may have off-target effects on normal physiological function. This review describes recent research advances into ion channel inhibitors as anticancer therapeutics. A growing body of evidence suggests that a range of existing and novel Na+, K+, Ca2+ and Cl- channel inhibitors may be effective for suppressing cancer cell proliferation, migration and invasion, as well as enhancing apoptosis, leading to suppression of tumour growth and metastasis, either alone or in combination with standard-of-care therapies. The majority of evidence to date is based on preclinical in vitro and in vivo studies, although there are several examples of ion channel-targeting strategies now reaching early phase clinical trials. Given the strong links between ion channel function and regulation of tumour growth, metastasis and chemotherapy resistance, it is likely that further work in this area will facilitate the development of new therapeutic approaches which will reach the clinic in the future.
Collapse
Affiliation(s)
| | - Dimitris Lagos
- Hull York Medical School, York, UK
- York Biomedical Research Institute, University of York, York, UK
| | - William J Brackenbury
- Department of Biology, University of York, York, UK.
- York Biomedical Research Institute, University of York, York, UK.
| |
Collapse
|
32
|
Böhme I, Schönherr R, Eberle J, Bosserhoff AK. Membrane Transporters and Channels in Melanoma. Rev Physiol Biochem Pharmacol 2020; 181:269-374. [PMID: 32737752 DOI: 10.1007/112_2020_17] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Recent research has revealed that ion channels and transporters can be important players in tumor development, progression, and therapy resistance in melanoma. For example, members of the ABC family were shown to support cancer stemness-like features in melanoma cells, while several members of the TRP channel family were reported to act as tumor suppressors.Also, many transporter proteins support tumor cell viability and thus suppress apoptosis induction by anticancer therapy. Due to the high number of ion channels and transporters and the resulting high complexity of the field, progress in understanding is often focused on single molecules and is in total rather slow. In this review, we aim at giving an overview about a broad subset of ion transporters, also illustrating some aspects of the field, which have not been addressed in detail in melanoma. In context with the other chapters in this special issue on "Transportome Malfunctions in the Cancer Spectrum," a comparison between melanoma and these tumors will be possible.
Collapse
Affiliation(s)
- Ines Böhme
- Institute of Biochemistry, Emil Fischer Center, Friedrich-Alexander-University of Erlangen-Nürnberg, Erlangen, Germany
| | - Roland Schönherr
- Institute of Biochemistry and Biophysics, Friedrich Schiller University Jena and Jena University Hospital, Jena, Germany
| | - Jürgen Eberle
- Department of Dermatology, Venerology and Allergology, Skin Cancer Center Charité, University Medical Center Charité, Berlin, Germany
| | - Anja Katrin Bosserhoff
- Institute of Biochemistry, Emil Fischer Center, Friedrich-Alexander-University of Erlangen-Nürnberg, Erlangen, Germany. .,Comprehensive Cancer Center (CCC) Erlangen-EMN, Erlangen, Germany.
| |
Collapse
|
33
|
Lima FJ, Lopes MLDDS, Barros CCDS, Nonaka CFW, Silveira ÉJDD. Modification in CLIC4 Expression is Associated with P53, TGF-β, TNF-α and Myofibroblasts in Lip Carcinogenesis. Braz Dent J 2020; 31:290-297. [PMID: 32667519 DOI: 10.1590/0103-6440202003104] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Accepted: 12/11/2019] [Indexed: 11/21/2022] Open
Abstract
Chloride intracellular channel-4 (CLIC4) is regulated by p53 and tumor necrosis factor-α (TNF-α), it is linked to the increase of transforming growth factor-β (TGF-β), and myofibroblastic differentiation in skin carcinogenesis. This study analyzed the immunoexpression of CLIC4, p53, TGF-β, TNF-α, and α-SMA in 50 actinic cheilitis (AC) and 50 lower lip squamous cell carcinoma (LLSCC). AC and LLSCC immunoexpression were categorized as score 1 (<5% positive cells), 2 (5-50%) or 3 (>50%). For CLIC4, nuclear and cytoplasmic immunostaining of epithelial cells was considered individually. For morphologic analysis, the World Health Organization criteria were used to epithelial dysplasia grade of ACs, and Bryne grading of malignancy system was applied for LLSCC. Higher nuclear CLIC4 (CLIC4n) and TGF-β were observed in ACs with low-risk of transformation, while cytoplasmic CLIC4 (CLIC4c), p53 and TNF-α were higher in the high-risk cases (p<0.05). In LLSCCs, CLIC4c was higher in cases with lymph node metastasis, advanced clinical stages, and histological high-grade malignancy. p53 expression was higher in high-grade LLSCCs, whereas TGF-β decreased as the clinical stage and morphological grade progressed (p<0.05). ACs showed an increased expression of CLIC4n and TGF-β, while CLIC4c and α-SMA were higher in LLSCCs (p<0.0001). Both lesions showed negative correlation between CLIC4n and CLIC4c, while in LLSCCs, negative correlation was also verified between CLIC4c and p53, as well as CLIC4c and TGF-β (p<0.05). Change of CLIC4 from the nucleus to cytoplasm and alterations in p53, TGF-β, TNF-α, and α-SMA expression are involved in lip carcinogenesis.
Collapse
Affiliation(s)
- Francisco Jadson Lima
- Department of Dentistry, Universidade Federal do Rio Grande do Norte, Natal, RN, Brazil
| | | | | | | | | |
Collapse
|
34
|
Yokoyama R, Kojima H, Takai R, Ohta T, Maeda H, Miyashita K, Mutoh M, Terasaki M. Effects of CLIC4 on Fucoxanthinol-Induced Apoptosis in Human Colorectal Cancer Cells. Nutr Cancer 2020; 73:889-898. [PMID: 33703973 DOI: 10.1080/01635581.2020.1779760] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Fucoxanthin is a marine xanthophyll found in edible brown algae, and a metabolite, fucoxanthinol (FxOH), possesses a potent apoptosis inducing effect in many cancer cells. Chloride intracellular channel 4 (CLIC4) is a member of the CLIC family that plays an important role in cancer development and apoptosis. However, the role of CLIC4 in FxOH-induced apoptosis is not well understood. In this study, we investigated whether CLIC4 affects the apoptotic properties of FxOH in human colorectal cancer (CRC) cells under FxOH treatment. Treating human CRC DLD-1 cells with 5.0 μmol/L FxOH significantly induced apoptosis. FxOH downregulated CLIC4, integrin β1, NHERF2 and pSmad2 (Ser465/467) by 0.6-, 0.7-, 0.7-, and 0.5-fold, respectively, compared with control cells without alteration of Rab35 expression. No colocalizing change was observed in CLIC4-related proteins in either control or FxOH-treated cells. CLIC4 knockdown suppressed cell growth and apoptosis. Interestingly, apoptosis induction by FxOH almost disappeared with CLIC4 knockdown. Our findings suggested that CLIC4 could be involved in FxOH-induced apoptosis in human CRC.
Collapse
Affiliation(s)
- Reo Yokoyama
- School of Pharmaceutical Sciences, Health Science University of Hokkaido, Ishikari-Tobetsu, Japan
| | - Hiroyuki Kojima
- School of Pharmaceutical Sciences, Health Science University of Hokkaido, Ishikari-Tobetsu, Japan
| | - Rie Takai
- Research Institute of Health Sciences, Health Science University of Hokkaido, Ishikari-Tobetsu, Japan
| | - Tohru Ohta
- Research Institute of Health Sciences, Health Science University of Hokkaido, Ishikari-Tobetsu, Japan
| | - Hayato Maeda
- Faculty of Agriculture and Life Science, Hirosaki University, Hirosaki, Aomori, Japan
| | - Kazuo Miyashita
- Laboratory of Biofunctional Material Chemistry, Division of Marine Bioscience, Graduate School of Fisheries Sciences, Hokkaido University, Hakodate, Hokkaido, Japan
| | - Michihiro Mutoh
- Epidemiology and Preventions Group, Center for Public Health Sciences, National Cancer Center, Chuo-ku, Tokyo, Japan
| | - Masaru Terasaki
- School of Pharmaceutical Sciences, Health Science University of Hokkaido, Ishikari-Tobetsu, Japan.,Cancer Prevention Laboratories, Health Sciences University of Hokkaido, Ishikari-Tobetsu, Hokkaido, Japan
| |
Collapse
|
35
|
Zhu L, Chen Q, Zhang L, Hu S, Zheng W, Wang C, Bai Y, Pan Y, Konishi T, Guan J, Shao C. CLIC4 regulates radioresistance of nasopharyngeal carcinoma by iNOS after γ-rays but not carbon ions irradiation. Am J Cancer Res 2020; 10:1400-1415. [PMID: 32509387 PMCID: PMC7269788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Accepted: 04/04/2020] [Indexed: 06/11/2023] Open
Abstract
Nasopharyngeal carcinoma (NPC) is a major health problem in the East and Southeast Asia, and the intensity modulated radiotherapy (IMRT) is the current preferred treatment method of NPC, but radioresistance-induced residual and recurrent tumors are the main cause of treatment failure. Till now, the mechanism of radioresistance and prognostic biomarkers of NPC are still unrevealed. In this study, we collected clinical NPC samples and established radioresistant NPC-R cell lines by irradiating NPC cells with fractionation doses of γ-rays. Using genechip assay between radioresistance and radiosensitive clinical samples and TMT assay between NPC and NPC-R cells, differential expressed genes were examined and the potential biomarker of radioresistance was screened. Immunohistochemical assay of NPC clinical specimens showed that CLIC4 was significantly up-regulated in radioresistance tumor tissues. In vitro studies confirmed that up-regulation of CLIC4 gene enhanced radioresistance in comparison with the alterations of intracellular oxidative metabolism of reactive oxygen species (ROS) and nitric oxide (NO) in an opposite way. Correspondingly, inhibition of CLIC4 sensitized NPC cells to irradiation and decreased nuclear translocation of iNOS and intracellular level of NO in NPC cells. Interestingly, the capacity for DNA repair had no difference between NPC and NPC-R cells. Moreover, because of great interests in using carbon ion irradiation to treat NPC effectively, we demonstrated that, after carbon ion irradiation, NPC-R and NPC cells had similar survival even under the status of up- or down-regulation of CLIC4. Conclusively, CLIC4 contributes to radioresistance of NPC to γ-rays but not carbon ions by regulating intracellular oxidative metabolism of nuclear translocation of iNOS.
Collapse
Affiliation(s)
- Lin Zhu
- Institute of Radiation Medicine, Shanghai Medical College, Fudan UniversityShanghai, China
| | - Qianping Chen
- Institute of Radiation Medicine, Shanghai Medical College, Fudan UniversityShanghai, China
| | - Longshan Zhang
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical UniversityGuangzhou, Guangdong, China
| | - Songling Hu
- Institute of Radiation Medicine, Shanghai Medical College, Fudan UniversityShanghai, China
| | - Wang Zheng
- Institute of Radiation Medicine, Shanghai Medical College, Fudan UniversityShanghai, China
| | - Chen Wang
- Institute of Radiation Medicine, Shanghai Medical College, Fudan UniversityShanghai, China
| | - Yang Bai
- Institute of Radiation Medicine, Shanghai Medical College, Fudan UniversityShanghai, China
| | - Yan Pan
- Institute of Radiation Medicine, Shanghai Medical College, Fudan UniversityShanghai, China
| | - Teruaki Konishi
- Single Cell Radiation Biology Group, Institute for Quantum Life Science, National Institutes for Quantum and Radiological Science and Technology (QST)Chiba, Japan
| | - Jian Guan
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical UniversityGuangzhou, Guangdong, China
| | - Chunlin Shao
- Institute of Radiation Medicine, Shanghai Medical College, Fudan UniversityShanghai, China
| |
Collapse
|
36
|
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
|
37
|
De Marchi U, Fernandez-Martinez S, de la Fuente S, Wiederkehr A, Santo-Domingo J. Mitochondrial ion channels in pancreatic β-cells: Novel pharmacological targets for the treatment of Type 2 diabetes. Br J Pharmacol 2020; 178:2077-2095. [PMID: 32056196 PMCID: PMC8246559 DOI: 10.1111/bph.15018] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Revised: 01/29/2020] [Accepted: 01/31/2020] [Indexed: 12/18/2022] Open
Abstract
Pancreatic beta‐cells are central regulators of glucose homeostasis. By tightly coupling nutrient sensing and granule exocytosis, beta‐cells adjust the secretion of insulin to the circulating blood glucose levels. Failure of beta‐cells to augment insulin secretion in insulin‐resistant individuals leads progressively to impaired glucose tolerance, Type 2 diabetes, and diabetes‐related diseases. Mitochondria play a crucial role in β‐cells during nutrient stimulation, linking the metabolism of glucose and other secretagogues to the generation of signals that promote insulin secretion. Mitochondria are double‐membrane organelles containing numerous channels allowing the transport of ions across both membranes. These channels regulate mitochondrial energy production, signalling, and cell death. The mitochondria of β‐cells express ion channels whose physio/pathological role is underappreciated. Here, we describe the mitochondrial ion channels identified in pancreatic β‐cells, we further discuss the possibility of targeting specific β‐cell mitochondrial channels for the treatment of Type 2 diabetes, and we finally highlight the evidence from clinical studies. LINKED ARTICLES This article is part of a themed issue on Cellular metabolism and diseases. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v178.10/issuetoc
Collapse
Affiliation(s)
| | - Silvia Fernandez-Martinez
- Division of Clinical Pharmacology and Toxicology, Centre de Recherche Clinique, HUG, Genève, Switzerland
| | - Sergio de la Fuente
- Center for Translational Medicine, Department of Medicine, Thomas Jefferson University, Philadelphia, Pennsylvania
| | | | | |
Collapse
|
38
|
Zhang X, Wang Z, Li W, Huang R, Zheng D, Bi G. MicroRNA-217-5p ameliorates endothelial cell apoptosis induced by ox-LDL by targeting CLIC4. Nutr Metab Cardiovasc Dis 2020; 30:523-533. [PMID: 31744714 DOI: 10.1016/j.numecd.2019.09.027] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Revised: 09/24/2019] [Accepted: 09/24/2019] [Indexed: 01/29/2023]
Abstract
BACKGROUND AND AIMS Endothelial cell apoptosis plays an essential role in the pathogenesis of atherosclerosis. MicroRNAs and chloride intracellular channels (CLICs) have been verified to participate in the endothelial cell apoptosis process, however, the underlying molecular mechanisms are still unclear. The main aim of this study was to investigate the biological effects of microRNA-217-5p (miR-217-5p) and CLIC4 on endothelial cell apoptosis in atherosclerosis. METHODS AND RESULTS An atherosclerotic mouse model (n = 18) was constructed by feeding apolipo protein E knockout ApoE(-/-) mice with high-fat diet for 12 weeks. An atherosclerotic cell model was established by treating human aortic endothelial cells with oxidized low-density lipoprotein (ox-LDL; 50 μg/mL) for 24 h. Quantitative real-time polymerase chain reaction and immunofluorescent staining confirmed the downregulation of miR-217-5p and upregulation of CLIC4 in atherosclerotic endothelial cells. Combined with western blot, flow cytometry assay and Hoechst staining, we demonstrated that miR-217-5p upregulation or CLIC4 knockdown regulated the apoptosis-related genes, ameliorated mitochondrial membrane permeability and therefore inhibited the apoptosis of aortic endothelial cells induced by ox-LDL. We further confirmed that miR-217-5p inhibited apoptosis of endothelial cells through targeting CLIC4 using luciferase report assay and rescue experiments. CONCLUSION We revealed for the first time that miR-217-5p inhibited apoptosis of endothelial cells in atherosclerosis and identified CLIC4 as a novel target of miR-217-5p. Our work provides a potential therapeutic approach for the treatment of atherosclerosis.
Collapse
Affiliation(s)
- Xiaotian Zhang
- Department of Neurology, Shengjing Hospital of China Medical University, 36 Sanhao Street, Shenyang 110004, People's Republic of China
| | - Zhimin Wang
- Department of Neurology, Shengjing Hospital of China Medical University, 36 Sanhao Street, Shenyang 110004, People's Republic of China
| | - Weishuai Li
- Department of Neurology, Shengjing Hospital of China Medical University, 36 Sanhao Street, Shenyang 110004, People's Republic of China
| | - Rui Huang
- Department of Neurology, Shengjing Hospital of China Medical University, 36 Sanhao Street, Shenyang 110004, People's Republic of China
| | - Dongming Zheng
- Department of Neurology, Shengjing Hospital of China Medical University, 36 Sanhao Street, Shenyang 110004, People's Republic of China
| | - Guorong Bi
- Department of Neurology, Shengjing Hospital of China Medical University, 36 Sanhao Street, Shenyang 110004, People's Republic of China.
| |
Collapse
|
39
|
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: 78] [Impact Index Per Article: 15.6] [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
|
40
|
Zajac M, Chakraborty K, Saha S, Mahadevan V, Infield DT, Accardi A, Qiu Z, Krishnan Y. What biologists want from their chloride reporters – a conversation between chemists and biologists. J Cell Sci 2020; 133:133/2/jcs240390. [DOI: 10.1242/jcs.240390] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
ABSTRACT
Impaired chloride transport affects diverse processes ranging from neuron excitability to water secretion, which underlie epilepsy and cystic fibrosis, respectively. The ability to image chloride fluxes with fluorescent probes has been essential for the investigation of the roles of chloride channels and transporters in health and disease. Therefore, developing effective fluorescent chloride reporters is critical to characterizing chloride transporters and discovering new ones. However, each chloride channel or transporter has a unique functional context that demands a suite of chloride probes with appropriate sensing characteristics. This Review seeks to juxtapose the biology of chloride transport with the chemistries underlying chloride sensors by exploring the various biological roles of chloride and highlighting the insights delivered by studies using chloride reporters. We then delineate the evolution of small-molecule sensors and genetically encoded chloride reporters. Finally, we analyze discussions with chloride biologists to identify the advantages and limitations of sensors in each biological context, as well as to recognize the key design challenges that must be overcome for developing the next generation of chloride sensors.
Collapse
Affiliation(s)
- Matthew Zajac
- Department of Chemistry, The University of Chicago, Chicago, IL 60637, USA
- Grossman Institute of Neuroscience, Quantitative Biology and Human Behavior, The University of Chicago, Chicago, IL 60637, USA
| | - Kasturi Chakraborty
- Department of Chemistry, The University of Chicago, Chicago, IL 60637, USA
- Grossman Institute of Neuroscience, Quantitative Biology and Human Behavior, The University of Chicago, Chicago, IL 60637, USA
- Ben May Department for Cancer Research, The University of Chicago, Chicago, IL 60637, USA
| | - Sonali Saha
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), 13125 Berlin, Germany
| | - Vivek Mahadevan
- Department of Cell & Systems Biology, University of Toronto, Toronto, ON M5S 3G5, Canada
| | - Daniel T. Infield
- Department of Molecular Physiology and Biophysics, The University of Iowa, Iowa City, IA 52242, USA
| | - Alessio Accardi
- Department of Anesthesiology, Weill Cornell Medical School, New York, NY 10065, USA
- Department of Physiology and Biophysics, Weill Cornell Medical School, New York, NY 10065, USA
- Department of Biochemistry, Weill Cornell Medical School, New York, NY 10065, USA
| | - Zhaozhu Qiu
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD 21218, USA
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21218, USA
| | - Yamuna Krishnan
- Department of Chemistry, The University of Chicago, Chicago, IL 60637, USA
- Grossman Institute of Neuroscience, Quantitative Biology and Human Behavior, The University of Chicago, Chicago, IL 60637, USA
| |
Collapse
|
41
|
Carofino BL, Dinshaw KM, Ho PY, Cataisson C, Michalowski AM, Ryscavage A, Alkhas A, Wong NW, Koparde V, Yuspa SH. Head and neck squamous cancer progression is marked by CLIC4 attenuation in tumor epithelium and reciprocal stromal upregulation of miR-142-3p, a novel post-transcriptional regulator of CLIC4. Oncotarget 2019; 10:7251-7275. [PMID: 31921386 PMCID: PMC6944452 DOI: 10.18632/oncotarget.27387] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Accepted: 12/02/2019] [Indexed: 02/06/2023] Open
Abstract
Chloride intracellular channel 4 (CLIC4) is a tumor suppressor implicated in processes including growth arrest, differentiation, and apoptosis. CLIC4 protein expression is diminished in the tumor parenchyma during progression in squamous cell carcinoma (SCC) and other neoplasms, but the underlying mechanisms have not been identified. Data from The Cancer Genome Atlas suggest this is not driven by genomic alterations. However, screening and functional assays identified miR-142-3p as a regulator of CLIC4. CLIC4 and miR-142-3p expression are inversely correlated in head and neck (HN) SCC and cervical SCC, particularly in advanced stage cancers. In situ localization revealed that stromal immune cells, not tumor cells, are the predominant source of miR-142-3p in HNSCC. Furthermore, HNSCC single-cell expression data demonstrated that CLIC4 is lower in tumor epithelial cells than in stromal fibroblasts and endothelial cells. Tumor-specific downregulation of CLIC4 was confirmed in an SCC xenograft model concurrent with immune cell infiltration and miR-142-3p upregulation. These findings provide the first evidence of CLIC4 regulation by miRNA. Furthermore, the distinct localization of CLIC4 and miR-142-3p within the HNSCC tumor milieu highlight the limitations of bulk tumor analysis and provide critical considerations for both future mechanistic studies and use of miR-142-3p as a HNSCC biomarker.
Collapse
Affiliation(s)
- Brandi L. Carofino
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Kayla M. Dinshaw
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
- Department of Molecular and Cellular Biology, University of California, Berkeley, Berkeley, CA, USA
| | - Pui Yan Ho
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
- Department of Pediatrics, Division of Stem Cell Transplantation and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Christophe Cataisson
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Aleksandra M. Michalowski
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Andrew Ryscavage
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | | | - Nathan W. Wong
- CCR Collaborative Bioinformatics Resource (CCBR), Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
- Advanced Biomedical Computational Science, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Vishal Koparde
- CCR Collaborative Bioinformatics Resource (CCBR), Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
- Advanced Biomedical Computational Science, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Stuart H. Yuspa
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| |
Collapse
|
42
|
Identification of two novel breast cancer loci through large-scale genome-wide association study in the Japanese population. Sci Rep 2019; 9:17332. [PMID: 31757997 PMCID: PMC6874604 DOI: 10.1038/s41598-019-53654-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Accepted: 10/26/2019] [Indexed: 12/21/2022] Open
Abstract
Genome-wide association studies (GWAS) have successfully identified about 70 genomic loci associated with breast cancer. Owing to the complexity of linkage disequilibrium and environmental exposures in different populations, it is essential to perform regional GWAS for better risk prediction. This study aimed to investigate the genetic architecture and to assess common genetic risk model of breast cancer with 6,669 breast cancer patients and 21,930 female controls in the Japanese population. This GWAS identified 11 genomic loci that surpass genome-wide significance threshold of P < 5.0 × 10−8 with nine previously reported loci and two novel loci that include rs9862599 on 3q13.11 (ALCAM) and rs75286142 on 21q22.12 (CLIC6-RUNX1). Validation study was carried out with 981 breast cancer cases and 1,394 controls from the Aichi Cancer Center. Pathway analyses of GWAS signals identified association of dopamine receptor medicated signaling and protein amino acid deacetylation with breast cancer. Weighted genetic risk score showed that individuals who were categorized in the highest risk group are approximately 3.7 times more likely to develop breast cancer compared to individuals in the lowest risk group. This well-powered GWAS is a representative study to identify SNPs that are associated with breast cancer in the Japanese population.
Collapse
|
43
|
Li B, Zhao Y, Song M, Cui H, Feng X, Yang T, Fan HG. Role of c-Myc/chloride intracellular channel 4 pathway in lipopolysaccharide-induced neurodegenerative diseases. Toxicology 2019; 429:152312. [PMID: 31693917 DOI: 10.1016/j.tox.2019.152312] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Revised: 09/21/2019] [Accepted: 10/16/2019] [Indexed: 12/21/2022]
Abstract
LPS-induced neuronal apoptosis leads to neurodegenerative diseases (NDs). However, the mechanisms underlying NDs pathogenesis remains unclear. The apoptotic response to activation of the c-Myc/chloride intracellular channel (CLIC4) pathway is directed through a mitochondrial pathway. In this study, we aimed to explore the c-Myc/CLIC4 pathway in the progression of NDs induced by lipopolysaccharide (LPS). In an in vivo experiment, the results of HE staining, transmission electron microscopic, immunofluorescence microscopy of cleaved caspase-3 and Bax and the increasing expression of apoptotic pathway related proteins in mitochondria showed that LPS (10 mg/kg) administration damaged mitochondrial and induced hippocampal neuron apoptosis. The Western blot and RT-PCR indicated that LPS induced the activation of c-Myc/CLIC4 pathway. Furthermore, in an in vitro experiment, PC12 cells were exposed to LPS to induce cell injuries to mimic the model of NDs. To further confirm the role of the c-Myc/CLIC4 pathway in LPS-induced neuronal apoptosis, the gene knockout of c-Myc and CLIC4 were performed by CRISPR/Cas9. The results of the flow cytometry assay and Annexin V-FITC/PI showed that knocking out c-Myc and CLIC4 significantly reduced cell apoptosis. The results of Western blot and dual immunofluorescence with Cyt c and TOM20 showed that knocking out c-Myc and CLIC4 significantly reduced the expression of mitochondrial apoptosis-related proteins. Our data confirmed that LPS-induced apoptosis is regulated by the activation of c-Myc/CLIC4 pathway. These results support further research mechanisms underlying neurodegenerative diseases and can provide effective pharmacodynamic targets for the clinical development of therapeutic drugs for neurodegenerative diseases.
Collapse
Affiliation(s)
- Bei Li
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, PR China
| | - Yuan Zhao
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, PR China
| | - ManYu Song
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, PR China
| | - HaiLin Cui
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, PR China
| | - XiuJing Feng
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, PR China
| | - TianYuan Yang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, PR China
| | - Hong-Gang Fan
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, PR China.
| |
Collapse
|
44
|
Saberbaghi T, Wong R, Rutka JT, Wang GL, Feng ZP, Sun HS. Role of Cl− channels in primary brain tumour. Cell Calcium 2019; 81:1-11. [DOI: 10.1016/j.ceca.2019.05.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Revised: 04/28/2019] [Accepted: 05/13/2019] [Indexed: 12/20/2022]
|
45
|
de Araujo CB, Heimann AS, Remer RA, Russo LC, Colquhoun A, Forti FL, Ferro ES. Intracellular Peptides in Cell Biology and Pharmacology. Biomolecules 2019; 9:biom9040150. [PMID: 30995799 PMCID: PMC6523763 DOI: 10.3390/biom9040150] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Revised: 04/02/2019] [Accepted: 04/12/2019] [Indexed: 12/11/2022] Open
Abstract
Intracellular peptides are produced by proteasomes following degradation of nuclear, cytosolic, and mitochondrial proteins, and can be further processed by additional peptidases generating a larger pool of peptides within cells. Thousands of intracellular peptides have been sequenced in plants, yeast, zebrafish, rodents, and in human cells and tissues. Relative levels of intracellular peptides undergo changes in human diseases and also when cells are stimulated, corroborating their biological function. However, only a few intracellular peptides have been pharmacologically characterized and their biological significance and mechanism of action remains elusive. Here, some historical and general aspects on intracellular peptides' biology and pharmacology are presented. Hemopressin and Pep19 are examples of intracellular peptides pharmacologically characterized as inverse agonists to cannabinoid type 1 G-protein coupled receptors (CB1R), and hemopressin fragment NFKF is shown herein to attenuate the symptoms of pilocarpine-induced epileptic seizures. Intracellular peptides EL28 (derived from proteasome 26S protease regulatory subunit 4; Rpt2), PepH (derived from Histone H2B type 1-H), and Pep5 (derived from G1/S-specific cyclin D2) are examples of peptides that function intracellularly. Intracellular peptides are suggested as biological functional molecules, and are also promising prototypes for new drug development.
Collapse
Affiliation(s)
- Christiane B de Araujo
- Special Laboratory of Cell Cycle, Center of Toxins, Immune Response and Cell Signaling - CeTICS, Butantan Institute, São Paulo SP 05503-900, Brazil.
| | | | | | - Lilian C Russo
- Department of Biochemistry, Chemistry Institute, University of São Paulo 1111, São Paulo 05508-000, Brazil.
| | - Alison Colquhoun
- Department of Cell and Developmental Biology, University of São Paulo (USP), São Paulo 05508-000, Brazil.
| | - Fábio L Forti
- Department of Biochemistry, Chemistry Institute, University of São Paulo 1111, São Paulo 05508-000, Brazil.
| | - Emer S Ferro
- Department of Pharmacology, Biomedical Sciences Institute, University of São Paulo (USP), São Paulo 05508-000, Brazil.
| |
Collapse
|
46
|
Yang X, Xu W, Huang K, Zhang B, Wang H, Zhang X, Gong L, Luo Y, He X. Precision toxicology shows that troxerutin alleviates ochratoxin A-induced renal lipotoxicity. FASEB J 2018; 33:2212-2227. [PMID: 30247986 DOI: 10.1096/fj.201800742r] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Lipotoxicity is the most common cause of severe kidney disease, with few treatment options available today. Precision toxicology can improve detection of subtle intracellular changes in response to exogenous substrates; thus, it facilitates in-depth research on bioactive molecules that may interfere with the onset of certain diseases. In the current study, troxerutin significantly relieved nephrotoxicity, increased endurance, and improved systemic energy metabolism and renal inflammation in OTA-induced nephrotic mice. Lipidomics showed that troxerutin effectively reduced the levels of triglycerides, phosphatidylcholines, and phosphatidylethanolamines in nephropathy. The mechanism was partly attributable to troxerutin in alleviating the aberrantly up-regulated expression of sphingomyelinase, the cystic fibrosis transmembrane conductance regulator, and chloride channel 2. Renal tubular epithelial cells, the main site of toxin-induced accumulation of lipids in the kidney, were subjected to transcriptomic profiling, which uncovered several metabolic factors relevant to aberrant lipid and lipoprotein metabolism. Our work provides new insights into the molecular features of toxin-induced lipotoxicity in renal tubular epithelial cells in vivo and demonstrates the function of troxerutin in alleviating OTA-induced nephrosis and associated systemic energy metabolism disorders.-Yang, X., Xu, W., Huang, K., Zhang, B., Wang, H., Zhang, X., Gong, L., Luo, Y., He, X. Precision toxicology shows that troxerutin alleviates ochratoxin A-induced renal lipotoxicity.
Collapse
Affiliation(s)
- Xuan Yang
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
| | - Wentao Xu
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China.,Beijing Laboratory for Food Quality and Safety, Beijing, China
| | - Kunlun Huang
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China.,Beijing Laboratory for Food Quality and Safety, Beijing, China
| | - Boyang Zhang
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
| | - Haomiao Wang
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
| | - Xueqin Zhang
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
| | - Lijing Gong
- China Academy of Sport and Health Sciences, Beijing Sport University, Beijing, China
| | - Yunbo Luo
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China.,Key Laboratory of Safety Assessment of Genetically Modified Organism-Food Safety, Ministry of Agriculture, China
| | - Xiaoyun He
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China.,Key Laboratory of Safety Assessment of Genetically Modified Organism-Food Safety, Ministry of Agriculture, China
| |
Collapse
|
47
|
Cyclin-dependent kinase 5-mediated phosphorylation of chloride intracellular channel 4 promotes oxidative stress-induced neuronal death. Cell Death Dis 2018; 9:951. [PMID: 30237421 PMCID: PMC6147799 DOI: 10.1038/s41419-018-0983-1] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Revised: 08/06/2018] [Accepted: 08/20/2018] [Indexed: 01/06/2023]
Abstract
Oxidative stress can cause apoptosis in neurons and may result in neurodegenerative diseases. However, the signaling mechanisms leading to oxidative stress–induced neuronal apoptosis are not fully understood. Oxidative stress stimulates aberrant activation of cyclin-dependent kinase 5 (CDK5), thought to promote neuronal apoptosis by phosphorylating many cell death-related substrates. Here, using protein pulldown methods, immunofluorescence experiments and in vitro kinase assays, we identified chloride intracellular channel 4 (CLIC4), the expression of which increases during neuronal apoptosis, as a CDK5 substrate. We found that activated CDK5 phosphorylated serine 108 in CLIC4, increasing CLIC4 protein stability, and accumulation. Pharmacological inhibition or shRNA-mediated silencing of CDK5 decreased CLIC4 levels in neurons. Moreover, CLIC4 overexpression led to neuronal apoptosis, whereas knockdown or pharmacological inhibition of CLIC4 attenuated H2O2-induced neuronal apoptosis. These results implied that CLIC4, by acting as a substrate of CDK5, mediated neuronal apoptosis induced by aberrant CDK5 activation. Targeting CLIC4 in neurons may therefore provide a therapeutic approach for managing progressive neurodegenerative diseases that arise from neuronal apoptosis.
Collapse
|
48
|
Huang MC, Chu IT, Wang ZF, Lin S, Chang TC, Chen CT. A G-Quadruplex Structure in the Promoter Region of CLIC4 Functions as a Regulatory Element for Gene Expression. Int J Mol Sci 2018; 19:ijms19092678. [PMID: 30201851 PMCID: PMC6165315 DOI: 10.3390/ijms19092678] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Revised: 09/05/2018] [Accepted: 09/06/2018] [Indexed: 12/25/2022] Open
Abstract
The differential transcriptional expression of CLIC4 between tumor cells and the surrounding stroma during cancer progression has been suggested to have a tumor-promoting effect. However, little is known about the transcriptional regulation of CLIC4. To better understand how this gene is regulated, the promoter region of CLIC4 was analyzed. We found that a high GC content near the transcriptional start site (TSS) might form an alternative G-quadruplex (G4) structure. Nuclear magnetic resonance spectroscopy (NMR) confirmed their formation in vitro. The reporter assay showed that one of the G4 structures exerted a regulatory role in gene transcription. When the G4-forming sequence was mutated to disrupt the G4 structure, the transcription activity dropped. To examine whether this G4 structure actually has an influence on gene transcription in the chromosome, we utilized the CRISPR/Cas9 system to edit the G4-forming sequence within the CLIC4 promoter in the cell genome. The pop-in/pop-out strategy was adopted to isolate the precisely-edited A375 cell clone. In CRISPR-modified A375 cell clones whose G4 was disrupted, there was a decrease in the endogenous CLIC4 messenger RNA (mRNA) expression level. In conclusion, we found that the G4 structure in the CLIC4 promoter might play an important role in regulating the level of transcription.
Collapse
Affiliation(s)
- Mu-Ching Huang
- Department of Biochemical Science and Technology, National Taiwan University, Taipei 106, Taiwan.
| | - I-Te Chu
- Institute of Atomic and Molecular Sciences, Academia Sinica, Taipei 115, Taiwan.
| | - Zi-Fu Wang
- Institute of Atomic and Molecular Sciences, Academia Sinica, Taipei 115, Taiwan.
| | - Steven Lin
- Institute of Biological Chemistry, Academia Sinica, Taipei 115, Taiwan.
| | - Ta-Chau Chang
- Institute of Atomic and Molecular Sciences, Academia Sinica, Taipei 115, Taiwan.
| | - Chin-Tin Chen
- Department of Biochemical Science and Technology, National Taiwan University, Taipei 106, Taiwan.
| |
Collapse
|
49
|
Lu J, Wu L, Wang X, Zhu J, Du J, Shen B. Detection of Mitochondria Membrane Potential to Study CLIC4 Knockdown-induced HN4 Cell Apoptosis In Vitro. J Vis Exp 2018. [PMID: 30080203 DOI: 10.3791/56317] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Depletion of the mitochondrial membrane potential (MMP, ΔΨm) is considered the earliest event in the apoptotic cascade. It even occurs ahead of nuclear apoptotic characteristics, including chromatin condensation and DNA breakage. Once the MMP collapses, cell apoptosis will initiate irreversibly. A series of lipophilic cationic dyes can pass through the cell membrane and aggregate inside the matrix of mitochondrion, and serve as fluorescence marker to evaluate MMP change. As one of the six members of the Cl- intracellular channel (CLIC) family, CLIC4 participates in the cell apoptotic process mainly through the mitochondrial pathway. Here we describe a detailed protocol to measure MMP via monitoring the fluorescence fluctuation of Rhodamine 123 (Rh123), through which we study apoptosis induced by CLIC4 knockdown. We discuss the advantages and limitations of the application of confocal laser scanning and normal fluorescence microscope in detail, and also compare it with other methods.
Collapse
Affiliation(s)
- Jinsen Lu
- School of Basic Medical Sciences, Anhui Medical University
| | - Lele Wu
- Department of Orthopedics, Anhui Provincial Hospital, Anhui Medical University
| | - Xiaoke Wang
- Department of Orthopedics, Anhui Provincial Hospital, Anhui Medical University
| | - Jinhang Zhu
- Department of Orthopedics, Anhui Provincial Hospital, Anhui Medical University
| | - Juan Du
- Department of Orthopedics, Anhui Provincial Hospital, Anhui Medical University
| | - Bing Shen
- Department of Orthopedics, Anhui Provincial Hospital, Anhui Medical University;
| |
Collapse
|
50
|
Allawzi AM, Vang A, Clements RT, Jhun BS, Kue NR, Mancini TJ, Landi AK, Terentyev D, O-Uchi J, Comhair SA, Erzurum SC, Choudhary G. Activation of Anoctamin-1 Limits Pulmonary Endothelial Cell Proliferation via p38-Mitogen-activated Protein Kinase-Dependent Apoptosis. Am J Respir Cell Mol Biol 2018; 58:658-667. [PMID: 29100477 PMCID: PMC5946325 DOI: 10.1165/rcmb.2016-0344oc] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2016] [Accepted: 09/15/2017] [Indexed: 11/24/2022] Open
Abstract
Hyperproliferative endothelial cells (ECs) play an important role in the pathogenesis of pulmonary arterial hypertension (PAH). Anoctamin (Ano)-1, a calcium-activated chloride channel, can regulate cell proliferation and cell cycle in multiple cell types. However, the expression and function of Ano1 in the pulmonary endothelium is unknown. We examined whether Ano1 was expressed in pulmonary ECs and if altering Ano1 activity would affect EC survival. Expression and localization of Ano1 in rat lung microvascular ECs (RLMVECs) was assessed using immunoblot, immunofluorescence, and subcellular fractionation. Cell counts, flow cytometry, and caspase-3 activity were used to assess changes in cell number and apoptosis in response to the small molecule Ano1 activator, Eact. Changes in mitochondrial membrane potential and mitochondrial reactive oxygen species (mtROS) were assessed using 5,5',6,6'-tetrachloro-1,1',3,3'-tetraethylbenzimidazolylcarbocyanine, iodide (mitochondrial membrane potential dye) and mitochondrial ROS dye, respectively. Ano1 is expressed in RLMVECs and is enriched in the mitochondria. Activation of Ano1 with Eact reduced RLMVEC counts through increased apoptosis. Ano1 knockdown blocked the effects of Eact. Ano1 activation increased mtROS, reduced mitochondrial membrane potential, increased p38 phosphorylation, and induced release of apoptosis-inducing factor. mtROS inhibition attenuated Eact-mediated p38 phosphorylation. Pulmonary artery ECs isolated from patients with idiopathic PAH (IPAH) had higher expression of Ano1 and increased cell counts compared with control subjects. Eact treatment reduced cell counts in IPAH cells, which was associated with increased apoptosis. In summary, Ano1 is expressed in lung EC mitochondria. Activation of Ano1 promotes apoptosis of pulmonary ECs and human IPAH-pulmonary artery ECs, likely via increased mtROS and p38 phosphorylation, leading to apoptosis.
Collapse
Affiliation(s)
- Ayed M. Allawzi
- Vascular Research Laboratory, Providence Veterans Affairs Medical Center, Providence, Rhode Island
- Department of Molecular Pharmacology, Physiology, and Biotechnology, Brown University, Providence, Rhode Island
| | - Alexander Vang
- Vascular Research Laboratory, Providence Veterans Affairs Medical Center, Providence, Rhode Island
| | - Richard T. Clements
- Vascular Research Laboratory, Providence Veterans Affairs Medical Center, Providence, Rhode Island
- Department of Surgery and
| | - Bong Sook Jhun
- Department of Medicine, Cardiovascular Research Center, Rhode Island Hospital and Alpert Medical School of Brown University, Providence, Rhode Island
| | - Nouaying R. Kue
- Vascular Research Laboratory, Providence Veterans Affairs Medical Center, Providence, Rhode Island
| | - Thomas J. Mancini
- Vascular Research Laboratory, Providence Veterans Affairs Medical Center, Providence, Rhode Island
| | - Amy K. Landi
- Department of Medicine, Cardiovascular Research Center, Rhode Island Hospital and Alpert Medical School of Brown University, Providence, Rhode Island
| | - Dmitry Terentyev
- Department of Medicine, Cardiovascular Research Center, Rhode Island Hospital and Alpert Medical School of Brown University, Providence, Rhode Island
| | - Jin O-Uchi
- Department of Medicine, Cardiovascular Research Center, Rhode Island Hospital and Alpert Medical School of Brown University, Providence, Rhode Island
| | - Suzy A. Comhair
- Lerner Research Institute, Cleveland Clinic, Cleveland Ohio; and
| | | | - Gaurav Choudhary
- Vascular Research Laboratory, Providence Veterans Affairs Medical Center, Providence, Rhode Island
- Department of Medicine, Warren Alpert Medical School of Brown University, Providence, Rhode Island
| |
Collapse
|