1
|
Kaitsuka T. The Unique Roles of Ion Channels in Pluripotent Stem Cells in Response to Biological Stimuli. BIOLOGY 2024; 13:1043. [PMID: 39765710 PMCID: PMC11673299 DOI: 10.3390/biology13121043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/17/2024] [Revised: 12/08/2024] [Accepted: 12/11/2024] [Indexed: 01/11/2025]
Abstract
Ion channels are essential for mineral ion homeostasis in mammalian cells, and these are activated or inhibited by environmental stimuli such as heat, cold, mechanical, acidic, or basic stresses. These expressions and functions are quite diverse between cell types. The function and importance of ion channels are well-studied in neurons and cardiac cells, while those functions in pluripotent stem cells (PSCs) were not fully understood. Some sodium, potassium, chloride, calcium, transient receptor potential channels and mechanosensitive Piezo channels are found to be expressed and implicated in pluripotency and self-renewal capacity in PSCs. This review summarizes present and previous reports about ion channels and their response to environmental stimuli in PSCs. Furthermore, we compare the expressions and roles between PSCs and their differentiated embryoid bodies. We then discuss those contributions to pluripotency and differentiation.
Collapse
Affiliation(s)
- Taku Kaitsuka
- School of Pharmacy at Fukuoka, International University of Health and Welfare, Enokizu 137-1, Okawa 831-8501, Fukuoka, Japan
| |
Collapse
|
2
|
Luo J, Wang J, Liu H, Jiang W, Pan L, Huang W, Liu C, Qu X, Liu C, Qin X, Xiang Y. Chloride intracellular channel 4 participates in the regulation of lipopolysaccharide-induced inflammatory responses in human bronchial epithelial cells. Respir Physiol Neurobiol 2024; 327:104303. [PMID: 39029565 DOI: 10.1016/j.resp.2024.104303] [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: 03/19/2024] [Revised: 07/11/2024] [Accepted: 07/15/2024] [Indexed: 07/21/2024]
Abstract
The airway epithelium is located at the interactional boundary between the external and internal environments of the organism and is often exposed to harmful environmental stimuli. Inflammatory response that occurs after airway epithelial stress is the basis of many lung and systemic diseases. Chloride intracellular channel 4 (CLIC4) is abundantly expressed in epithelial cells. The purpose of this study was to investigate whether CLIC4 is involved in the regulation of lipopolysaccharide (LPS)-induced inflammatory response in airway epithelial cells and to clarify its potential mechanism. Our results showed that LPS induced inflammatory response and decreased CLIC4 levels in vivo and in vitro. CLIC4 silencing aggravated the inflammatory response in epithelial cells, while overexpression of CLIC4 combined with LPS exposure significantly decreased the inflammatory response compared with cells exposed to LPS without CLIC4 overexpression. By labeling intracellular chloride ions with chloride fluorescent probe MQAE, we showed that CLIC4 mediated intracellular chloride ion-regulated LPS-induced cellular inflammatory response.
Collapse
Affiliation(s)
- Jinhua Luo
- Department of Physiology, School of Basic Medicine, Central South University, Changsha 410000, China
| | - Jia Wang
- Department of Physiology, School of Basic Medicine, Central South University, Changsha 410000, China; Hunan Provincial People's Hospital, The First-affiliated Hospital of Hunan Normal University, Changsha 410016, China
| | - Huijun Liu
- Department of Physiology, School of Basic Medicine, Central South University, Changsha 410000, China
| | - Wang Jiang
- Department of Physiology, School of Basic Medicine, Central South University, Changsha 410000, China
| | - Lang Pan
- Department of Physiology, School of Basic Medicine, Central South University, Changsha 410000, China; Department of Microbiology and Immunology, School of Medicine, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Wenjie Huang
- Department of Physiology, School of Basic Medicine, Central South University, Changsha 410000, China; Department of Reproductive Medicine, Liuzhou maternity and Child Healthcare Hospital, Liuzhou, Guangxi 545001, China
| | - Caixia Liu
- Department of Physiology, School of Basic Medicine, Central South University, Changsha 410000, China; Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Xiangping Qu
- Department of Physiology, School of Basic Medicine, Central South University, Changsha 410000, China
| | - Chi Liu
- Department of Physiology, School of Basic Medicine, Central South University, Changsha 410000, China
| | - Xiaoqun Qin
- Department of Physiology, School of Basic Medicine, Central South University, Changsha 410000, China
| | - Yang Xiang
- Department of Physiology, School of Basic Medicine, Central South University, Changsha 410000, China.
| |
Collapse
|
3
|
Mazzucchelli S, Signati L, Messa L, Franceschini A, Bonizzi A, Castagnoli L, Gasparini P, Consolandi C, Mangano E, Pelucchi P, Cifola I, Camboni T, Severgnini M, Villani L, Tagliaferri B, Carelli S, Pupa SM, Cereda C, Corsi F. Breast cancer patient-derived organoids for the investigation of patient-specific tumour evolution. Cancer Cell Int 2024; 24:220. [PMID: 38926706 PMCID: PMC11210105 DOI: 10.1186/s12935-024-03375-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Accepted: 05/16/2024] [Indexed: 06/28/2024] Open
Abstract
BACKGROUND A reliable preclinical model of patient-derived organoids (PDOs) was developed in a case study of a 69-year-old woman diagnosed with breast cancer (BC) to investigate the tumour evolution before and after neoadjuvant chemotherapy and surgery. The results were achieved due to the development of PDOs from tissues collected before (O-PRE) and after (O-POST) treatment. METHODS PDO cultures were characterized by histology, immunohistochemistry (IHC), transmission electron microscopy (TEM), scanning electron microscopy (SEM), confocal microscopy, flow cytometry, real-time PCR, bulk RNA-seq, single-cell RNA sequencing (scRNA-seq) and drug screening. RESULTS Both PDO cultures recapitulated the histological and molecular profiles of the original tissues, and they showed typical mammary gland organization, confirming their reliability as a personalized in vitro model. Compared with O-PRE, O-POST had a greater proliferation rate with a significant increase in the Ki67 proliferation index. Moreover O-POST exhibited a more stem-like and aggressive phenotype, with increases in the CD24low/CD44low and EPCAMlow/CD49fhigh cell populations characterized by increased tumour initiation potential and multipotency and metastatic potential in invasive lobular carcinoma. Analysis of ErbB receptor expression indicated a decrease in HER-2 expression coupled with an increase in EGFR expression in O-POST. In this context, deregulation of the PI3K/Akt signalling pathway was assessed by transcriptomic analysis, confirming the altered transcriptional profile. Finally, transcriptomic single-cell analysis identified 11 cell type clusters, highlighting the selection of the luminal component and the decrease in the number of Epithelial-mesenchymal transition cell types in O-POST. CONCLUSION Neoadjuvant treatment contributed to the enrichment of cell populations with luminal phenotypes that were more resistant to chemotherapy in O-POST. PDOs represent an excellent 3D cell model for assessing disease evolution.
Collapse
Affiliation(s)
- Serena Mazzucchelli
- Dipartimento di Scienze Biomediche e Cliniche, Università di Milano, Via G. B. Grassi 74, 20157, Milan, Italy.
| | - Lorena Signati
- Dipartimento di Scienze Biomediche e Cliniche, Università di Milano, Via G. B. Grassi 74, 20157, Milan, Italy
| | - Letizia Messa
- Department of Electronics, Information and Bioengineering (DEIB), Politecnico di Milano, 20133, Milan, Italy
- Pediatric Research Center "Romeo and Enrica Invernizzi", Università di Milano, 20157, Milan, Italy
- Center of Functional Genomics and Rare Diseases, Buzzi Children's Hospital, 20154, Milan, Italy
| | - Alma Franceschini
- Microenvironment and Biomarkers of Solid Tumors, Department of Experimental Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, 20133, Milan, Italy
| | - Arianna Bonizzi
- Istituti Clinici Scientifici Maugeri IRCCS, 27100, Pavia, Italy
| | - Lorenzo Castagnoli
- Microenvironment and Biomarkers of Solid Tumors, Department of Experimental Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, 20133, Milan, Italy
| | - Patrizia Gasparini
- Epigenomics and Biomarkers of Solid Tumors, Department of Experimental Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, 20133, Milan, Italy
| | - Clarissa Consolandi
- Institute for Biomedical Technologies, National Research Council (ITB-CNR), Via F. lli Cervi 93, 20054, Segrate, Italy
| | - Eleonora Mangano
- Institute for Biomedical Technologies, National Research Council (ITB-CNR), Via F. lli Cervi 93, 20054, Segrate, Italy
| | - Paride Pelucchi
- Institute for Biomedical Technologies, National Research Council (ITB-CNR), Via F. lli Cervi 93, 20054, Segrate, Italy
| | - Ingrid Cifola
- Institute for Biomedical Technologies, National Research Council (ITB-CNR), Via F. lli Cervi 93, 20054, Segrate, Italy
| | - Tania Camboni
- Institute for Biomedical Technologies, National Research Council (ITB-CNR), Via F. lli Cervi 93, 20054, Segrate, Italy
| | - Marco Severgnini
- Institute for Biomedical Technologies, National Research Council (ITB-CNR), Via F. lli Cervi 93, 20054, Segrate, Italy
| | - Laura Villani
- Istituti Clinici Scientifici Maugeri IRCCS, 27100, Pavia, Italy
| | | | - Stephana Carelli
- Pediatric Research Center "Romeo and Enrica Invernizzi", Università di Milano, 20157, Milan, Italy
- Center of Functional Genomics and Rare Diseases, Buzzi Children's Hospital, 20154, Milan, Italy
| | - Serenella M Pupa
- Microenvironment and Biomarkers of Solid Tumors, Department of Experimental Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, 20133, Milan, Italy
| | - Cristina Cereda
- Center of Functional Genomics and Rare Diseases, Buzzi Children's Hospital, 20154, Milan, Italy
| | - Fabio Corsi
- Dipartimento di Scienze Biomediche e Cliniche, Università di Milano, Via G. B. Grassi 74, 20157, Milan, Italy.
- Istituti Clinici Scientifici Maugeri IRCCS, 27100, Pavia, Italy.
| |
Collapse
|
4
|
Wojtera B, Ostrowska K, Szewczyk M, Masternak MM, Golusiński W. Chloride intracellular channels in oncology as potential novel biomarkers and personalized therapy targets: a systematic review. Rep Pract Oncol Radiother 2024; 29:258-270. [PMID: 39143969 PMCID: PMC11321771 DOI: 10.5603/rpor.99674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Accepted: 02/29/2024] [Indexed: 08/16/2024] Open
Abstract
Background The chloride intracellular channels (CLICs) family includes six ion channels (CLIC1-CLIC6) expressed on the cellular level and secreted into interstitial fluid and blood. They are involved in the physiological functioning of multiple systems as well as the pathogenetic processes of cancer. CLICs play essential roles in the tumor microenvironment. The current systematic review aimed at identifying and summarizing the research of CLICs in oncology on clinical material to assess CLICs' potential as novel biomarkers and personalized therapy targets. Materials and methods The authors systematically searched the PubMed database for original articles concerning CLIC research on clinical material of all types of cancer - fluids and tissues. Results Fifty-three articles investigating in summary 3944 clinical samples were qualified for the current review. Studied material included 3438 tumor samples (87%), 437 blood samples (11%), and 69 interstitial fluid samples (2%). Studies investigated 21 cancer types, mostly hepatocellular carcinoma, colorectal, ovarian, and gastric cancer. Importantly, CLIC1, CLIC2, CLIC3, CLIC4, and CLIC5 were differently expressed in cancerous tissues and patients' blood compared to healthy controls. Moreover, CLICs were found to be involved in several cancer-associated signaling pathways, such as PI3K/AKT, MAPK/ERK, and MAPK/p38. Conclusion CLIC family members may be candidates for potential novel cancer biomarkers due to the contrast in their expression between cancerous and healthy tissues and secretion to the interstitial fluid and blood. CLICs are investigated as potential therapeutic targets because of their involvement in cancer pathogenesis and tumor microenvironment.
Collapse
Affiliation(s)
- Bartosz Wojtera
- Department of Head and Neck Surgery, Greater Poland Cancer Centre, Poznan University of Medical Sciences, Poznan, Poland
- Radiobiology Laboratory, Department of Medical Physics, Greater Poland Cancer Centre, Poznan University of Medical Sciences, Poznan, Poland
| | - Kamila Ostrowska
- Department of Head and Neck Surgery, Greater Poland Cancer Centre, Poznan University of Medical Sciences, Poznan, Poland
- Radiobiology Laboratory, Department of Medical Physics, Greater Poland Cancer Centre, Poznan University of Medical Sciences, Poznan, Poland
| | - Mateusz Szewczyk
- Department of Head and Neck Surgery, Greater Poland Cancer Centre, Poznan University of Medical Sciences, Poznan, Poland
| | - Michał M. Masternak
- Department of Head and Neck Surgery, Greater Poland Cancer Centre, Poznan University of Medical Sciences, Poznan, Poland
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, Florida, United States
| | - Wojciech Golusiński
- Department of Head and Neck Surgery, Greater Poland Cancer Centre, Poznan University of Medical Sciences, Poznan, Poland
| |
Collapse
|
5
|
Sanchez VC, Craig‐Lucas A, Cataisson C, Carofino BL, Yuspa SH. Crosstalk between tumor and stroma modifies CLIC4 cargo in extracellular vesicles. JOURNAL OF EXTRACELLULAR BIOLOGY 2023; 2:e118. [PMID: 38264628 PMCID: PMC10803055 DOI: 10.1002/jex2.118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Revised: 09/08/2023] [Accepted: 09/11/2023] [Indexed: 01/25/2024]
Abstract
Mouse models of breast cancer have revealed that tumor-bearing hosts must express the oxidoreductase CLIC4 to develop lung metastases. In the absence of host CLIC4, primary tumors grow but the lung premetastatic niche is defective for metastatic seeding. Primary breast cancer cells release EVs that incorporate CLIC4 as cargo and circulate in plasma of wildtype tumor-bearing hosts. CLIC4-deficient breast cancer cells also form tumors in wildtype hosts and release EVs in plasma, but these EVs lack CLIC4, suggesting that the tumor is the source of the plasma-derived EVs that carry CLIC4 as cargo. Paradoxically, circulating EVs are also devoid of CLIC4 when CLIC4-expressing primary tumors are grown in CLIC4 knockout hosts. Thus, the incorporation of CLIC4 (and perhaps other factors) as EV cargo released from tumors involves specific signals from the surrounding stroma determined by its genetic composition. Since CLIC4 is also detected in circulating EVs from human breast cancer patients, future studies will address its association with disease.
Collapse
Affiliation(s)
- Vanesa C. Sanchez
- Center for Drug Evaluation and ResearchU.S. Food and Drug AdministrationSilver SpringMarylandUSA
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer InstituteNational Institutes of HealthBethesdaMarylandUSA
| | - Alayna Craig‐Lucas
- Department of SurgeryLehigh Valley Health NetworkAllentownPennsylvaniaUSA
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer InstituteNational Institutes of HealthBethesdaMarylandUSA
| | - Christophe Cataisson
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer InstituteNational Institutes of HealthBethesdaMarylandUSA
| | - Brandi L. Carofino
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer InstituteNational Institutes of HealthBethesdaMarylandUSA
| | - Stuart H. Yuspa
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer InstituteNational Institutes of HealthBethesdaMarylandUSA
| |
Collapse
|