1
|
Mierke CT. Extracellular Matrix Cues Regulate Mechanosensing and Mechanotransduction of Cancer Cells. Cells 2024; 13:96. [PMID: 38201302 PMCID: PMC10777970 DOI: 10.3390/cells13010096] [Citation(s) in RCA: 34] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2023] [Revised: 12/29/2023] [Accepted: 01/01/2024] [Indexed: 01/12/2024] Open
Abstract
Extracellular biophysical properties have particular implications for a wide spectrum of cellular behaviors and functions, including growth, motility, differentiation, apoptosis, gene expression, cell-matrix and cell-cell adhesion, and signal transduction including mechanotransduction. Cells not only react to unambiguously mechanical cues from the extracellular matrix (ECM), but can occasionally manipulate the mechanical features of the matrix in parallel with biological characteristics, thus interfering with downstream matrix-based cues in both physiological and pathological processes. Bidirectional interactions between cells and (bio)materials in vitro can alter cell phenotype and mechanotransduction, as well as ECM structure, intentionally or unintentionally. Interactions between cell and matrix mechanics in vivo are of particular importance in a variety of diseases, including primarily cancer. Stiffness values between normal and cancerous tissue can range between 500 Pa (soft) and 48 kPa (stiff), respectively. Even the shear flow can increase from 0.1-1 dyn/cm2 (normal tissue) to 1-10 dyn/cm2 (cancerous tissue). There are currently many new areas of activity in tumor research on various biological length scales, which are highlighted in this review. Moreover, the complexity of interactions between ECM and cancer cells is reduced to common features of different tumors and the characteristics are highlighted to identify the main pathways of interaction. This all contributes to the standardization of mechanotransduction models and approaches, which, ultimately, increases the understanding of the complex interaction. Finally, both the in vitro and in vivo effects of this mechanics-biology pairing have key insights and implications for clinical practice in tumor treatment and, consequently, clinical translation.
Collapse
Affiliation(s)
- Claudia Tanja Mierke
- Biological Physics Division, Peter Debye Institute of Soft Matter Physics, Faculty of Physics and Earth Science, Leipzig University, Linnéstraße 5, 04103 Leipzig, Germany
| |
Collapse
|
2
|
Toader C, Tataru CP, Florian IA, Covache-Busuioc RA, Dumitrascu DI, Glavan LA, Costin HP, Bratu BG, Ciurea AV. From Homeostasis to Pathology: Decoding the Multifaceted Impact of Aquaporins in the Central Nervous System. Int J Mol Sci 2023; 24:14340. [PMID: 37762642 PMCID: PMC10531540 DOI: 10.3390/ijms241814340] [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: 09/02/2023] [Revised: 09/15/2023] [Accepted: 09/18/2023] [Indexed: 09/29/2023] Open
Abstract
Aquaporins (AQPs), integral membrane proteins facilitating selective water and solute transport across cell membranes, have been the focus of extensive research over the past few decades. Particularly noteworthy is their role in maintaining cellular homeostasis and fluid balance in neural compartments, as dysregulated AQP expression is implicated in various degenerative and acute brain pathologies. This article provides an exhaustive review on the evolutionary history, molecular classification, and physiological relevance of aquaporins, emphasizing their significance in the central nervous system (CNS). The paper journeys through the early studies of water transport to the groundbreaking discovery of Aquaporin 1, charting the molecular intricacies that make AQPs unique. It delves into AQP distribution in mammalian systems, detailing their selective permeability through permeability assays. The article provides an in-depth exploration of AQP4 and AQP1 in the brain, examining their contribution to fluid homeostasis. Furthermore, it elucidates the interplay between AQPs and the glymphatic system, a critical framework for waste clearance and fluid balance in the brain. The dysregulation of AQP-mediated processes in this system hints at a strong association with neurodegenerative disorders such as Parkinson's Disease, idiopathic normal pressure hydrocephalus, and Alzheimer's Disease. This relationship is further explored in the context of acute cerebral events such as stroke and autoimmune conditions such as neuromyelitis optica (NMO). Moreover, the article scrutinizes AQPs at the intersection of oncology and neurology, exploring their role in tumorigenesis, cell migration, invasiveness, and angiogenesis. Lastly, the article outlines emerging aquaporin-targeted therapies, offering a glimpse into future directions in combatting CNS malignancies and neurodegenerative diseases.
Collapse
Affiliation(s)
- Corneliu Toader
- Department of Neurosurgery, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania; (C.T.); (R.-A.C.-B.); (D.-I.D.); (L.A.G.); (H.P.C.); (B.-G.B.); (A.V.C.)
- Department of Vascular Neurosurgery, National Institute of Neurology and Neurovascular Diseases, 077160 Bucharest, Romania
| | - Calin Petru Tataru
- Department of Opthamology, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania
- Central Military Emergency Hospital “Dr. Carol Davila”, 010825 Bucharest, Romania
| | - Ioan-Alexandru Florian
- Department of Neurosciences, “Iuliu Hatieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
| | - Razvan-Adrian Covache-Busuioc
- Department of Neurosurgery, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania; (C.T.); (R.-A.C.-B.); (D.-I.D.); (L.A.G.); (H.P.C.); (B.-G.B.); (A.V.C.)
| | - David-Ioan Dumitrascu
- Department of Neurosurgery, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania; (C.T.); (R.-A.C.-B.); (D.-I.D.); (L.A.G.); (H.P.C.); (B.-G.B.); (A.V.C.)
| | - Luca Andrei Glavan
- Department of Neurosurgery, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania; (C.T.); (R.-A.C.-B.); (D.-I.D.); (L.A.G.); (H.P.C.); (B.-G.B.); (A.V.C.)
| | - Horia Petre Costin
- Department of Neurosurgery, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania; (C.T.); (R.-A.C.-B.); (D.-I.D.); (L.A.G.); (H.P.C.); (B.-G.B.); (A.V.C.)
| | - Bogdan-Gabriel Bratu
- Department of Neurosurgery, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania; (C.T.); (R.-A.C.-B.); (D.-I.D.); (L.A.G.); (H.P.C.); (B.-G.B.); (A.V.C.)
| | - Alexandru Vlad Ciurea
- Department of Neurosurgery, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania; (C.T.); (R.-A.C.-B.); (D.-I.D.); (L.A.G.); (H.P.C.); (B.-G.B.); (A.V.C.)
- Neurosurgery Department, Sanador Clinical Hospital, 010991 Bucharest, Romania
| |
Collapse
|
3
|
Petrillo S, De Giorgio F, Bertino F, Garello F, Bitonto V, Longo DL, Mercurio S, Ammirata G, Allocco AL, Fiorito V, Chiabrando D, Altruda F, Terreno E, Provero P, Munaron L, Genova T, Nóvoa A, Carlos AR, Cardoso S, Mallo M, Soares MP, Tolosano E. Endothelial cells require functional FLVCR1a during developmental and adult angiogenesis. Angiogenesis 2023; 26:365-384. [PMID: 36631598 PMCID: PMC10328904 DOI: 10.1007/s10456-023-09865-w] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Accepted: 01/06/2023] [Indexed: 01/13/2023]
Abstract
The Feline Leukemia Virus Subgroup C Receptor 1a (FLVCR1a) is a transmembrane heme exporter essential for embryonic vascular development. However, the exact role of FLVCR1a during blood vessel development remains largely undefined. Here, we show that FLVCR1a is highly expressed in angiogenic endothelial cells (ECs) compared to quiescent ECs. Consistently, ECs lacking FLVCR1a give rise to structurally and functionally abnormal vascular networks in multiple models of developmental and pathologic angiogenesis. Firstly, zebrafish embryos without FLVCR1a displayed defective intersegmental vessels formation. Furthermore, endothelial-specific Flvcr1a targeting in mice led to a reduced radial expansion of the retinal vasculature associated to decreased EC proliferation. Moreover, Flvcr1a null retinas showed defective vascular organization and loose attachment of pericytes. Finally, adult neo-angiogenesis is severely affected in murine models of tumor angiogenesis. Tumor blood vessels lacking Flvcr1a were disorganized and dysfunctional. Collectively, our results demonstrate the critical role of FLVCR1a as a regulator of developmental and pathological angiogenesis identifying FLVCR1a as a potential therapeutic target in human diseases characterized by aberrant neovascularization.
Collapse
Affiliation(s)
- Sara Petrillo
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center (MBC) "Guido Tarone", University of Torino, Via Nizza, 52, 10126, Turin, Italy.
| | - F De Giorgio
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center (MBC) "Guido Tarone", University of Torino, Via Nizza, 52, 10126, Turin, Italy
| | - F Bertino
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center (MBC) "Guido Tarone", University of Torino, Via Nizza, 52, 10126, Turin, Italy
| | - F Garello
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center (MBC) "Guido Tarone", University of Torino, Via Nizza, 52, 10126, Turin, Italy
| | - V Bitonto
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center (MBC) "Guido Tarone", University of Torino, Via Nizza, 52, 10126, Turin, Italy
| | - D L Longo
- Institute of Biostructures and Bioimaging (IBB), Italian National Research Council (CNR), Via Nizza, 52, 10126, Turin, Italy
| | - S Mercurio
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center (MBC) "Guido Tarone", University of Torino, Via Nizza, 52, 10126, Turin, Italy
| | - G Ammirata
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center (MBC) "Guido Tarone", University of Torino, Via Nizza, 52, 10126, Turin, Italy
| | - A L Allocco
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center (MBC) "Guido Tarone", University of Torino, Via Nizza, 52, 10126, Turin, Italy
| | - V Fiorito
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center (MBC) "Guido Tarone", University of Torino, Via Nizza, 52, 10126, Turin, Italy
| | - D Chiabrando
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center (MBC) "Guido Tarone", University of Torino, Via Nizza, 52, 10126, Turin, Italy
| | - F Altruda
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center (MBC) "Guido Tarone", University of Torino, Via Nizza, 52, 10126, Turin, Italy
| | - E Terreno
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center (MBC) "Guido Tarone", University of Torino, Via Nizza, 52, 10126, Turin, Italy
| | - P Provero
- Department of Molecular Biotechnology and Health Sciences, and GenoBiToUS, Genomics and Bioinformatics Service, University of Torino, Turin, Italy
- Center for Translational Genomics and Bioinformatics, San Raffaele Scientific Institute IRCCS, Milan, Italy
| | - L Munaron
- Department of Life Sciences and Systems Biology, University of Torino, Via Accademia Albertina 13, 10123, Turin, Italy
| | - T Genova
- Department of Life Sciences and Systems Biology, University of Torino, Via Accademia Albertina 13, 10123, Turin, Italy
| | - A Nóvoa
- Instituto Gulbenkian de Ciência, Oeiras, Portugal
| | - A R Carlos
- Instituto Gulbenkian de Ciência, Oeiras, Portugal
| | - S Cardoso
- Instituto Gulbenkian de Ciência, Oeiras, Portugal
| | - M Mallo
- Instituto Gulbenkian de Ciência, Oeiras, Portugal
| | - M P Soares
- Instituto Gulbenkian de Ciência, Oeiras, Portugal
| | - E Tolosano
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center (MBC) "Guido Tarone", University of Torino, Via Nizza, 52, 10126, Turin, Italy
| |
Collapse
|
4
|
The Impact of Plasma Membrane Ion Channels on Bone Remodeling in Response to Mechanical Stress, Oxidative Imbalance, and Acidosis. Antioxidants (Basel) 2023; 12:antiox12030689. [PMID: 36978936 PMCID: PMC10045377 DOI: 10.3390/antiox12030689] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 03/01/2023] [Accepted: 03/03/2023] [Indexed: 03/14/2023] Open
Abstract
The extracellular milieu is a rich source of different stimuli and stressors. Some of them depend on the chemical–physical features of the matrix, while others may come from the ‘outer’ environment, as in the case of mechanical loading applied on the bones. In addition to these forces, a plethora of chemical signals drives cell physiology and fate, possibly leading to dysfunctions when the homeostasis is disrupted. This variety of stimuli triggers different responses among the tissues: bones represent a particular milieu in which a fragile balance between mechanical and metabolic demands should be tuned and maintained by the concerted activity of cell biomolecules located at the interface between external and internal environments. Plasma membrane ion channels can be viewed as multifunctional protein machines that act as rapid and selective dual-nature hubs, sensors, and transducers. Here we focus on some multisensory ion channels (belonging to Piezo, TRP, ASIC/EnaC, P2XR, Connexin, and Pannexin families) actually or potentially playing a significant role in bone adaptation to three main stressors, mechanical forces, oxidative stress, and acidosis, through their effects on bone cells including mesenchymal stem cells, osteoblasts, osteoclasts, and osteocytes. Ion channel-mediated bone remodeling occurs in physiological processes, aging, and human diseases such as osteoporosis, cancer, and traumatic events.
Collapse
|
5
|
Deng B, Zhao Z, Kong W, Han C, Shen X, Zhou C. Biological role of matrix stiffness in tumor growth and treatment. J Transl Med 2022; 20:540. [PMID: 36419159 PMCID: PMC9682678 DOI: 10.1186/s12967-022-03768-y] [Citation(s) in RCA: 112] [Impact Index Per Article: 37.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Accepted: 11/09/2022] [Indexed: 11/24/2022] Open
Abstract
In recent years, the biological role of changes in physical factors in carcinogenesis and progression has attracted increasing attention. Matrix stiffness, also known as ECM stress, is a critical physical factor of tumor microenvironment and remains alternating during carcinogenesis as a result of ECM remodeling through activation of cancer-associated fibroblasts and extracellular collagen accumulation, crosslinking and fibrosis. Different content and density of extracellular collagen in ECM endows matrix with varying stiffness. Physical signals induced by matrix stiffness are transmitted to tumor cells primarily by the integrins receptor family and trigger a series of mechanotransduction that result in changes in tumor cell morphology, proliferative capacity, and invasive ability. Importantly, accumulating evidence revealed that changes in matrix stiffness in tumor tissues greatly control the sensitivity of tumor cells in response to chemotherapy, radiotherapy, and immunotherapy through integrin signaling, YAP signaling, and related signaling pathways. Here, the present review analyzes the current research advances on matrix stiffness and tumor cell behavior with a view to contributing to tumor cell growth and treatment, with the hope of improving the understanding of the biological role of matrix stiffness in tumors.
Collapse
Affiliation(s)
- Boer Deng
- grid.24696.3f0000 0004 0369 153XDepartment of Gynecologic Oncology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, People’s Republic of China ,grid.10698.360000000122483208Division of Gynecologic Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599 USA
| | - Ziyi Zhao
- grid.24696.3f0000 0004 0369 153XDepartment of Gynecologic Oncology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, People’s Republic of China ,grid.10698.360000000122483208Division of Gynecologic Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599 USA
| | - Weimin Kong
- grid.24696.3f0000 0004 0369 153XDepartment of Gynecologic Oncology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, People’s Republic of China ,grid.10698.360000000122483208Division of Gynecologic Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599 USA
| | - Chao Han
- grid.24696.3f0000 0004 0369 153XDepartment of Gynecologic Oncology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, People’s Republic of China
| | - Xiaochang Shen
- grid.24696.3f0000 0004 0369 153XDepartment of Gynecologic Oncology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, People’s Republic of China ,grid.10698.360000000122483208Division of Gynecologic Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599 USA
| | - Chunxiao Zhou
- grid.10698.360000000122483208Division of Gynecologic Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599 USA ,grid.10698.360000000122483208Lineberger Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599 USA
| |
Collapse
|
6
|
Chatterjee A, Bararia A, Ganguly D, Mondal PK, Roy P, Banerjee S, Ghosh S, Gulati S, Ghatak S, Chattopadhay BK, Basu P, Chatterjee A, Sikdar N. DNA methylome in pancreatic cancer identified novel promoter hyper-methylation in NPY and FAIM2 genes associated with poor prognosis in Indian patient cohort. Cancer Cell Int 2022; 22:334. [PMID: 36329447 PMCID: PMC9635159 DOI: 10.1186/s12935-022-02737-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 09/17/2022] [Indexed: 11/06/2022] Open
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) is one of the leading cancers worldwide and has a poor survival, with a 5-year survival rate of only 8.5%. In this study we investigated altered DNA methylation associated with PDAC severity and prognosis. METHODS Methylome data, generated using 450 K bead array, was compared between paired PDAC and normal samples in the TCGA cohort (n = 9) and our Indian cohort (n = 7). The total Indian Cohort (n = 75) was split into cohort 1 (n = 7), cohort 2 (n = 22), cohort 3 (n = 26) and cohort 4 (n = 20).Validation of differential methylation (6 selected CpG loci) and associated gene expression for differentially methylated genes (10 selected gDMs) were carried out in separate validation cohorts, using MSP, RT-PCR and IHC correlations between methylation and gene expression were observed in TCGA, GTEx cohorts and in validation cohorts. Kaplan-Meier survival analysis was done to study differential prognosis, during 2-5 years of follow-up. RESULTS We identified 156 DMPs, mapped to 91 genes (gDMs), in PDAC; 68 (43.5%) DMPs were found to be differentially methylated both in TCGA cohort and our cohort, with significant concordance at hypo- and hyper-methylated loci. Enrichments of "regulation of ion transport", "Interferon alpha/beta signalling", "morphogenesis and development" and "transcriptional dysregulation" pathways were observed among 91 gDMs. Hyper-methylation of NPY and FAIM2 genes with down-regulated expression in PDAC, were significantly associated with poor prognosis in the Indian patient cohort. CONCLUSIONS Ethnic variations among populations may determine the altered epigenetic landscape in the PDAC patients of the Indian cohort. Our study identified novel differentially methylated genes (mainly NPY and FAIM2) and also validated the previously identified differentially methylated CpG sites associated with PDAC cancer patient's survival. Comparative analysis of our data with TCGA and CPTAC cohorts showed that both NPY and FAIM2 hyper-methylation and down-regulations can be novel epigenetically regulated genes in the Indian patient population, statistically significantly associated with poor survival and advanced tumour stages.
Collapse
Affiliation(s)
| | - Akash Bararia
- Biological Sciences Division, Human Genetics Unit, Indian Statistical Institute, 203, B. T. Road, Kolkata, West Bengal, 700108, India
| | | | - Pronoy Kanti Mondal
- Biological Sciences Division, Human Genetics Unit, Indian Statistical Institute, 203, B. T. Road, Kolkata, West Bengal, 700108, India
| | - Paromita Roy
- Department of Pathology & Department of Gastrointestinal Surgery, Tata Medical Center, Rajarhat, Kolkata, India
| | - Sudeep Banerjee
- Department of Pathology & Department of Gastrointestinal Surgery, Tata Medical Center, Rajarhat, Kolkata, India
| | - Shibajyoti Ghosh
- Department of General Surgery, Medical College and Hospital, Kolkata, India
| | - Sumit Gulati
- Department of HPB Surgery, Apollo Multispecialty Hospital, Kolkata, India
| | - Supriyo Ghatak
- Department of HPB Surgery, Apollo Multispecialty Hospital, Kolkata, India
| | | | | | - Aniruddha Chatterjee
- Department of Pathology, Dunedin School of Medicine, University of Otago, Dunedin, New Zealand
| | - Nilabja Sikdar
- Biological Sciences Division, Human Genetics Unit, Indian Statistical Institute, 203, B. T. Road, Kolkata, West Bengal, 700108, India.
| |
Collapse
|
7
|
Chang L, Yang P, Zhang C, Zhu J, Zhang Y, Wang Y, Ding J, Wang K. Long intergenic non-protein-coding RNA 467 promotes tumor progression and angiogenesis via the microRNA-128-3p/vascular endothelial growth factor C axis in colorectal cancer. Bioengineered 2022; 13:12392-12408. [PMID: 35587748 PMCID: PMC9275949 DOI: 10.1080/21655979.2022.2074666] [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] [Indexed: 11/28/2022] Open
Abstract
Long non-coding RNAs (lncRNAs) are important regulators and biomarkers of tumorigenesis and tumor metastasis. Long intergenic non-protein-coding RNA 467 (LINC00467) is associated with various cancers. However, the role and mechanism of LINC00467 in colorectal cancer (CRC) promotion are poorly understood. This study aimed to present new details of LINC00467 in the progression of CRC. Reverse transcription–polymerase chain reaction demonstrated that the expression level of LINC00467 in CRC tissues and cell lines was significantly upregulated, which was closely related to the clinical features of CRC. Cell and animal studies showed that the downregulation of LINC00467 expression in CRC cells significantly inhibited cell proliferation, metastasis, and angiogenesis. Moreover, the overexpression of LINC00467 accelerated CRC promotion. Bioinformatics analysis and luciferase reporter assay confirmed that LINC00467 binds to miR-128-3p. Rescue experiments manifested that decreased miR-128-3p level reversed CRC cell inhibition by silencing LINC00467. Furthermore, vascular endothelial growth factor C (VEGFC) was identified as a target of miR-128-3p that could reverse the inhibition of cell growth that is mediated by miR-128-3p. Altogether, our results showed that LINC00467 contributes to CRC progression and angiogenesis via the miR-128-3p/VEGFC axis. Our findings expand the understanding of the mechanisms underlying CRC and suggest potential targets for clinical strategies against CRC.
Collapse
Affiliation(s)
- Lisha Chang
- Department of Oncology, Second Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Peipei Yang
- Department of Oncology, Second Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Chun Zhang
- Department of Oncology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jing Zhu
- Department of Oncology, Second Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yirao Zhang
- Department of Oncology, Second Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yang Wang
- Department of Oncology, Second Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jie Ding
- Department of Oncology, Second Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Keming Wang
- Department of Oncology, Second Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| |
Collapse
|
8
|
Zhang Y, Wang H, Oliveira RHM, Zhao C, Popel AS. Systems biology of angiogenesis signaling: Computational models and omics. WIREs Mech Dis 2021; 14:e1550. [PMID: 34970866 PMCID: PMC9243197 DOI: 10.1002/wsbm.1550] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 12/03/2021] [Accepted: 12/06/2021] [Indexed: 01/10/2023]
Abstract
Angiogenesis is a highly regulated multiscale process that involves a plethora of cells, their cellular signal transduction, activation, proliferation, differentiation, as well as their intercellular communication. The coordinated execution and integration of such complex signaling programs is critical for physiological angiogenesis to take place in normal growth, development, exercise, and wound healing, while its dysregulation is critically linked to many major human diseases such as cancer, cardiovascular diseases, and ocular disorders; it is also crucial in regenerative medicine. Although huge efforts have been devoted to drug development for these diseases by investigation of angiogenesis‐targeted therapies, only a few therapeutics and targets have proved effective in humans due to the innate multiscale complexity and nonlinearity in the process of angiogenic signaling. As a promising approach that can help better address this challenge, systems biology modeling allows the integration of knowledge across studies and scales and provides a powerful means to mechanistically elucidate and connect the individual molecular and cellular signaling components that function in concert to regulate angiogenesis. In this review, we summarize and discuss how systems biology modeling studies, at the pathway‐, cell‐, tissue‐, and whole body‐levels, have advanced our understanding of signaling in angiogenesis and thereby delivered new translational insights for human diseases. This article is categorized under:Cardiovascular Diseases > Computational Models Cancer > Computational Models
Collapse
Affiliation(s)
- Yu Zhang
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Hanwen Wang
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Rebeca Hannah M Oliveira
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Chen Zhao
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,School of Pharmacy, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Aleksander S Popel
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
9
|
Gao B, Feng C, Chai F, Wei S, Hong N, Ye Y, Wang Y, Cheng J. CT-detected extramural venous invasion-related gene signature for the overall survival prediction in patients with gastric cancer. Cancer Med 2021; 10:7816-7830. [PMID: 34510798 PMCID: PMC8559479 DOI: 10.1002/cam4.4266] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2021] [Revised: 07/25/2021] [Accepted: 09/01/2021] [Indexed: 12/17/2022] Open
Abstract
Background Computed tomography (CT)‐detected extramural venous invasion (EMVI) has been identified as an independent factor that can be used for risk stratification and prediction of prognosis in patients with gastric cancer (GC). Overall survival (OS) is identified as the most important prognostic indicator for GC patients. However, the molecular mechanism of EMVI development and its potential relationship with OS in GC are not fully understood. In this radiogenomics‐based study, we sought to investigate the molecular mechanism underlying CT‐detected EMVI in patients with GC, and aimed to construct a genomic signature based on EMVI‐related genes with the goal of using this signature to predict the OS. Materials and Methods Whole mRNA genome sequencing of frozen tumor samples from 13 locally advanced GC patients was performed to identify EMVI‐related genes. EMVI‐prognostic hub genes were selected based on overlapping EMVI‐related differentially expressed genes and OS‐related genes, using a training cohort of 176 GC patients who were included in The Cancer Genome Atlas database. Another 174 GC patients from this database comprised the external validation cohort. A risk stratification model using a seven‐gene signature was constructed through the use of a least absolute shrinkage and selection operator Cox regression model. Results Patients with high risk score showed significantly reduced OS (training cohort, p = 1.143e‐04; validation cohort, p = 2.429e‐02). Risk score was an independent predictor of OS in multivariate Cox regression analyses (training cohort, HR = 2.758; 95% CI: 1.825–4.169; validation cohort, HR = 2.173; 95% CI: 1.347–3.505; p < 0.001 for both). Gene functions/pathways of the seven‐gene signature mainly included cell proliferation, cell adhesion, regulation of metal ion transport, and epithelial to mesenchymal transition. Conclusions A CT‐detected EMVI‐related gene model could be used to predict the prognosis in GC patients, potentially providing clinicians with additional information regarding appropriate therapeutic strategy and medical decision‐making.
Collapse
Affiliation(s)
- Bo Gao
- Department of General Surgery, Peking University People's Hospital, Beijing, China
| | - Caizhen Feng
- Department of Radiology, Peking University People's Hospital, Beijing, China
| | - Fan Chai
- Department of Radiology, Peking University People's Hospital, Beijing, China
| | - Shengcai Wei
- Department of Radiology, Peking University People's Hospital, Beijing, China
| | - Nan Hong
- Department of Radiology, Peking University People's Hospital, Beijing, China
| | - Yingjiang Ye
- Department of Gastrointestinal Surgery, Peking University People's Hospital, Beijing, China
| | - Yi Wang
- Department of Radiology, Peking University People's Hospital, Beijing, China
| | - Jin Cheng
- Department of Radiology, Peking University People's Hospital, Beijing, China
| |
Collapse
|
10
|
Kamili C, Kandoti HS, Radhakrishnan S, Konde A, Vattikutti UMR. Anti-angiogenic activity of chloride and potassium channel modulators: repurposing ion channel modulators. FUTURE JOURNAL OF PHARMACEUTICAL SCIENCES 2020. [DOI: 10.1186/s43094-020-00041-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
Excessive angiogenesis can be the root cause of many pathological conditions. Various types of ion channels are found on the endothelial cells. These ion channels play a vital role in the multi-stepped process of angiogenesis. The study aims to investigate the anti-angiogenic effects of specific ion channel modulators mefloquine (volume-regulated chloride channel blocker), lubiprostone (ClC-2 channel agonist), and 4-aminopyridine (voltage-gated potassium channel blocker).
Results
The anti-angiogenic activity of ion channel modulators was screened by measuring its effects on the area of neovascularization and histopathological studies by in vivo (corneal neovascularization) method and by in vitro assays, endothelial cell proliferation assay, cell migration assay, and matrigel cord-like morphogenesis assay. The test and standard drug (bevacizumab) groups were compared with the control group using one-way ANOVA, followed by post hoc test, and Dunnett’s test to compare the mean of all the groups with the control mean. The results revealed that mefloquine at the dose of 0.6% w/v and 1.0% w/v, lubiprostone at the dose of 0.5% w/v and 1.0% w/v, and 4-aminopyridine at the dose of 2% w/v and 4% w/v showed significant anti-angiogenic property. In the studies on human umbilical vein endothelial cells, the test drugs (100 nM) showed significant inhibition of proliferation, migration, and decrease in network length of cord-like tubes.
Conclusion
The scientific findings indicate that the test drugs have potent anti-angiogenic activity by inhibiting the cell proliferation, inhibiting the cell volume increase, arresting the cell cycle progression and by causing membrane hyperpolarization. The potent anti-angiogenic drugs obtained by repurposing these ion channel modulators, in the further studies, will be able to treat the diseases due to excess angiogenesis from the root cause.
Graphical abstract
Collapse
|
11
|
Kamili C, Kakaraparthy RS, Vattikuti UM. Anti-Angiogenic Activity of Flunarizine by In Ovo, In Vitro, and In Vivo Assays. Turk J Pharm Sci 2020; 16:303-309. [PMID: 32454728 DOI: 10.4274/tjps.galenos.2018.29981] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2018] [Accepted: 05/31/2018] [Indexed: 12/01/2022]
Abstract
Objectives The involvement of T-type calcium channels in cell proliferation and the role of sodium channels in cell migration have been extensively studied in angiogenesis. In the present study, flunarizine, a dual sodium/calcium channel blocker; was selected to evaluate its anti-angiogenic potential. This can be therapeutically beneficial in diseases caused by pathologically excessive angiogenesis. Materials and Methods The anti-angiogenic activity of ion channel blocker was screened by chick chorioallantoic membrane assay (in ovo), rat aortic ring assay, endothelial cell proliferation assay, transwell migration assay, Matrigel cord-like morphogenesis assay (in vitro), and sponge implantation method (in vivo). The anti-angiogenic activity of the test drug was compared with the standard anti-angiogenic drug bevacizumab and, in addition, the test responses were compared with the angiogenic factor vascular endothelial growth factor at a maximal concentration of 500 pM. Results All the groups were compared with the control group using one-way ANOVA, followed by a post hoc test, Dunnett's test, to compare the mean of all the groups with the control mean. In the chick chorioallantoic membrane assay, the number of branching points and angiogenic score were evaluated and significant results were observed at 10-5 M and 10-4 M. In the aortic ring assay a reduction in the area of sprouts was observed with 5-10 μM and significant reductions in the weight of sponges, number of blood vessels formed, and hemoglobin content were observed at all three tested concentrations of flunarizine in the sponge implantation method. In the studies on human umbilical vein endothelial cells the test drug (1-100 nM) showed significant inhibition of proliferation and migration and a decrease in the network length of cord-like tubes in a dose-dependent manner. Conclusion Flunarizine has significant anti-angiogenic action by inhibiting cell proliferation, migration, and cord-like tube formation, which resulted from blocking of the T-type calcium and sodium channels. Further studies on the structural modifications of flunarizine for repurposing this ion channel modulator will lead to treatment of the diseases due to excessive angiogenesis from the root cause.
Collapse
Affiliation(s)
| | - Ravi Shankar Kakaraparthy
- Sri Sai Aditya Institute of Pharmaceutical Sciences and Research, Department of Pharmacology, Surampalem, Andra Pradesh, India
| | | |
Collapse
|
12
|
Wang J, Zhang C, Wu Y, He W, Gou X. Identification and analysis of long non-coding RNA related miRNA sponge regulatory network in bladder urothelial carcinoma. Cancer Cell Int 2019; 19:327. [PMID: 31827401 PMCID: PMC6892182 DOI: 10.1186/s12935-019-1052-2] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Accepted: 11/27/2019] [Indexed: 12/11/2022] Open
Abstract
Background The aim of this study was to investigate the regulatory network of lncRNAs as competing endogenous RNAs (ceRNA) in bladder urothelial carcinoma (BUC) based on gene expression data derived from The Cancer Genome Atlas (TCGA). Materials and methods RNA sequence profiles and clinical information from 414 BUC tissues and 19 non-tumor adjacent tissues were downloaded from TCGA. Differentially expressed RNAs derived from BUC and non-tumor adjacent samples were identified using the R package “edgeR”. Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis was performed using the “clusterProfiler” package. Gene ontology and protein–protein interaction (PPI) networks were analyzed for the differentially expressed mRNAs using the “STRING” database. The network for the dysregulated lncRNA associated ceRNAs was then constructed for BUC using miRcode, miRTarBase, miRDB, and TargetScan. Cox regression analysis was performed to identify independent prognostic RNAs associated with BUC overall survival (OS). Survival analysis for the independent prognostic RNAs within the ceRNA network was calculated using Kaplan–Meier curves. Results Based on our analysis, a total of 666, 1819 and 157 differentially expressed lncRNAs, mRNAs and miRNAs were identified respectively. The ceRNA network was then constructed and contained 59 lncRNAs, 23 DEmiRNAs, and 52 DEmRNAs. In total, 5 lncRNAs (HCG22, ADAMTS9-AS1, ADAMTS9-AS2, AC078778.1, and AC112721.1), 2 miRNAs (hsa-mir-145 and hsa-mir-141) and 6 mRNAs (ZEB1, TMEM100, MAP1B, DUSP2, JUN, and AIFM3) were found to be related to OS. Two lncRNAs (ADAMTS9-AS1 and ADAMTS9-AS2) and 4 mRNA (DUSP2, JUN, MAP1B, and TMEM100) were validated using GEPIA. Thirty key hub genes were identified using the ranking method of degree. KEGG analysis demonstrated that the majority of the DEmRNAs were involved in pathways associated with cancer. Conclusion Our findings provide an understanding of the important role of lncRNA–related ceRNAs in BUC. Additional experimental and clinical validations are required to support our findings.
Collapse
Affiliation(s)
- Jiawu Wang
- 1Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Yuzhong District, Chongqing, China
| | - Chengyao Zhang
- 2Department of Head and Neck Cancer Center, Chongqing University Cancer Hospital & Chongqing Cancer Institute & Chongqing Cancer Hospital, Shapingba District, Chongqing, China
| | - Yan Wu
- 3Department of General Surgery, University-Town Hospital of Chongqing Medical University, Shapingba District, Chongqing, China
| | - Weiyang He
- 1Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Yuzhong District, Chongqing, China
| | - Xin Gou
- 1Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Yuzhong District, Chongqing, China
| |
Collapse
|
13
|
Prevarskaya N, Skryma R, Shuba Y. Ion Channels in Cancer: Are Cancer Hallmarks Oncochannelopathies? Physiol Rev 2018; 98:559-621. [PMID: 29412049 DOI: 10.1152/physrev.00044.2016] [Citation(s) in RCA: 311] [Impact Index Per Article: 44.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Genomic instability is a primary cause and fundamental feature of human cancer. However, all cancer cell genotypes generally translate into several common pathophysiological features, often referred to as cancer hallmarks. Although nowadays the catalog of cancer hallmarks is quite broad, the most common and obvious of them are 1) uncontrolled proliferation, 2) resistance to programmed cell death (apoptosis), 3) tissue invasion and metastasis, and 4) sustained angiogenesis. Among the genes affected by cancer, those encoding ion channels are present. Membrane proteins responsible for signaling within cell and among cells, for coupling of extracellular events with intracellular responses, and for maintaining intracellular ionic homeostasis ion channels contribute to various extents to pathophysiological features of each cancer hallmark. Moreover, tight association of these hallmarks with ion channel dysfunction gives a good reason to classify them as special type of channelopathies, namely oncochannelopathies. Although the relation of cancer hallmarks to ion channel dysfunction differs from classical definition of channelopathies, as disease states causally linked with inherited mutations of ion channel genes that alter channel's biophysical properties, in a broader context of the disease state, to which pathogenesis ion channels essentially contribute, such classification seems absolutely appropriate. In this review the authors provide arguments to substantiate such point of view.
Collapse
Affiliation(s)
- Natalia Prevarskaya
- INSERM U-1003, Equipe Labellisée par la Ligue Nationale contre le Cancer et LABEX, Université Lille1 , Villeneuve d'Ascq , France ; Bogomoletz Institute of Physiology and International Center of Molecular Physiology, NASU, Kyiv-24, Ukraine
| | - Roman Skryma
- INSERM U-1003, Equipe Labellisée par la Ligue Nationale contre le Cancer et LABEX, Université Lille1 , Villeneuve d'Ascq , France ; Bogomoletz Institute of Physiology and International Center of Molecular Physiology, NASU, Kyiv-24, Ukraine
| | - Yaroslav Shuba
- INSERM U-1003, Equipe Labellisée par la Ligue Nationale contre le Cancer et LABEX, Université Lille1 , Villeneuve d'Ascq , France ; Bogomoletz Institute of Physiology and International Center of Molecular Physiology, NASU, Kyiv-24, Ukraine
| |
Collapse
|
14
|
Endothelial Ca 2+ Signaling and the Resistance to Anticancer Treatments: Partners in Crime. Int J Mol Sci 2018; 19:ijms19010217. [PMID: 29324706 PMCID: PMC5796166 DOI: 10.3390/ijms19010217] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Revised: 01/08/2018] [Accepted: 01/10/2018] [Indexed: 02/06/2023] Open
Abstract
Intracellular Ca2+ signaling drives angiogenesis and vasculogenesis by stimulating proliferation, migration, and tube formation in both vascular endothelial cells and endothelial colony forming cells (ECFCs), which represent the only endothelial precursor truly belonging to the endothelial phenotype. In addition, local Ca2+ signals at the endoplasmic reticulum (ER)-mitochondria interface regulate endothelial cell fate by stimulating survival or apoptosis depending on the extent of the mitochondrial Ca2+ increase. The present article aims at describing how remodeling of the endothelial Ca2+ toolkit contributes to establish intrinsic or acquired resistance to standard anti-cancer therapies. The endothelial Ca2+ toolkit undergoes a major alteration in tumor endothelial cells and tumor-associated ECFCs. These include changes in TRPV4 expression and increase in the expression of P2X7 receptors, Piezo2, Stim1, Orai1, TRPC1, TRPC5, Connexin 40 and dysregulation of the ER Ca2+ handling machinery. Additionally, remodeling of the endothelial Ca2+ toolkit could involve nicotinic acetylcholine receptors, gasotransmitters-gated channels, two-pore channels and Na⁺/H⁺ exchanger. Targeting the endothelial Ca2+ toolkit could represent an alternative adjuvant therapy to circumvent patients' resistance to current anti-cancer treatments.
Collapse
|
15
|
Chen TJ, He HL, Shiue YL, Yang CC, Lin LC, Tian YF, Chen SH. High chloride channel accessory 1 expression predicts poor prognoses in patients with rectal cancer receiving chemoradiotherapy. Int J Med Sci 2018; 15:1171-1178. [PMID: 30123054 PMCID: PMC6097263 DOI: 10.7150/ijms.26685] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2018] [Accepted: 06/30/2018] [Indexed: 01/01/2023] Open
Abstract
Background: Concurrent chemoradiotherapy (CCRT) has now become the standard of treatments for advanced rectal cancer before surgery. To search the biological molecules with prognostic and therapeutic potential of CCRT could be beneficial for these patients. Recently, aberrant expression of chloride channels has been linked to radio-resistance in glioblastoma; however, its clinical implication has not been well-studied in rectal cancers. Therefore, we examined the clinical significance of targetable drivers associated with chloride channel activity in patients with rectal cancer receiving CCRT. Methods: After datamining from a published transcriptome of rectal cancers, upregulation of CLCA1 gene was recognized to be significantly correlated with non-responders of CCRT. In validation cohort of rectal cancers, the expression levels of CLCA1 were accessed by using immunohistochemistry assays in 172 tumor specimens that were obtained before any treatment. Expression levels of CLCA1 were statistically analyzed with principal clinicopathological features and survival outcomes in this substantial cohort. Results: In validation cohort, high expression of CLCA1 was significantly associated with higher pre-treatment tumor nodal stages (P=0.032), vascular invasion (P=0.028), and inferior tumor regression grade (P=0.042). In survival evaluations, high expression of CLCA1 was significantly correlated with worse local recurrence-free survival (LRFS; P=0.0012), metastasis-free survival (MeFS; P =0.0114), and disease-specific survival (DSS; P=0.0041). Furthermore, high expression of CLCA1 remained an independent prognosticator of shorter LRFS (P=0.029, hazard ratio=2.555), MeFS (P=0.044, hazard ratio=2.125) and DSS (P=0.044, hazard ratio=2.172). Conclusions: High expression of CLCA1 is significantly associated with poor therapeutic response and survival outcomes in rectal cancer patients with CCRT treatment before surgery. With the development of specific inhibitors, our findings indicate not only prognostic but also therapeutic potential of CLCA1 in rectal cancers.
Collapse
Affiliation(s)
- Tzu-Ju Chen
- Department of Pathology, Chi Mei Medical Center, Tainan, Taiwan.,Department of Optometry, Chung Hwa University of Medical Technology, Tainan, Taiwan.,Institute of Biomedical Science, National Sun Yat-sen University, Kaohsiung, Taiwan
| | - Hong-Lin He
- Department of Pathology, Chi Mei Medical Center, Tainan, Taiwan
| | - Yow-Ling Shiue
- Institute of Biomedical Science, National Sun Yat-sen University, Kaohsiung, Taiwan
| | - Ching-Chieh Yang
- Department of Radiation Oncology, Chi Mei Medical Center, Liouying, Tainan, Taiwan.,Institute of Biomedical Sciences, National Sun Yat-Sen University, Kaohsiung, Taiwan.,Department of Pharmacy, Chia-Nan University of Pharmacy and Science, Tainan, Taiwan
| | - Li-Ching Lin
- Department of Radiation Oncology, Chi Mei Medical Center, Liouying, Tainan, Taiwan
| | - Yu-Feng Tian
- Division of General Surgery, Department of Surgery, Chi Mei Medical Center, Tainan, Taiwan.,Department of Health & Nutrition, Chia Nan University of Pharmacy and Science, Tainan, Taiwan
| | - Shang-Hung Chen
- National Institute of Cancer Research, National Health Research Institutes, Tainan, Taiwan.,Division of Hematology and Oncology, Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| |
Collapse
|
16
|
Kamili C, Kakataparthy RS, Vattikutti UM, Chidrawar V, Ammineni S. Anti-proliferative and anti-angiogenic activities of ion-channel modulators: In-ovo , in-vitro and in-vivo study. Asian Pac J Trop Biomed 2017. [DOI: 10.1016/j.apjtb.2017.05.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
|
17
|
Harguindey S, Stanciu D, Devesa J, Alfarouk K, Cardone RA, Polo Orozco JD, Devesa P, Rauch C, Orive G, Anitua E, Roger S, Reshkin SJ. Cellular acidification as a new approach to cancer treatment and to the understanding and therapeutics of neurodegenerative diseases. Semin Cancer Biol 2017; 43:157-179. [PMID: 28193528 DOI: 10.1016/j.semcancer.2017.02.003] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Accepted: 02/06/2017] [Indexed: 12/27/2022]
Abstract
During the last few years, the understanding of the dysregulated hydrogen ion dynamics and reversed proton gradient of cancer cells has resulted in a new and integral pH-centric paradigm in oncology, a translational model embracing from cancer etiopathogenesis to treatment. The abnormalities of intracellular alkalinization along with extracellular acidification of all types of solid tumors and leukemic cells have never been described in any other disease and now appear to be a specific hallmark of malignancy. As a consequence of this intracellular acid-base homeostatic failure, the attempt to induce cellular acidification using proton transport inhibitors and other intracellular acidifiers of different origins is becoming a new therapeutic concept and selective target of cancer treatment, both as a metabolic mediator of apoptosis and in the overcoming of multiple drug resistance (MDR). Importantly, there is increasing data showing that different ion channels contribute to mediate significant aspects of cancer pH regulation and etiopathogenesis. Finally, we discuss the extension of this new pH-centric oncological paradigm into the opposite metabolic and homeostatic acid-base situation found in human neurodegenerative diseases (HNDDs), which opens novel concepts in the prevention and treatment of HNDDs through the utilization of a cohort of neural and non-neural derived hormones and human growth factors.
Collapse
Affiliation(s)
- Salvador Harguindey
- Institute of Clinical Biology and Metabolism, c) Postas 13, 01004 Vitoria, Spain.
| | - Daniel Stanciu
- Institute of Clinical Biology and Metabolism, c) Postas 13, 01004 Vitoria, Spain
| | - Jesús Devesa
- Department of Physiology, School of Medicine, University of Santiago de Compostela, Spain and Scientific Director of Foltra Medical Centre, Teo, Spain
| | - Khalid Alfarouk
- Al-Ghad International Colleges for Applied Medical Sciences, Al-Madinah Al-Munawarah, Saudi Arabia
| | - Rosa Angela Cardone
- Department of Biosciences, Biotechnology and Biopharmaceutics, University of Bari, Via E. Orabona 4, 70125 Bari, Italy
| | | | - Pablo Devesa
- Research and Development, Medical Centre Foltra, Teo, Spain
| | - Cyril Rauch
- School of Veterinary Medicine and Science, University of Nottingham,College Road, Sutton Bonington, LE12 5RD, UK
| | - Gorka Orive
- Laboratory of Pharmacy and Pharmaceutical Technology, Faculty of Pharmacy, University of the Basque Country, Networking Biomedical Research Center on Bioengineering, Biomaterials and Nanomedicine, CIBER-BBN, SLFPB-EHU, 01006 Vitoria, Spain
| | - Eduardo Anitua
- BTI Biotechnology Institute ImasD, S.L. C/Jacinto Quincoces, 39, 01007 Vitoria, Spain
| | - Sébastien Roger
- Inserm UMR1069, University François-Rabelais of Tours,10 Boulevard Tonnellé, 37032 Tours, France; Institut Universitaire de France, 1 Rue Descartes, Paris 75231, France
| | - Stephan J Reshkin
- Department of Biosciences, Biotechnology and Biopharmaceutics, University of Bari, Via E. Orabona 4, 70125 Bari, Italy
| |
Collapse
|
18
|
Tomita Y, Dorward H, Yool AJ, Smith E, Townsend AR, Price TJ, Hardingham JE. Role of Aquaporin 1 Signalling in Cancer Development and Progression. Int J Mol Sci 2017; 18:ijms18020299. [PMID: 28146084 PMCID: PMC5343835 DOI: 10.3390/ijms18020299] [Citation(s) in RCA: 84] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Revised: 01/19/2017] [Accepted: 01/23/2017] [Indexed: 02/07/2023] Open
Abstract
Cancer is a major health burden worldwide. Despite the advances in our understanding of its pathogenesis and continued improvement in cancer management and outcomes, there remains a strong clinical demand for more accurate and reliable biomarkers of metastatic progression and novel therapeutic targets to abrogate angiogenesis and tumour progression. Aquaporin 1 (AQP1) is a small hydrophobic integral transmembrane protein with a predominant role in trans-cellular water transport. Recently, over-expression of AQP1 has been associated with many types of cancer as a distinctive clinical prognostic factor. This has prompted researchers to evaluate the link between AQP1 and cancer biological functions. Available literature implicates the role of AQP1 in tumour cell migration, invasion and angiogenesis. This article reviews the current understanding of AQP1-facilitated tumour development and progression with a focus on regulatory mechanisms and downstream signalling pathways.
Collapse
Affiliation(s)
- Yoko Tomita
- Molecular Oncology, Basil Hetzel Institute, The Queen Elizabeth Hospital & Discipline of Physiology, School of Medicine, University of Adelaide, Adelaide, SA 5000, Australia.
| | - Hilary Dorward
- Molecular Oncology, Basil Hetzel Institute, The Queen Elizabeth Hospital, Woodville South, SA 5011, Australia.
| | - Andrea J Yool
- Discipline of Physiology, School of Medicine, University of Adelaide, Adelaide, SA 5000, Australia.
| | - Eric Smith
- Molecular Oncology, Basil Hetzel Institute, The Queen Elizabeth Hospital, Woodville South, SA 5011, Australia.
| | - Amanda R Townsend
- Medical Oncology, The Queen Elizabeth Hospital & School of Medicine, University of Adelaide, Adelaide, SA 5000, Australia.
| | - Timothy J Price
- Medical Oncology, The Queen Elizabeth Hospital & School of Medicine, University of Adelaide, Adelaide, SA 5000, Australia.
| | - Jennifer E Hardingham
- Molecular Oncology, Basil Hetzel Institute, The Queen Elizabeth Hospital & Discipline of Physiology, School of Medicine, University of Adelaide, Adelaide, SA 5000, Australia.
| |
Collapse
|
19
|
Peruzzo R, Biasutto L, Szabò I, Leanza L. Impact of intracellular ion channels on cancer development and progression. EUROPEAN BIOPHYSICS JOURNAL : EBJ 2016; 45:685-707. [PMID: 27289382 PMCID: PMC5045486 DOI: 10.1007/s00249-016-1143-0] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Revised: 05/13/2016] [Accepted: 05/17/2016] [Indexed: 12/13/2022]
Abstract
Cancer research is nowadays focused on the identification of possible new targets in order to try to develop new drugs for curing untreatable tumors. Ion channels have emerged as "oncogenic" proteins, since they have an aberrant expression in cancers compared to normal tissues and contribute to several hallmarks of cancer, such as metabolic re-programming, limitless proliferative potential, apoptosis-resistance, stimulation of neo-angiogenesis as well as cell migration and invasiveness. In recent years, not only the plasma membrane but also intracellular channels and transporters have arisen as oncological targets and were proposed to be associated with tumorigenesis. Therefore, the research is currently focusing on understanding the possible role of intracellular ion channels in cancer development and progression on one hand and, on the other, on developing new possible drugs able to modulate the expression and/or activity of these channels. In a few cases, the efficacy of channel-targeting drugs in reducing tumors has already been demonstrated in vivo in preclinical mouse models.
Collapse
Affiliation(s)
| | - Lucia Biasutto
- CNR Institute of Neuroscience, Padua, Italy
- Department of Biomedical Sciences, University of Padua, Padua, Italy
| | - Ildikò Szabò
- Department of Biology, University of Padua, Padua, Italy
- CNR Institute of Neuroscience, Padua, Italy
| | - Luigi Leanza
- Department of Biology, University of Padua, Padua, Italy.
| |
Collapse
|
20
|
Kovalenko I, Glasauer A, Schöckel L, Sauter DRP, Ehrmann A, Sohler F, Hägebarth A, Novak I, Christian S. Identification of KCa3.1 Channel as a Novel Regulator of Oxidative Phosphorylation in a Subset of Pancreatic Carcinoma Cell Lines. PLoS One 2016; 11:e0160658. [PMID: 27494181 PMCID: PMC4975431 DOI: 10.1371/journal.pone.0160658] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2015] [Accepted: 07/22/2016] [Indexed: 02/06/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) represents the most common form of pancreatic cancer with rising incidence in developing countries and overall 5-year survival rates of less than 5%. The most frequent mutations in PDAC are gain-of-function mutations in KRAS as well as loss-of-function mutations in p53. Both mutations have severe impacts on the metabolism of tumor cells. Many of these metabolic changes are mediated by transporters or channels that regulate the exchange of metabolites and ions between the intracellular compartment and the tumor microenvironment. In the study presented here, our goal was to identify novel transporters or channels that regulate oxidative phosphorylation (OxPhos) in PDAC in order to characterize novel potential drug targets for the treatment of these cancers. We set up a Seahorse Analyzer XF based siRNA screen and identified previously described as well as novel regulators of OxPhos. The siRNA that resulted in the greatest change in cellular oxygen consumption was targeting the KCNN4 gene, which encodes for the Ca2+-sensitive K+ channel KCa3.1. This channel has not previously been reported to regulate OxPhos. Knock-down experiments as well as the use of a small molecule inhibitor confirmed its role in regulating oxygen consumption, ATP production and cellular proliferation. Furthermore, PDAC cell lines sensitive to KCa3.1 inhibition were shown to express the channel protein in the plasma membrane as well as in the mitochondria. These differences in the localization of KCa3.1 channels as well as differences in the regulation of cellular metabolism might offer opportunities for targeted therapy in subsets of PDAC.
Collapse
Affiliation(s)
- Ilya Kovalenko
- Drug Discovery, Therapeutic Research Groups / Onc II, Bayer Pharma AG, Berlin, Germany
- Department of Bioanalytics, Institute of Biotechnology, Technische Universität Berlin, Berlin, Germany
| | - Andrea Glasauer
- Drug Discovery, Therapeutic Research Groups / Onc II, Bayer Pharma AG, Berlin, Germany
| | - Laura Schöckel
- Drug Discovery, Therapeutic Research Groups / Onc II, Bayer Pharma AG, Berlin, Germany
| | - Daniel R. P. Sauter
- Section for Cell Biology and Physiology, Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Alexander Ehrmann
- Drug Discovery, Lead Discovery / Cell Biology, Bayer Pharma AG, Berlin, Germany
| | - Florian Sohler
- Drug Discovery, Therapeutic Research Groups / Bioinformatics, Bayer Pharma AG, Leverkusen, Germany
| | - Andrea Hägebarth
- Drug Discovery, Therapeutic Research Groups / Onc II, Bayer Pharma AG, Berlin, Germany
| | - Ivana Novak
- Section for Cell Biology and Physiology, Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Sven Christian
- Drug Discovery, Therapeutic Research Groups / Onc II, Bayer Pharma AG, Berlin, Germany
- * E-mail:
| |
Collapse
|
21
|
Pharmacological targeting of ion channels for cancer therapy: In vivo evidences. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2016; 1863:1385-97. [DOI: 10.1016/j.bbamcr.2015.11.032] [Citation(s) in RCA: 85] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Revised: 11/25/2015] [Accepted: 11/26/2015] [Indexed: 12/29/2022]
|
22
|
Ghanghoria R, Tekade RK, Mishra AK, Chuttani K, Jain NK. Luteinizing hormone-releasing hormone peptide tethered nanoparticulate system for enhanced antitumoral efficacy of paclitaxel. Nanomedicine (Lond) 2016; 11:797-816. [DOI: 10.2217/nnm.16.19] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Aim: Paclitaxel (PTX) is an effective anticancer agent used in the therapy of a wide variety of cancers. However, the drug is difficult to formulate due to its low solubility, and therefore, it is administered under slow infusion with castor oil/ethanol solution as surfactant that causes serious side effects. This investigation investigates leutinizing hormone releasing hormone (LHRH)-tethered nanparticulate system as modality for cancer-specific delivery of PTX and therefore minimizing the adverse effects. Materials & methods: LHRH-tethered poly(lactic-co-glycolic acid) copolymer with poly ethylene glycol side chain was synthesized, characterized and employed to formulate PTX-loaded nanoparticulate system. Results & conclusion: The developed nanoparticulate appears to be proficient in carrying as well as targeted delivery of PTX with improved therapeutic efficacy and better safety.
Collapse
Affiliation(s)
- Raksha Ghanghoria
- Pharmaceutics Research Laboratory, Department of Pharmaceutical Sciences, Dr. Hari Singh Gour University, Sagar 470003 MP, India
| | - Rakesh Kumar Tekade
- The University of Texas Southwestern Medical Center, Advanced Imaging Research Centre, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA
- The International Medical University, School of Pharmacy, Department of Pharmaceutical Technology, Jalan Jalil Perkasa 19, 57000 Kuala Lumpur, Malaysia
| | - Anil Kumar Mishra
- Division of Cyclotron & Radiopharmaceutical Sciences, Molecular Imaging Research Center, Institute of Nuclear Medicine & Allied Sciences (INMAS), DRDO, 110054 Delhi, India
| | - Krishna Chuttani
- Division of Cyclotron & Radiopharmaceutical Sciences, Molecular Imaging Research Center, Institute of Nuclear Medicine & Allied Sciences (INMAS), DRDO, 110054 Delhi, India
| | - Narendra Kumar Jain
- Pharmaceutics Research Laboratory, Department of Pharmaceutical Sciences, Dr. Hari Singh Gour University, Sagar 470003 MP, India
| |
Collapse
|
23
|
Martial S. Involvement of ion channels and transporters in carcinoma angiogenesis and metastasis. Am J Physiol Cell Physiol 2016; 310:C710-27. [PMID: 26791487 DOI: 10.1152/ajpcell.00218.2015] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Angiogenesis is a finely tuned process, which is the result of the equilibrium between pro- and antiangiogenic factors. In solid tumor angiogenesis, the balance is highly in favor of the production of new, but poorly functional blood vessels, initially intended to provide growing tumors with nutrients and oxygen. Among the numerous proteins involved in tumor development, several types of ion channels are overexpressed in tumor cells, as well as in stromal and endothelial cells. Ion channels thus actively participate in the different hallmarks of cancer, especially in tumor angiogenesis and metastasis. Indeed, from their strategic localization in the plasma membrane, ion channels are key operators of cell signaling, as they sense and respond to environmental changes. This review aims to decipher how ion channels of different families are intricately involved in the fundamental angiogenesis and metastasis hallmarks, which lead from a nascent tumor to systemic dissemination. An overview of the possible use of ion channels as therapeutic targets will also be given, showing that ion channel inhibitors or specific antibodies may provide effective tools, in the near future, in the treatment of carcinomas.
Collapse
Affiliation(s)
- Sonia Martial
- Institut de Recherche sur le Cancer et le Vieillissement, CNRS UMR 7284, Inserm U1081, Université Nice-Sophia Antipolis, Nice, France
| |
Collapse
|
24
|
Hervé JC. Membrane channels and transporters in cancers. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2015; 1848:2473-6. [PMID: 26100062 DOI: 10.1016/j.bbamem.2015.06.017] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Affiliation(s)
- Jean Claude Hervé
- INSERM U1082, Université de Poitiers, Pôle Biologie Santé Bâtiment B36, 1 rue Georges Bonnet, 86073 Poitiers Cédex, France.
| |
Collapse
|
25
|
Moccia F, Zuccolo E, Soda T, Tanzi F, Guerra G, Mapelli L, Lodola F, D'Angelo E. Stim and Orai proteins in neuronal Ca(2+) signaling and excitability. Front Cell Neurosci 2015; 9:153. [PMID: 25964739 PMCID: PMC4408853 DOI: 10.3389/fncel.2015.00153] [Citation(s) in RCA: 132] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2015] [Accepted: 04/03/2015] [Indexed: 02/01/2023] Open
Abstract
Stim1 and Orai1 are ubiquitous proteins that have long been known to mediate Ca2+ release-activated Ca2+ (CRAC) current (ICRAC) and store-operated Ca2+ entry (SOCE) only in non-excitable cells. SOCE is activated following the depletion of the endogenous Ca2+ stores, which are mainly located within the endoplasmic reticulum (ER), to replete the intracellular Ca2+ reservoir and engage specific Ca2+-dependent processes, such as proliferation, migration, cytoskeletal remodeling, and gene expression. Their paralogs, Stim2, Orai2 and Orai3, support SOCE in heterologous expression systems, but their physiological role is still obscure. Ca2+ inflow in neurons has long been exclusively ascribed to voltage-operated and receptor-operated channels. Nevertheless, recent work has unveiled that Stim1–2 and Orai1-2, but not Orai3, proteins are also expressed and mediate SOCE in neurons. Herein, we survey current knowledge about the neuronal distribution of Stim and Orai proteins in rodent and human brains; we further discuss that Orai2 is the main pore-forming subunit of CRAC channels in central neurons, in which it may be activated by either Stim1 or Stim2 depending on species, brain region and physiological stimuli. We examine the functions regulated by SOCE in neurons, where this pathway is activated under resting conditions to refill the ER, control spinogenesis and regulate gene transcription. Besides, we highlighted the possibility that SOCE also controls neuronal excitation and regulate synaptic plasticity. Finally, we evaluate the involvement of Stim and Orai proteins in severe neurodegenerative and neurological disorders, such as Alzheimer’s disease and epilepsy.
Collapse
Affiliation(s)
- Francesco Moccia
- Laboratory of General Physiology, Department of Biology and Biotechnology "Lazzaro Spallanzani", University of Pavia Pavia, Italy
| | - Estella Zuccolo
- Laboratory of General Physiology, Department of Biology and Biotechnology "Lazzaro Spallanzani", University of Pavia Pavia, Italy
| | - Teresa Soda
- Neurophysiology Unit, Department of Brain and Behavioral Sciences, University of Pavia Pavia, Italy
| | - Franco Tanzi
- Laboratory of General Physiology, Department of Biology and Biotechnology "Lazzaro Spallanzani", University of Pavia Pavia, Italy
| | - Germano Guerra
- Department of Medicine and Health Sciences, University of Molise Campobasso, Italy
| | - Lisa Mapelli
- Neurophysiology Unit, Department of Brain and Behavioral Sciences, University of Pavia Pavia, Italy ; Museo Storico della Fisica e Centro di Studi e Ricerche Enrico Fermi Roma, Italy
| | - Francesco Lodola
- Laboratory of Molecular Cardiology, IRCCS Fondazione Salvatore Maugeri Pavia, Italy
| | - Egidio D'Angelo
- Neurophysiology Unit, Department of Brain and Behavioral Sciences, University of Pavia Pavia, Italy ; Brain Connectivity Center, C. Mondino National Neurological Institute, Fondazione IRCCS Policlinico San Matteo Pavia Pavia, Italy
| |
Collapse
|