1
|
Singh A, Choudhury SD, Singh P, Singh VV, Singh SN, Sharma A. Ionic reverberation modulates the cellular fate of CD8 +tissue resident memory T cells (TRMs) in patients with renal cell carcinoma: A novel mechanism. Clin Immunol 2024; 264:110256. [PMID: 38762062 DOI: 10.1016/j.clim.2024.110256] [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: 03/02/2024] [Revised: 05/02/2024] [Accepted: 05/14/2024] [Indexed: 05/20/2024]
Abstract
In metastatic renal cell carcinoma (mRCC), existing treatments including checkpoint inhibitors are failed to cure and/or prevent recurrence of the disease. Therefore, in-depth understanding of tumor tissue resident memory T cells (TRMs) dysfunction are necessitated to enrich efficacy of immunotherapies and increasing disease free survival in treated patients. In patients, we observed dysregulation of K+, Ca2+, Na2+ and Zn2+ ion channels leads to excess infiltration of their respective ions in tumor TRMs, thus ionic gradients are disturbed and cells became hyperpolarized. Moreover, overloaded intramitochondrial calcium caused mitochondrial depolarization and trigger apoptosis of tumor TRMs. Decreased prevalence of activated tumor TRMs reflected our observations. Furthermore, disruptions in ionic concentrations impaired the functional activities and/or suppressed anti-tumor action of circulating and tumor TRMs in RCC. Collectively, these findings revealed novel mechanism behind dysfunctionality of tumor TRMs. Implicating enrichment of activated TRMs within tumor would be beneficial for better management of RCC patients.
Collapse
Affiliation(s)
- Ashu Singh
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - Saumitra Dey Choudhury
- Central core Research facility, All India Institute of Medical Sciences, New Delhi, India
| | - Prabhjot Singh
- Department of Urology, All India Institute of Medical Sciences, New Delhi, India
| | | | - Som Nath Singh
- Defence Institute of Physiology and Allied Sciences, New Delhi, India
| | - Alpana Sharma
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India.
| |
Collapse
|
2
|
Ai LJ, Li GD, Chen G, Sun ZQ, Zhang JN, Liu M. Molecular subtyping and the construction of a predictive model of colorectal cancer based on ion channel genes. Eur J Med Res 2024; 29:219. [PMID: 38576045 PMCID: PMC10993535 DOI: 10.1186/s40001-024-01819-2] [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: 11/28/2023] [Accepted: 03/29/2024] [Indexed: 04/06/2024] Open
Abstract
PURPOSE Colorectal cancer (CRC) is a highly heterogeneous malignancy with an unfavorable prognosis. The purpose of this study was to address the heterogeneity of CRC by categorizing it into ion channel subtypes, and to develop a predictive modeling based on ion channel genes to predict the survival and immunological states of patients with CRC. The model will provide guidance for personalized immunotherapy and drug treatment. METHODS A consistent clustering method was used to classify 619 CRC samples based on the expression of 279 ion channel genes. Such a method was allowed to investigate the relationship between molecular subtypes, prognosis, and immune infiltration. Furthermore, a predictive modeling was constructed for ion channels to evaluate the ion channel properties of individual tumors using the least absolute shrinkage and selection operator. The expression patterns of the characteristic genes were validated through molecular biology experiments. The effect of potassium channel tetramerization domain containing 9 (KCTD9) on CRC was verified by cellular functional experiments. RESULTS Four distinct ion channel subtypes were identified in CRC, each characterized by unique prognosis and immune infiltration patterns. Notably, Ion Cluster3 exhibited high levels of immune infiltration and a favorable prognosis, while Ion Cluster4 showed relatively lower levels of immune infiltration and a poorer prognosis. The ion channel score could predict overall survival, with lower scores correlated with longer survival. This score served as an independent prognostic factor and presented an excellent predictive efficacy in the nomogram. In addition, the score was closely related to immune infiltration, immunotherapy response, and chemotherapy sensitivity. Experimental evidence further confirmed that low expression of KCTD9 in tumor tissues was associated with an unfavorable prognosis in patients with CRC. The cellular functional experiments demonstrated that KCTD9 inhibited the proliferation, migration and invasion capabilities of LOVO cells. CONCLUSIONS Ion channel subtyping and scoring can effectively predict the prognosis and evaluate the immune microenvironment, immunotherapy response, and drug sensitivity in patients with CRC.
Collapse
Affiliation(s)
- Lian-Jie Ai
- Colorectal Tumor Surgery, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, 150001, Heilongjiang, China
| | - Guo-Dong Li
- General Surgery, The 4th Affiliated Hospital of Harbin Medical University, Harbin, 150001, Heilongjiang, China
| | - Gang Chen
- General Surgery, The 4th Affiliated Hospital of Harbin Medical University, Harbin, 150001, Heilongjiang, China
| | - Zi-Quan Sun
- Colorectal Tumor Surgery, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, 150001, Heilongjiang, China
| | - Jin-Ning Zhang
- Colorectal Tumor Surgery, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, 150001, Heilongjiang, China
| | - Ming Liu
- General Surgery, The 4th Affiliated Hospital of Harbin Medical University, Harbin, 150001, Heilongjiang, China.
| |
Collapse
|
3
|
Jusztus V, Medyouni G, Bagosi A, Lampé R, Panyi G, Matolay O, Maka E, Krasznai ZT, Vörös O, Hajdu P. Activity of Potassium Channels in CD8 + T Lymphocytes: Diagnostic and Prognostic Biomarker in Ovarian Cancer? Int J Mol Sci 2024; 25:1949. [PMID: 38396628 PMCID: PMC10888402 DOI: 10.3390/ijms25041949] [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: 11/30/2023] [Revised: 01/31/2024] [Accepted: 02/03/2024] [Indexed: 02/25/2024] Open
Abstract
CD8+ T cells play a role in the suppression of tumor growth and immunotherapy. Ion channels control the Ca2+-dependent function of CD8+ lymphocytes such as cytokine/granzyme production and tumor killing. Kv1.3 and KCa3.1 K+ channels stabilize the negative membrane potential of T cells to maintain Ca2+ influx through CRAC channels. We assessed the expression of Kv1.3, KCa3.1 and CRAC in CD8+ cells from ovarian cancer (OC) patients (n = 7). We found that the expression level of Kv1.3 was higher in patients with malignant tumors than in control or benign tumor groups while the KCa3.1 activity was lower in the malignant tumor group as compared to the others. We demonstrated that the Ca2+ response in malignant tumor patients is higher compared to control groups. We propose that altered Kv1.3 and KCa3.1 expression in CD8+ cells in OC could be a reporter and may serve as a biomarker in diagnostics and that increased Ca2+ response through CRAC may contribute to the impaired CD8+ function.
Collapse
Affiliation(s)
- Vivien Jusztus
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, Egyetem tér 1., H-4032 Debrecen, Hungary; (V.J.); (G.M.); (A.B.); (G.P.); (O.V.)
| | - Ghofrane Medyouni
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, Egyetem tér 1., H-4032 Debrecen, Hungary; (V.J.); (G.M.); (A.B.); (G.P.); (O.V.)
| | - Adrienn Bagosi
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, Egyetem tér 1., H-4032 Debrecen, Hungary; (V.J.); (G.M.); (A.B.); (G.P.); (O.V.)
| | - Rudolf Lampé
- Department of Gynecology and Obstetrics, Faculty of Medicine, University of Debrecen, Egyetem tér 1., H-4032 Debrecen, Hungary; (R.L.); (O.M.); (E.M.); (Z.T.K.)
| | - György Panyi
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, Egyetem tér 1., H-4032 Debrecen, Hungary; (V.J.); (G.M.); (A.B.); (G.P.); (O.V.)
| | - Orsolya Matolay
- Department of Gynecology and Obstetrics, Faculty of Medicine, University of Debrecen, Egyetem tér 1., H-4032 Debrecen, Hungary; (R.L.); (O.M.); (E.M.); (Z.T.K.)
| | - Eszter Maka
- Department of Gynecology and Obstetrics, Faculty of Medicine, University of Debrecen, Egyetem tér 1., H-4032 Debrecen, Hungary; (R.L.); (O.M.); (E.M.); (Z.T.K.)
| | - Zoárd Tibor Krasznai
- Department of Gynecology and Obstetrics, Faculty of Medicine, University of Debrecen, Egyetem tér 1., H-4032 Debrecen, Hungary; (R.L.); (O.M.); (E.M.); (Z.T.K.)
| | - Orsolya Vörös
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, Egyetem tér 1., H-4032 Debrecen, Hungary; (V.J.); (G.M.); (A.B.); (G.P.); (O.V.)
| | - Péter Hajdu
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, Egyetem tér 1., H-4032 Debrecen, Hungary; (V.J.); (G.M.); (A.B.); (G.P.); (O.V.)
- Division of Dental Biochemistry, Department of Basic Medical Sciences, Faculty of Dentistry, University of Debrecen, Egyetem tér 1., H-4032 Debrecen, Hungary
| |
Collapse
|
4
|
Navarro-Pérez M, Capera J, Benavente-Garcia A, Cassinelli S, Colomer-Molera M, Felipe A. Kv1.3 in the spotlight for treating immune diseases. Expert Opin Ther Targets 2024; 28:67-82. [PMID: 38316438 DOI: 10.1080/14728222.2024.2315021] [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: 09/28/2023] [Accepted: 02/02/2024] [Indexed: 02/07/2024]
Abstract
INTRODUCTION Kv1.3 is the main voltage-gated potassium channel of leukocytes from both the innate and adaptive immune systems. Channel function is required for common processes such as Ca2+ signaling but also for cell-specific events. In this context, alterations in Kv1.3 are associated with multiple immune disorders. Excessive channel activity correlates with numerous autoimmune diseases, while reduced currents result in increased cancer prevalence and immunodeficiencies. AREAS COVERED This review offers a general view of the role of Kv1.3 in every type of leukocyte. Moreover, diseases stemming from dysregulations of the channel are detailed, as well as current advances in their therapeutic research. EXPERT OPINION Kv1.3 arises as a potential immune target in a variety of diseases. Several lines of research focused on channel modulation have yielded positive results. However, among the great variety of specific channel blockers, only one has reached clinical trials. Future investigations should focus on developing simpler administration routes for channel inhibitors to facilitate their entrance into clinical trials. Prospective Kv1.3-based treatments will ensure powerful therapies while minimizing undesired side effects.
Collapse
Affiliation(s)
- María Navarro-Pérez
- Molecular Physiology Laboratory, Departament de Bioquímica i Biomedicina Molecular, Institut de Biomedicina (IBUB), Universitat de Barcelona, Barcelona, Spain
| | - Jesusa Capera
- Molecular Physiology Laboratory, Departament de Bioquímica i Biomedicina Molecular, Institut de Biomedicina (IBUB), Universitat de Barcelona, Barcelona, Spain
- The Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics Rheumatology & Musculoskeletal Sciences, University of Oxford, Oxford, UK
| | - Anna Benavente-Garcia
- Molecular Physiology Laboratory, Departament de Bioquímica i Biomedicina Molecular, Institut de Biomedicina (IBUB), Universitat de Barcelona, Barcelona, Spain
| | - Silvia Cassinelli
- Molecular Physiology Laboratory, Departament de Bioquímica i Biomedicina Molecular, Institut de Biomedicina (IBUB), Universitat de Barcelona, Barcelona, Spain
| | - Magalí Colomer-Molera
- Molecular Physiology Laboratory, Departament de Bioquímica i Biomedicina Molecular, Institut de Biomedicina (IBUB), Universitat de Barcelona, Barcelona, Spain
| | - Antonio Felipe
- Molecular Physiology Laboratory, Departament de Bioquímica i Biomedicina Molecular, Institut de Biomedicina (IBUB), Universitat de Barcelona, Barcelona, Spain
| |
Collapse
|
5
|
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: 0] [Impact Index Per Article: 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
|
6
|
Li M, Tian P, Zhao Q, Ma X, Zhang Y. Potassium channels: Novel targets for tumor diagnosis and chemoresistance. Front Oncol 2023; 12:1074469. [PMID: 36703789 PMCID: PMC9872028 DOI: 10.3389/fonc.2022.1074469] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Accepted: 12/21/2022] [Indexed: 01/11/2023] Open
Abstract
In recent years, the role of potassium channels in tumors has been intensively studied. Potassium channel proteins are widely involved in various physiological and pathological processes of cells. The expression and dysfunction of potassium channels are closely related to tumor progression. Potassium channel blockers or activators present antitumor effects by directly inhibiting tumor growth or enhancing the potency of classical antitumor agents in combination therapy. This article reviews the mechanisms by which potassium channels contribute to tumor development in various tumors in recent years, introduces the potential of potassium channels as diagnostic targets and therapeutic means for tumors, and provides further ideas for the proper individualized treatment of tumors.
Collapse
Affiliation(s)
- Meizeng Li
- School of Basic Medical Science, Weifang Medical University, Weifang, China
| | - Peijie Tian
- School of Basic Medical Science, Weifang Medical University, Weifang, China
| | - Qing Zhao
- School of Basic Medical Science, Weifang Medical University, Weifang, China
| | - Xialin Ma
- School of Basic Medical Science, Weifang Medical University, Weifang, China
| | - Yunxiang Zhang
- Department of Pathology, Weifang People’ s Hospital, Weifang, China,*Correspondence: Yunxiang Zhang,
| |
Collapse
|
7
|
Chimote AA, Alshwimi AO, Chirra M, Gawali VS, Powers-Fletcher MV, Hudock KM, Conforti L. Immune and ionic mechanisms mediating the effect of dexamethasone in severe COVID-19. Front Immunol 2023; 14:1143350. [PMID: 37033961 PMCID: PMC10080085 DOI: 10.3389/fimmu.2023.1143350] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Accepted: 03/15/2023] [Indexed: 04/11/2023] Open
Abstract
Introduction Severe COVID-19 is characterized by cytokine storm, an excessive production of proinflammatory cytokines that contributes to acute lung damage and death. Dexamethasone is routinely used to treat severe COVID-19 and has been shown to reduce patient mortality. However, the mechanisms underlying the beneficial effects of dexamethasone are poorly understood. Methods We conducted transcriptomic analysis of peripheral blood mononuclear cells (PBMCs) from COVID-19 patients with mild disease, and patients with severe COVID-19 with and without dexamethasone treatment. We then treated healthy donor PBMCs in vitro with dexamethasone and investigated the effects of dexamethasone treatment ion channel abundance (by RT-qPCR and flow cytometry) and function (by electrophysiology, Ca2+ influx measurements and cytokine release) in T cells. Results We observed that dexamethasone treatment in severe COVID-19 inhibited pro-inflammatory and immune exhaustion pathways, circulating cytotoxic and Th1 cells, interferon (IFN) signaling, genes involved in cytokine storm, and Ca2+ signaling. Ca2+ influx is regulated by Kv1.3 potassium channels, but their role in COVID-19 pathogenesis remains elusive. Kv1.3 mRNA was increased in PBMCs of severe COVID-19 patients, and was significantly reduced in the dexamethasone-treated group. In agreement with these findings, in vitro treatment of healthy donor PBMCs with dexamethasone reduced Kv1.3 abundance in T cells and CD56dimNK cells. Furthermore, functional studies showed that dexamethasone treatment significantly reduced Kv1.3 activity, Ca2+ influx and IFN-g production in T cells. Conclusion Our findings suggest that dexamethasone attenuates inflammatory cytokine release via Kv1.3 suppression, and this mechanism contributes to dexamethasone-mediated immunosuppression in severe COVID-19.
Collapse
Affiliation(s)
- Ameet A. Chimote
- Department of Internal Medicine, Division of Nephrology, University of Cincinnati, Cincinnati, OH, United States
| | - Abdulaziz O. Alshwimi
- Department of Internal Medicine, Division of Nephrology, University of Cincinnati, Cincinnati, OH, United States
| | - Martina Chirra
- Department of Internal Medicine, Division of Nephrology, University of Cincinnati, Cincinnati, OH, United States
| | - Vaibhavkumar S. Gawali
- Department of Internal Medicine, Division of Nephrology, University of Cincinnati, Cincinnati, OH, United States
| | - Margaret V. Powers-Fletcher
- Department of Internal Medicine, Division of Infectious Diseases, University of Cincinnati, Cincinnati, OH, United States
| | - Kristin M. Hudock
- Department of Internal Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, University of Cincinnati, Cincinnati, OH, United States
- Department of Pediatrics, Division of Pulmonary Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States
| | - Laura Conforti
- Department of Internal Medicine, Division of Nephrology, University of Cincinnati, Cincinnati, OH, United States
- *Correspondence: Laura Conforti,
| |
Collapse
|
8
|
Han Y, Shi Y, Chen B, Wang J, Liu Y, Sheng S, Fu Z, Shen C, Wang X, Yin S, Li H. An ion-channel-gene-based prediction model for head and neck squamous cell carcinoma: Prognostic assessment and treatment guidance. Front Immunol 2022; 13:961695. [PMID: 36389709 PMCID: PMC9650652 DOI: 10.3389/fimmu.2022.961695] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2022] [Accepted: 10/12/2022] [Indexed: 09/18/2023] Open
Abstract
PURPOSE Head and neck squamous cell carcinoma (HNSCC) is a very diverse malignancy with a poor prognosis. The purpose of this study was to develop a new signature based on 12 ion channel genes to predict the outcome and immune status of HNSCC patients. METHODS Clinicopathological information and gene sequencing data of HNSCC patients were generated from the Cancer Genome Atlas and Gene Expression Omnibus databases. A set of 323 ion channel genes was obtained from the HUGO Gene Nomenclature Committee database and literature review. Using univariate Cox regression analysis, the ion channel genes related to HNSCC prognosis were identified. A prognostic signature and nomogram were then created using machine learning methods. Kaplan-Meier analysis was used to explore the relevance of the risk scores and overall survival (OS). We also investigated the association between risk scores, tumor immune infiltration, and gene mutational status. Finally, we detected the expression levels of the signature genes by quantitative real-time polymerase chain reaction, western blotting, and immunohistochemistry. RESULTS We separated the patients into high- and low-risk groups according to the risk scores computed based on these 12 ion channel genes, and the OS of the low-risk group was significantly longer (p<0.001). The area under the curve for predicting 3-year survival was 0.729. Univariate and multivariate analyses showed that the 12-ion-channel-gene risk model was an independent prognostic factor. We also developed a nomogram model based on risk scores and clinicopathological variables to forecast outcomes. Furthermore, immune cell infiltration, gene mutation status, immunotherapy response, and chemotherapeutic treatment sensitivity were all linked to risk scores. Moreover, high expression levels of ANO1, AQP9, and BEST2 were detected in HNSCC tissues, whereas AQP5, SCNN1G, and SCN4A expression was low in HNSCC tissues, as determined by experiments. CONCLUSION The 12-ion-channel-gene prognostic signatures have been demonstrated to be highly efficient in predicting the prognosis, immune microenvironment, gene mutation status, immunotherapy response, and chemotherapeutic sensitivity of HNSCC patients.
Collapse
Affiliation(s)
- Yanxun Han
- Department of Otolaryngology, Head and Neck Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Anhui Medical University, Hefei, Anhui, China
| | - Yangyang Shi
- Department of Emergency Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Bangjie Chen
- Anhui Medical University, Hefei, Anhui, China
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | | | - Yuchen Liu
- Department of Otolaryngology, Head and Neck Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Anhui Medical University, Hefei, Anhui, China
| | | | - Ziyue Fu
- Anhui Medical University, Hefei, Anhui, China
| | | | - Xinyi Wang
- Anhui Medical University, Hefei, Anhui, China
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Siyue Yin
- Anhui Medical University, Hefei, Anhui, China
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Haiwen Li
- Department of Gastroenterology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
9
|
Imaging Effector Memory T-Cells Predicts Response to PD1-Chemotherapy Combinations in Colon Cancer. Biomedicines 2022; 10:biomedicines10102343. [PMID: 36289605 PMCID: PMC9598730 DOI: 10.3390/biomedicines10102343] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 09/14/2022] [Accepted: 09/18/2022] [Indexed: 11/16/2022] Open
Abstract
Often, patients fail to respond to immune checkpoint inhibitor (ICI) treatment despite favourable biomarker status. Numerous chemotherapeutic agents have been shown to promote tumour immunogenicity when used in conjunction with ICIs; however, little is known about whether such combination therapies lead to a lasting immune response. Given the potential toxicity of ICI–chemotherapy combinations, identification of biomarkers that accurately predict how individuals respond to specific treatment combinations and whether these responses will be long lasting is of paramount importance. In this study, we explored [18F]AlF-NOTA-KCNA3P, a peptide radiopharmaceutical that targets the Kv1.3 potassium channel overexpressed on T-effector memory (TEM) cells as a PET imaging biomarker for lasting immunological memory response. The first-line colon cancer chemotherapies oxaliplatin and 5-fluorouracil were assessed in a syngeneic colon cancer model, either as monotherapies or in combination with PD1, comparing radiopharmaceutical uptake to memory-associated immune cells in the tumour. [18F]AlF-NOTA-KCNA3P reliably separated tumours with immunological memory responses from non-responding tumours and could be used to measure Kv1.3-expressing TEM cells responsible for durable immunological memory response to combination therapy in vivo.
Collapse
|
10
|
How the Potassium Channel Response of T Lymphocytes to the Tumor Microenvironment Shapes Antitumor Immunity. Cancers (Basel) 2022; 14:cancers14153564. [PMID: 35892822 PMCID: PMC9330401 DOI: 10.3390/cancers14153564] [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: 07/06/2022] [Revised: 07/18/2022] [Accepted: 07/20/2022] [Indexed: 12/10/2022] Open
Abstract
Competent antitumor immune cells are fundamental for tumor surveillance and combating active cancers. Once established, tumors generate a tumor microenvironment (TME) consisting of complex cellular and metabolic elements that serve to suppress the function of antitumor immune cells. T lymphocytes are key cellular elements of the TME. In this review, we explore the role of ion channels, particularly K+ channels, in mediating the suppressive effects of the TME on T cells. First, we will review the complex network of ion channels that mediate Ca2+ influx and control effector functions in T cells. Then, we will discuss how multiple features of the TME influence the antitumor capabilities of T cells via ion channels. We will focus on hypoxia, adenosine, and ionic imbalances in the TME, as well as overexpression of programmed cell death ligand 1 by cancer cells that either suppress K+ channels in T cells and/or benefit from regulating these channels’ activity, ultimately shaping the immune response. Finally, we will review some of the cancer treatment implications related to ion channels. A better understanding of the effects of the TME on ion channels in T lymphocytes could promote the development of more effective immunotherapies, especially for resistant solid malignancies.
Collapse
|
11
|
Store-Operated Ca2+ Entry Is Up-Regulated in Tumour-Infiltrating Lymphocytes from Metastatic Colorectal Cancer Patients. Cancers (Basel) 2022; 14:cancers14143312. [PMID: 35884372 PMCID: PMC9315763 DOI: 10.3390/cancers14143312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 07/03/2022] [Accepted: 07/04/2022] [Indexed: 11/29/2022] Open
Abstract
Simple Summary Store-operated Ca2+ entry (SOCE) has long been known to regulate the differentiation and effector functions of T cells as well as to be instrumental to the ability of cytotoxic T lymphocytes to target cancer cells. Currently, no information is available regarding the expression and function of SOCE in tumour-infiltrating lymphocytes (TILs) that have been expanded in vitro for adoptive cell therapy (ACT). This study provides the first evidence that SOCE is up-regulated in ex vivo-expanded TILs from metastatic colorectal cancer (mCRC) patients. The up-regulation of SOCE mainly depends on diacylglycerol kinase (DGK), which prevents the protein kinase C-dependent inhibition of Ca2+ entry in normal T cells. Of note, the pharmacological blockade of SOCE with the selective inhibitor, BTP-2, during target cell killing significantly increases cytotoxic activity at low TIL density, i.e., when TILs-mediated cancer cell death is rarer. This study, albeit preliminary, could lay the foundation to propose an alternative strategy to effect ACT. It has been shown that ex vivo-expanded TILs did not improve the disease-free survival rate in mCRC patients. Our results strongly suggest that pre-treating autologous TILs with a SOCE or DGK inhibitor before being infused into the patient could improve their cytotoxic activity against cancer cells. Abstract (1) Background: Store-operated Ca2+ entry (SOCE) drives the cytotoxic activity of cytotoxic T lymphocytes (CTLs) against cancer cells. However, SOCE can be enhanced in cancer cells due to an increase in the expression and/or function of its underlying molecular components, i.e., STIM1 and Orai1. Herein, we evaluated the SOCE expression and function in tumour-infiltrating lymphocytes (TILs) from metastatic colorectal cancer (mCRC) patients. (2) Methods: Functional studies were conducted in TILs expanded ex vivo from CRC liver metastases. Peripheral blood T cells from healthy donors (hPBTs) and mCRC patients (cPBTs) were used as controls. (3) Results: SOCE amplitude is enhanced in TILs compared to hPBTs and cPBTs, but the STIM1 protein is only up-regulated in TILs. Pharmacological manipulation showed that the increase in SOCE mainly depends on tonic modulation by diacylglycerol kinase, which prevents the protein kinase C-dependent inhibition of SOCE activity. The larger SOCE caused a stronger Ca2+ response to T-cell receptor stimulation by autologous mCRC cells. Reducing Ca2+ influx with BTP-2 during target cell killing significantly increases cytotoxic activity at low target:effector ratios. (4) Conclusions: SOCE is enhanced in ex vivo-expanded TILs deriving from mCRC patients but decreasing Ca2+ influx with BTP-2 increases cytotoxic activity at a low TIL density.
Collapse
|
12
|
Naseem MU, Carcamo-Noriega E, Beltrán-Vidal J, Borrego J, Szanto TG, Zamudio FZ, Delgado-Prudencio G, Possani LD, Panyi G. Cm28, a scorpion toxin having a unique primary structure, inhibits KV1.2 and KV1.3 with high affinity. J Gen Physiol 2022; 154:213282. [PMID: 35699659 PMCID: PMC9202693 DOI: 10.1085/jgp.202213146] [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: 03/09/2022] [Accepted: 05/23/2022] [Indexed: 02/03/2023] Open
Abstract
The Cm28 in the venom of Centruroides margaritatus is a short peptide consisting of 27 amino acid residues with a mol wt of 2,820 D. Cm28 has <40% similarity with other known α-KTx from scorpions and lacks the typical functional dyad (lysine-tyrosine) required to block KV channels. However, its unique sequence contains the three disulfide-bond traits of the α-KTx scorpion toxin family. We propose that Cm28 is the first example of a new subfamily of α-KTxs, registered with the systematic number α-KTx32.1. Cm28 inhibited voltage-gated K+ channels KV1.2 and KV1.3 with Kd values of 0.96 and 1.3 nM, respectively. There was no significant shift in the conductance-voltage (G-V) relationship for any of the channels in the presence of toxin. Toxin binding kinetics showed that the association and dissociation rates are consistent with a bimolecular interaction between the peptide and the channel. Based on these, we conclude that Cm28 is not a gating modifier but rather a pore blocker. In a selectivity assay, Cm28 at 150 nM concentration (>100× Kd value for KV1.3) did not inhibit KV1.5, KV11.1, KCa1.1, and KCa3.1 K+ channels; NaV1.5 and NaV1.4 Na+ channels; or the hHV1 H+ channel but blocked ∼27% of the KV1.1 current. In a biological functional assay, Cm28 strongly inhibited the expression of the activation markers interleukin-2 receptor and CD40 ligand in anti-CD3-activated human CD4+ effector memory T lymphocytes. Cm28, due to its unique structure, may serve as a template for the generation of novel peptides targeting KV1.3 in autoimmune diseases.
Collapse
Affiliation(s)
- Muhammad Umair Naseem
- Department of Biophysics and Cell Biology, Faculty of Medicine, Research Center for Molecular Medicine, University of Debrecen, Debrecen, Hungary
| | - Edson Carcamo-Noriega
- Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnologia, Universidad Nacional Autónoma de México, Cuernavaca, Mexico
| | - José Beltrán-Vidal
- Grupo de Investigaciones Herpetológicas y Toxinológicas, Centro de Investigaciones Biomédicas, Departamento de Biología, Facultad de Ciencias Naturales, Exactas y de la Educación, Universidad del Cauca, Popayán, Colombia
| | - Jesus Borrego
- Department of Biophysics and Cell Biology, Faculty of Medicine, Research Center for Molecular Medicine, University of Debrecen, Debrecen, Hungary
| | - Tibor G. Szanto
- Department of Biophysics and Cell Biology, Faculty of Medicine, Research Center for Molecular Medicine, University of Debrecen, Debrecen, Hungary
| | - Fernando Z. Zamudio
- Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnologia, Universidad Nacional Autónoma de México, Cuernavaca, Mexico
| | - Gustavo Delgado-Prudencio
- Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnologia, Universidad Nacional Autónoma de México, Cuernavaca, Mexico
| | - Lourival D. Possani
- Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnologia, Universidad Nacional Autónoma de México, Cuernavaca, Mexico
| | - Gyorgy Panyi
- Department of Biophysics and Cell Biology, Faculty of Medicine, Research Center for Molecular Medicine, University of Debrecen, Debrecen, Hungary,Correspondence to Gyorgy Panyi:
| |
Collapse
|
13
|
sVmKTx, a transcriptome analysis-based synthetic peptide analogue of Vm24, inhibits Kv1.3 channels of human T cells with improved selectivity. Biochem Pharmacol 2022; 199:115023. [PMID: 35358481 DOI: 10.1016/j.bcp.2022.115023] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 03/22/2022] [Accepted: 03/23/2022] [Indexed: 12/13/2022]
Abstract
Kv1.3 K+ channels play a central role in the regulation of T cell activation and Ca2+ signaling under physiological and pathophysiological conditions. Peptide toxins targeting Kv1.3 have a significant therapeutic potential in the treatment of autoimmune diseases; thus, the discovery of new toxins is highly motivated. Based on the transcriptome analysis of the venom gland of V. mexicanus smithi a novel synthetic peptide, sVmKTx was generated, containing 36 amino acid residues. sVmKTx shows high sequence similarity to Vm24, a previously characterized peptide from the same species, but contains a Glu at position 32 as opposed to Lys32 in Vm24. Vm24 inhibits Kv1.3 with high affinity (Kd = 2.9 pM). However, it has limited selectivity (~1,500-fold) for Kv1.3 over hKv1.2, hKCa3.1, and mKv1.1. sVmKTx displays reduced Kv1.3 affinity (Kd = 770 pM) but increased selectivity for Kv1.3 over hKv1.2 (~9,000-fold) as compared to Vm24, other channels tested in the panel (hKCa3.1, hKv1.1, hKv1.4, hKv1.5, rKv2.1, hKv11.1, hKCa1.1, hNav1.5) were practically insensitive to the toxin at 2.5 μM. Molecular dynamics simulations showed that introduction of a Glu instead of Lys at position 32 led to a decreased structural fluctuation of the N-terminal segment of sVmKTx, which may explain its increased selectivity for Kv1.3. sVmKTx at 100 nM concentration decreased the expression level of the Ca2+ -dependent T cell activation marker, CD40 ligand. The high affinity block of Kv1.3 and increased selectivity over the natural peptide makes sVmKTx a potential candidate for Kv1.3 blockade-mediated treatment of autoimmune diseases.
Collapse
|
14
|
Ginefra P, Carrasco Hope H, Spagna M, Zecchillo A, Vannini N. Ionic Regulation of T-Cell Function and Anti-Tumour Immunity. Int J Mol Sci 2021; 22:ijms222413668. [PMID: 34948472 PMCID: PMC8705279 DOI: 10.3390/ijms222413668] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 12/15/2021] [Accepted: 12/17/2021] [Indexed: 12/02/2022] Open
Abstract
The capacity of T cells to identify and kill cancer cells has become a central pillar of immune-based cancer therapies. However, T cells are characterized by a dysfunctional state in most tumours. A major obstacle for proper T-cell function is the metabolic constraints posed by the tumour microenvironment (TME). In the TME, T cells compete with cancer cells for macronutrients (sugar, proteins, and lipid) and micronutrients (vitamins and minerals/ions). While the role of macronutrients in T-cell activation and function is well characterized, the contribution of micronutrients and especially ions in anti-tumour T-cell activities is still under investigation. Notably, ions are important for most of the signalling pathways regulating T-cell anti-tumour function. In this review, we discuss the role of six biologically relevant ions in T-cell function and in anti-tumour immunity, elucidating potential strategies to adopt to improve immunotherapy via modulation of ion metabolism.
Collapse
|
15
|
Gawali VS, Chimote AA, Newton HS, Feria-Garzón MG, Chirra M, Janssen EM, Wise-Draper TM, Conforti L. Immune Checkpoint Inhibitors Regulate K + Channel Activity in Cytotoxic T Lymphocytes of Head and Neck Cancer Patients. Front Pharmacol 2021; 12:742862. [PMID: 34512366 PMCID: PMC8429813 DOI: 10.3389/fphar.2021.742862] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Accepted: 08/16/2021] [Indexed: 12/27/2022] Open
Abstract
Programmed death receptor-1 (PD-1) and its ligand (PD-L1) interaction negatively regulates T cell function in head and neck squamous cell carcinoma (HNSCC). Overexpression of PD-1 reduces intracellular Ca2+ fluxes, and thereby T cell effector functions. In HNSCC patients, PD-1 blockade increases KCa3.1 and Kv1.3 activity along with Ca2+ signaling and mobility in CD8+ peripheral blood T cells (PBTs). The mechanism by which PD-L1/PD-1 interaction regulates ion channel function is not known. We investigated the effects of blocking PD-1 and PD-L1 on ion channel functions and intracellular Ca2+ signaling in CD8+ PBTs of HNSCC patients and healthy donors (HDs) using single-cell electrophysiology and live microscopy. Anti-PD-1 and anti-PD-L1 antibodies increase KCa3.1 and Kv1.3 function in CD8+ PBTs of HNSCC patients. Anti-PD-1 treatment increases Ca2+ fluxes in a subset of HSNCC patients. In CD8+ PBTs of HDs, exposure to PD-L1 reduces KCa3.1 activity and Ca2+ signaling, which were restored by anti-PD-1 treatment. The PD-L1-induced inhibition of KCa3.1 channels was rescued by the intracellular application of the PI3 kinase modulator phosphatidylinositol 3-phosphate (PI3P) in patch-clamp experiments. In HNSCC CD8+ PBTs, anti-PD-1 treatment did not affect the expression of KCa3.1, Kv1.3, Ca2+ release activated Ca2+ (CRAC) channels, and markers of cell activation (CD69) and exhaustion (LAG-3 and TIM-3). Our data show that immune checkpoint blockade improves T cell function by increasing KCa3.1 and Kv1.3 channel activity in HNSCC patients.
Collapse
Affiliation(s)
- Vaibhavkumar S Gawali
- Department of Internal Medicine, Division of Nephrology, University of Cincinnati, Cincinnati, OH, United States
| | - Ameet A Chimote
- Department of Internal Medicine, Division of Nephrology, University of Cincinnati, Cincinnati, OH, United States
| | - Hannah S Newton
- Department of Internal Medicine, Division of Nephrology, University of Cincinnati, Cincinnati, OH, United States
| | - Manuel G Feria-Garzón
- Department of Internal Medicine, Division of Nephrology, University of Cincinnati, Cincinnati, OH, United States
| | - Martina Chirra
- Department of Internal Medicine, Division of Nephrology, University of Cincinnati, Cincinnati, OH, United States
| | - Edith M Janssen
- Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States.,Immunology, Janssen Research and Development, Spring House, PA, United States
| | - Trisha M Wise-Draper
- Department of Internal Medicine, Division of Hematology Oncology, University of Cincinnati, Cincinnati, OH, United States
| | - Laura Conforti
- Department of Internal Medicine, Division of Nephrology, University of Cincinnati, Cincinnati, OH, United States
| |
Collapse
|
16
|
Newton HS, Chimote AA, Arnold MJ, Wise-Draper TM, Conforti L. Targeted knockdown of the adenosine A 2A receptor by lipid NPs rescues the chemotaxis of head and neck cancer memory T cells. Mol Ther Methods Clin Dev 2021; 21:133-143. [PMID: 33816646 PMCID: PMC8005736 DOI: 10.1016/j.omtm.2021.03.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Accepted: 03/01/2021] [Indexed: 12/13/2022]
Abstract
In solid malignancies, including head and neck squamous cell carcinoma (HNSCC), the immunosuppressive molecule adenosine, which accumulates in the tumor, suppresses cytotoxic CD8+ T cell functions including chemotaxis and tumor infiltration. Adenosine functions through binding to the adenosine A2A receptor (A2AR) present on T cells. In order to increase T cell migration into the tumor, the negative effect of adenosine must be abrogated. Systemic drug treatments targeting A2AR are available; however, they could lead to negative toxicities due to the broad expression of this receptor. Herein, we developed a lipid nanoparticle (NP)-based targeted delivery approach to knock down A2AR in T cells in order to increase their chemotaxis in the presence of adenosine. By using flow cytometry, immunofluorescence, qRT-PCR, and 3D-chemotaxis, we demonstrated that CD45RO-labeled nanoparticles delivering ADORA2A gene-silencing-RNAs decreased ADORA2A mRNA expression and rescued the chemotaxis of HNSCC CD8+ memory T cells. Overall, the data indicate that targeting the adenosine signaling pathway with lipid NPs is successful at suppressing the inhibitory effect of adenosine on the chemotaxis of HNSCC memory T cells, which could ultimately help increase T cell infiltration into the tumor.
Collapse
Affiliation(s)
- Hannah S. Newton
- Department of Internal Medicine, Division of Nephrology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Ameet A. Chimote
- Department of Internal Medicine, Division of Nephrology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Michael J. Arnold
- Department of Internal Medicine, Division of Nephrology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Trisha M. Wise-Draper
- Department of Internal Medicine, Division of Hematology/Oncology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Laura Conforti
- Department of Internal Medicine, Division of Nephrology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| |
Collapse
|
17
|
Zhang Y, Wang J, Ye M, Li G, Zhong M, Guan X. The effect of mechanical stimulation on the expression of apoptosis-related genes in cardiomyocytes. APPLIED NANOSCIENCE 2021. [DOI: 10.1007/s13204-021-01901-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
18
|
Liu J, Lv XW, Zhang L, Wang H, Li J, Wu B. Review on Biological Characteristics of Kv1.3 and Its Role in Liver Diseases. Front Pharmacol 2021; 12:652508. [PMID: 34093186 PMCID: PMC8176307 DOI: 10.3389/fphar.2021.652508] [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: 01/14/2021] [Accepted: 04/23/2021] [Indexed: 01/30/2023] Open
Abstract
The liver accounts for the largest proportion of macrophages in all solid organs of the human body. Liver macrophages are mainly composed of cytolytic cells inherent in the liver and mononuclear macrophages recruited from the blood. Monocytes recruitment occurs mainly in the context of liver injury and inflammation and can be recruited into the liver and achieve a KC-like phenotype. During the immune response of the liver, macrophages/KC cells release inflammatory cytokines and infiltrate into the liver, which are considered to be the common mechanism of various liver diseases in the early stage. Meanwhile, macrophages/KC cells form an interaction network with other liver cells, which can affect the occurrence and progression of liver diseases. From the perspective of liver disease treatment, knowing the full spectrum of macrophage activation, the underlying molecular mechanisms, and their implication in either promoting liver disease progression or repairing injured liver tissue is highly relevant from a therapeutic point of view. Kv1.3 is a subtype of the voltage-dependent potassium channel, whose function is closely related to the regulation of immune cell function. At present, there are few studies on the relationship between Kv1.3 and liver diseases, and the application of its blockers as a potential treatment for liver diseases has not been reported. This manuscript reviewed the physiological characteristics of Kv1.3, the relationship between Kv1.3 and cell proliferation and apoptosis, and the role of Kv1.3 in a variety of liver diseases, so as to provide new ideas and strategies for the prevention and treatment of liver diseases. In short, by understanding the role of Kv1.3 in regulating the functions of immune cells such as macrophages, selective blockers of Kv1.3 or compounds with similar functions can be applied to alleviate the progression of liver diseases and provide new ideas for the prevention and treatment of liver diseases.
Collapse
Affiliation(s)
- Junda Liu
- First Affiliated Hospital of Anhui Medical University, Hefei, China.,School of Pharmacy, Anhui Medical University, Hefei, China.,Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, China
| | - Xiong-Wen Lv
- School of Pharmacy, Anhui Medical University, Hefei, China.,Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, China
| | - Lei Zhang
- School of Pharmacy, Anhui Medical University, Hefei, China.,Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, China
| | - Hua Wang
- School of Pharmacy, Anhui Medical University, Hefei, China.,Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, China
| | - Jun Li
- School of Pharmacy, Anhui Medical University, Hefei, China.,Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, China
| | - Baoming Wu
- School of Pharmacy, Anhui Medical University, Hefei, China.,Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, China
| |
Collapse
|
19
|
Abstract
Neoplastic transformation is reportedly associated with alterations of the potassium transport across plasma and intracellular membranes. These alterations have been identified as crucial elements of the tumourigenic reprogramming of cells. Potassium channels may contribute to cancer initiation, malignant progression and therapy resistance of tumour cells. The book chapter focusses on (oncogenic) potassium channels frequently upregulated in different tumour entities, upstream and downstream signalling of these channels, their contribution to the maintenance of cancer stemness and the formation of an immunosuppressive tumour microenvironment. In addition, their role in adaptation to tumour hypoxia, metabolic reprogramming, as well as tumour spreading and metastasis is discussed. Finally, we discuss how (oncogenic) potassium channels may confer treatment resistance of tumours against radiation and chemotherapy and thus might be harnessed for new therapy strategies, for instance, by repurposing approved drugs known to target potassium channels.
Collapse
|
20
|
Xu C, Zhang Y, Shen Y, Shi Y, Zhang M, Zhou L. Integrated Analysis Reveals ENDOU as a Biomarker in Head and Neck Squamous Cell Carcinoma Progression. Front Oncol 2021; 10:522332. [PMID: 33614471 PMCID: PMC7894080 DOI: 10.3389/fonc.2020.522332] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2019] [Accepted: 12/07/2020] [Indexed: 12/15/2022] Open
Abstract
Background Head and neck squamous cell carcinoma (HNSCC) is a leading cancer with high morbidity and mortality worldwide. The aim is to identify genes with clinical significance by integrated bioinformatics analysis and investigate their function in HNSCC. Methods We downloaded and analyzed two gene expression datasets of GSE6631 and GSE107591 to screen differentially expressed genes (DEGs) in HNSCC. Common DEGs were functionally analyzed by Gene ontology and KEGG pathway enrichment analysis. Protein-protein interaction (PPI) network was constructed with STRING database and Cytoscape. ENDOU was overexpressed in FaDu and Cal-27 cell lines, and cell proliferation and migration capability were evaluated with MTT, scratch and transwell assay. The prognostic performance of ENDOU and expression correlation with tumor infiltrates in HNSCC were validated with TCGA HNSCC datasets. Results Ninety-eight genes shared common differential expression in both datasets, with core functions like extracellular matrix organization significantly enriched. 15 genes showed prognostic significance, and COBL and ENDOU serve as independent survival markers in HNSCC. In-vitro ENDOU overexpression inhibited FaDu and Cal-27 cells proliferation and migration, indicating its tumor-suppressing role in HNSCC progression. GSEA analysis indicated ENDOU down-stream pathways like DNA replication, mismatch repair, cell cycle and IL-17 signaling pathway. ENDOU showed relative lower expression in HNSCC, especially HPV-positive HNSCC samples. At last, ENDOU showed negative correlation with tumor purity and tumor infiltrating macrophages, especially M2 macrophages. Conclusion This study identified ENDOU as a biomarker with prognostic significance in HNSCC progression.
Collapse
Affiliation(s)
- Chengzhi Xu
- Department of Otolaryngology-Head and Neck Surgery, Eye Ear Nose and Throat Hospital, Fudan University, Shanghai, China
| | - Yunbin Zhang
- Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China.,Department of Respirology, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Yupeng Shen
- Department of Otolaryngology-Head and Neck Surgery, Bethune International Peace Hospital, Shijiazhuang, China
| | - Yong Shi
- Department of Otolaryngology-Head and Neck Surgery, Eye Ear Nose and Throat Hospital, Fudan University, Shanghai, China
| | - Ming Zhang
- Department of Otolaryngology-Head and Neck Surgery, Eye Ear Nose and Throat Hospital, Fudan University, Shanghai, China
| | - Liang Zhou
- Department of Otolaryngology-Head and Neck Surgery, Eye Ear Nose and Throat Hospital, Fudan University, Shanghai, China
| |
Collapse
|
21
|
Sun J, Yu X, Xue L, Li S, Li J, Tong D, Du Y. TP53-Associated Ion Channel Genes Serve as Prognostic Predictor and Therapeutic Targets in Head and Neck Squamous Cell Carcinoma. Technol Cancer Res Treat 2020; 19:1533033820972344. [PMID: 33243093 PMCID: PMC7705194 DOI: 10.1177/1533033820972344] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
TP53 mutations are the most occurred mutation in HNSCC which might affect the ion channel genes. We aim to investigate the ion channel gene alteration under TP53 mutation and their prognostic implication. The overall mutation status of HNSCC were explored. By screening the TP53-associated ion channel genes (TICGs), an ion channel prognostic signature (ICPS) was established through a series of machine learning algorithms. The ICPS was then evaluated and its clinical significance was explored. 82 TICGs differentially expressed between TP53WT and TP53MUT were screened. Using univariate regression analysis and LASSO regression analysis and multivariate regression analysis, an ICPS containing 7 ion channel genes was established. A series of evaluation was carried out which proved the predictive ability of ICPS. Functional analysis of ICPS revealed that cancer-related pathways were enriched in high-risk group. Next, for clinical application, a nomogram was constructed based on ICPS and other independent clinicopathological factors. TP53 mutation status strongly affects the expression of ion channel genes. The ICPM we have identified is a strong indicator for HNSCC prognosis and could help with patient stratification as well as identification of novel drug targets.
Collapse
Affiliation(s)
- Jing Sun
- Department of Periodontology, Jinan Stomatological Hospital, Jinan, Shandong, China.,Jing Sun and Xijiao Yu contributed equally to this work
| | - Xijiao Yu
- Department of Endodontics, Jinan Stomatological Hospital, Jinan, Shandong, China.,Jing Sun and Xijiao Yu contributed equally to this work
| | - Lande Xue
- Department of Periodontology, Jinan Stomatological Hospital, Jinan, Shandong, China
| | - Shu Li
- Hospital of Stomatology, 12589Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Jinan, Shandong, China
| | - Jianxia Li
- Department of Periodontology, Jinan Stomatological Hospital, Jinan, Shandong, China
| | - Dongdong Tong
- Department of Oral and Maxillofacial, School and Hospital of Stomatology, 12589Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Jinan, Shandong, China
| | - Yi Du
- Department of Endodontics, Jinan Stomatological Hospital, Jinan, Shandong, China
| |
Collapse
|
22
|
Xie Z, Zhao Y, Yang W, Li W, Wu Y, Chen Z. Methotrexate, a small molecular scaffold targeting Kv1.3 channel extracellular pore region. Biochem Biophys Res Commun 2020; 532:265-270. [PMID: 32863001 DOI: 10.1016/j.bbrc.2020.08.050] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 08/14/2020] [Accepted: 08/18/2020] [Indexed: 11/18/2022]
Abstract
Methotrexate (MTX) has been widely used for the treatment of many types of autoimmune diseases, such as rheumatoid arthritis, psoriasis and dermatomyositis. However, its pharmacological mechanism is still unclear completely. In this study, we found that MTX is a potent and selective inhibitor of the Kv1.3 channel, a class of potassium channels highly associated with autoimmune diseases. Electrophysiological experiments showed that MTX inhibited human Kv1.3 channel with an IC50 of 41.5 ± 24.9 nM, and 1 μM MTX inhibited 32.6 ± 1.3% and 25.6 ± 2.2% of human Kv1.1 and Kv1.2 channel currents, respectively. These data implied the unique selectivity of MTX towards the Kv1.3 channel. Excitingly, using channel activation and chimeric experiments, we found that MTX bound to the outer pore region of Kv1.3 channel. Mutagenesis experiments in the Kv.3 channel extracellular pore region further showed that the Dsp371, Thr373 and His399 residues of outer pore region of Kv1.3 channel played important roles in MTX inhibiting activities. In conclusion, MTX inhibited Kv1.3 channel by targeting extracellular pore region, which is different form all the report small molecules, such as PAP-1 and 4-AP, but similar with many natural animal toxin peptides, such as ChTX, ShK and BmKTX. To the best of our knowledge, MTX is the first small molecular scaffold targeting the Kv1.3 channel extracellular pore region, suggesting its potential applications for designing novel Kv1.3 lead drugs and treating Kv1.3 channel-associated autoimmune diseases.
Collapse
Affiliation(s)
- Zili Xie
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, 430072, China
| | - Yonghui Zhao
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, 430072, China
| | - Weishan Yang
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, 430072, China
| | - Wenxin Li
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, 430072, China; Biodrug Research Center, Wuhan University, Wuhan, 430072, China
| | - Yingliang Wu
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, 430072, China; Biodrug Research Center, Wuhan University, Wuhan, 430072, China.
| | - Zongyun Chen
- Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, College of Basic Medicine, Hubei University of Medicine, Shiyan, 442000, China; State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, 430072, China; Biodrug Research Center, Wuhan University, Wuhan, 430072, China.
| |
Collapse
|
23
|
Khodoun M, Chimote AA, Ilyas FZ, Duncan HJ, Moncrieffe H, Kant KS, Conforti L. Targeted knockdown of Kv1.3 channels in T lymphocytes corrects the disease manifestations associated with systemic lupus erythematosus. SCIENCE ADVANCES 2020; 6:6/47/eabd1471. [PMID: 33208373 PMCID: PMC7673800 DOI: 10.1126/sciadv.abd1471] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Accepted: 10/02/2020] [Indexed: 05/16/2023]
Abstract
Lupus nephritis (LN) is an autoimmune disease with substantial morbidity/mortality and limited efficacy of available therapies. Memory T (Tm) lymphocytes infiltrate LN kidneys, contributing to organ damage. Analysis of LN, diabetic nephropathy, and healthy donor kidney biopsies revealed high infiltration of active CD8+ Tm cells expressing high voltage-dependent Kv1.3 potassium channels-key T cell function regulators-in LN. Nanoparticles that selectively down-regulate Kv1.3 in Tm cells (Kv1.3-NPs) reduced CD40L and interferon-γ (IFNγ) in Tm cells from LN patients in vitro. Kv1.3-NPs were tested in humanized LN mice obtained by engrafting peripheral blood mononuclear cells (PBMCs) from LN patients into immune-deficient mice. LN mice exhibited features of the disease: increased IFNγ and CD3+CD8+ T cell renal infiltration, and reduced survival versus healthy donor PBMC engrafted mice. Kv1.3-NP treatment of patient PBMCs before engraftment decreased CD40L/IFNγ and prolonged survival of LN mice. These data show the potential benefits of targeting Kv1.3 in LN.
Collapse
Affiliation(s)
- Marat Khodoun
- Division of Rheumatology, Department of Internal Medicine, University of Cincinnati, Cincinnati, OH, USA
- Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Ameet A Chimote
- Division of Nephrology, Department of Internal Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Farhan Z Ilyas
- Division of Nephrology, Department of Internal Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Heather J Duncan
- Division of Nephrology, Department of Internal Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Halima Moncrieffe
- Center for Autoimmune Genomics and Etiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Department of Pediatrics, University of Cincinnati, Cincinnati, OH, USA
| | - K Shashi Kant
- Division of Nephrology, Department of Internal Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Laura Conforti
- Division of Nephrology, Department of Internal Medicine, University of Cincinnati, Cincinnati, OH, USA.
| |
Collapse
|
24
|
Newton HS, Gawali VS, Chimote AA, Lehn MA, Palackdharry SM, Hinrichs BH, Jandarov R, Hildeman D, Janssen EM, Wise-Draper TM, Conforti L. PD1 blockade enhances K + channel activity, Ca 2+ signaling, and migratory ability in cytotoxic T lymphocytes of patients with head and neck cancer. J Immunother Cancer 2020; 8:e000844. [PMID: 33060146 PMCID: PMC7566435 DOI: 10.1136/jitc-2020-000844] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/10/2020] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Immunotherapy has emerged as a promising treatment modality for head and neck squamous cell carcinoma (HNSCC). Pembrolizumab, an anti-programmed death 1 antibody, is an immunotherapy agent currently approved for metastatic HNSCC and curative intent clinical trials. Although clinical responses to pembrolizumab are promising, many patients fail to respond. However, it is well known that T cell cytotoxicity and chemotaxis are critically important in the elimination of HNSCC tumors. These functions depend on ion channel activity and downstream Ca2+ fluxing abilities, which are defective in patients with HNSCC. The purpose of this study was to elucidate the effects of pembrolizumab on potassium (K+) channel (KCa3.1 and Kv1.3) activity, Ca2+ fluxes, and chemotaxis in the cytotoxic T cells of patients with HNSCC and to determine their correlation with treatment response. METHODS Functional studies were conducted in CD8+ peripheral blood T cells (PBTs) and tumor infiltrating lymphocytes (TILs) from patients with HNSCC treated with pembrolizumab. Untreated patients with HNSCC were used as controls. The ion channel activity of CD8+ T cells was measured by patch-clamp electrophysiology; single-cell Ca2+ fluxing abilities were measured by live microscopy. Chemotaxis experiments were conducted in a three-dimensional collagen matrix. Pembrolizumab patients were stratified as responders or non-responders based on pathological response (percent of viable tumor remaining at resection; responders: ≤80% viable tumor; non-responders: >80% viable tumor). RESULTS Pembrolizumab increased K+ channel activity and Ca2+ fluxes in TILs independently of treatment response. However, in PBTs from responder patients there was an increased KCa3.1 activity immediately after pembrolizumab treatment that was accompanied by a characteristic increase in Kv1.3 and Ca2+ fluxes as compared with PBTs from non-responder patients. The effects on Kv1.3 and Ca2+ were prolonged and persisted after tumor resection. Chemotaxis was also improved in responder patients' PBTs. Unlike non-responders' PBTs, pembrolizumab increased their ability to chemotax in a tumor-like, adenosine-rich microenvironment immediately after treatment, and additionally they maintained an efficient chemotaxis after tumor resection. CONCLUSIONS Pembrolizumab enhanced K+ channel activity, Ca2+ fluxes and chemotaxis of CD8+ T cells in patients with HNSCC, with a unique pattern of response in responder patients that is conducive to the heightened functionality of their cytotoxic T cells.
Collapse
Affiliation(s)
- Hannah S Newton
- Department of Internal Medicine, Division of Nephrology, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Vaibhavkumar S Gawali
- Department of Internal Medicine, Division of Nephrology, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Ameet A Chimote
- Department of Internal Medicine, Division of Nephrology, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Maria A Lehn
- Department of Internal Medicine, Division of Hematology/Oncology, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
- Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Sarah M Palackdharry
- Department of Internal Medicine, Division of Hematology/Oncology, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Benjamin H Hinrichs
- Department of Pathology and Laboratory Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Roman Jandarov
- Department of Environmental Health, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - David Hildeman
- Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Edith M Janssen
- Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
- Immunology, Janssen Research and Development, Spring House, Pennsylvania, USA
| | - Trisha M Wise-Draper
- Department of Internal Medicine, Division of Hematology/Oncology, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Laura Conforti
- Department of Internal Medicine, Division of Nephrology, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| |
Collapse
|
25
|
Pethő Z, Najder K, Carvalho T, McMorrow R, Todesca LM, Rugi M, Bulk E, Chan A, Löwik CWGM, Reshkin SJ, Schwab A. pH-Channeling in Cancer: How pH-Dependence of Cation Channels Shapes Cancer Pathophysiology. Cancers (Basel) 2020; 12:E2484. [PMID: 32887220 PMCID: PMC7565548 DOI: 10.3390/cancers12092484] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 08/25/2020] [Accepted: 08/26/2020] [Indexed: 12/20/2022] Open
Abstract
Tissue acidosis plays a pivotal role in tumor progression: in particular, interstitial acidosis promotes tumor cell invasion, and is a major contributor to the dysregulation of tumor immunity and tumor stromal cells. The cell membrane and integral membrane proteins commonly act as important sensors and transducers of altered pH. Cell adhesion molecules and cation channels are prominent membrane proteins, the majority of which is regulated by protons. The pathophysiological consequences of proton-sensitive ion channel function in cancer, however, are scarcely considered in the literature. Thus, the main focus of this review is to highlight possible events in tumor progression and tumor immunity where the pH sensitivity of cation channels could be of great importance.
Collapse
Affiliation(s)
- Zoltán Pethő
- Institute of Physiology II, University Münster, 48147 Münster, Germany; (K.N.); (L.M.T.); (M.R.); (E.B.); (A.S.)
| | - Karolina Najder
- Institute of Physiology II, University Münster, 48147 Münster, Germany; (K.N.); (L.M.T.); (M.R.); (E.B.); (A.S.)
| | - Tiago Carvalho
- Department of Biosciences, Biotechnologies, and Biopharmaceutics, University of Bari, 90126 Bari, Italy; (T.C.); (S.J.R.)
| | - Roisin McMorrow
- Department of Radiology and Nuclear Medicine, Erasmus Medical Center, 3035 GD Rotterdam, The Netherlands; (R.M.); (C.W.G.M.L.)
| | - Luca Matteo Todesca
- Institute of Physiology II, University Münster, 48147 Münster, Germany; (K.N.); (L.M.T.); (M.R.); (E.B.); (A.S.)
| | - Micol Rugi
- Institute of Physiology II, University Münster, 48147 Münster, Germany; (K.N.); (L.M.T.); (M.R.); (E.B.); (A.S.)
| | - Etmar Bulk
- Institute of Physiology II, University Münster, 48147 Münster, Germany; (K.N.); (L.M.T.); (M.R.); (E.B.); (A.S.)
| | - Alan Chan
- Percuros B.V., 2333 CL Leiden, The Netherlands;
| | - Clemens W. G. M. Löwik
- Department of Radiology and Nuclear Medicine, Erasmus Medical Center, 3035 GD Rotterdam, The Netherlands; (R.M.); (C.W.G.M.L.)
- Department of Oncology CHUV, UNIL and Ludwig Cancer Center, 1011 Lausanne, Switzerland
| | - Stephan J. Reshkin
- Department of Biosciences, Biotechnologies, and Biopharmaceutics, University of Bari, 90126 Bari, Italy; (T.C.); (S.J.R.)
| | - Albrecht Schwab
- Institute of Physiology II, University Münster, 48147 Münster, Germany; (K.N.); (L.M.T.); (M.R.); (E.B.); (A.S.)
| |
Collapse
|
26
|
Fewer tumour-specific PD-1 +CD8 + TILs in high-risk "Infiltrating" HPV - HNSCC. Br J Cancer 2020; 123:932-941. [PMID: 32616847 PMCID: PMC7492364 DOI: 10.1038/s41416-020-0966-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2020] [Revised: 05/24/2020] [Accepted: 06/12/2020] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND The prognosis of HPV- HNSCC was worse than that of HPV+ HNSCC. Analysis of tumours and tumour-infiltrating lymphocytes (TILs) may provide insight into the progression of HPV- HNSCC. METHODS The tumour and TIL phenotypic characteristics of 134 HNSCC specimens (HPV- tumours were classified into "Infiltrating" and "Pushing" subtypes based on their different tumour nest configuration and prognosis) were retrospectively analysed. HNSCC data from the Cancer Genome Atlas (n = 263) were analysed for CD8α, HPV and overall survival (OS). A murine HNSCC model was used to verify the antitumour role of PD-1+CD8+ TILs. RESULTS The "Infiltrating" HPV- subtype showed shorter OS than the "Pushing" subtype. Moreover, there is a tendency from "Pushing" to "Infiltrating" subtype from the primary to the recurrent lesion. Different from total CD8+ TILs, tumour-specific PD-1+CD8+ TILs were fewer in invasive margin (IM) of "Infiltrating" HPV- tumours. PD-1+CD8+ TILs recognised autologous HNSCC cells and showed stronger inhibition of tumour growth in a murine HNSCC model resistant to PD-1 blockade. CONCLUSIONS Coevolution of HPV- HNSCC and TILs is characterised by an "Infiltrating" phenotype and less tumour-specific PD-1+CD8+ TILs, which may provide a framework for further translational studies and patient stratification.
Collapse
|
27
|
Chimote AA, Gawali VS, Newton HS, Wise-Draper TM, Conforti L. A Compartmentalized Reduction in Membrane-Proximal Calmodulin Reduces the Immune Surveillance Capabilities of CD8 + T Cells in Head and Neck Cancer. Front Pharmacol 2020; 11:143. [PMID: 32184726 PMCID: PMC7059094 DOI: 10.3389/fphar.2020.00143] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Accepted: 02/04/2020] [Indexed: 01/28/2023] Open
Abstract
The limited ability of cytotoxic CD8+ T cells to infiltrate solid tumors and function within the tumor microenvironment presents a major roadblock to effective immunotherapy. Ion channels and Ca2+-dependent signaling events control the activity of T cells and are implicated in the failure of immune surveillance in cancer. Reduced KCa3.1 channel activity mediates the heightened inhibitory effect of adenosine on the chemotaxis of circulating T cells from head and neck squamous cell carcinoma (HNSCC) patients. Herein, we conducted experiments that elucidate the mechanisms of KCa3.1 dysfunction and impaired chemotaxis in HNSCC CD8+ T cells. The Ca2+ sensor calmodulin (CaM) controls multiple cellular functions including KCa3.1 activation. Our data showed that CaM expression is lower in HNSCC than healthy donor (HD) T cells. This reduction was due to an intrinsic decrease in the genes encoding CaM combined to the failure of HNSCC T cells to upregulate CaM upon activation. Furthermore, the reduction in CaM was confined to the plasma membrane and resulted in decreased CaM-KCa3.1 association and KCa3.1 activity (which was rescued by the delivery of CaM). IFNγ production, also Ca2+- and CaM-dependent, was instead not reduced in HNSCC T cells, which maintained intact cytoplasmic CaM and Ca2+ fluxing ability. Knockdown of CaM in HD T cells decreased KCa3.1 activity, but not IFNγ production, and reduced their chemotaxis in the presence of adenosine, thus recapitulating HNSCC T cell dysfunction. Activation of KCa3.1 with 1-EBIO restored the ability of CaM knockdown HD T cells to chemotax in the presence of adenosine. Additionally, 1-EBIO enhanced INFγ production. Our data showed a localized downregulation of membrane-proximal CaM that suppressed KCa3.1 activity in HNSCC circulating T cells and limited their ability to infiltrate adenosine-rich tumor-like microenvironments. Furthermore, they indicate that KCa3.1 activators could be used as positive CD8+ T cell modulators in cancers.
Collapse
Affiliation(s)
- Ameet A. Chimote
- Division of Nephrology, Department of Internal Medicine, University of Cincinnati, Cincinnati, OH, United States
| | - Vaibhavkumar S. Gawali
- Division of Nephrology, Department of Internal Medicine, University of Cincinnati, Cincinnati, OH, United States
| | - Hannah S. Newton
- Division of Nephrology, Department of Internal Medicine, University of Cincinnati, Cincinnati, OH, United States
| | - Trisha M. Wise-Draper
- Division of Hematology Oncology, Department of Internal Medicine, University of Cincinnati, Cincinnati, OH, United States
| | - Laura Conforti
- Division of Nephrology, Department of Internal Medicine, University of Cincinnati, Cincinnati, OH, United States
| |
Collapse
|
28
|
Ion Channel Dysregulation in Head and Neck Cancers: Perspectives for Clinical Application. Rev Physiol Biochem Pharmacol 2020; 181:375-427. [PMID: 32789787 DOI: 10.1007/112_2020_38] [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: 12/24/2022]
Abstract
Head and neck cancers are a highly complex and heterogeneous group of malignancies that involve very diverse anatomical structures and distinct aetiological factors, treatments and clinical outcomes. Among them, head and neck squamous cell carcinomas (HNSCC) are predominant and the sixth most common cancer worldwide with still low survival rates. Omic technologies have unravelled the intricacies of tumour biology, harbouring a large diversity of genetic and molecular changes to drive the carcinogenesis process. Nonetheless, this remarkable heterogeneity of molecular alterations opens up an immense opportunity to discover novel biomarkers and develop molecular-targeted therapies. Increasing evidence demonstrates that dysregulation of ion channel expression and/or function is frequently and commonly observed in a variety of cancers from different origin. As a consequence, the concept of ion channels as potential membrane therapeutic targets and/or biomarkers for cancer diagnosis and prognosis has attracted growing attention. This chapter intends to comprehensively and critically review the current state-of-art ion channel dysregulation specifically focusing on head and neck cancers and to formulate the major challenges and research needs to translate this knowledge into clinical application. Based on current reported data, various voltage-gated potassium (Kv) channels (i.e. Kv3.4, Kv10.1 and Kv11.1) have been found frequently aberrantly expressed in HNSCC as well as precancerous lesions and are highlighted as clinically and biologically relevant features in both early stages of tumourigenesis and late stages of disease progression. More importantly, they also emerge as promising candidates as cancer risk markers, tumour markers and potential anti-proliferative and anti-metastatic targets for therapeutic interventions; however, the oncogenic properties seem to be independent of their ion-conducting function.
Collapse
|
29
|
Lowinus T, Heidel FH, Bose T, Nimmagadda SC, Schnöder T, Cammann C, Schmitz I, Seifert U, Fischer T, Schraven B, Bommhardt U. Memantine potentiates cytarabine-induced cell death of acute leukemia correlating with inhibition of K v1.3 potassium channels, AKT and ERK1/2 signaling. Cell Commun Signal 2019; 17:5. [PMID: 30651113 PMCID: PMC6335768 DOI: 10.1186/s12964-018-0317-z] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Accepted: 12/28/2018] [Indexed: 12/23/2022] Open
Abstract
Background Treatment of acute leukemia is challenging and long-lasting remissions are difficult to induce. Innovative therapy approaches aim to complement standard chemotherapy to improve drug efficacy and decrease toxicity. Promising new therapeutic targets in cancer therapy include voltage-gated Kv1.3 potassium channels, but their role in acute leukemia is unclear. We reported that Kv1.3 channels of lymphocytes are blocked by memantine, which is known as an antagonist of neuronal N-methyl-D-aspartate type glutamate receptors and clinically applied in therapy of advanced Alzheimer disease. Here we evaluated whether pharmacological targeting of Kv1.3 channels by memantine promotes cell death of acute leukemia cells induced by chemotherapeutic cytarabine. Methods We analyzed acute lymphoid (Jurkat, CEM) and myeloid (HL-60, Molm-13, OCI-AML-3) leukemia cell lines and patients’ acute leukemic blasts after treatment with either drug alone or the combination of cytarabine and memantine. Patch-clamp analysis was performed to evaluate inhibition of Kv1.3 channels and membrane depolarization by memantine. Cell death was determined with propidium iodide, Annexin V and SYTOX staining and cytochrome C release assay. Molecular effects of memantine co-treatment on activation of Caspases, AKT, ERK1/2, and JNK signaling were analysed by Western blot. Kv1.3 channel expression in Jurkat cells was downregulated by shRNA. Results Our study demonstrates that memantine inhibits Kv1.3 channels of acute leukemia cells and in combination with cytarabine potentiates cell death of acute lymphoid and myeloid leukemia cell lines as well as primary leukemic blasts from acute leukemia patients. At molecular level, memantine co-application fosters concurrent inhibition of AKT, S6 and ERK1/2 and reinforces nuclear down-regulation of MYC, a common target of AKT and ERK1/2 signaling. In addition, it augments mitochondrial dysfunction resulting in enhanced cytochrome C release and activation of Caspase-9 and Caspase-3 leading to amplified apoptosis. Conclusions Our study underlines inhibition of Kv1.3 channels as a therapeutic strategy in acute leukemia and proposes co-treatment with memantine, a licensed and safe drug, as a potential approach to promote cytarabine-based cell death of various subtypes of acute leukemia. Electronic supplementary material The online version of this article (10.1186/s12964-018-0317-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Theresa Lowinus
- Institute of Molecular and Clinical Immunology, Health Campus Immunology, Infectiology and Inflammation (GC-I3), Otto-von-Guericke-University Magdeburg, Leipziger Str. 44, 39120, Magdeburg, Germany.,Present address: Department of Hematology, Oncology, and Stem Cell Transplantation, Faculty of Medicine, Freiburg University Medical Center, Freiburg, Germany
| | - Florian H Heidel
- Department of Hematology and Oncology, GC-I3, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany.,Leibniz Institute on Aging, Fritz-Lipmann Institute, Jena, Germany.,Innere Medizin II, Universitätsklinikum Jena, Jena, Germany
| | - Tanima Bose
- Leibniz Institute of Neurobiology, Magdeburg, Germany.,Present address: Institute for Clinical Neuroimmunology, Ludwigs-Maximilians-University, Munich, Germany
| | - Subbaiah Chary Nimmagadda
- Department of Hematology and Oncology, GC-I3, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany
| | - Tina Schnöder
- Department of Hematology and Oncology, GC-I3, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany.,Leibniz Institute on Aging, Fritz-Lipmann Institute, Jena, Germany.,Innere Medizin II, Universitätsklinikum Jena, Jena, Germany
| | - Clemens Cammann
- Friedrich Loeffler Institute for Medical Microbiology, University Medicine Greifswald, Greifswald, Germany
| | - Ingo Schmitz
- Institute of Molecular and Clinical Immunology, Health Campus Immunology, Infectiology and Inflammation (GC-I3), Otto-von-Guericke-University Magdeburg, Leipziger Str. 44, 39120, Magdeburg, Germany.,Systems-Oriented Immunology and Inflammation Research Group, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Ulrike Seifert
- Institute of Molecular and Clinical Immunology, Health Campus Immunology, Infectiology and Inflammation (GC-I3), Otto-von-Guericke-University Magdeburg, Leipziger Str. 44, 39120, Magdeburg, Germany.,Friedrich Loeffler Institute for Medical Microbiology, University Medicine Greifswald, Greifswald, Germany
| | - Thomas Fischer
- Department of Hematology and Oncology, GC-I3, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany
| | - Burkhart Schraven
- Institute of Molecular and Clinical Immunology, Health Campus Immunology, Infectiology and Inflammation (GC-I3), Otto-von-Guericke-University Magdeburg, Leipziger Str. 44, 39120, Magdeburg, Germany.,Department of Immune Control, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Ursula Bommhardt
- Institute of Molecular and Clinical Immunology, Health Campus Immunology, Infectiology and Inflammation (GC-I3), Otto-von-Guericke-University Magdeburg, Leipziger Str. 44, 39120, Magdeburg, Germany.
| |
Collapse
|
30
|
Marshall HT, Djamgoz MBA. Immuno-Oncology: Emerging Targets and Combination Therapies. Front Oncol 2018; 8:315. [PMID: 30191140 PMCID: PMC6115503 DOI: 10.3389/fonc.2018.00315] [Citation(s) in RCA: 225] [Impact Index Per Article: 32.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2018] [Accepted: 07/24/2018] [Indexed: 12/20/2022] Open
Abstract
Host immunity recognizes and eliminates most early tumor cells, yet immunological checkpoints, exemplified by CTLA-4, PD-1, and PD-L1, pose a significant obstacle to effective antitumor immune responses. T-lymphocyte co-inhibitory pathways influence intensity, inflammation and duration of antitumor immunity. However, tumors and their immunosuppressive microenvironments exploit them to evade immune destruction. Recent PD-1 checkpoint inhibitors yielded unprecedented efficacies and durable responses across advanced-stage melanoma, showcasing potential to replace conventional radiotherapy regimens. Neverthless, many clinical problems remain in terms of efficacy, patient-to-patient variability, and undesirable outcomes and side effects. In this review, we evaluate recent advances in the immuno-oncology field and discuss ways forward. First, we give an overview of current immunotherapy modalities, involving mainy single agents, including inhibitor monoclonal antibodies (mAbs) targeting T-cell checkpoints of PD-1 and CTLA-4. However, neoantigen recognition alone cannot eliminate tumors effectively in vivo given their inherent complex micro-environment, heterogeneous nature and stemness. Then, based mainly upon CTLA-4 and PD-1 checkpoint inhibitors as a "backbone," we cover a range of emerging ("second-generation") therapies incorporating other immunotherapies or non-immune based strategies in synergistic combination. These include targeted therapies such as tyrosine kinase inhibitors, co-stimulatory mAbs, bifunctional agents, epigenetic modulators (such as inhibitors of histone deacetylases or DNA methyltransferase), vaccines, adoptive-T-cell therapy, nanoparticles, oncolytic viruses, and even synthetic "gene circuits." A number of novel immunotherapy co-targets in pre-clinical development are also introduced. The latter include metabolic components, exosomes and ion channels. We discuss in some detail of the personalization of immunotherapy essential for ultimate maximization of clinical outcomes. Finally, we outline possible future technical and conceptual developments including realistic in vitro and in vivo models and inputs from physics, engineering, and artificial intelligence. We conclude that the breadth and quality of immunotherapeutic approaches and the types of cancers that can be treated will increase significantly in the foreseeable future.
Collapse
Affiliation(s)
- Henry T Marshall
- Neuroscience Solutions to Cancer Research Group, Department of Life Sciences, Imperial College London, London, United Kingdom
| | - Mustafa B A Djamgoz
- Neuroscience Solutions to Cancer Research Group, Department of Life Sciences, Imperial College London, London, United Kingdom
| |
Collapse
|
31
|
Caruana I, Simula L, Locatelli F, Campello S. T lymphocytes against solid malignancies: winning ways to defeat tumours. Cell Stress 2018; 2:200-212. [PMID: 31225487 PMCID: PMC6551626 DOI: 10.15698/cst2018.07.148] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
In the last decades, a novel field has emerged in the cure of cancer, by boosting the ability of the patient’s immune system to recognize and kill tumour cells. Although excellent and encouraging results, exploiting the effect of genetically modified T cells, have been obtained, it is now evident that tumour malignancies can evolve several mechanisms to escape such immune responses, thus continuing their growth in the body. These mechanisms are in part due to tumour cell metabolic or genetic alterations, which can render the target invisible to the immune system or can favour the generation of an extracellular milieu preventing immune cell infiltration or cytotoxicity. Such mechanisms may also involve the accumulation inside the tumour microenvironment of different immune-suppressive cell types, which further down-regulate the activity of cytotoxic immune cells either directly by interacting with them or indirectly by releasing suppressive molecules. In this review, we will first focus on describing several mechanisms by which tumour cells may dampen or abrogate the immune response inside the tumour microenvironment and, second, on current strategies that are adopted to cope with and possibly overcome such alterations, thus ameliorating the efficacy of the current-in-use anti-cancer immuno-therapies.
Collapse
Affiliation(s)
- Ignazio Caruana
- Dept. of Pediatric Onco-Hematology and cell and gene therapy, IRCCS Bambino Gesù Children's Hospital, Rome, Italy
| | - Luca Simula
- Dept. of Pediatric Onco-Hematology and cell and gene therapy, IRCCS Bambino Gesù Children's Hospital, Rome, Italy.,Dept. of Biology, University of Rome Tor Vergata, Rome, Italy
| | - Franco Locatelli
- Dept. of Pediatric Onco-Hematology and cell and gene therapy, IRCCS Bambino Gesù Children's Hospital, Rome, Italy
| | - Silvia Campello
- Dept. of Biology, University of Rome Tor Vergata, Rome, Italy.,IRCCS, Santa Lucia Foundation, Rome, Italy
| |
Collapse
|
32
|
Kv1.3 activity perturbs the homeostatic properties of astrocytes in glioma. Sci Rep 2018; 8:7654. [PMID: 29769580 PMCID: PMC5955950 DOI: 10.1038/s41598-018-25940-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Accepted: 05/02/2018] [Indexed: 01/06/2023] Open
Abstract
Glial cells actively maintain the homeostasis of brain parenchyma, regulating neuronal excitability and preserving the physiological composition of the extracellular milieu. Under pathological conditions, some functions of glial cells could be compromised, exacerbating the neurotoxic processes. We investigated if the homeostatic activities of astrocytes and microglia could be modulated by the voltage-gated K+ channel Kv1.3. To this end we used in vitro and in vivo systems to model cell-to-cell interactions in tumoral conditions, using a specific inhibitor of Kv1.3 channels, 5-(4-phenoxybutoxy) psoralen (PAP-1). We demonstrated that PAP-1 increases astrocytic glutamate uptake, reduces glioma-induced neurotoxicity, and decreases microglial migration and phagocytosis. We also found in a tumor blood brain barrier model that Kv1.3 activity is required for its integrity. The crucial role of Kv1.3 channels as modulators of glial cell activity was confirmed in a mouse model of glioma, where PAP-1 treatment reduces tumor volume only in the presence of active glutamate transporters GLT-1. In the same mouse model, PAP-1 reduces astrogliosis and microglial infiltration. PAP-1 also reduces tumor cell invasion. All these findings point to Kv1.3 channels as potential targets to re-instruct glial cells toward their homeostatic functions, in the context of brain tumors.
Collapse
|
33
|
Chimote AA, Balajthy A, Arnold MJ, Newton HS, Hajdu P, Qualtieri J, Wise-Draper T, Conforti L. A defect in KCa3.1 channel activity limits the ability of CD8 + T cells from cancer patients to infiltrate an adenosine-rich microenvironment. Sci Signal 2018; 11:11/527/eaaq1616. [PMID: 29692361 DOI: 10.1126/scisignal.aaq1616] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The limited ability of cytotoxic T cells to infiltrate solid tumors hampers immune surveillance and the efficacy of immunotherapies in cancer. Adenosine accumulates in solid tumors and inhibits tumor-specific T cells. Adenosine inhibits T cell motility through the A2A receptor (A2AR) and suppression of KCa3.1 channels. We conducted three-dimensional chemotaxis experiments to elucidate the effect of adenosine on the migration of peripheral blood CD8+ T cells from head and neck squamous cell carcinoma (HNSCC) patients. The chemotaxis of HNSCC CD8+ T cells was reduced in the presence of adenosine, and the effect was greater on HNSCC CD8+ T cells than on healthy donor (HD) CD8+ T cells. This response correlated with the inability of CD8+ T cells to infiltrate tumors. The effect of adenosine was mimicked by an A2AR agonist and prevented by an A2AR antagonist. We found no differences in A2AR expression, 3',5'-cyclic adenosine monophosphate abundance, or protein kinase A type 1 activity between HNSCC and HD CD8+ T cells. We instead detected a decrease in KCa3.1 channel activity, but not expression, in HNSCC CD8+ T cells. Activation of KCa3.1 channels by 1-EBIO restored the ability of HNSCC CD8+ T cells to chemotax in the presence of adenosine. Our data highlight the mechanism underlying the increased sensitivity of HNSCC CD8+ T cells to adenosine and the potential therapeutic benefit of KCa3.1 channel activators, which could increase infiltration of these T cells into tumors.
Collapse
Affiliation(s)
- Ameet A Chimote
- Division of Nephrology, Department of Internal Medicine, University of Cincinnati, Cincinnati, OH 45267, USA
| | - Andras Balajthy
- Division of Nephrology, Department of Internal Medicine, University of Cincinnati, Cincinnati, OH 45267, USA
| | - Michael J Arnold
- Division of Nephrology, Department of Internal Medicine, University of Cincinnati, Cincinnati, OH 45267, USA
| | - Hannah S Newton
- Division of Nephrology, Department of Internal Medicine, University of Cincinnati, Cincinnati, OH 45267, USA
| | - Peter Hajdu
- Division of Nephrology, Department of Internal Medicine, University of Cincinnati, Cincinnati, OH 45267, USA
| | - Julianne Qualtieri
- Department of Pathology, University of Cincinnati, Cincinnati, OH 45267, USA
| | - Trisha Wise-Draper
- Division of Hematology Oncology, Department of Internal Medicine, University of Cincinnati, Cincinnati, OH 45267, USA
| | - Laura Conforti
- Division of Nephrology, Department of Internal Medicine, University of Cincinnati, Cincinnati, OH 45267, USA.
| |
Collapse
|
34
|
Gurusamy D, Clever D, Eil R, Restifo NP. Novel "Elements" of Immune Suppression within the Tumor Microenvironment. Cancer Immunol Res 2018; 5:426-433. [PMID: 28576921 DOI: 10.1158/2326-6066.cir-17-0117] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Revised: 03/30/2017] [Accepted: 04/19/2017] [Indexed: 01/05/2023]
Abstract
Adaptive evolution has prompted immune cells to use a wide variety of inhibitory signals, many of which are usurped by tumor cells to evade immune surveillance. Although tumor immunologists often focus on genes and proteins as mediators of immune function, here we highlight two elements from the periodic table-oxygen and potassium-that suppress the immune system in previously unappreciated ways. While both are key to the maintenance of T-cell function and tissue homeostasis, they are exploited by tumors to suppress immuno-surveillance and promote metastatic spread. We discuss the temporal and spatial roles of these elements within the tumor microenvironment and explore possible therapeutic interventions for effective and promising anticancer therapies. Cancer Immunol Res; 5(6); 426-33. ©2017 AACR.
Collapse
Affiliation(s)
- Devikala Gurusamy
- Surgery Branch, National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, Maryland.,Center for Cell-Based Therapy, National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, MD
| | - David Clever
- Medical Scientist Training Program, The Ohio State University College of Medicine, Columbus, Ohio
| | - Robert Eil
- Department of Surgery, Oregon Health and Sciences University, Portland, Oregon
| | - Nicholas P Restifo
- Surgery Branch, National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, Maryland. .,Center for Cell-Based Therapy, National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, MD
| |
Collapse
|
35
|
Massa C, Seliger B. The tumor microenvironment: Thousand obstacles for effector T cells. Cell Immunol 2017; 343:103730. [PMID: 29249298 DOI: 10.1016/j.cellimm.2017.12.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2017] [Revised: 12/04/2017] [Accepted: 12/07/2017] [Indexed: 12/24/2022]
Abstract
The immune system is endowed with the capability to recognize and destroy transformed cells, but even in the presence of an immune infiltrate many tumors do progress. In the last decades new discoveries have shed light into (some of) the underlying mechanisms. Immune effector cells are not only under the influence of immune suppressive cell subsets, but also intrinsically regulated by immune check point molecules that under physiological condition avoid attach of healthy tissue. Moreover, tumor cells are modifying the surrounding microenvironment through secretion of immune modulators as well as via their own metabolism, thus further impairing the development of immune effector functions. Different approaches are currently being evaluated in the clinic to overcome those regulatory mechanisms and to unleash effector T cells.
Collapse
Affiliation(s)
- Chiara Massa
- Institute for Medical Immunology, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Barbara Seliger
- Institute for Medical Immunology, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany.
| |
Collapse
|
36
|
Venom-derived peptide inhibitors of voltage-gated potassium channels. Neuropharmacology 2017; 127:124-138. [PMID: 28689025 DOI: 10.1016/j.neuropharm.2017.07.002] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Revised: 07/02/2017] [Accepted: 07/04/2017] [Indexed: 12/11/2022]
Abstract
Voltage-gated potassium channels play a key role in human physiology and pathology. Reflecting their importance, numerous channelopathies have been characterised that arise from mutations in these channels or from autoimmune attack on the channels. Voltage-gated potassium channels are also the target of a broad range of peptide toxins from venomous organisms, including sea anemones, scorpions, spiders, snakes and cone snails; many of these peptides bind to the channels with high potency and selectivity. In this review we describe the various classes of peptide toxins that block these channels and illustrate the broad range of three-dimensional structures that support channel blockade. The therapeutic opportunities afforded by these peptides are also highlighted. This article is part of the Special Issue entitled 'Venom-derived Peptides as Pharmacological Tools.'
Collapse
|
37
|
Leanza L, Romio M, Becker KA, Azzolini M, Trentin L, Managò A, Venturini E, Zaccagnino A, Mattarei A, Carraretto L, Urbani A, Kadow S, Biasutto L, Martini V, Severin F, Peruzzo R, Trimarco V, Egberts JH, Hauser C, Visentin A, Semenzato G, Kalthoff H, Zoratti M, Gulbins E, Paradisi C, Szabo I. Direct Pharmacological Targeting of a Mitochondrial Ion Channel Selectively Kills Tumor Cells In Vivo. Cancer Cell 2017; 31:516-531.e10. [PMID: 28399409 DOI: 10.1016/j.ccell.2017.03.003] [Citation(s) in RCA: 131] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Revised: 02/03/2017] [Accepted: 03/07/2017] [Indexed: 12/13/2022]
Abstract
The potassium channel Kv1.3 is highly expressed in the mitochondria of various cancerous cells. Here we show that direct inhibition of Kv1.3 using two mitochondria-targeted inhibitors alters mitochondrial function and leads to reactive oxygen species (ROS)-mediated death of even chemoresistant cells independently of p53 status. These inhibitors killed 98% of ex vivo primary chronic B-lymphocytic leukemia tumor cells while sparing healthy B cells. In orthotopic mouse models of melanoma and pancreatic ductal adenocarcinoma, the compounds reduced tumor size by more than 90% and 60%, respectively, while sparing immune and cardiac functions. Our work provides direct evidence that specific pharmacological targeting of a mitochondrial potassium channel can lead to ROS-mediated selective apoptosis of cancer cells in vivo, without causing significant side effects.
Collapse
Affiliation(s)
- Luigi Leanza
- Department of Biology, University of Padova, viale G. Colombo 3, 35121 Padova, Italy
| | - Matteo Romio
- Department of Chemical Sciences, University of Padova, via F. Marzolo 1, 35121 Padova, Italy
| | - Katrin Anne Becker
- Department of Molecular Biology, University of Duisburg-Essen, Hufelandstrasse 55, 45122 Essen, Germany
| | - Michele Azzolini
- Department of Biomedical Sciences, University of Padova, viale G. Colombo 3, 35121 Padova, Italy; CNR Institute of Neuroscience, viale G. Colombo 3, 35121 Padova, Italy
| | - Livio Trentin
- Department of Medicine, Hematology and Immunological Branch, University of Padova, and Venetian Institute for Molecular Medicine (VIMM), via G. Orus 2, 35129 Padova, Italy
| | - Antonella Managò
- Department of Biology, University of Padova, viale G. Colombo 3, 35121 Padova, Italy
| | - Elisa Venturini
- Department of Molecular Biology, University of Duisburg-Essen, Hufelandstrasse 55, 45122 Essen, Germany
| | - Angela Zaccagnino
- Institute for Experimental Cancer Research, Medical Faculty, CAU, Kiel, and Department of Surgery, UKSH, Campus Kiel, Arnold-Heller-Strasse 3 (Haus 17), 24105 Kiel, Germany
| | - Andrea Mattarei
- Department of Chemical Sciences, University of Padova, via F. Marzolo 1, 35121 Padova, Italy
| | - Luca Carraretto
- Department of Biology, University of Padova, viale G. Colombo 3, 35121 Padova, Italy
| | - Andrea Urbani
- Department of Biology, University of Padova, viale G. Colombo 3, 35121 Padova, Italy
| | - Stephanie Kadow
- Department of Molecular Biology, University of Duisburg-Essen, Hufelandstrasse 55, 45122 Essen, Germany
| | - Lucia Biasutto
- Department of Biomedical Sciences, University of Padova, viale G. Colombo 3, 35121 Padova, Italy; CNR Institute of Neuroscience, viale G. Colombo 3, 35121 Padova, Italy
| | - Veronica Martini
- Department of Medicine, Hematology and Immunological Branch, University of Padova, and Venetian Institute for Molecular Medicine (VIMM), via G. Orus 2, 35129 Padova, Italy
| | - Filippo Severin
- Department of Medicine, Hematology and Immunological Branch, University of Padova, and Venetian Institute for Molecular Medicine (VIMM), via G. Orus 2, 35129 Padova, Italy
| | - Roberta Peruzzo
- Department of Biology, University of Padova, viale G. Colombo 3, 35121 Padova, Italy
| | - Valentina Trimarco
- Department of Medicine, Hematology and Immunological Branch, University of Padova, and Venetian Institute for Molecular Medicine (VIMM), via G. Orus 2, 35129 Padova, Italy
| | - Jan-Hendrik Egberts
- Institute for Experimental Cancer Research, Medical Faculty, CAU, Kiel, and Department of Surgery, UKSH, Campus Kiel, Arnold-Heller-Strasse 3 (Haus 17), 24105 Kiel, Germany
| | - Charlotte Hauser
- Institute for Experimental Cancer Research, Medical Faculty, CAU, Kiel, and Department of Surgery, UKSH, Campus Kiel, Arnold-Heller-Strasse 3 (Haus 17), 24105 Kiel, Germany
| | - Andrea Visentin
- Department of Medicine, Hematology and Immunological Branch, University of Padova, and Venetian Institute for Molecular Medicine (VIMM), via G. Orus 2, 35129 Padova, Italy
| | - Gianpietro Semenzato
- Department of Medicine, Hematology and Immunological Branch, University of Padova, and Venetian Institute for Molecular Medicine (VIMM), via G. Orus 2, 35129 Padova, Italy
| | - Holger Kalthoff
- Institute for Experimental Cancer Research, Medical Faculty, CAU, Kiel, and Department of Surgery, UKSH, Campus Kiel, Arnold-Heller-Strasse 3 (Haus 17), 24105 Kiel, Germany
| | - Mario Zoratti
- Department of Biomedical Sciences, University of Padova, viale G. Colombo 3, 35121 Padova, Italy; CNR Institute of Neuroscience, viale G. Colombo 3, 35121 Padova, Italy
| | - Erich Gulbins
- Department of Molecular Biology, University of Duisburg-Essen, Hufelandstrasse 55, 45122 Essen, Germany; Department of Surgery, University of Cincinnati, 231 Albert Sabin Way, Cincinnati, OH 45267-0558, USA.
| | - Cristina Paradisi
- Department of Chemical Sciences, University of Padova, via F. Marzolo 1, 35121 Padova, Italy.
| | - Ildiko Szabo
- Department of Biology, University of Padova, viale G. Colombo 3, 35121 Padova, Italy; CNR Institute of Neuroscience, viale G. Colombo 3, 35121 Padova, Italy.
| |
Collapse
|
38
|
Conforti L. Potassium channels of T lymphocytes take center stage in the fight against cancer. J Immunother Cancer 2017; 5:2. [PMID: 28105369 PMCID: PMC5240326 DOI: 10.1186/s40425-016-0202-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Accepted: 12/06/2016] [Indexed: 01/17/2023] Open
Abstract
A recent study by Eil at al. published in Nature in September 2016 provides evidence that alterations of the K+ homeostasis of tumor infiltrating lymphocytes (TILs) in necrotic areas of the tumor microenvironment (TME) suppress the function of effector T cells. Furthermore, they establish that overexpression of K+ channels in T lymphocytes counterbalances this negative effect of the TME and restores the ability of TILs to function, ultimately leading to increased survival of tumor bearing mice. Thus, K+ channels in T lymphocytes become interesting new targets for novel immunotherapies in cancer. This Commentary discusses Eil's finding in the context of the central role that K+ channels play in the suppressed state of TILs as they mediate the immunosuppressive effects of multiple conditions of the TME including hypoxia and adenosine.
Collapse
Affiliation(s)
- Laura Conforti
- Department of Internal Medicine, Division of Nephrology, University of Cincinnati, Cincinnati, OH USA
| |
Collapse
|