1
|
Rezayat F, Esmaeil N, Nikpour P, Feizi A, Rezaei A. Different behavior of NK cells isolated from healthy women and women with recurrent spontaneous abortion after treatment with human amniotic epithelial cells. J Leukoc Biol 2025; 117:qiaf020. [PMID: 40276927 DOI: 10.1093/jleuko/qiaf020] [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: 06/26/2024] [Revised: 12/03/2024] [Indexed: 04/26/2025] Open
Abstract
Maternal immunotolerance during pregnancy is heavily dependent on the critical properties of human amniotic epithelial cells (hAECs). Recurrent spontaneous abortion (RSA) is one of the most common diseases in women and is caused by feto-maternal immunotolerance disruption. The objective of this study is to investigate how hAEECs affect pNK cells isolated from RSA and healthy women in terms of immunomodulation. Peripheral blood NK cells were isolated from 20 women with RSA and 20 healthy women. Purified NK cells were co-cultured with hAECs, obtained from full-term healthy pregnant women at different cellular ratios. After 24 and 72 h of incubation, the expression of immunomodulatory genes in hAECs, immunophenotype, and cytotoxicity of NK cells, and cytokine production were investigated using real-time PCR, flow cytometry, and ELISA techniques, respectively. We observed a significant increase in TGF-β and IL-10 production, and CD56bright CD16+ subpopulation in pNK cells, a significant decrease in IFN-γ production and CD107a and FasL expression on NK cells. Also, NK cells' cytotoxicity against K562 cells was diminished after co-culture with hAECs. The expression of TGF-β and HLA-G genes by hAECs was diminished after co-culture with NK cells isolated from women with RSA. Our research indicates that the interaction between NK cells and hAECs influences the phenotype and function of both cells. Also, NK cells belonging to women with RSA and healthy women exhibit different behavior during treatment with hAECs, possibly due to NK cell dysfunction. However, extensive research is required to assess NK cell defects and their mutual interaction with hAECs.
Collapse
Affiliation(s)
- Fatemeh Rezayat
- Department of Immunology, School of Medicine, Isfahan University of Medical Sciences, Hezar Jerib Street, Isfahan 8174673461, Iran
| | - Nafiseh Esmaeil
- Department of Immunology, School of Medicine, Isfahan University of Medical Sciences, Hezar Jerib Street, Isfahan 8174673461, Iran
- Pooya Zist-Mabna Hakim Company, Isfahan Health Center, Aghababaei Highway, Isfahan 8159611119, Iran
| | - Parvaneh Nikpour
- Department of Genetics and Molecular Biology, Faculty of Medicine, Isfahan University of Medical Sciences, Hezar Jerib Street, Isfahan 8174673461, Iran
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy at University of Gothenburg, Post box 100 405 30, Gothenburg, Sweden
| | - Awat Feizi
- Department of Epidemiology and Biostatistics, Isfahan Endocrine and Metabolism Research Center, School of Public Health, Isfahan University of Medical Sciences, Hezar Jerib Street, Isfahan 8174673461, Iran
| | - Abbas Rezaei
- Department of Immunology, School of Medicine, Isfahan University of Medical Sciences, Hezar Jerib Street, Isfahan 8174673461, Iran
- Immunodeficiency Diseases Research Center, Isfahan University of Medical Sciences, Hezar Jerib Street, Isfahan 8174673461, Iran
| |
Collapse
|
2
|
Rea A, Santana-Hernández S, Villanueva J, Sanvicente-García M, Cabo M, Suarez-Olmos J, Quimis F, Qin M, Llorens E, Blasco-Benito S, Torralba-Raga L, Perez L, Bhattarai B, Alari-Pahissa E, Georgoudaki AM, Balaguer F, Juan M, Pardo J, Celià-Terrassa T, Rovira A, Möker N, Zhang C, Colonna M, Spanholtz J, Malmberg KJ, Montagut C, Albanell J, Güell M, López-Botet M, Muntasell A. Enhancing human NK cell antitumor function by knocking out SMAD4 to counteract TGFβ and activin A suppression. Nat Immunol 2025; 26:582-594. [PMID: 40119192 PMCID: PMC11957989 DOI: 10.1038/s41590-025-02103-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Accepted: 01/30/2025] [Indexed: 03/24/2025]
Abstract
Transforming growth factor beta (TGFβ) and activin A suppress natural killer (NK) cell function and proliferation, limiting the efficacy of adoptive NK cell therapies. Inspired by the partial resistance to TGFβ of NK cells with SMAD4 haploinsufficiency, we used CRISPR-Cas9 for knockout of SMAD4 in human NK cells. Here we show that SMAD4KO NK cells were resistant to TGFβ and activin A inhibition, retaining their cytotoxicity, cytokine secretion and interleukin-2/interleukin-15-driven proliferation. They showed enhanced tumor penetration and tumor growth control, both as monotherapy and in combination with tumor-targeted therapeutic antibodies. Notably, SMAD4KO NK cells outperformed control NK cells treated with a TGFβ inhibitor, underscoring the benefit of maintaining SMAD4-independent TGFβ signaling. SMAD4KO conferred TGFβ resistance across diverse NK cell platforms, including CD19-CAR NK cells, stem cell-derived NK cells and ADAPT-NK cells. These findings position SMAD4 knockout as a versatile and compelling strategy to enhance NK cell antitumor activity, providing a new avenue for improving NK cell-based cancer immunotherapies.
Collapse
Grants
- 765104 EC | EC Seventh Framework Programm | FP7 People: Marie-Curie Actions (FP7-PEOPLE - Specific Programme "People" Implementing the Seventh Framework Programme of the European Community for Research, Technological Development and Demonstration Activities (2007 to 2013))
- ICI24/00041 Ministry of Economy and Competitiveness | Instituto de Salud Carlos III (Institute of Health Carlos III)
- SGR863 Generalitat de Catalunya (Government of Catalonia)
- 765104 EC | EU Framework Programme for Research and Innovation H2020 | H2020 Priority Excellent Science | H2020 Marie Sklodowska-Curie Actions (H2020 Excellent Science - Marie Sklodowska-Curie Actions)
- PI21/00002 Ministry of Economy and Competitiveness | Instituto de Salud Carlos III (Institute of Health Carlos III)
- PI22/00040 Ministry of Economy and Competitiveness | Instituto de Salud Carlos III (Institute of Health Carlos III)
- 2024PROD00089 Departament d'Innovació, Universitats i Empresa, Generalitat de Catalunya (Department of Innovation, Education and Enterprise, Government of Catalonia)
- FI23/00075 Ministry of Economy and Competitiveness | Instituto de Salud Carlos III (Institute of Health Carlos III)
- P01 CA111412 NCI NIH HHS
- Ministerio de Ciencia, Innovación y Universidades/FEDER CNS2023-144487
- AECC postdoctoral fellowship POSTD234709BLAS
- Ministerio de Ciencia, Innovación y Universidades PID2020-113963RBI00 Gobierno de Aragón B29-23R
- Ministerio de Ciencia, Innovación y Universidades PID2023-147310OB-I00
- Research Council of Norway 275469, 237579, the Research Council of Norway through its Centres of Excellence scheme 332727, the Norwegian Cancer Society-190386, 223310, The South-Eastern Norway Regional Health Authority 2021-073, 2024-053, Knut and Alice Wallenberg Foundation 2018.0106, Swedish Foundation for Strategic Research, and the US National Cancer Institute P01 CA111412, P009500901.
- CRIS EXCELLENCE 19-30, funded by CRIS Contra el Cáncer
- CIBERONC: CB16/12/00241
Collapse
Affiliation(s)
- Anna Rea
- University Pompeu Fabra (UPF), Barcelona, Spain
| | - Sara Santana-Hernández
- Hospital del Mar Research Institute (IMIM), Barcelona, Spain
- Institut de Biotecnologia i Biomedicina, Cell Biology, Physiology and Immunology Deptartments, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Javier Villanueva
- University Pompeu Fabra (UPF), Barcelona, Spain
- Institut de Biotecnologia i Biomedicina, Cell Biology, Physiology and Immunology Deptartments, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | | | - Mariona Cabo
- Hospital del Mar Research Institute (IMIM), Barcelona, Spain
| | | | - Fabricio Quimis
- Hospital del Mar Research Institute (IMIM), Barcelona, Spain
| | - Mengjuan Qin
- Hospital del Mar Research Institute (IMIM), Barcelona, Spain
| | - Eduard Llorens
- Hospital del Mar Research Institute (IMIM), Barcelona, Spain
| | | | - Lamberto Torralba-Raga
- Precision Immunotherapy Alliance, The University of Oslo, Oslo, Norway
- Department of Cancer Immunology, Institute for Cancer Research Oslo, Oslo University Hospital, Oslo, Norway
| | - Lorena Perez
- Department of Immunology, Hospital Clínic de Barcelona (HCB), Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Joint Platform of Immunotherapy Hospital Sant Joan de Deu - HCB, University of Barcelona, Barcelona, Spain
| | - Bishan Bhattarai
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | | | | | - Francesc Balaguer
- Department of Gastroenterology, Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), University of Barcelona, Barcelona, Spain
| | - Manel Juan
- Department of Immunology, Hospital Clínic de Barcelona (HCB), Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Joint Platform of Immunotherapy Hospital Sant Joan de Deu - HCB, University of Barcelona, Barcelona, Spain
| | - Julián Pardo
- IIS Aragon Foundation/ Dpt. Microbiology, Radiology Pediatry and Public Health, University of Zaragoza, Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERinfec), Zaragoza, Spain
| | - Toni Celià-Terrassa
- Hospital del Mar Research Institute (IMIM), Barcelona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
| | - Ana Rovira
- Hospital del Mar Research Institute (IMIM), Barcelona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
- Department of Oncology, Hospital del Mar, Barcelona, Spain
| | - Nina Möker
- Miltenyi Biotec B.V. & Co. KG, Bergisch Gladbach, Germany
| | - Congcong Zhang
- Miltenyi Biotec B.V. & Co. KG, Bergisch Gladbach, Germany
| | - Marco Colonna
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | | | - Karl-Johan Malmberg
- Precision Immunotherapy Alliance, The University of Oslo, Oslo, Norway
- Department of Cancer Immunology, Institute for Cancer Research Oslo, Oslo University Hospital, Oslo, Norway
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Clara Montagut
- University Pompeu Fabra (UPF), Barcelona, Spain
- Hospital del Mar Research Institute (IMIM), Barcelona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
- Department of Oncology, Hospital del Mar, Barcelona, Spain
| | - Joan Albanell
- University Pompeu Fabra (UPF), Barcelona, Spain
- Hospital del Mar Research Institute (IMIM), Barcelona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
- Department of Oncology, Hospital del Mar, Barcelona, Spain
| | - Marc Güell
- University Pompeu Fabra (UPF), Barcelona, Spain
- Institució Catalana de Recerca i Estudis Avançats, ICREA, Barcelona, Spain
| | - Miguel López-Botet
- University Pompeu Fabra (UPF), Barcelona, Spain
- Hospital del Mar Research Institute (IMIM), Barcelona, Spain
| | - Aura Muntasell
- Hospital del Mar Research Institute (IMIM), Barcelona, Spain.
- Institut de Biotecnologia i Biomedicina, Cell Biology, Physiology and Immunology Deptartments, Universitat Autònoma de Barcelona, Bellaterra, Spain.
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain.
| |
Collapse
|
3
|
Aftabi S, Barzegar Behrooz A, Cordani M, Rahiman N, Sadeghdoust M, Aligolighasemabadi F, Pistorius S, Alavizadeh SH, Taefehshokr N, Ghavami S. Therapeutic targeting of TGF-β in lung cancer. FEBS J 2025; 292:1520-1557. [PMID: 39083441 PMCID: PMC11970718 DOI: 10.1111/febs.17234] [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: 11/05/2023] [Revised: 05/22/2024] [Accepted: 07/19/2024] [Indexed: 08/02/2024]
Abstract
Transforming growth factor-β (TGF-β) plays a complex role in lung cancer pathophysiology, initially acting as a tumor suppressor by inhibiting early-stage tumor growth. However, its role evolves in the advanced stages of the disease, where it contributes to tumor progression not by directly promoting cell proliferation but by enhancing epithelial-mesenchymal transition (EMT) and creating a conducive tumor microenvironment. While EMT is typically associated with enhanced migratory and invasive capabilities rather than proliferation per se, TGF-β's influence on this process facilitates the complex dynamics of tumor metastasis. Additionally, TGF-β impacts the tumor microenvironment by interacting with immune cells, a process influenced by genetic and epigenetic changes within tumor cells. This interaction highlights its role in immune evasion and chemoresistance, further complicating lung cancer therapy. This review provides a critical overview of recent findings on TGF-β's involvement in lung cancer, its contribution to chemoresistance, and its modulation of the immune response. Despite the considerable challenges encountered in clinical trials and the development of new treatments targeting the TGF-β pathway, this review highlights the necessity for continued, in-depth investigation into the roles of TGF-β. A deeper comprehension of these roles may lead to novel, targeted therapies for lung cancer. Despite the intricate behavior of TGF-β signaling in tumors and previous challenges, further research could yield innovative treatment strategies.
Collapse
Affiliation(s)
- Sajjad Aftabi
- Department of Human Anatomy and Cell ScienceUniversity of Manitoba College of MedicineWinnipegCanada
- Paul Albrechtsen Research Institute, CancerCare ManitobaUniversity of ManitobaWinnipegCanada
- Department of Physics and AstronomyUniversity of ManitobaWinnipegCanada
| | - Amir Barzegar Behrooz
- Department of Human Anatomy and Cell ScienceUniversity of Manitoba College of MedicineWinnipegCanada
- Electrophysiology Research Center, Neuroscience InstituteTehran University of Medical SciencesIran
| | - Marco Cordani
- Department of Biochemistry and Molecular Biology, Faculty of BiologyComplutense UniversityMadridSpain
- Instituto de Investigaciones Sanitarias San Carlos (IdISSC)MadridSpain
| | - Niloufar Rahiman
- Nanotechnology Research Center, Pharmaceutical Technology InstituteMashhad University of Medical SciencesIran
- Department of Pharmaceutical Nanotechnology, School of PharmacyMashhad University of Medical SciencesIran
| | - Mohammadamin Sadeghdoust
- Division of BioMedical Sciences, Faculty of MedicineMemorial University of NewfoundlandSt. John'sCanada
| | - Farnaz Aligolighasemabadi
- Department of Human Anatomy and Cell ScienceUniversity of Manitoba College of MedicineWinnipegCanada
| | - Stephen Pistorius
- Department of Human Anatomy and Cell ScienceUniversity of Manitoba College of MedicineWinnipegCanada
- Paul Albrechtsen Research Institute, CancerCare ManitobaUniversity of ManitobaWinnipegCanada
- Department of Physics and AstronomyUniversity of ManitobaWinnipegCanada
| | - Seyedeh Hoda Alavizadeh
- Nanotechnology Research Center, Pharmaceutical Technology InstituteMashhad University of Medical SciencesIran
- Department of Pharmaceutical Nanotechnology, School of PharmacyMashhad University of Medical SciencesIran
| | - Nima Taefehshokr
- Apoptosis Research CentreChildren's Hospital of Eastern Ontario Research InstituteOttawaCanada
| | - Saeid Ghavami
- Department of Human Anatomy and Cell ScienceUniversity of Manitoba College of MedicineWinnipegCanada
- Paul Albrechtsen Research Institute, CancerCare ManitobaUniversity of ManitobaWinnipegCanada
- Faculty Academy of Silesia, Faculty of MedicineKatowicePoland
- Children Hospital Research Institute of ManitobaUniversity of ManitobaWinnipegCanada
| |
Collapse
|
4
|
Shin SH, Lee YE, Yoon HN, Yuk CM, An JY, Seo M, Yoon S, Oh MS, Shin SC, Kim JH, Kim YJ, Kim JC, Kim SC, Jang M. An innovative strategy harnessing self-activating CAR-NK cells to mitigate TGF-β1-driven immune suppression. Biomaterials 2025; 314:122888. [PMID: 39423512 DOI: 10.1016/j.biomaterials.2024.122888] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 09/19/2024] [Accepted: 10/09/2024] [Indexed: 10/21/2024]
Abstract
The dysfunction of natural killer (NK) cells, mediated by transforming growth factor β1 (TGFβ1) within the tumor microenvironment, impedes antitumor therapy and contributes to poor clinical outcomes. Our study introduces self-activating chimeric antigen receptor (CAR)-NK cells that block TGFβ1 signaling by releasing a specifically designed peptide, P6, which targets mesothelin in pancreatic tumors. P6 originates from the interaction sites between TGFβ1 and TGFβ receptor 1 and effectively disrupts TGFβ1's inhibitory signaling in NK cells. Our analysis demonstrates that P6 treatment interrupts the SMAD2/3 pathway in NK cells, mitigating TGFβ1-mediated suppression of NK cell activity, thereby enhancing their metabolic function and cytotoxic response against pancreatic tumors. These CAR-NK cells exhibit potent antitumor capabilities, as evidenced in spheroid cultures with cancer-associated fibroblasts and in vivo mouse models. Our approach marks a substantial advancement in overcoming TGFβ1-mediated immune evasion, offering a promising avenue for revolutionizing cancer immunotherapy.
Collapse
Affiliation(s)
- Seung Hun Shin
- Medicinal Materials Research Center, Biomedical Research Division, Korea Institute of Science and Technology, Seoul, Republic of Korea; Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, Republic of Korea
| | - Young Eun Lee
- Medicinal Materials Research Center, Biomedical Research Division, Korea Institute of Science and Technology, Seoul, Republic of Korea
| | - Han-Na Yoon
- Rare & Pediatric Cancer Branch, Division of Rare and Refractory Cancer, Research Institute, National Cancer Center, Goyang, Republic of Korea
| | - Chae Min Yuk
- Center for Advanced Biomolecular Recognition, Biomedical Research Division, Korea Institute of Science and Technology, Seoul, Republic of Korea
| | - Jun Yop An
- Corporate Research & Development Center, UCI Therapeutics, Seoul, Republic of Korea
| | - Minkoo Seo
- Corporate Research & Development Center, UCI Therapeutics, Seoul, Republic of Korea
| | - Sangwon Yoon
- Corporate Research & Development Center, UCI Therapeutics, Seoul, Republic of Korea
| | - Min-Suk Oh
- Corporate Research & Development Center, UCI Therapeutics, Seoul, Republic of Korea
| | - Sang Chul Shin
- Technological Convergence Center, Korea Institute of Science and Technology, Seoul, Republic of Korea
| | - Ji Hyung Kim
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, Republic of Korea
| | - Yong Jun Kim
- Department of Pathology, College of Medicine, Kyung Hee University, Seoul, Republic of Korea; KHU-KIST Department of Converging Science and Technology, Kyung Hee University, Seoul, Republic of Korea
| | - Jin-Chul Kim
- Natural Products Research Institute, Korea Institute of Science and Technology, Gangneung, Republic of Korea
| | - Song Cheol Kim
- Division of Hepato-Biliary and Pancreatic Surgery, Department of Surgery, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Republic of Korea
| | - Mihue Jang
- Medicinal Materials Research Center, Biomedical Research Division, Korea Institute of Science and Technology, Seoul, Republic of Korea; KHU-KIST Department of Converging Science and Technology, Kyung Hee University, Seoul, Republic of Korea.
| |
Collapse
|
5
|
Hosseinalizadeh H, Wang LS, Mirzaei H, Amoozgar Z, Tian L, Yu J. Emerging combined CAR-NK cell therapies in cancer treatment: Finding a dancing partner. Mol Ther 2025:S1525-0016(24)00895-5. [PMID: 39754357 DOI: 10.1016/j.ymthe.2024.12.057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 09/21/2024] [Accepted: 12/31/2024] [Indexed: 01/06/2025] Open
Abstract
In recent decades, immunotherapy with chimeric antigen receptors (CARs) has revolutionized cancer treatment and given hope where other cancer therapies have failed. CAR-natural killer (NK) cells are NK cells that have been engineered ex vivo with a CAR on the cell membrane with high specificity for specific target antigens of tumor cells. The impressive results of several studies suggest that CAR-NK cell therapy has significant potential and successful performance in cancer treatment. Despite its effectiveness, CAR-NK cell therapy can have significant challenges when it comes to treating cancer. These challenges include tumor heterogeneity, antigen escape, an immunosuppressive tumor microenvironment, limited tissue migration from blood, exhaustion of CAR-NK cells, and inhibition by immunosuppressive checkpoint molecule signaling, etc. In CAR-T cell therapy, the use of combined approaches has shown encouraging outcomes for tumor regression and improved cancer treatment compared to single therapies. Therefore, to overcome these significant challenges in CAR-NK cells, innovative combination therapies of CAR-NK cells with other conventional therapies (e.g., chemotherapy and radiotherapy) or other immunotherapies are needed to counteract the above challenges and thereby increase the activity of CAR-NK cells. This review comprehensively discusses various cancer-treatment approaches in combination with CAR-NK cell therapy in the hope of providing valuable insights that may improve cancer treatment in the near future.
Collapse
Affiliation(s)
- Hamed Hosseinalizadeh
- Department of Medical Biotechnology, Faculty of Paramedicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Li-Shu Wang
- Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Hamed Mirzaei
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Zohreh Amoozgar
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Lei Tian
- Division of Hematology/Oncology, Department of Medicine, School of Medicine, University of California Irvine, Irvine, CA, USA; Chao Family Comprehensive Cancer Center, University of California Irvine, Irvine, CA, USA.
| | - Jianhua Yu
- Division of Hematology/Oncology, Department of Medicine, School of Medicine, University of California Irvine, Irvine, CA, USA; Chao Family Comprehensive Cancer Center, University of California Irvine, Irvine, CA, USA.
| |
Collapse
|
6
|
Liao Y, Zheng Y, Zhang R, Chen X, Huang J, Liu J, Zhao Y, Zheng Y, Zhang X, Gao Z, Gao X, Bu J, Peng T, Li X, Shen E. Regulatory roles of transcription factors T-bet and Eomes in group 1 ILCs. Int Immunopharmacol 2024; 143:113229. [PMID: 39357208 DOI: 10.1016/j.intimp.2024.113229] [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: 07/06/2024] [Revised: 09/18/2024] [Accepted: 09/19/2024] [Indexed: 10/04/2024]
Abstract
T-bet and Eomes, both T-box transcription factors, have been extensively studied for their critical roles in the differentiation and functional maintenance of various immune cells. In this review, we provide a focused overview of their contributions to the transcriptional activation and differentiation, development, and terminal maturation of natural killer cells and innate lymphoid cell 1 cells. Furthermore, the interplay between T-bet and Eomes in regulating NK cell function, and its subsequent implications for immune responses against infections and tumors, is thoroughly examined. The review explores the ramifications of dysregulated transcription factor expression, examining its impact on homeostatic balance and its role in a spectrum of disease models. Expression variances among distinct NK cell subsets resident in different tissues are highlighted to underscore the complexity of their biological roles. Collectively, this work aims to expand the current understanding of NK cell biology, thereby paving the way for innovative approaches in the realm of NK cell-based immunotherapies.
Collapse
Affiliation(s)
- Yue Liao
- Sino-French Hoffmann Institute, School of Basic Medical Sciences, The Second Affiliated Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Guangzhou Medical University, Guangzhou, China
| | - Yanling Zheng
- Sino-French Hoffmann Institute, School of Basic Medical Sciences, The Second Affiliated Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Guangzhou Medical University, Guangzhou, China; Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou, China
| | - Ruizhi Zhang
- Department of Emergency Medicine, the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Xiangming Chen
- Sino-French Hoffmann Institute, School of Basic Medical Sciences, The Second Affiliated Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Guangzhou Medical University, Guangzhou, China
| | - Jijun Huang
- Sino-French Hoffmann Institute, School of Basic Medical Sciences, The Second Affiliated Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Guangzhou Medical University, Guangzhou, China
| | - Jiamin Liu
- Sino-French Hoffmann Institute, School of Basic Medical Sciences, The Second Affiliated Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Guangzhou Medical University, Guangzhou, China
| | - Yuyang Zhao
- Sino-French Hoffmann Institute, School of Basic Medical Sciences, The Second Affiliated Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Guangzhou Medical University, Guangzhou, China
| | - Yu Zheng
- National Center for STD Control, Chinese Center for Disease Control and Prevention, Nanjing, Jiangsu, China; Hospital for Skin Disease (Institute of Dermatology), Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, Jiangsu, China
| | - Xueyan Zhang
- Sino-French Hoffmann Institute, School of Basic Medical Sciences, The Second Affiliated Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Guangzhou Medical University, Guangzhou, China
| | - Zhiyan Gao
- Sino-French Hoffmann Institute, School of Basic Medical Sciences, The Second Affiliated Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Guangzhou Medical University, Guangzhou, China
| | - Xiaojing Gao
- National Center for STD Control, Chinese Center for Disease Control and Prevention, Nanjing, Jiangsu, China; Hospital for Skin Disease (Institute of Dermatology), Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, Jiangsu, China
| | - Jin Bu
- National Center for STD Control, Chinese Center for Disease Control and Prevention, Nanjing, Jiangsu, China; Hospital for Skin Disease (Institute of Dermatology), Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, Jiangsu, China.
| | - Tieli Peng
- The Affiliated Qingyuan Hospital (Qingyuan People's Hospital), Guangzhou Medical University, China.
| | - Xiaomin Li
- Sino-French Hoffmann Institute, School of Basic Medical Sciences, The Second Affiliated Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Guangzhou Medical University, Guangzhou, China.
| | - Erxia Shen
- Sino-French Hoffmann Institute, School of Basic Medical Sciences, The Second Affiliated Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Guangzhou Medical University, Guangzhou, China.
| |
Collapse
|
7
|
Kim HW, Shin S, Park SH, Park JH, Kim SM, Lee YH, Lee MJ. Next-generation adjuvant systems containing furfurman drives potent adaptive immunity and host defense as a foot-and-mouth disease vaccine adjuvant. Front Immunol 2024; 15:1491043. [PMID: 39742276 PMCID: PMC11687127 DOI: 10.3389/fimmu.2024.1491043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Accepted: 11/26/2024] [Indexed: 01/03/2025] Open
Abstract
Introduction Many countries use commercial foot-and-mouth disease (FMD) vaccines to prevent FMD pandemics, but these vaccines have disadvantages, such as repeated vaccinations due to the short persistence of antibody (Ab) titers and incomplete host defense despite high Ab titers. To address these shortcomings, we aimed to develop a novel FMD vaccine containing furfurman as an adjuvant. Method To demonstrate the efficacy of the test vaccine, adaptive immunity was evaluated by measuring Ab and neutralizing Ab titers and host defense against viral infections in experimental and target animals. In addition, the expression levels of cytokines [interferon (IFN)α, IFNβ, IFNγ, interleukin (IL)-1β, IL-2, and IL-12p40] were evaluated at the early stages of vaccination to confirm the simultaneous induction of cellular and humoral immune responses induced by the test vaccine. Result The groups that received vaccine containing furfurman showed a strong early, mid-term, and long-term immune response and host defense against viral infections compared to the control groups. The significant upregulation observed in cytokine levels in the furfurman group compared to those in the control groups strongly suggest that the test vaccine strengthens cellular immune response and effectively induces a humoral immune response. Conclusion Our study demonstrated that furfurman, as an FMD vaccine adjuvant, achieves long-lasting immunity and host defense against viral infections by eliciting potent cellular and humoral immune responses. Therefore, our findings contribute to the design of next-generation FMD vaccines and highlight the potential application of furfurman as an adjuvant for other viral diseases.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Min Ja Lee
- Center for Foot-and-Mouth Disease Vaccine Research, Animal and Plant Quarantine Agency, Gimcheon-si, Gyeongsangbuk-do, Republic of Korea
| |
Collapse
|
8
|
Du F, Li J, Zhong X, Zhang Z, Zhao Y. Endothelial-to-mesenchymal transition in the tumor microenvironment: Roles of transforming growth factor-β and matrix metalloproteins. Heliyon 2024; 10:e40118. [PMID: 39568849 PMCID: PMC11577214 DOI: 10.1016/j.heliyon.2024.e40118] [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: 08/28/2023] [Revised: 10/26/2024] [Accepted: 11/03/2024] [Indexed: 11/22/2024] Open
Abstract
Cancer is a leading cause of global morbidity and mortality. Tumor cells grow in a complex microenvironment, comprising immune cells, stromal cells, and vascular cells, collaborating to support tumor growth and facilitate metastasis. Transforming growth factor-beta (TGF-β) is a multipotent factor that can not only affect fibrosis promotion but also assume distinct roles in the early and late stages of the tumor. Matrix metalloproteinases (MMPs) primarily function to degrade the extracellular matrix, a pivotal cellular player in tumor progression. Moreover, endothelial-to-mesenchymal transition (EndMT), similar to epithelial-to-mesenchymal transition, is associated with cancer progression by promoting angiogenesis, disrupting the endothelial barrier, and leading to cancer-associated fibroblasts. Recent studies have underscored the pivotal roles of TGF-β and MMPs in EndMT. This review delves into the contributions of TGF-β and MMPs, as well as their regulatory mechanisms, within the tumor microenvironment. This collective understanding offers fresh insights into the potential for combined targeted therapies in the fight against cancer.
Collapse
Affiliation(s)
- Fei Du
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
- Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan, China
- South Sichuan Institute of Translational Medicine, Luzhou, Sichuan, China
- Department of Pharmacy, Meishan TianFu New Area People's Hospital, Meishan, Sichuan, China
| | - Jing Li
- Department of Oncology and Hematology, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, China
| | - Xiaolin Zhong
- Department of Gastroenterology, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan, China
| | - Zhuo Zhang
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
- Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan, China
| | - Yueshui Zhao
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
- Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan, China
- South Sichuan Institute of Translational Medicine, Luzhou, Sichuan, China
| |
Collapse
|
9
|
Dhall A, Patiyal S, Raghava GPS. A hybrid method for discovering interferon-gamma inducing peptides in human and mouse. Sci Rep 2024; 14:26859. [PMID: 39501025 PMCID: PMC11538504 DOI: 10.1038/s41598-024-77957-8] [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: 07/26/2024] [Accepted: 10/28/2024] [Indexed: 11/08/2024] Open
Abstract
Interferon-gamma (IFN-γ) is a versatile pleiotropic cytokine essential for both innate and adaptive immune responses. It exhibits both pro-inflammatory and anti-inflammatory properties, making it a promising therapeutic candidate for treating various infectious diseases and cancers. We present IFNepitope2, a host-specific technique to annotate IFN-γ inducing peptides, it is an updated version of IFNepitope introduced by Dhanda et al. In this study, dataset used for developing prediction method contain experimentally validated 25,492 and 7983 IFN-γ inducing peptides in human and mouse host, respectively. In initial phase, machine learning techniques have been exploited to develop classification model using wide range of peptide features. Further, to improve machine learning based models or alignment free models, we explore potential of similarity-based technique BLAST. Finally, a hybrid model has been developed that combine best machine learning based model with BLAST. In most of the case, models based on extra tree perform better than other machine learning techniques. In case of peptide features, compositional feature particularly dipeptide composition performs better than one-hot encoding or binary profile. Our best machine learning based models achieved AUROC 0.89 and 0.83 for human and mouse host, respectively. The hybrid model achieved the AUROC 0.90 and 0.85 for human and mouse host, respectively. All models have been evaluated on an independent/validation dataset not used for training or testing these models. Newly developed method performs better than existing method on independent dataset. The major objective of this study is to predict, design and scan IFN-γ inducing peptides, thus server/software have been developed ( https://webs.iiitd.edu.in/raghava/ifnepitope2/ ). This method is also available as standalone at https://github.com/raghavagps/ifnepitope2 and python package index at https://pypi.org/project/ifnepitope2/ .
Collapse
Affiliation(s)
- Anjali Dhall
- Department of Computational Biology, Indraprastha Institute of Information Technology, Okhla Industrial Estate, Phase III, (Near Govind Puri Metro Station), New Delhi, 110020, India
| | - Sumeet Patiyal
- Department of Computational Biology, Indraprastha Institute of Information Technology, Okhla Industrial Estate, Phase III, (Near Govind Puri Metro Station), New Delhi, 110020, India
| | - Gajendra P S Raghava
- Department of Computational Biology, Indraprastha Institute of Information Technology, Okhla Industrial Estate, Phase III, (Near Govind Puri Metro Station), New Delhi, 110020, India.
| |
Collapse
|
10
|
Villageliu DN, Cunningham KC, Smith DR, Knoell DL, Mandolfo M, Wyatt TA, Samuelson DR. Natural killer cell effector function is critical for host defense against alcohol-associated bacterial pneumonia. NPJ Biofilms Microbiomes 2024; 10:79. [PMID: 39227647 PMCID: PMC11372167 DOI: 10.1038/s41522-024-00558-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Accepted: 08/25/2024] [Indexed: 09/05/2024] Open
Abstract
Alcohol use is an independent risk factor for the development of bacterial pneumonia due, in part, to impaired mucus-facilitated clearance, macrophage phagocytosis, and recruitment of neutrophils. Alcohol consumption is also known to reduce peripheral natural killer (NK) cell numbers and compromise NK cell cytolytic activity, especially NK cells with a mature phenotype. However, the role of innate lymphocytes, such as NK cells during host defense against alcohol-associated bacterial pneumonia is essentially unknown. We have previously shown that indole supplementation mitigates increases in pulmonary bacterial burden and improves pulmonary NK cell recruitment in alcohol-fed mice, which were dependent on aryl hydrocarbon receptor (AhR) signaling. Employing a binge-on-chronic alcohol-feeding model we sought to define the role and interaction of indole and NK cells during pulmonary host defense against alcohol-associated pneumonia. We demonstrate that alcohol dysregulates NK cell effector function and pulmonary recruitment via alterations in two key signaling pathways. We found that alcohol increases transforming growth factor beta (TGF-β) signaling while suppressing AhR signaling. We further demonstrated that NK cells isolated from alcohol-fed mice have a reduced ability to kill Klebsiella pneumoniae. NK cell migratory capacity to chemokines was also significantly altered by alcohol, as NK cells isolated from alcohol-fed mice exhibited preferential migration in response to CXCR3 chemokines but exhibited reduced migration in response to CCR2, CXCR4, and CX3CR1 chemokines. Together this data suggests that alcohol disrupts NK cell-specific TGF-β and AhR signaling pathways leading to decreased pulmonary recruitment and cytolytic activity thereby increasing susceptibility to alcohol-associated bacterial pneumonia.
Collapse
Affiliation(s)
- Daniel N Villageliu
- Department of Internal Medicine, Division of Pulmonary, Critical Care, & Sleep, University of Nebraska Medical Center, Omaha, NE, USA
| | - Kelly C Cunningham
- Department of Internal Medicine, Division of Pulmonary, Critical Care, & Sleep, University of Nebraska Medical Center, Omaha, NE, USA
| | - Deandra R Smith
- Department of Pharmacy Practice and Science, College of Pharmacy, University of Nebraska Medical Center, Omaha, NE, USA
| | - Daren L Knoell
- Department of Pharmacy Practice and Science, College of Pharmacy, University of Nebraska Medical Center, Omaha, NE, USA
| | - Mason Mandolfo
- Department of Internal Medicine, Division of Pulmonary, Critical Care, & Sleep, University of Nebraska Medical Center, Omaha, NE, USA
| | - Todd A Wyatt
- Department of Internal Medicine, Division of Pulmonary, Critical Care, & Sleep, University of Nebraska Medical Center, Omaha, NE, USA
- Department of Environmental, Agricultural and Occupational Health, College of Public Health, University of Nebraska Medical Center, Omaha, NE, USA
- Department of Veterans Affairs Nebraska, University of Nebraska Medical Center, Western Iowa Health Care System, Omaha, NE, USA
| | - Derrick R Samuelson
- Department of Internal Medicine, Division of Pulmonary, Critical Care, & Sleep, University of Nebraska Medical Center, Omaha, NE, USA.
- Nebraska Food for Health Center, University of Nebraska-Lincoln, Lincoln, NE, USA.
| |
Collapse
|
11
|
Mohammad Taheri M, Javan F, Poudineh M, Athari SS. Beyond CAR-T: The rise of CAR-NK cell therapy in asthma immunotherapy. J Transl Med 2024; 22:736. [PMID: 39103889 PMCID: PMC11302387 DOI: 10.1186/s12967-024-05534-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Accepted: 07/23/2024] [Indexed: 08/07/2024] Open
Abstract
Asthma poses a major public health burden. While existing asthma drugs manage symptoms for many, some patients remain resistant. The lack of a cure, especially for severe asthma, compels exploration of novel therapies. Cancer immunotherapy successes with CAR-T cells suggest its potential for asthma treatment. Researchers are exploring various approaches for allergic diseases including membrane-bound IgE, IL-5, PD-L2, and CTLA-4 for asthma, and Dectin-1 for fungal asthma. NK cells offer several advantages over T cells for CAR-based immunotherapy. They offer key benefits: (1) HLA compatibility, meaning they can be used in a wider range of patients without the need for matching tissue types. (2) Minimal side effects (CRS and GVHD) due to their limited persistence and cytokine profile. (3) Scalability for "off-the-shelf" production from various sources. Several strategies have been introduced that highlight the superiority and challenges of CAR-NK cell therapy for asthma treatment including IL-10, IFN-γ, ADCC, perforin-granzyme, FASL, KIR, NCRs (NKP46), DAP, DNAM-1, TGF-β, TNF-α, CCL, NKG2A, TF, and EGFR. Furthermore, we advocate for incorporating AI for CAR design optimization and CRISPR-Cas9 gene editing technology for precise gene manipulation to generate highly effective CAR constructs. This review will delve into the evolution and production of CAR designs, explore pre-clinical and clinical studies of CAR-based therapies in asthma, analyze strategies to optimize CAR-NK cell function, conduct a comparative analysis of CAR-T and CAR-NK cell therapy with their respective challenges, and finally present established novel CAR designs with promising potential for asthma treatment.
Collapse
Affiliation(s)
| | - Fatemeh Javan
- Student Research Committee, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Mohadeseh Poudineh
- Student Research Committee, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Seyed Shamseddin Athari
- Cancer Gene Therapy Research Center, Zanjan University of Medical Sciences, Zanjan, Iran.
- Department of Immunology, Zanjan School of Medicine, Zanjan University of Medical Sciences, 12th Street, Shahrake Karmandan, Zanjan, 45139-561111, Iran.
| |
Collapse
|
12
|
Bahramloo M, Shahabi SA, Kalarestaghi H, Rafat A, Mazloumi Z, Samimifar A, Asl KD. CAR-NK cell therapy in AML: Current treatment, challenges, and advantage. Biomed Pharmacother 2024; 177:117024. [PMID: 38941897 DOI: 10.1016/j.biopha.2024.117024] [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: 05/21/2024] [Accepted: 06/21/2024] [Indexed: 06/30/2024] Open
Abstract
Over the last decade, discovery of novel therapeutic method has been attention by the researchers and has changed the therapeutic perspective of hematological malignancies. Although NK cell play a pivotal role in the elimination of abnormal and cancerous cells, there are evidence that NK cell are disarm in hematological malignancy. Chimeric antigen receptor NK (CAR-NK) cell therapy, which includes the engineering of NK cells to detect tumor-specific antigens and, as a result, clear of cancerous cells, has created various clinical advantage for several human malignancies treatment. In the current review, we summarized NK cell dysfunction and CAR-NK cell based immunotherapy to treat AML patient.
Collapse
Affiliation(s)
- Mohammadmahdi Bahramloo
- Department of Medical Sciences, Student Research Committee, Islamic Azad University, Tabriz Branch, Tabriz, Iran
| | - Sina Alinejad Shahabi
- Department of Medical Sciences, Student Research Committee, Islamic Azad University, Tabriz Branch, Tabriz, Iran
| | - Hossein Kalarestaghi
- Research Laboratory for Embryology and Stem Cell, Department of Anatomical Sciences, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Ali Rafat
- Anatomical Sciences Research Center, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Zeinab Mazloumi
- Department of Applied Cell Sciences, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Arian Samimifar
- Department of Medical Sciences, Student Research Committee, Islamic Azad University, Tabriz Branch, Tabriz, Iran
| | - Khadijeh Dizaji Asl
- Department of Histopathology and Anatomy, Faculty of Medical Sciences, Tabriz Medical Sciences, Islamic Azad University, Tabriz, Iran.
| |
Collapse
|
13
|
Bakhtiyaridovvombaygi M, Yazdanparast S, Kheyrandish S, Safdari SM, Amiri Samani F, Sohani M, Jaafarian AS, Damirchiloo F, Izadpanah A, Parkhideh S, Mikanik F, Roshandel E, Hajifathali A, Gharehbaghian A. Harnessing natural killer cells for refractory/relapsed non-Hodgkin lymphoma: biological roles, clinical trials, and future prospective. Biomark Res 2024; 12:66. [PMID: 39020411 PMCID: PMC11253502 DOI: 10.1186/s40364-024-00610-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Accepted: 06/28/2024] [Indexed: 07/19/2024] Open
Abstract
Non-Hodgkin lymphomas (NHLs) are heterogeneous and are among the most common hematological malignancies worldwide. Despite the advances in the treatment of patients with NHLs, relapse or resistance to treatment is anticipated in several patients. Therefore, novel therapeutic approaches are needed. Recently, natural killer (NK) cell-based immunotherapy alone or in combination with monoclonal antibodies, chimeric antigen receptors, or bispecific killer engagers have been applied in many investigations for NHL treatment. The functional defects of NK cells and the ability of cancerous cells to escape NK cell-mediated cytotoxicity within the tumor microenvironment of NHLs, as well as the beneficial results from previous studies in the context of NK cell-based immunotherapy in NHLs, direct our attention to this therapeutic strategy. This review aims to summarize clinical studies focusing on the applications of NK cells in the immunotherapy of patients with NHL.
Collapse
Affiliation(s)
- Mehdi Bakhtiyaridovvombaygi
- Student Research Committee, Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Hematopoietic Stem Cell Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Somayeh Yazdanparast
- Department of Hematology and Blood Banking, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Setare Kheyrandish
- Student Research Committee, Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Seyed Mehrab Safdari
- Departments of Hematology and Blood Transfusion, School of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Fateme Amiri Samani
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Iranian Blood Transfusion Organization (IBTO), Tehran, Iran
| | - Mahsa Sohani
- Student Research Committee, Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Akram Sadat Jaafarian
- Departments of Hematology and Blood Transfusion, School of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Fateme Damirchiloo
- Departments of Hematology and Blood Transfusion, School of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Amirhossein Izadpanah
- Hematopoietic Stem Cell Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sahar Parkhideh
- Hematopoietic Stem Cell Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fatemeh Mikanik
- Laboratory Hematology and Blood Bank Department, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Elham Roshandel
- Hematopoietic Stem Cell Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Abbas Hajifathali
- Hematopoietic Stem Cell Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Ahmad Gharehbaghian
- Laboratory Hematology and Blood Bank Department, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
- Pediatric Congenital Hematologic Disorders Research Center, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
14
|
Wang D, Dou L, Sui L, Xue Y, Xu S. Natural killer cells in cancer immunotherapy. MedComm (Beijing) 2024; 5:e626. [PMID: 38882209 PMCID: PMC11179524 DOI: 10.1002/mco2.626] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 05/30/2024] [Accepted: 05/30/2024] [Indexed: 06/18/2024] Open
Abstract
Natural killer (NK) cells, as innate lymphocytes, possess cytotoxic capabilities and engage target cells through a repertoire of activating and inhibitory receptors. Particularly, natural killer group 2, member D (NKG2D) receptor on NK cells recognizes stress-induced ligands-the MHC class I chain-related molecules A and B (MICA/B) presented on tumor cells and is key to trigger the cytolytic response of NK cells. However, tumors have developed sophisticated strategies to evade NK cell surveillance, which lead to failure of tumor immunotherapy. In this paper, we summarized these immune escaping strategies, including the downregulation of ligands for activating receptors, upregulation of ligands for inhibitory receptors, secretion of immunosuppressive compounds, and the development of apoptosis resistance. Then, we focus on recent advancements in NK cell immune therapies, which include engaging activating NK cell receptors, upregulating NKG2D ligand MICA/B expression, blocking inhibitory NK cell receptors, adoptive NK cell therapy, chimeric antigen receptor (CAR)-engineered NK cells (CAR-NK), and NKG2D CAR-T cells, especially several vaccines targeting MICA/B. This review will inspire the research in NK cell biology in tumor and provide significant hope for improving cancer treatment outcomes by harnessing the potent cytotoxic activity of NK cells.
Collapse
Affiliation(s)
- DanRu Wang
- National Key Lab of Immunity and Inflammation and Institute of Immunology Naval Medical University Shanghai China
| | - LingYun Dou
- National Key Lab of Immunity and Inflammation and Institute of Immunology Naval Medical University Shanghai China
| | - LiHao Sui
- National Key Lab of Immunity and Inflammation and Institute of Immunology Naval Medical University Shanghai China
| | - Yiquan Xue
- National Key Lab of Immunity and Inflammation and Institute of Immunology Naval Medical University Shanghai China
| | - Sheng Xu
- National Key Lab of Immunity and Inflammation and Institute of Immunology Naval Medical University Shanghai China
- Shanghai Institute of Stem Cell Research and Clinical Translation Dongfang Hospital Shanghai China
| |
Collapse
|
15
|
Wang M, Wu H, Jiang W, Ren Y, Yuan X, Wang Y, Zhou J, Feng W, Wang Y, Xu T, Zhang D, Fang Y, He C, Li W. Differences in nature killer cell response and interference with mitochondrial DNA induced apoptosis in moxifloxacin environment. Int Immunopharmacol 2024; 132:111970. [PMID: 38608472 DOI: 10.1016/j.intimp.2024.111970] [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: 12/18/2023] [Revised: 03/22/2024] [Accepted: 03/27/2024] [Indexed: 04/14/2024]
Abstract
OBJECTIVES As antibiotics become more prevalent, accuracy and safety are critical. Moxifloxacin (MXF) have been reported to have immunomodulatory effects on a variety of immune cells and even anti-proliferative and pro-apoptotic effects, but the mechanism of action is not fully clear. METHODS Peripheral blood mononuclear cells (PBMC) from experimental groups of healthy adults (n = 3) were treated with MXF (10ug/ml) in vitro for 24 h. Single-cell sequencing was performed to investigate differences in the response of each immune cell to MXF. Flow cytometry determined differential gene expression in subsets of most damaged NK cells. Pseudo-time analysis identified drivers that influence MXF-stimulated cell differentiation. Detection of mitochondrial DNA and its involvement in the mitochondrial respiratory chain pathway clarifies the origin of MXF-induced stress injury. RESULTS Moxifloxacin-environmental NK cells are markedly reduced: a new subset of NK cells emerges, and immediate-early-response genes in this subset indicate the presence of an early activation response. The inhibitory receptor-dominant subset shows enhanced activation, leading to increased expression of cytokines and chemokines. The near-mature subset showed greater cytotoxicity and the most pronounced cellular damage. CD56bright cells responded by antagonizing the regulation of activation and inhibitory signals, demonstrating a strong cleavage capacity. The severe depletion of mitochondrial genes was focused on apoptosis induced by the mitochondrial respiratory chain complex. CONCLUSION NK cells exhibit heightened sensitivity to the MXF environment. Different NK subsets upregulate the expression of cytokines and chemokines through different activation pathways. Concurrently, MXF induces impairment of the mitochondrial oxidative phosphorylation system, culminating in apoptosis.
Collapse
Affiliation(s)
- Mengqing Wang
- Department of Emergency and Critical Care, Second Affiliated Hospital of Naval Medical University, Shanghai 200003, China
| | - Hao Wu
- Department of Emergency and Critical Care, Second Affiliated Hospital of Naval Medical University, Shanghai 200003, China
| | - Weiwei Jiang
- Department of Emergency and Critical Care, Second Affiliated Hospital of Naval Medical University, Shanghai 200003, China
| | - Yunfei Ren
- Department of Emergency and Critical Care, Second Affiliated Hospital of Naval Medical University, Shanghai 200003, China
| | - Xiaowei Yuan
- Department of Emergency and Critical Care, Second Affiliated Hospital of Naval Medical University, Shanghai 200003, China
| | - Yanan Wang
- Department of Emergency and Critical Care, Second Affiliated Hospital of Naval Medical University, Shanghai 200003, China
| | - Jian Zhou
- Department of Emergency and Critical Care, Second Affiliated Hospital of Naval Medical University, Shanghai 200003, China
| | - Wei Feng
- Department of Emergency and Critical Care, Second Affiliated Hospital of Naval Medical University, Shanghai 200003, China
| | - Yusen Wang
- Department of Emergency and Critical Care, Second Affiliated Hospital of Naval Medical University, Shanghai 200003, China
| | - Tianpeng Xu
- Department of Emergency and Critical Care, Second Affiliated Hospital of Naval Medical University, Shanghai 200003, China
| | - Danying Zhang
- Department of Emergency and Critical Care, Second Affiliated Hospital of Naval Medical University, Shanghai 200003, China
| | - Yunhao Fang
- Department of Emergency and Critical Care, Second Affiliated Hospital of Naval Medical University, Shanghai 200003, China
| | - Chao He
- Department of Emergency and Critical Care, Second Affiliated Hospital of Naval Medical University, Shanghai 200003, China
| | - Wenfang Li
- Department of Emergency and Critical Care, Second Affiliated Hospital of Naval Medical University, Shanghai 200003, China.
| |
Collapse
|
16
|
Reggiani F, Talarico G, Gobbi G, Sauta E, Torricelli F, Manicardi V, Zanetti E, Orecchioni S, Falvo P, Piana S, Lococo F, Paci M, Bertolini F, Ciarrocchi A, Sancisi V. BET inhibitors drive Natural Killer activation in non-small cell lung cancer via BRD4 and SMAD3. Nat Commun 2024; 15:2567. [PMID: 38519469 PMCID: PMC10960013 DOI: 10.1038/s41467-024-46778-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Accepted: 03/11/2024] [Indexed: 03/25/2024] Open
Abstract
Non-small-cell lung carcinoma (NSCLC) is the most common lung cancer and one of the pioneer tumors in which immunotherapy has radically changed patients' outcomes. However, several issues are emerging and their implementation is required to optimize immunotherapy-based protocols. In this work, we investigate the ability of the Bromodomain and Extra-Terminal protein inhibitors (BETi) to stimulate a proficient anti-tumor immune response toward NSCLC. By using in vitro, ex-vivo, and in vivo models, we demonstrate that these epigenetic drugs specifically enhance Natural Killer (NK) cell cytotoxicity. BETi down-regulate a large set of NK inhibitory receptors, including several immune checkpoints (ICs), that are direct targets of the transcriptional cooperation between the BET protein BRD4 and the transcription factor SMAD3. Overall, BETi orchestrate an epigenetic reprogramming that leads to increased recognition of tumor cells and the killing ability of NK cells. Our results unveil the opportunity to exploit and repurpose these drugs in combination with immunotherapy.
Collapse
Affiliation(s)
- Francesca Reggiani
- Translational Research Laboratory, Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia, Reggio Emilia, Italy.
| | - Giovanna Talarico
- Laboratory of Hematology-Oncology, European Institute of Oncology IRCCS, Milan, Italy
- Onco-Tech Lab, European Institute of Oncology IRCCS and Politecnico di Milano, Milan, Italy
| | - Giulia Gobbi
- Translational Research Laboratory, Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | - Elisabetta Sauta
- Translational Research Laboratory, Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia, Reggio Emilia, Italy
- Humanitas Clinical and Research Center, IRCCS, Rozzano, Milan, Italy
| | - Federica Torricelli
- Translational Research Laboratory, Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | - Veronica Manicardi
- Translational Research Laboratory, Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia, Reggio Emilia, Italy
- Clinical and Experimental Medicine PhD Program, University of Modena and Reggio Emilia, Modena, Italy
| | - Eleonora Zanetti
- Pathology Unit, Azienda USL-IRCCS di Reggio Emilia, Reggio Emilia, Italy
- Biobank, Azienda USL-IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | - Stefania Orecchioni
- Laboratory of Hematology-Oncology, European Institute of Oncology IRCCS, Milan, Italy
- Onco-Tech Lab, European Institute of Oncology IRCCS and Politecnico di Milano, Milan, Italy
| | - Paolo Falvo
- Laboratory of Hematology-Oncology, European Institute of Oncology IRCCS, Milan, Italy
- Onco-Tech Lab, European Institute of Oncology IRCCS and Politecnico di Milano, Milan, Italy
| | - Simonetta Piana
- Pathology Unit, Azienda USL-IRCCS di Reggio Emilia, Reggio Emilia, Italy
- Biobank, Azienda USL-IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | - Filippo Lococo
- Università Cattolica del Sacro Cuore, Rome, Italy
- Department of General Thoracic Surgery, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Massimiliano Paci
- Thoracic Surgery Unit, Azienda USL-IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | - Francesco Bertolini
- Laboratory of Hematology-Oncology, European Institute of Oncology IRCCS, Milan, Italy
- Onco-Tech Lab, European Institute of Oncology IRCCS and Politecnico di Milano, Milan, Italy
| | - Alessia Ciarrocchi
- Translational Research Laboratory, Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | - Valentina Sancisi
- Translational Research Laboratory, Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia, Reggio Emilia, Italy.
| |
Collapse
|
17
|
He J, Yan Y, Zhang J, Wei Z, Li H, Xing L. Synergistic treatment strategy: combining CAR-NK cell therapy and radiotherapy to combat solid tumors. Front Immunol 2023; 14:1298683. [PMID: 38162672 PMCID: PMC10755030 DOI: 10.3389/fimmu.2023.1298683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Accepted: 11/20/2023] [Indexed: 01/03/2024] Open
Abstract
Immunotherapy, notably chimeric antigen receptor (CAR) modified natural killer (NK) cell therapy, has shown exciting promise in the treatment of hematologic malignancies due to its unique advantages including fewer side effects, diverse activation mechanisms, and wide availability. However, CAR-NK cell therapies have demonstrated limited efficacy against solid tumors, primarily due to challenges posed by the solid tumor microenvironment. In contrast, radiotherapy, a well-established treatment modality, has been proven to modulate the tumor microenvironment and facilitate immune cell infiltration. With these observations, we hypothesize that a novel therapeutic strategy integrating CAR-NK cell therapy with radiotherapy could enhance the ability to treat solid tumors. This hypothesis aims to address the obstacles CAR-NK cell therapies face within the solid tumor microenvironment and explore the potential efficacy of their combination with radiotherapy. By capitalizing on the synergistic advantages of CAR-NK cell therapy and radiotherapy, we posit that this could lead to improved prognoses for patients with solid tumors.
Collapse
Affiliation(s)
- Jie He
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Yushan Yan
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Jun Zhang
- Asclepius (Soochow) Technology Company Group, Suzhou, Jiangsu, China
| | - Zhiming Wei
- Asclepius (Soochow) Technology Company Group, Suzhou, Jiangsu, China
| | - Huashun Li
- Asclepius (Soochow) Technology Company Group, Suzhou, Jiangsu, China
| | - Ligang Xing
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| |
Collapse
|
18
|
Qiu J, Zhao L, Cheng Y, Chen Q, Xu Y, Lu Y, Gao J, Lei W, Yan C, Ling Z, Wu S. Exploring the gut mycobiome: differential composition and clinical associations in hypertension, chronic kidney disease, and their comorbidity. Front Immunol 2023; 14:1317809. [PMID: 38162661 PMCID: PMC10755858 DOI: 10.3389/fimmu.2023.1317809] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Accepted: 11/30/2023] [Indexed: 01/03/2024] Open
Abstract
Background Hypertension (HTN) and chronic kidney disease (CKD) pose significant global health challenges and often coexist, amplifying cardiovascular risks. Recent attention has turned to the gut mycobiome as a potential factor in their pathophysiology. Our study sought to examine the gut fungal profile in individuals with HTN, CKD, and the concurrent HTN+CKD condition, investigating its connections with serum cytokines, renal function, and blood pressure. Methods and materials We investigated three distinct participant groups: a cohort of 50 healthy controls (HC), 50 individuals diagnosed with HTN-only, and 50 participants suffering from both HTN and CKD (HTN+CKD). To facilitate our research, we gathered fecal and blood samples and conducted a comprehensive analysis of serum cytokines. Moreover, fungal DNA extraction was conducted with meticulous care, followed by sequencing of the Internal Transcribed Spacer (ITS) region. Results HTN+CKD patients displayed distinctive fungal composition with increased richness and diversity compared to controls. In contrast, HTN-only patients exhibited minimal fungal differences. Specific fungal genera were notably altered in HTN+CKD patients, characterized by increased Apiotrichum and Saccharomyces levels and reduced Candida abundance. Our correlation analyses revealed significant associations between fungal genera and serum cytokines. Moreover, certain fungal taxa, such as Apiotrichum and Saccharomyces, exhibited positive correlations with renal function, while others, including Septoria, Nakaseomyces, and Saccharomyces, were linked to blood pressure, particularly diastolic pressure. Conclusion Gut mycobiome dysbiosis in individuals with comorbid HTN and CKD differs significantly from that observed in HTN-only and healthy controls. The interactions between serum cytokines, renal function, and blood pressure emphasize the potential impact of the fungal microbiome on these conditions. Additional research is required to clarify the underlying mechanisms and identify therapeutic opportunities associated with mycobiome dysbiosis in HTN and CKD.
Collapse
Affiliation(s)
- Juan Qiu
- Prenatal Diagnosis Center, Longhua Maternity and Child Healthcare Hospital, Shenzhen, Guangdong, China
| | - Longyou Zhao
- Department of Laboratory Medicine, Lishui Second People’s Hospital, Lishui, Zhejiang, China
| | - Yiwen Cheng
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, Shandong, China
| | - Qiaoxia Chen
- Department of Laboratory Medicine, Lishui Second People’s Hospital, Lishui, Zhejiang, China
| | - Yiran Xu
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Yingfeng Lu
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Jie Gao
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Wenhui Lei
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, Shandong, China
- Department of Laboratory Medicine, Shandong First Medical University, Jinan, Shandong, China
| | - Chengmin Yan
- Department of Intensive Unit, Hangzhou Jiaye Rehabilitation Hospital, Hangzhou, Zhejing, China
| | - Zongxin Ling
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, Shandong, China
| | - Shaochang Wu
- Department of Laboratory Medicine, Lishui Second People’s Hospital, Lishui, Zhejiang, China
| |
Collapse
|
19
|
Bakhtiyaridovvombaygi M, Yazdanparast S, Mikanik F, Izadpanah A, Parkhideh S, Shahbaz Ghasabeh A, Roshandel E, Hajifathali A, Gharehbaghian A. Cytokine-Induced Memory-Like NK Cells: Emerging strategy for AML immunotherapy. Biomed Pharmacother 2023; 168:115718. [PMID: 37857247 DOI: 10.1016/j.biopha.2023.115718] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 10/10/2023] [Accepted: 10/11/2023] [Indexed: 10/21/2023] Open
Abstract
Acute myeloid leukemia (AML) is a heterogeneous disease developed from the malignant expansion of myeloid precursor cells in the bone marrow and peripheral blood. The implementation of intensive chemotherapy and hematopoietic stem cell transplantation (HSCT) has improved outcomes associated with AML, but relapse, along with suboptimal outcomes, is still a common scenario. In the past few years, exploring new therapeutic strategies to optimize treatment outcomes has occurred rapidly. In this regard, natural killer (NK) cell-based immunotherapy has attracted clinical interest due to its critical role in immunosurveillance and their capabilities to target AML blasts. NK cells are cytotoxic innate lymphoid cells that mediate anti-viral and anti-tumor responses by producing pro-inflammatory cytokines and directly inducing cytotoxicity. Although NK cells are well known as short-lived innate immune cells with non-specific responses that have limited their clinical applications, the discovery of cytokine-induced memory-like (CIML) NK cells could overcome these challenges. NK cells pre-activated with the cytokine combination IL-12/15/18 achieved a long-term life span with adaptive immunity characteristics, termed CIML-NK cells. Previous studies documented that using CIML-NK cells in cancer treatment is safe and results in promising outcomes. This review highlights the current application, challenges, and opportunities of CIML-NK cell-based therapy in AML.
Collapse
Affiliation(s)
- Mehdi Bakhtiyaridovvombaygi
- Student Research Committee, Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Hematopoietic Stem Cell Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Somayeh Yazdanparast
- Student Research Committee, Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fatemeh Mikanik
- Student Research Committee, Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Hematopoietic Stem Cell Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Amirhossein Izadpanah
- Hematopoietic Stem Cell Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sahar Parkhideh
- Hematopoietic Stem Cell Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Amin Shahbaz Ghasabeh
- Department of Hematology and Blood Bank, School of Allied Medical Science, Shahid Beheshti University of Medical Science, Tehran, Iran
| | - Elham Roshandel
- Hematopoietic Stem Cell Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Abbas Hajifathali
- Hematopoietic Stem Cell Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Ahmad Gharehbaghian
- Department of Hematology and Blood Bank, School of Allied Medical Science, Shahid Beheshti University of Medical Science, Tehran, Iran; Pediatric Congenital Hematologic Disorders Research Center, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
20
|
Chen H, Wang Q, Li J, Li Y, Chen A, Zhou J, Zhao J, Mao Z, Zhou Z, Zhang J, Wang Y, Wang R, Li Q, Zhang Y, Jiang R, Miao D, Jin J. IFNγ Transcribed by IRF1 in CD4+ Effector Memory T Cells Promotes Senescence-Associated Pulmonary Fibrosis. Aging Dis 2023; 14:2215-2237. [PMID: 37199578 PMCID: PMC10676796 DOI: 10.14336/ad.2023.0320] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Accepted: 03/20/2023] [Indexed: 05/19/2023] Open
Abstract
Physiologically aged lungs are prone to senescence-associated pulmonary diseases (SAPD). This study aimed to determine the mechanism and subtype of aged T cells affecting alveolar type II epithelial (AT2) cells, which promote the pathogenesis of senescence-associated pulmonary fibrosis (SAPF). Cell proportions, the relationship between SAPD and T cells, and the aging- and senescence-associated secretory phenotype (SASP) of T cells between young and aged mice were analyzed using lung single-cell transcriptomics. SAPD was monitored by markers of AT2 cells and found to be induced by T cells. Furthermore, IFNγ signaling pathways were activated and cell senescence, SASP, and T cell activation were shown in aged lungs. Physiological aging led to pulmonary dysfunction and TGF-β1/IL-11/MEK/ERK (TIME) signaling-mediated SAPF, which was induced by senescence and SASP of aged T cells. Especially, IFNγ was produced by the accumulated CD4+ effector memory T (TEM) cells in the aged lung. This study also found that physiological aging increased pulmonary CD4+ TEM cells, IFNγ was produced mainly by CD4+ TEM cells, and pulmonary cells had increased responsiveness to IFNγ signaling. Specific regulon activity was increased in T cell subclusters. IFNγ transcriptionally regulated by IRF1 in CD4+ TEM cells promoted the epithelial-to-mesenchymal transition by activating TIME signaling and cell senescence of AT2 cells with aging. Accumulated IRF1+CD4+ TEM produced IFNγ in lung with aging and anti-IRF1 primary antibody treatment inhibited the expression of IFNγ. Aging might drive T cell differentiation toward helper T cells with developmental trajectories and enhance cell interactions of pulmonary T cells with other surrounding cells. Thus, IFNγ transcribed by IRF1 in CD4+ effector memory T cells promotes SAPF. IFNγ produced by CD4+ TEM cells in physiologically aged lungs could be a therapeutic target for preventing SAPF.
Collapse
Affiliation(s)
- Haiyun Chen
- Department of Human Anatomy, Research Centre for Bone and Stem Cells
- Key Laboratory for Aging & Disease;
- Nanjing Medical University, Nanjing, Jiangsu, China. Medical School of Nanjing University, Jiangsu Laboratory of Molecular Medicine, Nanjing University, Nanjing, Jiangsu, China. Department of Orthopaedics, Xuzhou Central Hospital
| | - Qiuyi Wang
- Department of Human Anatomy, Research Centre for Bone and Stem Cells
| | - Jie Li
- The State Key Laboratory of Reproductive Medicine
| | - Yuan Li
- The Xuzhou Clinical School of Xuzhou Medical University
| | - Ao Chen
- Department of Human Anatomy, Research Centre for Bone and Stem Cells
| | - Jiawen Zhou
- Department of Human Anatomy, Research Centre for Bone and Stem Cells
| | - Jingyu Zhao
- Department of Human Anatomy, Research Centre for Bone and Stem Cells
| | - Zhiyuan Mao
- Department of Human Anatomy, Research Centre for Bone and Stem Cells
| | - Zihao Zhou
- Department of Human Anatomy, Research Centre for Bone and Stem Cells
| | - Jin’ge Zhang
- Department of Human Anatomy, Research Centre for Bone and Stem Cells
| | - Yue Wang
- Department of Human Anatomy, Research Centre for Bone and Stem Cells
| | - Rong Wang
- Department of Human Anatomy, Research Centre for Bone and Stem Cells
| | - Qing Li
- The Xuzhou School of Clinical Medicine of Nanjing Medical University, Xuzhou, Jiangsu, China. The Research Center for Aging, Affiliated Friendship Plastic Surgery Hospital of Nanjing Medical University, Nanjing, Jiangsu, China. Department of cardiology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, Jiangsu, China. Department of Science and Technology, Jiangsu Jiankang Vocational College, Nanjing, China.
| | - Yongjie Zhang
- Department of Human Anatomy, Research Centre for Bone and Stem Cells
| | | | - Dengshun Miao
- Department of Human Anatomy, Research Centre for Bone and Stem Cells
- Nanjing Medical University, Nanjing, Jiangsu, China. Medical School of Nanjing University, Jiangsu Laboratory of Molecular Medicine, Nanjing University, Nanjing, Jiangsu, China. Department of Orthopaedics, Xuzhou Central Hospital
| | - Jianliang Jin
- Department of Human Anatomy, Research Centre for Bone and Stem Cells
| |
Collapse
|
21
|
Hegewisch-Solloa E, Nalin AP, Freud AG, Mace EM. Deciphering the localization and trajectory of human natural killer cell development. J Leukoc Biol 2023; 114:487-506. [PMID: 36869821 DOI: 10.1093/jleuko/qiad027] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 02/07/2023] [Accepted: 02/12/2023] [Indexed: 03/05/2023] Open
Abstract
Innate immune cells represent the first line of cellular immunity, comprised of both circulating and tissue-resident natural killer cells and innate lymphoid cells. These innate lymphocytes arise from a common CD34+ progenitor that differentiates into mature natural killer cells and innate lymphoid cells. The successive stages in natural killer cell maturation are characterized by increased lineage restriction and changes to phenotype and function. Mechanisms of human natural killer cell development have not been fully elucidated, especially the role of signals that drive the spatial localization and maturation of natural killer cells. Cytokines, extracellular matrix components, and chemokines provide maturation signals and influence the trafficking of natural killer cell progenitors to peripheral sites of differentiation. Here we present the latest advances in our understanding of natural killer and innate lymphoid cell development in peripheral sites, including secondary lymphoid tissues (i.e. tonsil). Recent work in the field has provided a model for the spatial distribution of natural killer cell and innate lymphoid cell developmental intermediates in tissue and generated further insights into the developmental niche. In support of this model, future studies using multifaceted approaches seek to fully map the developmental trajectory of human natural killer cells and innate lymphoid cells in secondary lymphoid tissues.
Collapse
Affiliation(s)
- Everardo Hegewisch-Solloa
- Department of Pediatrics, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, 630 W 168th St. New York, NY 10032, USA
| | - Ansel P Nalin
- Biomedical Sciences Graduate Program, Medical Scientist Training Program, Comprehensive Cancer Center and The James Cancer Hospital and Solove Research Institute, The Ohio State University, 460 W 10th Ave. Columbus, OH 43210, USA
| | - Aharon G Freud
- Department of Pathology, Comprehensive Cancer Center and The James Cancer Hospital and Solove Research Institute, The Ohio State University, 460 W 12th Ave. Columbus, OH 43210, USA
| | - Emily M Mace
- Department of Pediatrics, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, 630 W 168th St. New York, NY 10032, USA
| |
Collapse
|
22
|
Samuelson D, Villageliu D, Cunningham K, Smith D, Knoell D, Mandolfo M, Wyatt T. Regulation of Natural Killer Cell TGF-β and AhR Signaling Pathways Via the Intestinal Microbiota is Critical for Host Defense Against Alcohol-Associated Bacterial Pneumonia. RESEARCH SQUARE 2023:rs.3.rs-3328953. [PMID: 37886455 PMCID: PMC10602187 DOI: 10.21203/rs.3.rs-3328953/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/28/2023]
Abstract
Alcohol use is an independent risk factor for the development of bacterial pneumonia due, in part, to impaired mucus-facilitated clearance, macrophage phagocytosis, and recruitment of neutrophils. Alcohol consumption is also known to reduce peripheral natural killer (NK) cell numbers and compromises NK cell cytolytic activity, especially NK cells with a mature phenotype. However, the role of innate lymphocytes, such as NK cells during host defense against alcohol-associated bacterial pneumonia is essentially unknown. We have previously shown that indole supplementation mitigates increases in pulmonary bacterial burden and improves pulmonary NK cell recruitment in alcohol-fed mice, which were dependent of aryl hydrocarbon receptor (AhR) signaling. Employing a binge-on-chronic alcohol-feeding model we sought to define the role and interaction of indole and NK cells during pulmonary host defense against alcohol-associated pneumonia. We demonstrate that alcohol dysregulates NK cell effector function and pulmonary recruitment via alterations in two key signaling pathways. We found that alcohol increases transforming growth factor beta (TGF-β) signaling, while suppressing AhR signaling. We further demonstrated that NK cells isolated from alcohol-fed mice have a reduced ability to kill Klebsiella pneumoniae. NK cell migratory capacity to chemokines was also significantly altered by alcohol, as NK cells isolated from alcohol-fed mice exhibited preferential migration in response to CXCR3 chemokines but exhibited reduced migration in response to CCR2, CXCR4, and CX3CR1 chemokines. Together this data suggests that alcohol disrupts NK cell specific TGF-β and AhR signaling pathways leading to decreased pulmonary recruitment and cytolytic activity thereby increasing susceptibility to alcohol-associated bacterial pneumonia.
Collapse
|
23
|
Ma S, Caligiuri MA, Yu J. Harnessing Natural Killer Cells for Lung Cancer Therapy. Cancer Res 2023; 83:3327-3339. [PMID: 37531223 DOI: 10.1158/0008-5472.can-23-1097] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 06/13/2023] [Accepted: 07/31/2023] [Indexed: 08/04/2023]
Abstract
Lung cancer is the leading cause of cancer-related death worldwide. Although natural killer (NK) cells are garnering interest as a potential anticancer therapy because they selectively recognize and eliminate cancer cells, their use in treating solid tumors, including lung cancer, has been limited due to impediments to their efficacy, such as their limited ability to reach tumor tissues, the reduced antitumor activity of tumor-infiltrating NK cells, and the suppressive tumor microenvironment (TME). This comprehensive review provides an in-depth analysis of the cross-talk between the lung cancer TME and NK cells. We highlight the various mechanisms used by the TME to modulate NK-cell phenotypes and limit infiltration, explore the role of the TME in limiting the antitumor activity of NK cells, and discuss the current challenges and obstacles that hinder the success of NK-cell-based immunotherapy for lung cancer. Potential opportunities and promising strategies to address these challenges have been implemented or are being developed to optimize NK-cell-based immunotherapy for lung cancer. Through critical evaluation of existing literature and emerging trends, this review provides a comprehensive outlook on the future of NK-cell-based immunotherapy for treating lung cancer.
Collapse
Affiliation(s)
- Shoubao Ma
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Los Angeles, California
- Hematologic Malignancies and Stem Cell Transplantation Institute, City of Hope National Medical Center, Los Angeles, California
| | - Michael A Caligiuri
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Los Angeles, California
- Hematologic Malignancies and Stem Cell Transplantation Institute, City of Hope National Medical Center, Los Angeles, California
- Comprehensive Cancer Center, City of Hope, Los Angeles, California
| | - Jianhua Yu
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Los Angeles, California
- Hematologic Malignancies and Stem Cell Transplantation Institute, City of Hope National Medical Center, Los Angeles, California
- Comprehensive Cancer Center, City of Hope, Los Angeles, California
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope, Los Angeles, California
| |
Collapse
|
24
|
Petillo S, Sproviero E, Loconte L, Cuollo L, Zingoni A, Molfetta R, Fionda C, Soriani A, Cerboni C, Petrucci MT, Fazio F, Paolini R, Santoni A, Cippitelli M. NEDD8-activating enzyme inhibition potentiates the anti-myeloma activity of natural killer cells. Cell Death Dis 2023; 14:438. [PMID: 37460534 DOI: 10.1038/s41419-023-05949-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 04/26/2023] [Accepted: 06/09/2023] [Indexed: 07/20/2023]
Abstract
Natural Killer (NK) cells act as important regulators in the development and progression of hematological malignancies and their suppressor activity against Multiple Myeloma (MM) cells has been confirmed in many studies. Significant changes in the distribution of NK cell subsets and dysfunctions of NK cell effector activities were described in MM patients and correlated with disease staging. Thus, restoring or enhancing the functionality of these effectors for the treatment of MM represents a critical need. Neddylation is a post-translational modification that adds a ubiquitin-like molecule, NEDD8, to the substrate protein. One of the outcomes is the activation of the Cullin Ring Ligases (CRLs), a class of ubiquitin-ligases that controls the degradation of about 20% of proteasome-regulated proteins. Overactivation of CRLs has been described in cancer and can lead to tumor growth and progression. Thus, targeting neddylation represents an attractive approach for cancer treatment. Our group has recently described how pharmacologic inhibition of neddylation increases the expression of the NKG2D activating receptor ligands, MICA and MICB, in MM cells, making these cells more susceptible to NK cell degranulation and killing. Here, we extended our investigation to the direct role of neddylation on NK cell effector functions exerted against MM. We observed that inhibition of neddylation enhanced NK cell-mediated degranulation and killing against MM cells and improved Daratumumab/Elotuzumab-mediated response. Mechanistically, inhibition of neddylation increased the expression of Rac1 and RhoA GTPases in NK cells, critical mediators for an efficient degranulation at the immunological synapse of cytotoxic lymphocytes, and augmented the levels of F-actin and perforin polarization in NK cells contacting target cells. Moreover, inhibition of neddylation partially abrogated TGFβ-mediated repression of NK cell effector activity. This study describes the role of neddylation on NK cell effector functions and highlights the positive immunomodulatory effects achieved by the inhibition of this pathway in MM.
Collapse
Affiliation(s)
- Sara Petillo
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Elena Sproviero
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Luisa Loconte
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Lorenzo Cuollo
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Alessandra Zingoni
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Rosa Molfetta
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Cinzia Fionda
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Alessandra Soriani
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Cristina Cerboni
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Maria Teresa Petrucci
- Hematology, Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Francesca Fazio
- Hematology, Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Rossella Paolini
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Angela Santoni
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
- Istituto Pasteur-Fondazione Cenci Bolognetti, Rome, Italy
- IRCCS, Neuromed, Pozzilli, Italy
| | - Marco Cippitelli
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy.
| |
Collapse
|
25
|
Ruf B, Greten TF, Korangy F. Innate lymphoid cells and innate-like T cells in cancer - at the crossroads of innate and adaptive immunity. Nat Rev Cancer 2023; 23:351-371. [PMID: 37081117 DOI: 10.1038/s41568-023-00562-w] [Citation(s) in RCA: 41] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/13/2023] [Indexed: 04/22/2023]
Abstract
Immunotherapies targeting conventional T cells have revolutionized systemic treatment for many cancers, yet only a subset of patients benefit from these approaches. A better understanding of the complex immune microenvironment of tumours is needed to design the next generation of immunotherapeutics. Innate lymphoid cells (ILCs) and innate-like T cells (ILTCs) are abundant, tissue-resident lymphocytes that have recently been shown to have critical roles in many types of cancers. ILCs and ILTCs rapidly respond to changes in their surrounding environment and act as the first responders to bridge innate and adaptive immunity. This places ILCs and ILTCs as pivotal orchestrators of the final antitumour immune response. In this Review, we outline hallmarks of ILCs and ILTCs and discuss their emerging role in antitumour immunity, as well as the pathophysiological adaptations leading to their pro-tumorigenic function. We explore the pleiotropic, in parts redundant and sometimes opposing, mechanisms that underlie the delicate interplay between the different subsets of ILCs and ILTCs. Finally, we highlight their role in amplifying and complementing conventional T cell functions and summarize immunotherapeutic strategies for targeting ILCs and ILTCs in cancer.
Collapse
Affiliation(s)
- Benjamin Ruf
- Gastrointestinal Malignancy Section, Thoracic and Gastrointestinal Malignancies Branch, Centre for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Tim F Greten
- Gastrointestinal Malignancy Section, Thoracic and Gastrointestinal Malignancies Branch, Centre for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
- NCI CCR Liver Cancer Program, National Institutes of Health, Bethesda, MD, USA
| | - Firouzeh Korangy
- Gastrointestinal Malignancy Section, Thoracic and Gastrointestinal Malignancies Branch, Centre for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
26
|
Ma S, Han J, Li Z, Xiao S, Zhang J, Yan J, Tang T, Barr T, Kraft AS, Caligiuri MA, Yu J. An XBP1s-PIM-2 positive feedback loop controls IL-15-mediated survival of natural killer cells. Sci Immunol 2023; 8:eabn7993. [PMID: 36897958 DOI: 10.1126/sciimmunol.abn7993] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/12/2023]
Abstract
Spliced X-box-binding protein 1 (XBP1s) is an essential transcription factor downstream of interleukin-15 (IL-15) and AKT signaling, which controls cell survival and effector functions of human natural killer (NK) cells. However, the precise mechanisms, especially the downstream targets of XBP1s, remain unknown. In this study, by using XBP1 conditional knockout mice, we found that XBP1s is critical for IL-15-mediated NK cell survival but not proliferation in vitro and in vivo. Mechanistically, XBP1s regulates homeostatic NK cell survival by targeting PIM-2, a critical anti-apoptotic gene, which in turn stabilizes XBP1s protein by phosphorylating it at Thr58. In addition, XBP1s enhances the effector functions and antitumor immunity of NK cells by recruiting T-bet to the promoter region of Ifng. Collectively, our findings identify a previously unknown mechanism by which IL-15-XBP1s signaling regulates the survival and effector functions of NK cells.
Collapse
Affiliation(s)
- Shoubao Ma
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Los Angeles, CA 91010, USA
- Hematologic Malignancies Research Institute, City of Hope National Medical Center, Los Angeles, CA 91010, USA
| | - Jingjing Han
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Los Angeles, CA 91010, USA
- Hematologic Malignancies Research Institute, City of Hope National Medical Center, Los Angeles, CA 91010, USA
| | - Zhenlong Li
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Los Angeles, CA 91010, USA
- Hematologic Malignancies Research Institute, City of Hope National Medical Center, Los Angeles, CA 91010, USA
| | - Sai Xiao
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Los Angeles, CA 91010, USA
- Hematologic Malignancies Research Institute, City of Hope National Medical Center, Los Angeles, CA 91010, USA
| | - Jianying Zhang
- Department of Computational and Quantitative Medicine, City of Hope National Medical Center, Los Angeles, CA 91010, USA
| | - Jiazhuo Yan
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Los Angeles, CA 91010, USA
| | - Tingting Tang
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Los Angeles, CA 91010, USA
| | - Tasha Barr
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Los Angeles, CA 91010, USA
| | - Andrew S Kraft
- University of Arizona Cancer Center, University of Arizona, Tucson, AZ 85724, USA
| | - Michael A Caligiuri
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Los Angeles, CA 91010, USA
- Hematologic Malignancies Research Institute, City of Hope National Medical Center, Los Angeles, CA 91010, USA
- Comprehensive Cancer Center, City of Hope, Los Angeles, CA 91010, USA
| | - Jianhua Yu
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Los Angeles, CA 91010, USA
- Hematologic Malignancies Research Institute, City of Hope National Medical Center, Los Angeles, CA 91010, USA
- Comprehensive Cancer Center, City of Hope, Los Angeles, CA 91010, USA
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope, Los Angeles, CA 91010, USA
| |
Collapse
|
27
|
Arianfar E, Khandoozi SR, Mohammadi S, Memarian A. Suppression of CD56 bright NK cells in breast cancer patients is associated with the PD-1 and TGF-βRII expression. Clin Transl Oncol 2023; 25:841-851. [PMID: 36414921 DOI: 10.1007/s12094-022-02997-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Accepted: 10/26/2022] [Indexed: 11/24/2022]
Abstract
BACKGROUND Natural killer (NK) cells, as professional cytotoxic cells, play a key role against cancer in the early and metastatic stages. Their functional defects are highly associated with the initiation or progression of breast cancer (BC). Here, we investigated the phenotypic characterization of NK cells in 26 newly diagnosed BC patients in comparison to 12 healthy counterparts. METHODS Expression of CXCR3 and PD-1, and also NKG2D, and TGF-βRII were studied on CD56dim and CD56bright NK cells from fresh peripheral blood (PB) samples using flow cytometry. The plasma levels of IFN-γ and soluble MIC-A levels were also assessed by ELISA. RESULTS Both CD56dim and CD56bright NK subtypes showed lower CXCR3 and NKG2D expression in BC patients than healthy subjects. Furthermore, patients' CD56bright NK cells significantly showed higher expression levels of TGF-βRII and PD-1. Interestingly, increased concentration of MIC-A level in plasma of BC patients was associated with the higher TGF-βRII and PD-1 expression in all NK cells, while the plasma level of IFN-γ was associated with the lower TGF-βRII expression on CD56bright NK cells in these patients. CONCLUSION Our results demonstrated phenotypically suppressed-NK cells, especially in the CD56bright subset of BC patients. It specifies their potential incompetence and outlines decrement of their anti-tumor activity, which could be interrelated with the tumor pathogenesis, TME immunosuppression, and so disease progression. The induction of compensatory mechanisms revives NK cells function and could be used in combination with the conventional treatments as a putative therapeutic approach for targeted immunotherapy.
Collapse
Affiliation(s)
- Elaheh Arianfar
- Student Research Committee, Golestan University of Medical Sciences, Gorgan, Iran.,Department of Immunology, Faculty of Medicine, Golestan University of Medical Sciences, Gorgan, Iran
| | | | - Saeed Mohammadi
- Stem Cell Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Ali Memarian
- Department of Immunology, Faculty of Medicine, Golestan University of Medical Sciences, Gorgan, Iran. .,Golestan Research Center of Gastroenterology and Hepatology, Golestan University of Medical Sciences, Gorgan, Iran.
| |
Collapse
|
28
|
Youlin K, Simin L, Jian K, Li Z. Inhibition of miR-20a by pterostilbene facilitates prostate cancer cells killed by NK cells via up-regulation of NKG2D ligands and TGF-β1down-regulation. Heliyon 2023; 9:e14957. [PMID: 37064475 PMCID: PMC10102449 DOI: 10.1016/j.heliyon.2023.e14957] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2022] [Revised: 03/19/2023] [Accepted: 03/22/2023] [Indexed: 03/31/2023] Open
Abstract
Natural killer (NK) cells play a potent role in antitumor immunity via spontaneously eliminating tumor directly. However, some tumors such as prostate cancer constantly escape this immune response by down-regulating cell surface molecule recognition and/or secreting immune impressive cytokines. Here, we found pterostilbene, a natural agent with potent anticancer activity, could enhance expression of major histocompatibility complex class I chain-related proteins A and B (MICA/B) on prostate cancer cells surface, which are ligands of the natural killer group 2 member D (NKG2D) expressed by NK cells, and inhibit TGF-β1 secretion by prostate cancer cells. Further, we discovered that these effects were caused by inhibition of miR-20a in prostate cancer cells by pterostilbene. MiR-20a could target the 3' untranslated region (UTR) of MICA/B, resulting in their expression down-regulation. Inhibition of TGF-β1 function by its specific antibody attenuated its impairment to NKG2D on NK cells. Finally, we observed that pterostilbene-treated prostate cancer cells were more easily to be killed by NK cells. Taken together, our findings demonstrated inhibition of miR-20a by pterostilbene in prostate cancer cells could increase MICA/B expression and decrease TGF-β1 secretion, which enhanced NK cell-mediated cytotoxicity againt prostate cancer cells, suggesting a potential approach for increasing anti-prostate cancer immune.
Collapse
Affiliation(s)
- Kuang Youlin
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Liang Simin
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Kang Jian
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Zhang Li
- Department of Gastroenterology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
- Corresponding author.
| |
Collapse
|
29
|
Ma S, Barr T, Yu J. Recent Advances of RNA m 6A Modifications in Cancer Immunoediting and Immunotherapy. Cancer Treat Res 2023; 190:49-94. [PMID: 38112999 DOI: 10.1007/978-3-031-45654-1_3] [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] [Indexed: 12/21/2023]
Abstract
Cancer immunotherapy, which modulates immune responses against tumors using immune-checkpoint inhibitors or adoptive cell transfer, has emerged as a novel and promising therapy for tumors. However, only a minority of patients demonstrate durable responses, while the majority of patients are resistant to immunotherapy. The immune system can paradoxically constrain and promote tumor development and progression. This process is referred to as cancer immunoediting. The mechanisms of resistance to immunotherapy seem to be that cancer cells undergo immunoediting to evade recognition and elimination by the immune system. RNA modifications, specifically N6-methyladenosine (m6A) methylation, have emerged as a key regulator of various post-transcriptional gene regulatory processes, such as RNA export, splicing, stability, and degradation, which play unappreciated roles in various physiological and pathological processes, including immune system development and cancer pathogenesis. Therefore, a deeper understanding of the mechanisms by which RNA modifications impact the cancer immunoediting process can provide insight into the mechanisms of resistance to immunotherapies and the strategies that can be used to overcome such resistance. In this chapter, we briefly introduce the background of cancer immunoediting and immunotherapy. We also review and discuss the roles and mechanisms of RNA m6A modifications in fine-tuning the innate and adaptive immune responses, as well as in regulating tumor escape from immunosurveillance. Finally, we summarize the current strategies targeting m6A regulators for cancer immunotherapy.
Collapse
Affiliation(s)
- Shoubao Ma
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Los Angeles, CA, 91010, USA
- Hematologic Malignancies and Stem Cell Transplantation Institute, City of Hope National Medical Center, Los Angeles, CA, 91010, USA
| | - Tasha Barr
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Los Angeles, CA, 91010, USA
- Hematologic Malignancies and Stem Cell Transplantation Institute, City of Hope National Medical Center, Los Angeles, CA, 91010, USA
| | - Jianhua Yu
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Los Angeles, CA, 91010, USA.
- Hematologic Malignancies and Stem Cell Transplantation Institute, City of Hope National Medical Center, Los Angeles, CA, 91010, USA.
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope, Los Angeles, CA, 91010, USA.
- Comprehensive Cancer Center, City of Hope, Los Angeles, CA, 91010, USA.
| |
Collapse
|
30
|
Talayero VC, Vicente-Manzanares M. A primer on cancer-associated fibroblast mechanics and immunosuppressive ability. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2023; 4:17-27. [PMID: 36937319 PMCID: PMC10017186 DOI: 10.37349/etat.2023.00120] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2022] [Accepted: 10/11/2022] [Indexed: 02/25/2023] Open
Abstract
Cancer-associated fibroblasts (CAFs) are a major point of interest in modern oncology. Their interest resides in their ability to favor tumor growth without carrying genetic mutations. From a translational standpoint, they are potential therapeutic targets, particularly for hard-to-treat solid cancers. CAFs can be defined as non-tumor cells within the tumor microenvironment that have the morphological traits of fibroblasts, are negative for lineage-specific markers (e.g., leukocyte, endothelium), and enhance tumor progression in a multi-pronged manner. Two often-mentioned aspects of CAF biology are their ability to alter the mechanics and architecture of the tumor microenvironment, and also to drive local immunosuppression. These two aspects are the specific focus of this work, which also contains a brief summary of novel therapeutic interventions under study to normalize or eliminate CAFs from the tumor microenvironment.
Collapse
Affiliation(s)
- Vanessa C. Talayero
- Molecular Mechanisms Program, Centro de Investigación del Cáncer and Instituto de Biología Molecular y Celular del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC)-Universidad de Salamanca, 37007 Salamanca, Spain
| | - Miguel Vicente-Manzanares
- Molecular Mechanisms Program, Centro de Investigación del Cáncer and Instituto de Biología Molecular y Celular del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC)-Universidad de Salamanca, 37007 Salamanca, Spain
- Correspondence: Miguel Vicente-Manzanares, Molecular Mechanisms Program, Centro de Investigación del Cáncer and Instituto de Biología Molecular y Celular del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC)-Universidad de Salamanca, 37007 Salamanca, Spain.
| |
Collapse
|
31
|
Krammer S, Yang Z, Mitländer H, Grund JC, Trump S, Mittler S, Zirlik S, Finotto S. Rhinovirus Suppresses TGF-β-GARP Presentation by Peripheral NK Cells. Cells 2022; 12:129. [PMID: 36611921 PMCID: PMC9818541 DOI: 10.3390/cells12010129] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 12/15/2022] [Accepted: 12/23/2022] [Indexed: 12/31/2022] Open
Abstract
Asthma is a chronic airway disease whose exacerbations are often triggered by rhinovirus infection. TGF-β1 induces rhinovirus replication in infected cells. Moreover, TGF-β1 is a pleiotropic mediator that is produced by many immune cells in the latent, inactive form bound to the latency-associated peptide (LAP) and to the transmembrane protein glycoprotein A repetitions predominant (GARP). In this study we wanted to investigate the effect of rhinovirus infection on the TGF-β secretion and the downstream signaling via TGF-βRI/RII in peripheral blood mononuclear cells from control and asthmatic patients after rhinovirus infection ex vivo. Here, we found a significant upregulation of TGF-βRII in untouched PBMCs of asthmatics as well as a suppression of TGF-β release in the rhinovirus-infected PBMC condition. Moreover, consistent with an effect of TGF-β on Tregs, PBMCs infected with RV induced Tregs, and TGF-βRII directly correlated with RV1b mRNA. Finally, we found via flow cytometry that NK cells expressed less GARP surface-bound TGF-β, while cytokine-producing NKbright cells were induced. In summary, we show that rhinovirus infection inhibits TGF-β release in PBMCs, which results in the activation of both Treg and NK cells.
Collapse
Affiliation(s)
- Susanne Krammer
- Department of Molecular Pneumology, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Universitätsklinikum Erlangen, 91054 Erlangen, Germany
| | - Zuqin Yang
- Department of Molecular Pneumology, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Universitätsklinikum Erlangen, 91054 Erlangen, Germany
| | - Hannah Mitländer
- Department of Molecular Pneumology, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Universitätsklinikum Erlangen, 91054 Erlangen, Germany
| | - Janina C. Grund
- Department of Molecular Pneumology, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Universitätsklinikum Erlangen, 91054 Erlangen, Germany
| | - Sonja Trump
- Department of Molecular Pneumology, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Universitätsklinikum Erlangen, 91054 Erlangen, Germany
| | - Susanne Mittler
- Department of Molecular Pneumology, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Universitätsklinikum Erlangen, 91054 Erlangen, Germany
| | - Sabine Zirlik
- Department of Medicine 1, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Universitätsklinikum Erlangen, 91054 Erlangen, Germany
| | - Susetta Finotto
- Department of Molecular Pneumology, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Universitätsklinikum Erlangen, 91054 Erlangen, Germany
| |
Collapse
|
32
|
Rana PS, Soler DC, Kort J, Driscoll JJ. Targeting TGF-β signaling in the multiple myeloma microenvironment: Steering CARs and T cells in the right direction. Front Cell Dev Biol 2022; 10:1059715. [PMID: 36578789 PMCID: PMC9790996 DOI: 10.3389/fcell.2022.1059715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2022] [Accepted: 11/29/2022] [Indexed: 12/14/2022] Open
Abstract
Multiple myeloma (MM) remains a lethal hematologic cancer characterized by the expansion of transformed plasma cells within the permissive bone marrow (BM) milieu. The emergence of relapsed and/or refractory MM (RRMM) is provoked through clonal evolution of malignant plasma cells that harbor genomic, metabolic and proteomic perturbations. For most patients, relapsed disease remains a major cause of overall mortality. Transforming growth factors (TGFs) have pleiotropic effects that regulate myelomagenesis as well as the emergence of drug resistance. Moreover, TGF-β modulates numerous cell types present with the tumor microenvironment, including many immune cell types. While numerous agents have been FDA-approved over the past 2 decades and significantly expanded the treatment options available for MM patients, the molecular mechanisms responsible for drug resistance remain elusive. Multiple myeloma is uniformly preceded by a premalignant state, monoclonal gammopathy of unknown significance, and both conditions are associated with progressive deregulation in host immunity characterized by reduced T cell, natural killer (NK) cell and antigen-presenting dendritic cell (DC) activity. TGF-β promotes myelomagenesis as well as intrinsic drug resistance by repressing anti-myeloma immunity to promote tolerance, drug resistance and disease progression. Hence, repression of TGF-β signaling is a prerequisite to enhance the efficacy of current and future immunotherapeutics. Novel strategies that incorporate T cells that have been modified to express chimeric antigen receptor (CARs), T cell receptors (TCRs) and bispecific T cell engagers (BiTEs) offer promise to block TGF-β signaling, overcome chemoresistance and enhance anti-myeloma immunity. Here, we describe the effects of TGF-β signaling on immune cell effectors in the bone marrow and emerging strategies to overcome TGF-β-mediated myeloma growth, drug resistance and survival.
Collapse
Affiliation(s)
- Priyanka S. Rana
- Division of Hematology and Oncology, Department of Medicine, Case Western Reserve University, Cleveland, OH, United States,Case Comprehensive Cancer Center, Cleveland, OH, United States
| | - David C. Soler
- The Brain Tumor and Neuro-Oncology Center, The Center of Excellence for Translational Neuro-Oncology, Department of Neurosurgery, Case Western Reserve University, Cleveland, OH, United States
| | - Jeries Kort
- Division of Hematology and Oncology, Department of Medicine, Case Western Reserve University, Cleveland, OH, United States,Case Comprehensive Cancer Center, Cleveland, OH, United States,Adult Hematologic Malignancies and Stem Cell Transplant Section, Seidman Cancer Center, University Hospitals Cleveland Medical Center, Cleveland, OH, United States
| | - James J. Driscoll
- Division of Hematology and Oncology, Department of Medicine, Case Western Reserve University, Cleveland, OH, United States,Case Comprehensive Cancer Center, Cleveland, OH, United States,Adult Hematologic Malignancies and Stem Cell Transplant Section, Seidman Cancer Center, University Hospitals Cleveland Medical Center, Cleveland, OH, United States,*Correspondence: James J. Driscoll,
| |
Collapse
|
33
|
Bu MT, Chandrasekhar P, Ding L, Hugo W. The roles of TGF-β and VEGF pathways in the suppression of antitumor immunity in melanoma and other solid tumors. Pharmacol Ther 2022; 240:108211. [PMID: 35577211 PMCID: PMC10956517 DOI: 10.1016/j.pharmthera.2022.108211] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 05/09/2022] [Accepted: 05/10/2022] [Indexed: 12/13/2022]
Abstract
Immune checkpoint blockade (ICB) has become well-known in cancer therapy, strengthening the body's antitumor immune response rather than directly targeting cancer cells. Therapies targeting immune inhibitory checkpoints, such as PD-1, PD-L1, and CTLA-4, have resulted in impressive clinical responses across different types of solid tumors. However, as with other types of cancer treatments, ICB-based immunotherapy is hampered by both innate and acquired drug resistance. We previously reported the enrichment of gene signatures associated with wound healing, epithelial-to-mesenchymal, and angiogenesis processes in the tumors of patients with innate resistance to PD-1 checkpoint antibody therapy; we termed these the Innate Anti-PD-1 Resistance Signatures (IPRES). The TGF-β and VEGFA pathways emerge as the dominant drivers of IPRES-associated processes. Here, we review these pathways' functions, their roles in immunosuppression, and the currently available therapies that target them. We also discuss recent developments in the targeting of TGF-β using a specific antibody class termed trap antibody. The application of trap antibodies opens the promise of localized targeting of the TGF-β and VEGFA pathways within the tumor microenvironment. Such specificity may offer an enhanced therapeutic window that enables suppression of the IPRES processes in the tumor microenvironment while sparing the normal homeostatic functions of TGF-β and VEGFA in healthy tissues.
Collapse
Affiliation(s)
- Melissa T Bu
- Department of Medicine/Dermatology, University of California Los Angeles, Los Angeles, CA 90095, USA; Department of Molecular, Cell, and Developmental Biology, University of California Los Angeles, Los Angeles, CA 90095, USA; David Geffen School of Medicine at UCLA, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Pallavi Chandrasekhar
- Department of Medicine/Dermatology, University of California Los Angeles, Los Angeles, CA 90095, USA; David Geffen School of Medicine at UCLA, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Lizhong Ding
- Department of Medicine/Dermatology, University of California Los Angeles, Los Angeles, CA 90095, USA; Parker Institute for Cancer Immunotherapy UCLA, USA; David Geffen School of Medicine at UCLA, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Willy Hugo
- Department of Medicine/Dermatology, University of California Los Angeles, Los Angeles, CA 90095, USA; Jonsson Comprehensive Cancer Center, University of California Los Angeles, Los Angeles, CA 90095, USA; Parker Institute for Cancer Immunotherapy UCLA, USA; David Geffen School of Medicine at UCLA, University of California Los Angeles, Los Angeles, CA 90095, USA.
| |
Collapse
|
34
|
Gauthier T, Chen W. IFN-γ and TGF-β, Crucial Players in Immune Responses: A Tribute to Howard Young. J Interferon Cytokine Res 2022; 42:643-654. [PMID: 36516375 PMCID: PMC9917322 DOI: 10.1089/jir.2022.0132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Accepted: 06/18/2022] [Indexed: 12/15/2022] Open
Abstract
Interferon gamma (IFN-γ) and transforming growth factor beta (TGF-β), both pleiotropic cytokines, have been long studied and described as critical mediators of the immune response, notably in T cells. One of the investigators who made seminal and critical discoveries in the field of IFN-γ biology is Dr. Howard Young. In this review, we provide an overview of the biology of IFN-γ as well as its role in cancer and autoimmunity with an emphasis on Dr. Young's critical work in the field. We also describe how Dr. Young's work influenced our own research studying the role of TGF-β in the modulation of immune responses.
Collapse
Affiliation(s)
- Thierry Gauthier
- Mucosal Immunology Section, National Institute of Dental and Craniofacial Research (NIDCR), National Institutes of Health, Bethesda, Maryland, USA
| | - WanJun Chen
- Mucosal Immunology Section, National Institute of Dental and Craniofacial Research (NIDCR), National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
35
|
Alluqmani N, Jirovec A, Taha Z, Varette O, Chen A, Serrano D, Maznyi G, Khan S, Forbes NE, Arulanandam R, Auer RC, Diallo JS. Vanadyl sulfate-enhanced oncolytic virus immunotherapy mediates the antitumor immune response by upregulating the secretion of pro-inflammatory cytokines and chemokines. Front Immunol 2022; 13:1032356. [PMID: 36532027 PMCID: PMC9749062 DOI: 10.3389/fimmu.2022.1032356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 11/09/2022] [Indexed: 11/29/2022] Open
Abstract
Oncolytic viruses (OVs) are promising anticancer treatments that specifically replicate in and kill cancer cells and have profound immunostimulatory effects. We previously reported the potential of vanadium-based compounds such as vanadyl sulfate (VS) as immunostimulatory enhancers of OV immunotherapy. These compounds, in conjunction with RNA-based OVs such as oncolytic vesicular stomatitis virus (VSVΔ51), improve viral spread and oncolysis, leading to long-term antitumor immunity and prolonged survival in resistant tumor models. This effect is associated with a virus-induced antiviral type I IFN response shifting towards a type II IFN response in the presence of vanadium. Here, we investigated the systemic impact of VS+VSVΔ51 combination therapy to understand the immunological mechanism of action leading to improved antitumor responses. VS+VSVΔ51 combination therapy significantly increased the levels of IFN-γ and IL-6, and improved tumor antigen-specific T-cell responses. Supported by immunological profiling and as a proof of concept for the design of more effective therapeutic regimens, we found that local delivery of IL-12 using VSVΔ51 in combination with VS further improved therapeutic outcomes in a syngeneic CT26WT colon cancer model.
Collapse
Affiliation(s)
- Nouf Alluqmani
- Centre for Innovative Cancer Research, Ottawa Hospital Research Institute, Ottawa, ON, Canada,Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, ON, Canada,Research Center, Molecular Oncology Department King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Anna Jirovec
- Centre for Innovative Cancer Research, Ottawa Hospital Research Institute, Ottawa, ON, Canada,Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, ON, Canada
| | - Zaid Taha
- Centre for Innovative Cancer Research, Ottawa Hospital Research Institute, Ottawa, ON, Canada,Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, ON, Canada
| | - Oliver Varette
- Centre for Innovative Cancer Research, Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - Andrew Chen
- Centre for Innovative Cancer Research, Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - Daniel Serrano
- Centre for Innovative Cancer Research, Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - Glib Maznyi
- Centre for Innovative Cancer Research, Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - Sarwat Khan
- Centre for Innovative Cancer Research, Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - Nicole E. Forbes
- Centre for Innovative Cancer Research, Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - Rozanne Arulanandam
- Centre for Innovative Cancer Research, Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - Rebecca C. Auer
- Centre for Innovative Cancer Research, Ottawa Hospital Research Institute, Ottawa, ON, Canada,Department of Surgery, University of Ottawa, Ottawa, ON, Canada
| | - Jean-Simon Diallo
- Centre for Innovative Cancer Research, Ottawa Hospital Research Institute, Ottawa, ON, Canada,Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, ON, Canada,*Correspondence: Jean-Simon Diallo,
| |
Collapse
|
36
|
Preventing Surgery-Induced NK Cell Dysfunction Using Anti-TGF-β Immunotherapeutics. Int J Mol Sci 2022; 23:ijms232314608. [PMID: 36498937 PMCID: PMC9737532 DOI: 10.3390/ijms232314608] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Revised: 11/11/2022] [Accepted: 11/18/2022] [Indexed: 11/24/2022] Open
Abstract
Natural Killer (NK) cell cytotoxicity and interferon-gamma (IFNγ) production are profoundly suppressed postoperatively. This dysfunction is associated with increased morbidity and cancer recurrence. NK activity depends on the integration of activating and inhibitory signals, which may be modulated by transforming growth factor-beta (TGF-β). We hypothesized that impaired postoperative NK cell IFNγ production is due to altered signaling pathways caused by postoperative TGF-β. NK cell receptor expression, downstream phosphorylated targets, and IFNγ production were assessed using peripheral blood mononuclear cells (PBMCs) from patients undergoing cancer surgery. Healthy NK cells were incubated in the presence of healthy/baseline/postoperative day (POD) 1 plasma and in the presence/absence of a TGF-β-blocking monoclonal antibody (mAb) or the small molecule inhibitor (smi) SB525334. Single-cell RNA sequencing (scRNA-seq) was performed on PBMCs from six patients with colorectal cancer having surgery at baseline/on POD1. Intracellular IFNγ, activating receptors (CD132, CD212, NKG2D, DNAM-1), and downstream target (STAT5, STAT4, p38 MAPK, S6) phosphorylation were significantly reduced on POD1. Furthermore, this dysfunction was phenocopied in healthy NK cells through incubation with rTGF-β1 or POD1 plasma and was prevented by the addition of anti-TGF-β immunotherapeutics (anti-TGF-β mAb or TGF-βR smi). Targeted gene analysis revealed significant decreases in S6 and FKBP12, an increase in Shp-2, and a reduction in NK metabolism-associated transcripts on POD1. pSmad2/3 was increased and pS6 was reduced in response to rTGF-β1 on POD1, changes that were prevented by anti-TGF-β immunotherapeutics. Together, these results suggest that both canonical and mTOR pathways downstream of TGF-β mediate phenotypic changes that result in postoperative NK cell dysfunction.
Collapse
|
37
|
Lei B, Song H, Xu F, Wei Q, Wang F, Tan G, Ma H. When does hepatitis B virus meet long-stranded noncoding RNAs? Front Microbiol 2022; 13:962186. [PMID: 36118202 PMCID: PMC9479684 DOI: 10.3389/fmicb.2022.962186] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Accepted: 07/22/2022] [Indexed: 01/16/2023] Open
Abstract
Hepatitis B virus (HBV) infection in humans and its associated diseases are long-standing problems. HBV can produce a large number of non-self-molecules during its life cycle, which acts as targets for innate immune recognition and initiation. Among these, interferon and its large number of downstream interferon-stimulated gene molecules are important early antiviral factors. However, the development of an effective antiviral immune response is not simple and depends not only on the delicate regulation of the immune response but also on the various mechanisms of virus-related immune escape and immune tolerance. Therefore, despite there being a relatively well-established consensus on the major pathways of the antiviral response and their component molecules, the complete clearance of HBV remains a challenge in both basic and clinical research. Long-noncoding RNAs (lncRNAs) are generally >200 bp in length and perform different functions in the RNA strand encoding the protein. As an important part of the IFN-inducible genes, interferon-stimulated lncRNAs are involved in the regulation of several HBV infection-related pathways. This review traces the basic elements of such pathways and characterizes the various recent targets of lncRNAs, which not only complement the regulatory mechanisms of pathways related to chronic HBV infection, fibrosis, and cancer promotion but also present with new potential therapeutic targets for controlling HBV infection and the malignant transformation of hepatocytes.
Collapse
Affiliation(s)
- Bingxin Lei
- Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, Department of Immunology, Center for Pathogen Biology and Infectious Diseases, The First Hospital of Jilin University, Changchun, Jilin, China
- Department of Anesthesiology, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Hongxiao Song
- Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, Department of Immunology, Center for Pathogen Biology and Infectious Diseases, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Fengchao Xu
- Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, Department of Immunology, Center for Pathogen Biology and Infectious Diseases, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Qi Wei
- Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, Department of Immunology, Center for Pathogen Biology and Infectious Diseases, The First Hospital of Jilin University, Changchun, Jilin, China
- Department of Anesthesiology, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Fei Wang
- Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, Department of Immunology, Center for Pathogen Biology and Infectious Diseases, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Guangyun Tan
- Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, Department of Immunology, Center for Pathogen Biology and Infectious Diseases, The First Hospital of Jilin University, Changchun, Jilin, China
- *Correspondence: Guangyun Tan,
| | - Haichun Ma
- Department of Anesthesiology, The First Hospital of Jilin University, Changchun, Jilin, China
- Haichun Ma,
| |
Collapse
|
38
|
Mukherjee A, Bilecz AJ, Lengyel E. The adipocyte microenvironment and cancer. Cancer Metastasis Rev 2022; 41:575-587. [PMID: 35941408 DOI: 10.1007/s10555-022-10059-x] [Citation(s) in RCA: 73] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Accepted: 08/01/2022] [Indexed: 02/08/2023]
Abstract
Many epithelial tumors grow in the vicinity of or metastasize to adipose tissue. As tumors develop, crosstalk between adipose tissue and cancer cells leads to changes in adipocyte function and paracrine signaling, promoting a microenvironment that supports tumor growth. Over the last decade, it became clear that tumor cells co-opt adipocytes in the tumor microenvironment, converting them into cancer-associated adipocytes (CAA). As adipocytes and cancer cells engage, a metabolic symbiosis ensues that is driven by bi-directional signaling. Many cancers (colon, breast, prostate, lung, ovarian cancer, and hematologic malignancies) stimulate lipolysis in adipocytes, followed by the uptake of fatty acids (FA) from the surrounding adipose tissue. The FA enters the cancer cell through specific fatty acid receptors and binding proteins (e.g., CD36, FATP1) and are used for membrane synthesis, energy metabolism (β-oxidation), or lipid-derived cell signaling molecules (derivatives of arachidonic and linolenic acid). Therefore, blocking adipocyte-derived lipid uptake or lipid-associated metabolic pathways in cancer cells, either with a single agent or in combination with standard of care chemotherapy, might prove to be an effective strategy against cancers that grow in lipid-rich tumor microenvironments.
Collapse
Affiliation(s)
- Abir Mukherjee
- Department of Obstetrics and Gynecology/Section of Gynecologic Oncology, Center for Integrative Science, University of Chicago, 5841 S. Maryland Avenue, Chicago, IL, 60637, USA
| | - Agnes J Bilecz
- Department of Obstetrics and Gynecology/Section of Gynecologic Oncology, Center for Integrative Science, University of Chicago, 5841 S. Maryland Avenue, Chicago, IL, 60637, USA
| | - Ernst Lengyel
- Department of Obstetrics and Gynecology/Section of Gynecologic Oncology, Center for Integrative Science, University of Chicago, 5841 S. Maryland Avenue, Chicago, IL, 60637, USA.
| |
Collapse
|
39
|
Zhu J, Sanford LD, Ren R, Zhang Y, Tang X. Multiple Machine Learning Methods Reveal Key Biomarkers of Obstructive Sleep Apnea and Continuous Positive Airway Pressure Treatment. Front Genet 2022; 13:927545. [PMID: 35910196 PMCID: PMC9326093 DOI: 10.3389/fgene.2022.927545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Accepted: 06/24/2022] [Indexed: 11/13/2022] Open
Abstract
Obstructive sleep apnea (OSA) is a worldwide health issue that affects more than 400 million people. Given the limitations inherent in the current conventional diagnosis of OSA based on symptoms report, novel diagnostic approaches are required to complement existing techniques. Recent advances in gene sequencing technology have made it possible to identify a greater number of genes linked to OSA. We identified key genes in OSA and CPAP treatment by screening differentially expressed genes (DEGs) using the Gene Expression Omnibus (GEO) database and employing machine learning algorithms. None of these genes had previously been implicated in OSA. Moreover, a new diagnostic model of OSA was developed, and its diagnostic accuracy was verified in independent datasets. By performing Single Sample Gene Set Enrichment Analysis (ssGSEA) and Counting Relative Subsets of RNA Transcripts (CIBERSORT), we identified possible immunologic mechanisms, which led us to conclude that patients with high OSA risk tend to have elevated inflammation levels that can be brought down by CPAP treatment.
Collapse
Affiliation(s)
- Jie Zhu
- Sleep Medicine Center, Department of Respiratory and Critical Care Medicine, Mental Health Center, West China Hospital, Sichuan University, Chengdu, China
| | - Larry D. Sanford
- Sleep Research Laboratory, Center for Integrative Neuroscience and Inflammatory Diseases, Pathology and Anatomy, Eastern Virginia Medical School, Norfolk, VA, United States
| | - Rong Ren
- Sleep Medicine Center, Department of Respiratory and Critical Care Medicine, Mental Health Center, West China Hospital, Sichuan University, Chengdu, China
| | - Ye Zhang
- Sleep Medicine Center, Department of Respiratory and Critical Care Medicine, Mental Health Center, West China Hospital, Sichuan University, Chengdu, China
| | - Xiangdong Tang
- Sleep Medicine Center, Department of Respiratory and Critical Care Medicine, Mental Health Center, West China Hospital, Sichuan University, Chengdu, China
- *Correspondence: Xiangdong Tang,
| |
Collapse
|
40
|
Ran GH, Lin YQ, Tian L, Zhang T, Yan DM, Yu JH, Deng YC. Natural killer cell homing and trafficking in tissues and tumors: from biology to application. Signal Transduct Target Ther 2022; 7:205. [PMID: 35768424 PMCID: PMC9243142 DOI: 10.1038/s41392-022-01058-z] [Citation(s) in RCA: 131] [Impact Index Per Article: 43.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 05/24/2022] [Accepted: 06/14/2022] [Indexed: 02/06/2023] Open
Abstract
Natural killer (NK) cells, a subgroup of innate lymphoid cells, act as the first line of defense against cancer. Although some evidence shows that NK cells can develop in secondary lymphoid tissues, NK cells develop mainly in the bone marrow (BM) and egress into the blood circulation when they mature. They then migrate to and settle down in peripheral tissues, though some special subsets home back into the BM or secondary lymphoid organs. Owing to its success in allogeneic adoptive transfer for cancer treatment and its "off-the-shelf" potential, NK cell-based immunotherapy is attracting increasing attention in the treatment of various cancers. However, insufficient infiltration of adoptively transferred NK cells limits clinical utility, especially for solid tumors. Expansion of NK cells or engineered chimeric antigen receptor (CAR) NK cells ex vivo prior to adoptive transfer by using various cytokines alters the profiles of chemokine receptors, which affects the infiltration of transferred NK cells into tumor tissue. Several factors control NK cell trafficking and homing, including cell-intrinsic factors (e.g., transcriptional factors), cell-extrinsic factors (e.g., integrins, selectins, chemokines and their corresponding receptors, signals induced by cytokines, sphingosine-1-phosphate (S1P), etc.), and the cellular microenvironment. Here, we summarize the profiles and mechanisms of NK cell homing and trafficking at steady state and during tumor development, aiming to improve NK cell-based cancer immunotherapy.
Collapse
Affiliation(s)
- Guang He Ran
- Department of Immunology, School of Basic Medical, Jiamusi University, 154007, Jiamusi, China
- Institute of Materia Medica, College of Pharmacy, Army Medical University, 400038, Chongqing, China
| | - Yu Qing Lin
- Department of Immunology, School of Basic Medical, Jiamusi University, 154007, Jiamusi, China
- Institute of Materia Medica, College of Pharmacy, Army Medical University, 400038, Chongqing, China
| | - Lei Tian
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Los Angeles, CA, 91010, USA
| | - Tao Zhang
- Department of Immunology, School of Basic Medical, Jiamusi University, 154007, Jiamusi, China.
| | - Dong Mei Yan
- Department of Immunology, School of Basic Medical, Jiamusi University, 154007, Jiamusi, China.
| | - Jian Hua Yu
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Los Angeles, CA, 91010, USA.
| | - You Cai Deng
- Institute of Materia Medica, College of Pharmacy, Army Medical University, 400038, Chongqing, China.
- Department of Clinical Hematology, College of Pharmacy, Army Medical University, 400038, Chongqing, China.
| |
Collapse
|
41
|
Challagundla N, Shah D, Yadav S, Agrawal-Rajput R. Saga of monokines in shaping tumour-immune microenvironment: Origin to execution. Cytokine 2022; 157:155948. [PMID: 35764025 DOI: 10.1016/j.cyto.2022.155948] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 06/09/2022] [Accepted: 06/13/2022] [Indexed: 11/19/2022]
Abstract
Cellular communication mediated by cytokines is an important mechanism dictating immune responses, their cross talk and final immune output. Cytokines play a major role in dictating the immune outcome to cancer by regulating the events of development, differentiation and activation of innate immune cells. Cytokines are pleiotropic in nature, hence understanding their role individually or as member of network cytokines is critical to delineate their role in tumour immunity. Tumour systemically manipulates the immune system to evade and escape immune recognition for their uncontrollable growth and metastasis. The developing tumour comprise a large and diverse set of myeloid cells which are vulnerable to manipulation by the tumour-microenvironment. The innate immune cells of the monocytic lineage skew the fate of the adaptive immune cells and thus dictating cancer elimination or progression. Targeting cells at tumour cite is preposterous owing to their tight network, poor reach and abundance of immunosuppressive mechanisms. Monocytic lineage-derived cytokines (monokines) play crucial role in tumour regression or progression by either directly killing the tumour cells with TNFα or promoting its growth by TGFβ. In addition, the monokines like IL-12, IL-1β, IL-6, IL-10 and TGFβ direct the adaptive immune cells to secrete anti-tumour cytokines, TNFα, IFNγ, perforin and granzyme or pro-tumour cytokines, IL-10 and TGFβ. In this review, we elucidate the roles of monokines in dictating the fate of tumour by regulating responses at various stages of generation, differentiation and activation of immune cells along with the extensive cross talk. We have attempted to delineate the synergy and antagonism of major monokines among themselves or with tumour-derived or adaptive immune cytokines. The review provides an update on the possibilities of placing monokines to potential practical use as cytokine therapy against cancer.
Collapse
Affiliation(s)
- Naveen Challagundla
- Immunology Lab, Indian Institute of Advanced Research, Koba Institutional Area, Gandhinagar, Gujarat 382426, India
| | - Dhruvi Shah
- Immunology Lab, Indian Institute of Advanced Research, Koba Institutional Area, Gandhinagar, Gujarat 382426, India
| | - Shivani Yadav
- Immunology Lab, Indian Institute of Advanced Research, Koba Institutional Area, Gandhinagar, Gujarat 382426, India
| | - Reena Agrawal-Rajput
- Immunology Lab, Indian Institute of Advanced Research, Koba Institutional Area, Gandhinagar, Gujarat 382426, India.
| |
Collapse
|
42
|
Rasmi Y, Heidari N, Kübra Kırboğa K, Hatamkhani S, Tekin B, Alipour S, Naderi R, Farnamian Y, Akca I. The importance of neopterin in COVID-19: The prognostic value and relation with the disease severity. Clin Biochem 2022; 104:1-12. [PMID: 35307400 PMCID: PMC8929545 DOI: 10.1016/j.clinbiochem.2022.03.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 03/10/2022] [Accepted: 03/12/2022] [Indexed: 02/07/2023]
Abstract
Coronavirus Disease 2019 [COVID-19], caused by severe acute respiratory syndrome coronavirus 2 [SARS-CoV-2], has rapidly evolved into a global health emergency. Neopterin [NPT], produced by macrophages when stimulated with interferon [IFN-]gamma, is an essential cytokine in the antiviral immune response. NPT has been used as a marker for the early assessment of disease severity in different diseases. The leading cause of NPT production is the pro-inflammatory cytokine IFN-. Macrophage activation has also been revealed to be linked with disease severity in SARS-CoV-2 patients. We demonstrate the importance of NPT in the pathogenesis of SARS-CoV-2 and suggest that targeting NPT in SARS-CoV-2 infection may be critical in the early prediction of disease progression and provision of timely management of infected individuals.
Collapse
Affiliation(s)
- Yousef Rasmi
- Cellular and Molecular Research Center, Cellular and Molecular Medicine Institute, Urmia University of Medical Sciences, Urmia, Iran; Department of Biochemistry, School of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Nadia Heidari
- Department of Biochemistry, School of Medicine, Gorgan University of Medical Sciences, Urmia, Iran
| | | | - Shima Hatamkhani
- Experimental and Applied Pharmaceutical Sciences Research Center, Department of Clinical Pharmacy, School of Pharmacy, Urmia University of Medical Sciences, Urmia, Iran
| | - Burcu Tekin
- Izmir Institute of Technology, Biotechnology Department, Izmir, Turkey
| | - Shahryar Alipour
- Department of Biochemistry and Applied Cell, School of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Roya Naderi
- Department of Physiology, School of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Yeghaneh Farnamian
- Student Research Center, School of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Ilknur Akca
- Mersin University, Faculty of Sciences, Department of Biotechnology, Mersin, Turkey
| |
Collapse
|
43
|
Xu H, Yin L, Xu Q, Xiang J, Xu R. N6-methyladenosine methylation modification patterns reveal immune profiling in pancreatic adenocarcinoma. Cancer Cell Int 2022; 22:199. [PMID: 35606813 PMCID: PMC9125922 DOI: 10.1186/s12935-022-02614-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Accepted: 05/06/2022] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Several studies have revealed that N6-methyladenosine (m6A) regulation is involved in various biological processes and cancer progression. Nevertheless, the potential effects of m6A modifications in the tumor immune microenvironment (TIME) and on immune regulation in pancreatic adenocarcinoma (PAAD) remains unclear. METHODS A consensus clustering algorithm was used to identify different m6A modification patterns and construct an m6A-associated gene signature based on 23 m6A regulators in PAAD. The CIBERSORT and ssGSEA algorithms were used to estimate the components of the immune cells in each sample. The PCA algorithm was used to develop the m6Ascore system for the evaluation of m6A modification patterns in each sample. RESULTS Two m6A modification patterns with different biological properties and prognoses were identified in 176 PAAD patient samples. The features of TIME between the two patterns were similar, with two definite immune phenotypes: immune-inflamed and immune-excluded. Based on the m6A phenotype-associated signature genes, we constructed an m6Ascore system to investigate the m6A modification pattern of each sample, profile the dissection of physiological processes, immune infiltration, clinical prognosis, immunotherapy, and genetic variation. Patients with low m6Ascore scores had better clinical outcomes, enhanced immune infiltration, and lower expression of immunotherapeutic drug targets, such as CD274 and PDCD1LG2. Further research indicated that the m6Ascore and tumor mutation burden were significantly correlated, and patients with low m6Ascore had higher mutation rates in SMAD4 and TTN. Moreover, TNFRSF21 was significantly upregulated in PAAD tumor tissues and cell lines. Lower expression of TNFRSF21 had a prominent advantage in survival and was correlated with a low level of immune infiltration. PAAD samples with different TNFRSF21 expression levels showed significantly distinct sensitivities to chemotherapeutic agents. CONCLUSIONS This study revealed that m6A modification patterns could play an important role in the diversity and complexity of TIME, and the m6Ascore system could serve as an independent and powerful prognostic biomarker and is latently related to PAAD immunotherapies. Quantitative determination of m6A modification patterns in individual patients will be instrumental in mapping the TIME landscape and further optimizing precision immunotherapy.
Collapse
Affiliation(s)
- Hao Xu
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang China
| | - Lu Yin
- Department of Pathology, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang China
| | - Qianhui Xu
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang China
| | - Jingjing Xiang
- Department of Pathology, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang China
| | - Rujun Xu
- Department of Pathology, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang China
| |
Collapse
|
44
|
Rauch J, Jochum J, Eisermann P, Gisbrecht J, Völker K, Hunstig F, Mehlhoop U, Muntau B, Tappe D. Inflammatory cytokine profile and T cell responses in African tick bite fever patients. Med Microbiol Immunol 2022; 211:143-152. [PMID: 35543881 PMCID: PMC9092931 DOI: 10.1007/s00430-022-00738-5] [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: 09/22/2021] [Accepted: 04/05/2022] [Indexed: 11/29/2022]
Abstract
African tick bite fever, an acute febrile illness, is caused by the obligate intracellular bacterium Rickettsia africae. Immune responses to rickettsial infections have so far mainly been investigated in vitro with infected endothelial cells as the main target cells, and in mouse models. Patient studies are rare and little is known about the immunology of human infections. In this study, inflammatory mediators and T cell responses were examined in samples from 13 patients with polymerase chain reaction-confirmed R. africae infections at different time points of illness. The Th1-associated cytokines IFNγ and IL-12 were increased in the acute phase of illness, as were levels of the T cell chemoattractant cytokine CXCL-10. In addition, the anti-inflammatory cytokine IL-10 and also IL-22 were elevated. IL-22 but not IFNγ was increasingly produced by CD4+ and CD8+ T cells during illness. Besides IFNγ, IL-22 appears to play a protective role in rickettsial infections.
Collapse
Affiliation(s)
- Jessica Rauch
- Bernhard Nocht Institute for Tropical Medicine, Bernhard-Nocht-Str. 74, 20359, Hamburg, Germany.
| | - Johannes Jochum
- University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Philip Eisermann
- Bernhard Nocht Institute for Tropical Medicine, Bernhard-Nocht-Str. 74, 20359, Hamburg, Germany
| | - Jana Gisbrecht
- Bernhard Nocht Institute for Tropical Medicine, Bernhard-Nocht-Str. 74, 20359, Hamburg, Germany
| | | | | | - Ute Mehlhoop
- Bernhard Nocht Institute for Tropical Medicine, Bernhard-Nocht-Str. 74, 20359, Hamburg, Germany
| | - Birgit Muntau
- Bernhard Nocht Institute for Tropical Medicine, Bernhard-Nocht-Str. 74, 20359, Hamburg, Germany
| | - Dennis Tappe
- Bernhard Nocht Institute for Tropical Medicine, Bernhard-Nocht-Str. 74, 20359, Hamburg, Germany
| |
Collapse
|
45
|
Zhang Y, Wu Y, Shen W, Wang B, Yuan X. Crosstalk between NK cells and hepatic stellate cells in liver fibrosis (Review). Mol Med Rep 2022; 25:208. [PMID: 35506449 PMCID: PMC9133963 DOI: 10.3892/mmr.2022.12724] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2022] [Accepted: 04/19/2022] [Indexed: 01/18/2023] Open
Abstract
Liver fibrosis is a common pathological process of chronic liver diseases, including viral hepatitis and alcoholic liver disease, and ultimately progresses to irreversible cirrhosis and cancer. Hepatic stellate cells (HSCs) are activated to produce amounts of collagens in response to liver injury, thus triggering the initiation and progression of fibrogenesis. Natural killer (NK) cells serve as the essential component of hepatic innate immunity and are considered to alleviate fibrosis by killing activated HSCs. Current antifibrotic interventions have improved fibrosis, but fail to halt its progression in the advanced stage. Clarifying the interaction between NK cells and HSCs will provide clues to the pathogenesis and potential therapies for advanced liver fibrosis.
Collapse
Affiliation(s)
- Yang Zhang
- Department of Gastroenterology, The First Affiliated Hospital of Heilongjiang University of Traditional Chinese Medicine, Harbin, Heilongjiang 150001, P.R. China
| | - Yuan Wu
- The Graduate School, Heilongjiang University of Traditional Chinese Medicine, Harbin, Heilongjiang 150001, P.R. China
| | - Wenjuan Shen
- Department of Gynaecology, The First Affiliated Hospital of Heilongjiang University of Traditional Chinese Medicine, Harbin, Heilongjiang 150001, P.R. China
| | - Bingyu Wang
- Department of Gastroenterology, Heilongjiang Academy of Traditional Chinese Medicine, Harbin, Heilongjiang 150001, P.R. China
| | - Xingxing Yuan
- Department of Gastroenterology, Heilongjiang Academy of Traditional Chinese Medicine, Harbin, Heilongjiang 150001, P.R. China
| |
Collapse
|
46
|
Rocca Y, Pouxvielh K, Marotel M, Benezech S, Jaeger B, Allatif O, Bendriss-Vermare N, Marçais A, Walzer T. Combinatorial Expression of NK Cell Receptors Governs Cell Subset Reactivity and Effector Functions but Not Tumor Specificity. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2022; 208:1802-1812. [PMID: 35288470 DOI: 10.4049/jimmunol.2100874] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Accepted: 01/28/2022] [Indexed: 12/20/2022]
Abstract
NK cell receptors allow NK cells to recognize targets such as tumor cells. Many of them are expressed on a subset of NK cells, independently of each other, which creates a vast diversity of receptor combinations. Whether these combinations influence NK cell antitumor responses is not well understood. We addressed this question in the C57BL/6 mouse model and analyzed the individual effector response of 444 mouse NK cell subsets, defined by combinations of 12 receptors, against tumor cell lines originating from different tissues and mouse strains. We found a wide range of reactivity among NK subsets, but the same hierarchy of responses was observed for the different tumor types, showing that the repertoire of NK cell receptors does not encode for different tumor specificities but for different intrinsic reactivities. The coexpression of CD27, NKG2A, and DNAM-1 identified subsets with relative cytotoxic specialization, whereas reciprocally, CD11b and KLRG1 defined the best IFN-γ producers. The expression of educating receptors Ly49C, Ly49I, and NKG2A was also strongly correlated with IFN-γ production, but this effect was suppressed by unengaged receptors Ly49A, Ly49F, and Ly49G2. Finally, IL-15 coordinated NK cell effector functions, but education and unbound inhibitory receptors retained some influence on their response. Collectively, these data refine our understanding of the mechanisms governing NK cell reactivity, which could help design new NK cell therapy protocols.
Collapse
Affiliation(s)
- Yamila Rocca
- Centre International de Recherche en Infectiologie, INSERM U1111, Ecole Normale Supérieure de Lyon, Université Lyon 1, CNRS, UMR 5308, Lyon, France.,Centre de Recherche en Cancérologie de Lyon, INSERM U1052, CNRS UMR 5286, Centre Léon Bérard, Université Claude Bernard Lyon 1, Lyon, France
| | - Kevin Pouxvielh
- Centre International de Recherche en Infectiologie, INSERM U1111, Ecole Normale Supérieure de Lyon, Université Lyon 1, CNRS, UMR 5308, Lyon, France
| | - Marie Marotel
- Centre International de Recherche en Infectiologie, INSERM U1111, Ecole Normale Supérieure de Lyon, Université Lyon 1, CNRS, UMR 5308, Lyon, France
| | - Sarah Benezech
- Centre International de Recherche en Infectiologie, INSERM U1111, Ecole Normale Supérieure de Lyon, Université Lyon 1, CNRS, UMR 5308, Lyon, France
| | - Baptiste Jaeger
- Faculty of Medicine, Brain Research Institute, University of Zurich, Zurich, Switzerland; and.,Faculty of Science, Brain Research Institute, University of Zurich, Zurich, Switzerland
| | - Omran Allatif
- Centre International de Recherche en Infectiologie, INSERM U1111, Ecole Normale Supérieure de Lyon, Université Lyon 1, CNRS, UMR 5308, Lyon, France
| | - Nathalie Bendriss-Vermare
- Centre de Recherche en Cancérologie de Lyon, INSERM U1052, CNRS UMR 5286, Centre Léon Bérard, Université Claude Bernard Lyon 1, Lyon, France
| | - Antoine Marçais
- Centre International de Recherche en Infectiologie, INSERM U1111, Ecole Normale Supérieure de Lyon, Université Lyon 1, CNRS, UMR 5308, Lyon, France;
| | - Thierry Walzer
- Centre International de Recherche en Infectiologie, INSERM U1111, Ecole Normale Supérieure de Lyon, Université Lyon 1, CNRS, UMR 5308, Lyon, France;
| |
Collapse
|
47
|
Czaja AJ. Immune Inhibitory Properties and Therapeutic Prospects of Transforming Growth Factor-Beta and Interleukin 10 in Autoimmune Hepatitis. Dig Dis Sci 2022; 67:1163-1186. [PMID: 33835375 DOI: 10.1007/s10620-021-06968-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Accepted: 03/22/2021] [Indexed: 12/14/2022]
Abstract
Transforming growth factor-beta and interleukin 10 have diverse immune inhibitory properties that have restored homeostatic defense mechanisms in experimental models of autoimmune disease. The goals of this review are to describe the actions of each cytokine, review their investigational use in animal models and patients, and indicate their prospects as interventions in autoimmune hepatitis. English abstracts were identified in PubMed by multiple search terms. Full-length articles were selected for review, and secondary and tertiary bibliographies were developed. Transforming growth factor-beta expands the natural and inducible populations of regulatory T cells, limits the proliferation of natural killer cells, suppresses the activation of naïve CD8+ T cells, decreases the production of interferon-gamma, and stimulates fibrotic repair. Interleukin 10 selectively inhibits the CD28 co-stimulatory signal for antigen recognition and impairs antigen-specific activation of uncommitted CD4+ and CD8+ T cells. It also inhibits maturation of dendritic cells, suppresses Th17 cells, supports regulatory T cells, and limits production of diverse pro-inflammatory cytokines. Contradictory immune stimulatory effects have been associated with each cytokine and may relate to the dose and accompanying cytokine milieu. Experimental findings have not translated into successful early clinical trials. The recombinant preparation of each agent in low dosage has been safe in human studies. In conclusion, transforming growth factor-beta and interleukin 10 have powerful immune inhibitory actions of potential therapeutic value in autoimmune hepatitis. The keys to their therapeutic application will be to match their predominant non-redundant function with the pivotal pathogenic mechanism or cytokine deficiency and to avoid contradictory immune stimulatory actions.
Collapse
Affiliation(s)
- Albert J Czaja
- Professor Emeritus of Medicine, Division of Gastroenterology and Hepatology, Mayo Clinic College of Medicine and Science, 200 First Street S.W., Rochester, MN, 55905, USA.
| |
Collapse
|
48
|
Dong A, Lin CW, Echeveste CE, Huang YW, Oshima K, Yearsley M, Chen X, Yu J, Wang LS. Protocatechuic Acid, a Gut Bacterial Metabolite of Black Raspberries, Inhibits Adenoma Development and Alters Gut Microbiome Profiles in Apc Min/+ Mice. J Cancer Prev 2022; 27:50-57. [PMID: 35419306 PMCID: PMC8984655 DOI: 10.15430/jcp.2022.27.1.50] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 03/19/2022] [Accepted: 03/21/2022] [Indexed: 11/06/2022] Open
Abstract
Administration of black raspberries (BRBs) and their anthocyanin metabolites, including protocatechuic acid (PCA), has been demonstrated to exert chemopreventive effects against colorectal cancer through alteration of innate immune cell trafficking, modulation of metabolic and inflammatory pathways, etc. Previous research has shown that the gut microbiome is important in the effectiveness of chemoprevention of colorectal cancer. This study aimed to assess the potency of PCA versus BRB dietary administration for colorectal cancer prevention using an Apc Min/+ mouse model and determine how bacterial profiles change in response to PCA and BRBs. A control AIN-76A diet supplemented with 5% BRBs, 500 ppm PCA, or 1,000 ppm PCA was administered to Apc Min/+ mice. Changes in incidence, polyp number, and polyp size regarding adenomas of the small intestine and colon were assessed after completion of the diet regimen. There were significant decreases in adenoma development by dietary administration of PCA and BRBs in the small intestine and the 5% BRB-supplemented diet in the colon. Pro-inflammatory bacterial profiles were replaced with anti-inflammatory bacteria in all treatments, with the greatest effects in the 5% BRB and 500 ppm PCA-supplemented diets accompanied by decreased COX-2 and prostaglandin E2 levels in colonic mucosa. We further showed that 500 ppm PCA, but not 1,000 ppm PCA, increased IFN-γ and SMAD4 levels in primary cultured human natural killer cells. These results suggest that both BRBs and a lower dose PCA can benefit colorectal cancer patients by inhibiting the growth and proliferation of adenomas and promoting a more favorable gut microbiome condition.
Collapse
Affiliation(s)
- Athena Dong
- Division of Hematology and Oncology, Department of Medicine, Medical College of Wisconsin, WI, USA
| | - Chien-Wei Lin
- Division of Biostatistics, Medical College of Wisconsin, WI, USA
| | - Carla Elena Echeveste
- Division of Hematology and Oncology, Department of Medicine, Medical College of Wisconsin, WI, USA
| | - Yi-Wen Huang
- Department of Obstetrics and Gynecology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Kiyoko Oshima
- Department of Pathology, Johns Hopkins University, Baltimore, MD, USA
| | - Martha Yearsley
- Department of Pathology, The Ohio State University, Columbus, OH, USA
| | - Xiao Chen
- Division of Hematology and Oncology, Department of Medicine, Medical College of Wisconsin, WI, USA
| | - Jianhua Yu
- Department of Hematology & Hematopoietic Cell Transplantation, City of Hope National Medical Center and Beckman Research Institute, Duarte, CA, USA
| | - Li-Shu Wang
- Division of Hematology and Oncology, Department of Medicine, Medical College of Wisconsin, WI, USA
| |
Collapse
|
49
|
Martynova E, Rizvanov A, Urbanowicz RA, Khaiboullina S. Inflammasome Contribution to the Activation of Th1, Th2, and Th17 Immune Responses. Front Microbiol 2022; 13:851835. [PMID: 35369454 PMCID: PMC8969514 DOI: 10.3389/fmicb.2022.851835] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Accepted: 02/22/2022] [Indexed: 12/24/2022] Open
Abstract
Inflammasomes are cytosolic polyprotein complexes formed in response to various external and internal stimuli, including viral and bacterial antigens. The main product of the inflammasome is active caspase 1 which proteolytically cleaves, releasing functional interleukin-1 beta (IL-1β) and interleukin-18 (IL-18). These cytokines play a central role in shaping immune response to pathogens. In this review, we will focus on the mechanisms of inflammasome activation, as well as their role in development of Th1, Th2, and Th17 lymphocytes. The contribution of cytokines IL-1β, IL-18, and IL-33, products of activated inflammasomes, are summarized. Additionally, the role of cytokines released from tissue cells in promoting differentiation of lymphocyte populations is discussed.
Collapse
Affiliation(s)
| | | | - Richard A. Urbanowicz
- Department of Infection Biology and Microbiomes, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, United Kingdom
| | | |
Collapse
|
50
|
Hrbac T, Kopkova A, Siegl F, Vecera M, Ruckova M, Kazda T, Jancalek R, Hendrych M, Hermanova M, Vybihal V, Fadrus P, Smrcka M, Sokol F, Kubes V, Lipina R, Slaby O, Kren L, Sana J. HLA-E and HLA-F Are Overexpressed in Glioblastoma and HLA-E Increased After Exposure to Ionizing Radiation. Cancer Genomics Proteomics 2022; 19:151-162. [PMID: 35181585 PMCID: PMC8865046 DOI: 10.21873/cgp.20311] [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: 11/05/2021] [Revised: 12/08/2021] [Accepted: 01/21/2022] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND/AIM Glioblastoma (GBM) is one of the deadliest human cancers responding very poorly to therapy. Although the central nervous system has been traditionally considered an immunologically privileged site with an enhanced immune response, GBM appears to benefit from this immunosuppressive milieu. Immunomodulatory molecules play an important role in immune tumor-host interactions. Non-classical human leukocyte antigens (HLA) class Ib molecules HLA-E, HLA-F, and HLA-G have been previously described to be involved in protecting semi-allogeneic fetal allografts from the maternal immune response and in transplant tolerance as well as tumoral immune escape. Unfortunately, their role in GBM remains poorly understood. Our study, therefore, aimed to characterize the relationship between the expression of these molecules in GBM on the transcriptional level and clinicopathological and molecular features of GBM as well as the effect of ionizing radiation. MATERIALS AND METHODS We performed the analysis of HLA-E, HLA-F, and HLA-G mRNA expression in 69 GBM tissue samples and 21 non-tumor brain tissue samples (controls) by reverse transcription polymerase chain reaction. Furthermore, two primary GBM cell cultures had been irradiated to identify the effect of ionizing radiation on the expression of non-classical HLA molecules. RESULTS Analyses revealed that both HLA-E and HLA-F are significantly up-regulated in GBM samples. Subsequent survival analysis showed a significant association between low expression of HLA-E and shorter survival of GBM patients. The dysregulated expression of both molecules was also observed between patients with methylated and unmethylated O-6-methylguanine-DNA methyltransferase (MGMT) promoter. Finally, we showed that ionizing radiation increased HLA-E expression level in GBM cells in vitro. CONCLUSION HLA-E and HLA-F play an important role in GBM biology and could be used as diagnostic biomarkers, and in the case of HLA-E also as a prognostic biomarker.
Collapse
Affiliation(s)
- Tomas Hrbac
- Department of Neurosurgery, University Hospital Ostrava, Ostrava, Czech Republic
| | - Alena Kopkova
- Central European Institute of Technology, Masaryk University, Brno, Czech Republic
| | - Frantisek Siegl
- Central European Institute of Technology, Masaryk University, Brno, Czech Republic
| | - Marek Vecera
- Central European Institute of Technology, Masaryk University, Brno, Czech Republic
| | - Michaela Ruckova
- Central European Institute of Technology, Masaryk University, Brno, Czech Republic
| | - Tomas Kazda
- Department of Radiation Oncology, Masaryk Memorial Cancer Institute and Faculty of Medicine of Masaryk University, Brno, Czech Republic
| | - Radim Jancalek
- Department of Neurosurgery, St. Anne's University Hospital and Faculty of Medicine of Masaryk University, Brno, Czech Republic
| | - Michal Hendrych
- First Department of Pathology, St. Anne's University Hospital and Faculty of Medicine of Masaryk University, Brno, Czech Republic
| | - Marketa Hermanova
- First Department of Pathology, St. Anne's University Hospital and Faculty of Medicine of Masaryk University, Brno, Czech Republic
| | - Vaclav Vybihal
- Department of Neurosurgery, University Hospital Brno, Brno, Czech Republic
| | - Pavel Fadrus
- Department of Neurosurgery, University Hospital Brno, Brno, Czech Republic
| | - Martin Smrcka
- Department of Neurosurgery, University Hospital Brno, Brno, Czech Republic
| | - Filip Sokol
- Department of Pathology, University Hospital Brno, Brno, Czech Republic
| | - Vaclav Kubes
- Department of Pathology, University Hospital Brno, Brno, Czech Republic
| | - Radim Lipina
- Department of Neurosurgery, University Hospital Ostrava, Ostrava, Czech Republic
| | - Ondrej Slaby
- Central European Institute of Technology, Masaryk University, Brno, Czech Republic
- Department of Biology, Faculty of Medicine of Masaryk University, Brno, Czech Republic
- Department of Comprehensive Cancer Care, Masaryk Memorial Cancer Institute, Brno, Czech Republic
| | - Leos Kren
- Department of Pathology, University Hospital Brno, Brno, Czech Republic;
| | - Jiri Sana
- Central European Institute of Technology, Masaryk University, Brno, Czech Republic;
- Department of Comprehensive Cancer Care, Masaryk Memorial Cancer Institute, Brno, Czech Republic
| |
Collapse
|