1
|
Hunzeker ZE, Zhao L, Kim AM, Parker JM, Zhu Z, Xiao H, Bai Q, Wakefield MR, Fang Y. The role of IL-22 in cancer. Med Oncol 2024; 41:240. [PMID: 39231878 DOI: 10.1007/s12032-024-02481-8] [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: 06/23/2024] [Accepted: 08/16/2024] [Indexed: 09/06/2024]
Abstract
Interleukin-22, discovered in the year of 2000, is a pleiotropic Th17 cytokine from the IL-10 family of cytokines. IL-22 signals through the type 2 cytokine receptor complex IL-22R and predominantly activates STAT3. This pathway leads to the transcription of several different types of genes, giving IL-22 context-specific functions ranging from inducing antimicrobial peptide expression to target cell proliferation. In recent years, it has been shown that IL-22 is involved in the pathogenesis of neoplasia in some cancers through its pro-proliferative and anti-apoptotic effects. This review highlights studies with recent discoveries and conclusions drawn on IL-22 and its involvement and function in various cancers. Such a study may be helpful to better understand the role of IL-22 in cancer so that new treatment could be developed targeting IL-22.
Collapse
Affiliation(s)
- Zachary E Hunzeker
- Department of Microbiology, Immunology & Pathology, Des Moines University College of Osteopathic Medicine, Des Moines, IA, 50312, USA
- Department of Surgery, University of Missouri School of Medicine, Columbia, MO, 65212, USA
- Department of Internal Medicine, University of Texas Houston Health Science Center, Houston, TX, USA
| | - Lei Zhao
- Department of Respiratory Medicine, the 2nd People's Hospital of Hefei and Hefei Hospital Affiliated to Anhui Medical University, Hefei, China
| | - Austin M Kim
- Department of Microbiology, Immunology & Pathology, Des Moines University College of Osteopathic Medicine, Des Moines, IA, 50312, USA
| | - Jacob M Parker
- Department of Microbiology, Immunology & Pathology, Des Moines University College of Osteopathic Medicine, Des Moines, IA, 50312, USA
| | - Ziwen Zhu
- Department of Surgery, University of Missouri School of Medicine, Columbia, MO, 65212, USA
| | - Huaping Xiao
- Department of Microbiology, Immunology & Pathology, Des Moines University College of Osteopathic Medicine, Des Moines, IA, 50312, USA
- Department of Surgery, University of Missouri School of Medicine, Columbia, MO, 65212, USA
| | - Qian Bai
- Department of Surgery, University of Missouri School of Medicine, Columbia, MO, 65212, USA
| | - Mark R Wakefield
- Department of Surgery, University of Missouri School of Medicine, Columbia, MO, 65212, USA
- Ellis Fischel Cancer Center, University of Missouri School of Medicine, Columbia, MO, 65212, USA
| | - Yujiang Fang
- Department of Microbiology, Immunology & Pathology, Des Moines University College of Osteopathic Medicine, Des Moines, IA, 50312, USA.
- Department of Surgery, University of Missouri School of Medicine, Columbia, MO, 65212, USA.
- Ellis Fischel Cancer Center, University of Missouri School of Medicine, Columbia, MO, 65212, USA.
| |
Collapse
|
2
|
Lee HK, Kim SY, Chung SH, Choi B, Kim JE, Yoon D, Jang SI, Yeo A, Kang HG, Lee J, Choi YH, Park JS, Sung Y, Kim JK, Chang EJ, Lee DK. Tumour-associated myeloid cells expressing IL-10R2/IL-22R1 as a potential biomarker for diagnosis and recurrence of pancreatic ductal adenocarcinoma. Br J Cancer 2024; 130:1979-1989. [PMID: 38643339 PMCID: PMC11183123 DOI: 10.1038/s41416-024-02676-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: 07/05/2023] [Revised: 03/05/2024] [Accepted: 04/02/2024] [Indexed: 04/22/2024] Open
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) is an aggressive malignancy with a poor survival rate, largely due to the lack of early diagnosis. Although myeloid cells are crucial in the tumour microenvironment, whether their specific subset can be a biomarker of PDAC progression is unclear. METHODS We analysed IL-22 receptor expression in PDAC and peripheral blood. Additionally, we analysed gene expression profiles of IL-10R2+/IL-22R1+ myeloid cells and the presence of these cells using single-cell RNA sequencing and murine orthotropic PDAC models, respectively, followed by examining the immunosuppressive function of IL-10R2+/IL-22R1+ myeloid cells. Finally, the correlation between IL-10R2 expression and PDAC progression was evaluated. RESULTS IL-10R2+/IL-22R1+ myeloid cells were present in PDAC and peripheral blood. Blood IL-10R2+ myeloid cells displayed a gene expression signature associated with tumour-educated circulating monocytes. IL-10R2+/IL-22R1+ myeloid cells from human myeloid cell culture inhibited T cell proliferation. By mouse models for PDAC, we found a positive correlation between pancreatic tumour growth and increased blood IL-10R2+/IL-22R1+ myeloid cells. IL-10R2+/IL-22R1+ myeloid cells from an early phase of the PDAC model suppressed T cell proliferation and cytotoxicity. IL-10R2+ myeloid cells indicated tumour recurrence 130 days sooner than CA19-9 in post-pancreatectomy patients. CONCLUSIONS IL-10R2+/IL-22R1+ myeloid cells in the peripheral blood might be an early marker of PDAC prognosis.
Collapse
MESH Headings
- Carcinoma, Pancreatic Ductal/genetics
- Carcinoma, Pancreatic Ductal/pathology
- Carcinoma, Pancreatic Ductal/diagnosis
- Carcinoma, Pancreatic Ductal/blood
- Humans
- Animals
- Pancreatic Neoplasms/pathology
- Pancreatic Neoplasms/genetics
- Pancreatic Neoplasms/diagnosis
- Pancreatic Neoplasms/blood
- Mice
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/blood
- Receptors, Interleukin/genetics
- Myeloid Cells/metabolism
- Myeloid Cells/pathology
- Neoplasm Recurrence, Local/genetics
- Neoplasm Recurrence, Local/pathology
- Interleukin-10 Receptor beta Subunit/genetics
- Female
- Male
- Tumor Microenvironment/genetics
- Cell Line, Tumor
Collapse
Affiliation(s)
- Hyung Keun Lee
- Severance Institute for Vascular and Metabolic Research, Yonsei University College of Medicine, Seoul, Korea
- Institute of Vision Research, Department of Ophthalmology, Yonsei University College of Medicine, Seoul, Korea
- College of Pharmacy, Yonsei University, Incheon, Korea
| | - So Young Kim
- Severance Institute for Vascular and Metabolic Research, Yonsei University College of Medicine, Seoul, Korea
- Institute of Biomedical Research, Yonsei University College of Medicine, Seoul, Korea
| | - Soo-Hyun Chung
- Department of Biochemistry and Molecular Biology, Brain Korea 21 Project, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
- Stem Cell Immunomodulation Research Center, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Bongkun Choi
- Department of Biochemistry and Molecular Biology, Brain Korea 21 Project, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
- Stem Cell Immunomodulation Research Center, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Ji-Eun Kim
- Department of Biochemistry and Molecular Biology, Brain Korea 21 Project, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
- Stem Cell Immunomodulation Research Center, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Dohee Yoon
- Department of Biochemistry and Molecular Biology, Brain Korea 21 Project, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
- Stem Cell Immunomodulation Research Center, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Sung Ill Jang
- Department of Internal Medicine, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Areum Yeo
- Severance Institute for Vascular and Metabolic Research, Yonsei University College of Medicine, Seoul, Korea
- Institute of Biomedical Research, Yonsei University College of Medicine, Seoul, Korea
| | - Hyun Goo Kang
- Institute of Vision Research, Department of Ophthalmology, Yonsei University College of Medicine, Seoul, Korea
| | - Jusung Lee
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang, Korea
- Department of New Biology, DGIST, Daegu, Korea
| | - Yoon Ha Choi
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang, Korea
| | - Joon Seong Park
- Department of Surgery, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
- Department of Surgery and Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Yoolim Sung
- Department of Biochemistry and Molecular Biology, Brain Korea 21 Project, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
- Stem Cell Immunomodulation Research Center, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Jong Kyoung Kim
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang, Korea.
- Institute for Convergence Research and Education in Advanced Technology, Yonsei University, Seoul, Korea.
| | - Eun-Ju Chang
- Department of Biochemistry and Molecular Biology, Brain Korea 21 Project, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea.
- Stem Cell Immunomodulation Research Center, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea.
| | - Dong Ki Lee
- Department of Internal Medicine, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea.
| |
Collapse
|
3
|
Mastini C, Campisi M, Patrucco E, Mura G, Ferreira A, Costa C, Ambrogio C, Germena G, Martinengo C, Peola S, Mota I, Vissio E, Molinaro L, Arigoni M, Olivero M, Calogero R, Prokoph N, Tabbò F, Shoji B, Brugieres L, Geoerger B, Turner SD, Cuesta-Mateos C, D’Aliberti D, Mologni L, Piazza R, Gambacorti-Passerini C, Inghirami GG, Chiono V, Kamm RD, Hirsch E, Koch R, Weinstock DM, Aster JC, Voena C, Chiarle R. Targeting CCR7-PI3Kγ overcomes resistance to tyrosine kinase inhibitors in ALK-rearranged lymphoma. Sci Transl Med 2023; 15:eabo3826. [PMID: 37379367 PMCID: PMC10804420 DOI: 10.1126/scitranslmed.abo3826] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Accepted: 06/02/2023] [Indexed: 06/30/2023]
Abstract
Anaplastic lymphoma kinase (ALK) tyrosine kinase inhibitors (TKIs) show potent efficacy in several ALK-driven tumors, but the development of resistance limits their long-term clinical impact. Although resistance mechanisms have been studied extensively in ALK-driven non-small cell lung cancer, they are poorly understood in ALK-driven anaplastic large cell lymphoma (ALCL). Here, we identify a survival pathway supported by the tumor microenvironment that activates phosphatidylinositol 3-kinase γ (PI3K-γ) signaling through the C-C motif chemokine receptor 7 (CCR7). We found increased PI3K signaling in patients and ALCL cell lines resistant to ALK TKIs. PI3Kγ expression was predictive of a lack of response to ALK TKI in patients with ALCL. Expression of CCR7, PI3Kγ, and PI3Kδ were up-regulated during ALK or STAT3 inhibition or degradation and a constitutively active PI3Kγ isoform cooperated with oncogenic ALK to accelerate lymphomagenesis in mice. In a three-dimensional microfluidic chip, endothelial cells that produce the CCR7 ligands CCL19/CCL21 protected ALCL cells from apoptosis induced by crizotinib. The PI3Kγ/δ inhibitor duvelisib potentiated crizotinib activity against ALCL lines and patient-derived xenografts. Furthermore, genetic deletion of CCR7 blocked the central nervous system dissemination and perivascular growth of ALCL in mice treated with crizotinib. Thus, blockade of PI3Kγ or CCR7 signaling together with ALK TKI treatment reduces primary resistance and the survival of persister lymphoma cells in ALCL.
Collapse
Affiliation(s)
- Cristina Mastini
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino 10126, Italy
| | - Marco Campisi
- Dana Farber Cancer Institute, Boston, MA 02115, USA
- Department of Pathology, Boston Children’s Hospital and Harvard Medical School, Boston, MA 02115, USA
- Department of Mechanical and Aerospace Engineering, Politecnico of Torino, Torino 10129, Italy
| | - Enrico Patrucco
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino 10126, Italy
| | - Giulia Mura
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino 10126, Italy
| | - Antonio Ferreira
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston MA 02115, USA
| | - Carlotta Costa
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino 10126, Italy
| | - Chiara Ambrogio
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino 10126, Italy
| | - Giulia Germena
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino 10126, Italy
| | - Cinzia Martinengo
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino 10126, Italy
| | - Silvia Peola
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino 10126, Italy
| | - Ines Mota
- Department of Pathology, Boston Children’s Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Elena Vissio
- Department of Oncology, University of Torino, Orbassano, Torino 10043, Italy
| | - Luca Molinaro
- Department of Medical Science, University of Torino, Torino 10126, Italy
| | - Maddalena Arigoni
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino 10126, Italy
| | - Martina Olivero
- Department of Oncology, University of Torino, Orbassano, Torino 10043, Italy
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Torino 10060, Italy
| | - Raffaele Calogero
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino 10126, Italy
| | - Nina Prokoph
- Division of Cellular and Molecular Pathology, Department of Pathology, University of Cambridge, Addenbrooke’s Hospital, Cambridge CB2 0QQ, UK
| | - Fabrizio Tabbò
- Department of Pathology, Cornell University, New York NY 10121, USA
| | - Brent Shoji
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston MA 02115, USA
| | - Laurence Brugieres
- Department of Pediatric and Adolescent Oncology, Gustave Roussy Cancer Center, Paris-Saclay University, Villejuif 94805, France
| | - Birgit Geoerger
- Department of Pediatric and Adolescent Oncology, Gustave Roussy Cancer Center, Paris-Saclay University, Villejuif 94805, France
- Université Paris-Saclay, INSERM U1015, Villejuif 94805, France
| | - Suzanne D. Turner
- Division of Cellular and Molecular Pathology, Department of Pathology, University of Cambridge, Addenbrooke’s Hospital, Cambridge CB2 0QQ, UK
- Faculty of Medicine, Masaryk University, Brno 601 77, Czech Republic
| | - Carlos Cuesta-Mateos
- Department of Pre-Clinical Development, Catapult Therapeutics B.V., 8243 RC, Lelystad, Netherlands
| | - Deborah D’Aliberti
- Department of Medicine and Surgery, University of Milan-Bicocca, Monza 20900, Italy
| | - Luca Mologni
- Department of Medicine and Surgery, University of Milan-Bicocca, Monza 20900, Italy
| | - Rocco Piazza
- Department of Medicine and Surgery, University of Milan-Bicocca, Monza 20900, Italy
| | | | | | - Valeria Chiono
- Department of Mechanical and Aerospace Engineering, Politecnico of Torino, Torino 10129, Italy
| | - Roger D. Kamm
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Emilio Hirsch
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino 10126, Italy
| | - Raphael Koch
- Dana Farber Cancer Institute, Boston, MA 02115, USA
- Harvard Medical School, Boston, MA 02115, USA
- University Medical Center Göttingen, 37075 Göttingen, Germany
| | - David M. Weinstock
- Dana Farber Cancer Institute, Boston, MA 02115, USA
- Harvard Medical School, Boston, MA 02115, USA
| | - Jon C. Aster
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston MA 02115, USA
| | - Claudia Voena
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino 10126, Italy
| | - Roberto Chiarle
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino 10126, Italy
- Department of Pathology, Boston Children’s Hospital and Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
4
|
Donnelly RP. Howard A. Young: Always Willing to Lend a Helping Hand. J Interferon Cytokine Res 2022; 42:608-610. [PMID: 35647936 DOI: 10.1089/jir.2022.0085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Affiliation(s)
- Raymond P Donnelly
- Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland, USA
| |
Collapse
|
5
|
Liang HC. IL-2/IL-2R signaling and IL-2Rα-targeted therapy in anaplastic large cell lymphoma. PATHOLOGIE (HEIDELBERG, GERMANY) 2022; 43:25-30. [PMID: 36094651 DOI: 10.1007/s00292-022-01108-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 08/22/2022] [Indexed: 06/15/2023]
Abstract
Anaplastic large cell lymphoma (ALCL) is a CD30-positive non-Hodgkin's T‑cell lymphoma. Despite the implementation of CD30 antibody-drug conjugate-targeted therapy into front-line treatment regimens, the prognosis of some subtypes of the disease remains unsatisfactory. In the relapsed/refractory setting, effective second-line treatment options are still lacking. However, it has been reported that blockade of direct downstream targets of activator protein‑1 (AP-1) transcription factors, which are highly dysregulated in ALCL, results in complete and sustained remission in late-stage relapsed/refractory anaplastic lymphoma kinase (ALK)-positive ALCL patients. Moreover, it has been identified that involvement of the BATF3/AP‑1 module promotes lymphomagenesis via oncogenic BATF3/IL-2/IL-2R signaling through hyperphosphorylation of ERK1/2, STAT1, and STAT5 in ALCL cells regardless of their ALK status. Therefore, targeting BATF3/IL-2/IL-2R signaling may represent a novel therapeutic alternative for ALCL patients.
Collapse
Affiliation(s)
- Huan-Chang Liang
- Human Oncology & Pathogenesis Program (HOPP), Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, USA.
| |
Collapse
|
6
|
Guo Y, Guo H, Zhang Y, Cui J. Anaplastic lymphoma kinase-special immunity and immunotherapy. Front Immunol 2022; 13:908894. [PMID: 35958559 PMCID: PMC9359062 DOI: 10.3389/fimmu.2022.908894] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 06/30/2022] [Indexed: 11/13/2022] Open
Abstract
Alterations in the anaplastic lymphoma kinase (ALK) gene play a key role in the development of various human tumors, and targeted therapy has transformed the treatment paradigm for these oncogene-driven tumors. However, primary or acquired resistance remains a challenge. ALK gene variants (such as gene rearrangements and mutations) also play a key role in the tumor immune microenvironment. Immunotherapy targeting the ALK gene has potential clinical applications. Here, we review the results of recent studies on the immunological relevance of ALK-altered tumors, which provides important insights into the development of tumor immunotherapies targeting this large class of tumors.
Collapse
Affiliation(s)
| | | | | | - Jiuwei Cui
- Cancer Center, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
7
|
Albayrak N, Orte Cano C, Karimi S, Dogahe D, Van Praet A, Godefroid A, Del Marmol V, Grimaldi D, Bondue B, Van Vooren JP, Mascart F, Corbière V. Distinct Expression Patterns of Interleukin-22 Receptor 1 on Blood Hematopoietic Cells in SARS-CoV-2 Infection. Front Immunol 2022; 13:769839. [PMID: 35422799 PMCID: PMC9004465 DOI: 10.3389/fimmu.2022.769839] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Accepted: 03/07/2022] [Indexed: 12/12/2022] Open
Abstract
The new pandemic virus SARS-CoV-2 is characterized by uncontrolled hyper-inflammation in severe cases. As the IL-22/IL-22R1 axis was reported to be involved in inflammation during viral infections, we characterized the expression of IL-22 receptor1, IL-22 and IL-22 binding protein in COVID-19 patients. Blood samples were collected from 19 non-severe and 14 severe patients on the day they presented (D0), at D14, and six months later, and from 6 non-infected controls. The IL-22R1 expression was characterized by flow cytometry. Results were related to HLA-DR expression of myeloid cells, to plasma concentrations of different cytokines and chemokines and NK cells and T lymphocytes functions characterized by their IFN-γ, IL-22, IL-17A, granzyme B and perforin content. The numbers of IL-22R1+ classical, intermediate, and non-classical monocytes and the proportions of IL-22R1+ plasmacytoid DC (pDC), myeloid DC1 and DC2 (mDC1, mDC2) were higher in patients than controls at D0. The proportions of IL-22R1+ classical and intermediate monocytes, and pDC and mDC2 remained high for six months. High proportions of IL-22R1+ non-classical monocytes and mDC2 displayed HLA-DRhigh expression and were thus activated. Multivariate analysis for all IL-22R1+ myeloid cells discriminated the severity of the disease (AUC=0.9023). However, correlation analysis between IL-22R1+ cell subsets and plasma chemokine concentrations suggested pro-inflammatory effects of some subsets and protective effects of others. The numbers of IL-22R1+ classical monocytes and pDC were positively correlated with pro-inflammatory chemokines MCP-1 and IP-10 in severe infections, whereas IL-22R1+ intermediate monocytes were negatively correlated with IL-6, IFN-α and CRP in non-severe infections. Moreover, in the absence of in vitro stimulation, NK and CD4+ T cells produced IFN-γ and IL-22, and CD4+ and CD8+ T cells produced IL-17A. CD4+ T lymphocytes also expressed IL-22R1, the density of its expression defining two different functional subsets. In conclusion, we provide the first evidence that SARS-CoV-2 infection is characterized by an abnormal expression of IL22R1 on blood myeloid cells and CD4+ T lymphocytes. Our results suggest that the involvement of the IL-22R1/IL-22 axis could be protective at the beginning of SARS-CoV-2 infection but could shift to a detrimental response over time.
Collapse
Affiliation(s)
- Nurhan Albayrak
- Laboratory of Vaccinology and Mucosal Immunity, Université Libre de Bruxelles, Brussels, Belgium
| | - Carmen Orte Cano
- Department of Dermatology, Hopital Erasme, Université Libre de Bruxelles, Brussels, Belgium
| | - Sina Karimi
- Department of Internal Medicine, Hopital Erasme, Université Libre de Bruxelles, Brussels, Belgium
| | - David Dogahe
- Department of Internal Medicine, Hopital Erasme, Université Libre de Bruxelles, Brussels, Belgium
| | - Anne Van Praet
- Laboratory of Vaccinology and Mucosal Immunity, Université Libre de Bruxelles, Brussels, Belgium
| | - Audrey Godefroid
- Laboratory of Vaccinology and Mucosal Immunity, Université Libre de Bruxelles, Brussels, Belgium
| | - Véronique Del Marmol
- Department of Dermatology, Hopital Erasme, Université Libre de Bruxelles, Brussels, Belgium
| | - David Grimaldi
- Department of Intensive Care Unit, Hopital Erasme, Université Libre de Bruxelles, Brussels, Belgium
| | - Benjamin Bondue
- Department of Pneumology, Hopital Erasme, Université Libre de Bruxelles, Brussels, Belgium
| | - Jean-Paul Van Vooren
- Laboratory of Vaccinology and Mucosal Immunity, Université Libre de Bruxelles, Brussels, Belgium.,Immunodeficiency Unit, Hopital Erasme, Université Libre de Bruxelles, Brussels, Belgium
| | - Françoise Mascart
- Laboratory of Vaccinology and Mucosal Immunity, Université Libre de Bruxelles, Brussels, Belgium
| | - Véronique Corbière
- Laboratory of Vaccinology and Mucosal Immunity, Université Libre de Bruxelles, Brussels, Belgium
| |
Collapse
|
8
|
Abstract
Sjögren's syndrome (SjS) is a systemic autoimmune disease marked by xerostomia (dry mouth), keratoconjunctivitis sicca (eye dryness), and other systematic disorders. Its pathogenesis involves an inflammatory process that is characterized by lymphocytic infiltration into exocrine glands and other tissues. Although the development of ectopic lymphoid tissue and overproduction of autoantibodies by hyperactive B cells suggest that they may promote SjS development, treatment directed towards them fails to induce significant laboratory or clinical improvement. T cells are overwhelming infiltrators in most phases of the disease, and the involvement of multiple T cell subsets of suggests the extraordinary complexity of SjS pathogenesis. The factors, including various cellular subtypes and molecules, regulate the activation and suppression of T cells. T cell activation induces inflammatory cell infiltration, B cell activation, tissue damage, and metabolic changes in SjS. Knowledge of the pathways that link these T cell subtypes and regulation of their activities are not completely understood. This review comprehensively summarizes the research progress and our understanding of T cells in SjS, including CD4+ T cells, CD8+ TRM cells, and innate T cells, to provide insights into for clinical treatment.
Collapse
|
9
|
Pawlicki JM, Cookmeyer DL, Maseda D, Everett JK, Wei F, Kong H, Zhang Q, Wang HY, Tobias JW, Walter DM, Zullo KM, Javaid S, Watkins A, Wasik MA, Bushman FD, Riley JL. NPM-ALK-Induced Reprogramming of Mature TCR-Stimulated T Cells Results in Dedifferentiation and Malignant Transformation. Cancer Res 2021; 81:3241-3254. [PMID: 33619116 PMCID: PMC8260452 DOI: 10.1158/0008-5472.can-20-2297] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 12/28/2020] [Accepted: 02/19/2021] [Indexed: 12/22/2022]
Abstract
Fusion genes including NPM-ALK can promote T-cell transformation, but the signals required to drive a healthy T cell to become malignant remain undefined. In this study, we introduce NPM-ALK into primary human T cells and demonstrate induction of the epithelial-to-mesenchymal transition (EMT) program, attenuation of most T-cell effector programs, reemergence of an immature epigenomic profile, and dynamic regulation of c-Myc, E2F, and PI3K/mTOR signaling pathways early during transformation. A mutant of NPM-ALK failed to bind several signaling complexes including GRB2/SOS, SHC1, SHC4, and UBASH3B and was unable to transform T cells. Finally, T-cell receptor (TCR)-generated signals were required to achieve T-cell transformation, explaining how healthy individuals can harbor T cells with NPM-ALK translocations. These findings describe the fundamental mechanisms of NPM-ALK-mediated oncogenesis and may serve as a model to better understand factors that regulate tumor formation. SIGNIFICANCE: This investigation into malignant transformation of T cells uncovers a requirement for TCR triggering, elucidates integral signaling complexes nucleated by NPM-ALK, and delineates dynamic transcriptional changes as a T cell transforms.See related commentary by Spasevska and Myklebust, p. 3160.
Collapse
MESH Headings
- Apoptosis
- Cell Dedifferentiation
- Cell Proliferation
- Cell Transformation, Neoplastic/immunology
- Cell Transformation, Neoplastic/metabolism
- Cell Transformation, Neoplastic/pathology
- Cellular Reprogramming
- Humans
- Lymphoma, Large-Cell, Anaplastic/genetics
- Lymphoma, Large-Cell, Anaplastic/immunology
- Lymphoma, Large-Cell, Anaplastic/metabolism
- Lymphoma, Large-Cell, Anaplastic/pathology
- Phosphorylation
- Protein-Tyrosine Kinases/genetics
- Protein-Tyrosine Kinases/metabolism
- Receptors, Antigen, T-Cell/genetics
- Receptors, Antigen, T-Cell/metabolism
- T-Lymphocytes/immunology
- TOR Serine-Threonine Kinases/genetics
- TOR Serine-Threonine Kinases/metabolism
- Tumor Cells, Cultured
Collapse
Affiliation(s)
- Jan M Pawlicki
- Department of Microbiology, University of Pennsylvania, Philadelphia, Pennsylvania
- Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, Pennsylvania
| | - David L Cookmeyer
- Department of Microbiology, University of Pennsylvania, Philadelphia, Pennsylvania
- Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Damian Maseda
- Department of Microbiology, University of Pennsylvania, Philadelphia, Pennsylvania
- Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, Pennsylvania
| | - John K Everett
- Department of Microbiology, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Fang Wei
- Department of Microbiology, University of Pennsylvania, Philadelphia, Pennsylvania
- Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Hong Kong
- Department of Microbiology, University of Pennsylvania, Philadelphia, Pennsylvania
- Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Qian Zhang
- Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Hong Y Wang
- Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - John W Tobias
- Penn Genomic Analysis Core, University of Pennsylvania, Philadelphia, Pennsylvania
| | - David M Walter
- Department of Microbiology, University of Pennsylvania, Philadelphia, Pennsylvania
- Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Kelly M Zullo
- Department of Microbiology, University of Pennsylvania, Philadelphia, Pennsylvania
- Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Sarah Javaid
- Merck Research Laboratories, Boston, Massachusetts
| | | | - Mariusz A Wasik
- Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Frederic D Bushman
- Department of Microbiology, University of Pennsylvania, Philadelphia, Pennsylvania
| | - James L Riley
- Department of Microbiology, University of Pennsylvania, Philadelphia, Pennsylvania.
- Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
10
|
Hossein-Khannazer N, Zian Z, Bakkach J, Kamali AN, Hosseinzadeh R, Anka AU, Yazdani R, Azizi G. Features and roles of T helper 22 cells in immunological diseases and malignancies. Scand J Immunol 2021; 93:e13030. [PMID: 33576072 DOI: 10.1111/sji.13030] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Revised: 02/08/2021] [Accepted: 02/09/2021] [Indexed: 12/23/2022]
Abstract
T helper 22 (Th22) cell populations are a newly identified subset of CD4+ T cells that primarily mediate biological effects on the epithelial barrier through interleukin (IL)-22. Although, new studies showed that both Th22 and IL-22 are closely associated with the pathogenesis of inflammatory, autoimmune and allergic disease as well as malignancies. In this review, we aim to describe the development and characteristics of Th22 cells as well as their roles in the immunopathogenesis of immune-related disorders and cancer.
Collapse
Affiliation(s)
- Nikoo Hossein-Khannazer
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Zeineb Zian
- Biomedical Genomics and Oncogenetics Research Laboratory, Faculty of Sciences and Techniques of Tangier, Abdelmalek Essaadi University, Tetouan, Morocco
| | - Joaira Bakkach
- Biomedical Genomics and Oncogenetics Research Laboratory, Faculty of Sciences and Techniques of Tangier, Abdelmalek Essaadi University, Tetouan, Morocco
| | - Ali N Kamali
- CinnaGen Medical Biotechnology Research Center, Alborz University of Medical Sciences, Karaj, Iran
- CinnaGen Research and Production Co, Alborz, Iran
| | - Ramin Hosseinzadeh
- Department of Medical Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Abubakar Umar Anka
- Department of Medical Laboratory Science, College of Medical Sciences, Ahmadu Bello University, Zaria, Nigeria
| | - Reza Yazdani
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Gholamreza Azizi
- Non-Communicable Diseases Research Center, Alborz University of Medical Sciences, Karaj, Iran
| |
Collapse
|
11
|
Xuan X, Zhang L, Tian C, Wu T, Ye H, Cao J, Chen F, Liang Y, Yang H, Huang C. Interleukin-22 and connective tissue diseases: emerging role in pathogenesis and therapy. Cell Biosci 2021; 11:2. [PMID: 33407883 PMCID: PMC7788945 DOI: 10.1186/s13578-020-00504-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2020] [Accepted: 11/27/2020] [Indexed: 01/14/2023] Open
Abstract
Interleukin-22 (IL-22), a member of the IL-10 family of cytokines, is produced by a number of immune cells involved in the immune microenvironment of the body. IL-22 plays its pivotal roles by binding to the IL-22 receptor complex (IL-22R) and subsequently activating the IL-22R downstream signalling pathway. It has recently been reported that IL-22 also contributes to the pathogenesis of many connective tissue diseases (CTDs). In this review, we will discuss the role of IL-22 in several CTDs, such as system lupus erythematosus, rheumatoid arthritis, Sjögren’s syndrome, systemic sclerosis and dermatomyositis, suggesting that IL-22 may be a potential therapeutic target in CTDs.
Collapse
Affiliation(s)
- Xiuyun Xuan
- Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, China
| | - Lin Zhang
- Department of Gerontology, Jinan City People's Hospital, Jinan, 271199, Shandong, China
| | - Chunxia Tian
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Ting Wu
- Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, China
| | - Haihua Ye
- Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, China
| | - Juanmei Cao
- Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, China
| | - Fangqi Chen
- Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, China
| | - Yan Liang
- Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, China
| | - Huilan Yang
- Department of Dermatology, General Hospital of Southern Theatre Command, Guangzhou, 510000, China.
| | - Changzheng Huang
- Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, China.
| |
Collapse
|
12
|
Jiang R, Sun B. IL-22 Signaling in the Tumor Microenvironment. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1290:81-88. [PMID: 33559856 DOI: 10.1007/978-3-030-55617-4_5] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Interleukin (IL)-22 belongs to the IL-10 cytokine family which performs biological functions by binding to heterodimer receptors comprising a type 1 receptor chain (R1) and a type 2 receptor chain (R2). IL-22 is mainly derived from CD4+ helper T cells, CD8+ cytotoxic T cells, innate lymphocytes, and natural killer T cells. It can activate downstream signaling pathways such as signal transducer and activator of transcription (STAT)1/3/5, nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB), mitogen-activated protein kinase (MAPK), and phosphoinositide 3-kinase (PI3K)-protein kinase B (AKT)-mammalian target of rapamycin (mTOR) through these heterodimer receptors. Although IL-22 is produced by immune cells, its specific receptor IL-22R1 is selectively expressed in nonimmune cells, such as hepatocytes, colonic epithelial cells, and pancreatic epithelial cells (Jiang et al. Hepatology 54(3):900-9, 2011; Jiang et al. BMC Cancer 13:59, 2013; Curd et al. Clin Exp Immunol 168(2):192-9, 2012). Immune cells do not respond to IL-22 stimulation directly within tumors, reports from different groups have revealed that IL-22 can indirectly regulate the tumor microenvironment (TME). In the present chapter, we discuss the roles of IL-22 in malignant cells and immunocytes within the TME, meanwhile, the potential roles of IL-22 as a target for drug discovery will be discussed.
Collapse
Affiliation(s)
- Runqiu Jiang
- Department of Hepatobiliary Surgery, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, People's Republic of China
- Medical School of Nanjing University, Nanjing, People's Republic of China
| | - Beicheng Sun
- Department of Hepatobiliary Surgery, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, People's Republic of China.
| |
Collapse
|
13
|
Allegra A, Musolino C, Tonacci A, Pioggia G, Casciaro M, Gangemi S. Clinico-Biological Implications of Modified Levels of Cytokines in Chronic Lymphocytic Leukemia: A Possible Therapeutic Role. Cancers (Basel) 2020; 12:cancers12020524. [PMID: 32102441 PMCID: PMC7072434 DOI: 10.3390/cancers12020524] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Revised: 02/08/2020] [Accepted: 02/22/2020] [Indexed: 12/19/2022] Open
Abstract
B-cell chronic lymphocytic leukemia (B-CLL) is the main cause of mortality among hematologic diseases in Western nations. B-CLL is correlated with an intense alteration of the immune system. The altered functions of innate immune elements and adaptive immune factors are interconnected in B-CLL and are decisive for its onset, evolution, and therapeutic response. Modifications in the cytokine balance could support the growth of the leukemic clone via a modulation of cellular proliferation and apoptosis, as some cytokines have been reported to be able to affect the life of B-CLL cells in vivo. In this review, we will examine the role played by cytokines in the cellular dynamics of B-CLL patients, interpret the contradictions sometimes present in the literature regarding their action, and evaluate the possibility of manipulating their production in order to intervene in the natural history of the disease.
Collapse
Affiliation(s)
- Alessandro Allegra
- Division of Haematology, Department of Human Pathology in Adulthood and Childhood “Gaetano Barresi”, University of Messina, 98125 Messina, Italy; (A.A.); (C.M.)
| | - Caterina Musolino
- Division of Haematology, Department of Human Pathology in Adulthood and Childhood “Gaetano Barresi”, University of Messina, 98125 Messina, Italy; (A.A.); (C.M.)
| | - Alessandro Tonacci
- Clinical Physiology Institute, National Research Council of Italy (IFC-CNR), 56124 Pisa, Italy;
| | - Giovanni Pioggia
- Institute for Biomedical Research and Innovation (IRIB), National Research Council of Italy (CNR), 98164 Messina, Italy;
| | - Marco Casciaro
- Operative Unit of Allergy and Clinical Immunology, Department of Clinical and Experimental Medicine, University of Messina, 98125 Messina, Italy;
| | - Sebastiano Gangemi
- Operative Unit of Allergy and Clinical Immunology, Department of Clinical and Experimental Medicine, University of Messina, 98125 Messina, Italy;
- Correspondence:
| |
Collapse
|
14
|
Wang L, Lui VWY. Emerging Roles of ALK in Immunity and Insights for Immunotherapy. Cancers (Basel) 2020; 12:E426. [PMID: 32059449 PMCID: PMC7072244 DOI: 10.3390/cancers12020426] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 01/29/2020] [Accepted: 02/06/2020] [Indexed: 12/24/2022] Open
Abstract
Anaplastic lymphoma kinase (ALK) is mostly known for its oncogenic role in several human cancers. Recent evidences clearly indicate new roles of ALK and its genetic aberrations (e.g. gene rearrangements and mutations) in immune evasion, innate and cell-mediated immunity. New ALK-related immunotherapy approaches are demonstrating both preclinical and clinical promises. Here, we provide a timely review on the most updated laboratory and patient-related findings on ALK and immunity, which would grant us important insights for the development of novel ALK immunotherapies for ALK-altered cancers.
Collapse
Affiliation(s)
| | - Vivian Wai Yan Lui
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR 999077, China;
| |
Collapse
|
15
|
Rudloff I, Jardé T, Bachmann M, Elgass KD, Kerr G, Engel R, Richards E, Oliva K, Wilkins S, McMurrick PJ, Abud HE, Mühl H, Nold MF. Molecular signature of interleukin-22 in colon carcinoma cells and organoid models. Transl Res 2020; 216:1-22. [PMID: 31734267 DOI: 10.1016/j.trsl.2019.10.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Revised: 10/18/2019] [Accepted: 10/22/2019] [Indexed: 12/18/2022]
Abstract
Interleukin (IL)-22 activates STAT (signal transducer and activator of transcription) 3 and antiapoptotic and proproliferative pathways; but beyond this, the molecular mechanisms by which IL-22 promotes carcinogenesis are poorly understood. Characterizing the molecular signature of IL-22 in human DLD-1 colon carcinoma cells, we observed increased expression of 26 genes, including NNMT (nicotinamide N-methyltransferase, ≤10-fold) and CEA (carcinoembryonic antigen, ≤7-fold), both known to promote intestinal carcinogenesis. ERP27 (endoplasmic reticulum protein-27, function unknown, ≤5-fold) and the proinflammatory ICAM1 (intercellular adhesion molecule-1, ≤4-fold) were also increased. The effect on CEA was partly STAT3-mediated, as STAT3-silencing reduced IL-22-induced CEA by ≤56%. Silencing of CEA or NNMT inhibited IL-22-induced proliferation/migration of DLD-1, Caco-2, and SW480 colon carcinoma cells. To validate these results in primary tissues, we assessed IL-22-induced gene expression in organoids from human healthy colon and colon cancer patients, and from normal mouse small intestine and colon. Gene regulation by IL-22 was similar in DLD-1 cells and human and mouse healthy organoids. CEA was an exception with no induction by IL-22 in organoids, indicating the 3-dimensional organization of the tissue may produce signals absent in 2D cell culture. Importantly, augmentation of NNMT was 5-14-fold greater in human cancerous compared to normal organoids, supporting a role for NNMT in IL-22-mediated colon carcinogenesis. Thus, NNMT and CEA emerge as mediators of the tumor-promoting effects of IL-22 in the intestine. These data advance our understanding of the multifaceted role of IL-22 in the gut and suggest the IL-22 pathway may represent a therapeutic target in colon cancer.
Collapse
Affiliation(s)
- Ina Rudloff
- Department of Paediatrics, Monash University, Clayton, Melbourne, Australia; Ritchie Centre, Hudson Institute of Medical Research, Clayton, Melbourne, Australia; Pharmazentrum Frankfurt/ZAFES, University Hospital Goethe University Frankfurt am Main, Frankfurt am Main, Germany.
| | - Thierry Jardé
- Department of Anatomy and Developmental Biology, Monash University, Clayton, Melbourne, Australia; Stem Cells and Development Program, Monash Biomedicine Discovery Institute, Monash University, Clayton, Melbourne, Australia; Centre for Cancer Research, Hudson Institute of Medical Research, Clayton, Melbourne, Australia
| | - Malte Bachmann
- Pharmazentrum Frankfurt/ZAFES, University Hospital Goethe University Frankfurt am Main, Frankfurt am Main, Germany
| | - Kirstin D Elgass
- Monash Micro Imaging, Hudson Institute of Medical Research, Clayton, Melbourne, Australia
| | - Genevieve Kerr
- Department of Anatomy and Developmental Biology, Monash University, Clayton, Melbourne, Australia; Stem Cells and Development Program, Monash Biomedicine Discovery Institute, Monash University, Clayton, Melbourne, Australia
| | - Rebekah Engel
- Department of Anatomy and Developmental Biology, Monash University, Clayton, Melbourne, Australia; Stem Cells and Development Program, Monash Biomedicine Discovery Institute, Monash University, Clayton, Melbourne, Australia; Cabrini Monash University Department of Surgery, Cabrini Hospital, Malvern, Melbourne, Australia
| | - Elizabeth Richards
- Department of Anatomy and Developmental Biology, Monash University, Clayton, Melbourne, Australia; Stem Cells and Development Program, Monash Biomedicine Discovery Institute, Monash University, Clayton, Melbourne, Australia
| | - Karen Oliva
- Cabrini Monash University Department of Surgery, Cabrini Hospital, Malvern, Melbourne, Australia
| | - Simon Wilkins
- Cabrini Monash University Department of Surgery, Cabrini Hospital, Malvern, Melbourne, Australia; Department of Epidemiology and Preventive Medicine, School of Public Health and Preventive Medicine, Monash University, Melbourne, Australia
| | - Paul J McMurrick
- Cabrini Monash University Department of Surgery, Cabrini Hospital, Malvern, Melbourne, Australia
| | - Helen E Abud
- Department of Anatomy and Developmental Biology, Monash University, Clayton, Melbourne, Australia; Stem Cells and Development Program, Monash Biomedicine Discovery Institute, Monash University, Clayton, Melbourne, Australia
| | - Heiko Mühl
- Pharmazentrum Frankfurt/ZAFES, University Hospital Goethe University Frankfurt am Main, Frankfurt am Main, Germany
| | - Marcel F Nold
- Department of Paediatrics, Monash University, Clayton, Melbourne, Australia; Ritchie Centre, Hudson Institute of Medical Research, Clayton, Melbourne, Australia.
| |
Collapse
|
16
|
Kim EY, Choi B, Kim JE, Park SO, Kim SM, Chang EJ. Interleukin-22 Mediates the Chemotactic Migration of Breast Cancer Cells and Macrophage Infiltration of the Bone Microenvironment by Potentiating S1P/SIPR Signaling. Cells 2020; 9:E131. [PMID: 31935914 PMCID: PMC7017200 DOI: 10.3390/cells9010131] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Revised: 12/30/2019] [Accepted: 01/03/2020] [Indexed: 12/16/2022] Open
Abstract
The interleukin-22 (IL-22) signaling pathway is well known to be involved in the progression of various cancer types but its role in bone metastatic breast cancer remains unclear. We demonstrate using human GEO profiling that bone metastatic breast cancer displays elevated interleukin-22 receptor 1 (IL-22R1) and sphingosine-1-phosphate receptor 1 (S1PR1) expression. Importantly, IL-22 stimuli promoted the expression of IL-22R1 and S1PR1 in aggressive MDA-MB-231 breast cancer cells. IL-22 treatment also increased sphingosine-1-phosphate production in mesenchymal stem cells (MSCs) and induced the sphingosine-1-phosphate (S1P)-mediated chemotactic migration of MDA-MB-231 cells. This effect was inhibited by an S1P antagonist. In addition to the S1PR1 axis, IL-22 stimulated the expression of matrix metalloproteinase-9 (MMP-9), thereby promoting breast cancer cell invasion. Moreover, IL-22 induced IL22R1 and S1PR1 expression in macrophages, myeloid cell, and MCP1 expression in MSCs to facilitate macrophage infiltration. Immunohistochemistry indicated that IL-22R1 and S1PR1 are overexpressed in invasive malignant breast cancers and that this correlates with the MMP-9 levels. Collectively, our present results indicate a potential role of IL-22 in driving the metastasis of breast cancers into the bone microenvironment through the IL22R1-S1PR1 axis.
Collapse
Affiliation(s)
- Eun-Young Kim
- Department of Biomedical Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Korea; (E.-Y.K.); (B.C.); (J.-E.K.); (S.-O.P.); (S.-M.K.)
- Stem Cell Immunomodulation Research Center, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Korea
| | - Bongkun Choi
- Department of Biomedical Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Korea; (E.-Y.K.); (B.C.); (J.-E.K.); (S.-O.P.); (S.-M.K.)
- Stem Cell Immunomodulation Research Center, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Korea
| | - Ji-Eun Kim
- Department of Biomedical Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Korea; (E.-Y.K.); (B.C.); (J.-E.K.); (S.-O.P.); (S.-M.K.)
- Stem Cell Immunomodulation Research Center, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Korea
| | - Si-On Park
- Department of Biomedical Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Korea; (E.-Y.K.); (B.C.); (J.-E.K.); (S.-O.P.); (S.-M.K.)
- Stem Cell Immunomodulation Research Center, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Korea
| | - Sang-Min Kim
- Department of Biomedical Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Korea; (E.-Y.K.); (B.C.); (J.-E.K.); (S.-O.P.); (S.-M.K.)
| | - Eun-Ju Chang
- Department of Biomedical Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Korea; (E.-Y.K.); (B.C.); (J.-E.K.); (S.-O.P.); (S.-M.K.)
- Stem Cell Immunomodulation Research Center, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Korea
- Department of Biochemistry and Molecular Biology, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Korea
| |
Collapse
|
17
|
Kim EY, Noh HM, Choi B, Park JE, Kim JE, Jang Y, Lee HK, Chang EJ. Interleukin-22 Induces the Infiltration of Visceral Fat Tissue by a Discrete Subset of Duffy Antigen Receptor for Chemokine-Positive M2-Like Macrophages in Response to a High Fat Diet. Cells 2019; 8:E1587. [PMID: 31817755 PMCID: PMC6953014 DOI: 10.3390/cells8121587] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Revised: 12/03/2019] [Accepted: 12/05/2019] [Indexed: 12/14/2022] Open
Abstract
Interleukin-22 (IL-22) is a cytokine with important functions in host defense and inflammatory responses and has recently been suggested to play a role in immune-inflammatory system in the context of obesity and its metabolic consequences. The specific cellular targets and mechanisms of IL-22-mediated obesity are largely unknown however. We here identified a previously unknown subset of monocyte-derived Duffy antigen receptors for chemokines (DARC)+ macrophages in epididymal fat adipose tissue and found that they are preferentially recruited into the crown-like structures of adipose tissue in the mouse upon high fat diet-induced obesity. Importantly, DARC+ macrophages highly express the IL-22 receptor (IL-22Ra1). Exposure to recombinant IL-22 shifts macrophages to an alternative M2 polarization pathway and augments DARC expression via a STAT5b signaling axis. STAT5b directly binds to the DARC promoter and a STAT5 inhibitor abrogates the IL-22-mediated induction of DARC. These M2-like DARC+ subpopulations of monocytes/macrophages were elevated in obese db/db mice compared to WT lean mice. Furthermore, subsets of CD14+ and/or CD16+ monocytes/macrophages within human peripheral blood mononuclear cell populations express DARC and the prevalence of these subsets is enhanced by IL-22 stimuli. This suggested that IL-22 is a critical cytokine that promotes the infiltration of adipose tissue macrophages, that regulate inflammatory processes. Taken together, our present findings provide important insights into the molecular mechanism by which IL-22 signal modulates DARC expression in M2-like macrophages.
Collapse
Affiliation(s)
- Eun-Young Kim
- Department of Biomedical Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Korea; (E.-Y.K.); (B.C.); (J.-E.P.); (J.-E.K.); (Y.J.)
- Stem Cell Immunomodulation Research Center, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Korea
| | - Hye Mi Noh
- Department of Ophthalmology, Institute of Vision Research, Yonsei University College of Medicine, Seoul 06273, Korea;
| | - Bongkun Choi
- Department of Biomedical Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Korea; (E.-Y.K.); (B.C.); (J.-E.P.); (J.-E.K.); (Y.J.)
- Stem Cell Immunomodulation Research Center, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Korea
| | - Ji-Eun Park
- Department of Biomedical Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Korea; (E.-Y.K.); (B.C.); (J.-E.P.); (J.-E.K.); (Y.J.)
- Stem Cell Immunomodulation Research Center, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Korea
| | - Ji-Eun Kim
- Department of Biomedical Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Korea; (E.-Y.K.); (B.C.); (J.-E.P.); (J.-E.K.); (Y.J.)
- Stem Cell Immunomodulation Research Center, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Korea
| | - Youngsaeng Jang
- Department of Biomedical Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Korea; (E.-Y.K.); (B.C.); (J.-E.P.); (J.-E.K.); (Y.J.)
| | - Hyung Keun Lee
- Department of Ophthalmology, Institute of Vision Research, Yonsei University College of Medicine, Seoul 06273, Korea;
- Department of Pharmacy, Integrated Science and Engineering Division, Yonsei University, Incheon 21983, Korea
| | - Eun-Ju Chang
- Department of Biomedical Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Korea; (E.-Y.K.); (B.C.); (J.-E.P.); (J.-E.K.); (Y.J.)
- Stem Cell Immunomodulation Research Center, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Korea
| |
Collapse
|
18
|
Prutsch N, Gurnhofer E, Suske T, Liang HC, Schlederer M, Roos S, Wu LC, Simonitsch-Klupp I, Alvarez-Hernandez A, Kornauth C, Leone DA, Svinka J, Eferl R, Limberger T, Aufinger A, Shirsath N, Wolf P, Hielscher T, Sternberg C, Aberger F, Schmoellerl J, Stoiber D, Strobl B, Jäger U, Staber PB, Grebien F, Moriggl R, Müller M, Inghirami GG, Sanda T, Look AT, Turner SD, Kenner L, Merkel O. Dependency on the TYK2/STAT1/MCL1 axis in anaplastic large cell lymphoma. Leukemia 2019; 33:696-709. [PMID: 30131584 PMCID: PMC8076043 DOI: 10.1038/s41375-018-0239-1] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2018] [Revised: 07/02/2018] [Accepted: 07/27/2018] [Indexed: 12/11/2022]
Abstract
TYK2 is a member of the JAK family of tyrosine kinases that is involved in chromosomal translocation-induced fusion proteins found in anaplastic large cell lymphomas (ALCL) that lack rearrangements activating the anaplastic lymphoma kinase (ALK). Here we demonstrate that TYK2 is highly expressed in all cases of human ALCL, and that in a mouse model of NPM-ALK-induced lymphoma, genetic disruption of Tyk2 delays the onset of tumors and prolongs survival of the mice. Lymphomas in this model lacking Tyk2 have reduced STAT1 and STAT3 phosphorylation and reduced expression of Mcl1, a pro-survival member of the BCL2 family. These findings in mice are mirrored in human ALCL cell lines, in which TYK2 is activated by autocrine production of IL-10 and IL-22 and by interaction with specific receptors expressed by the cells. Activated TYK2 leads to STAT1 and STAT3 phosphorylation, activated expression of MCL1 and aberrant ALCL cell survival. Moreover, TYK2 inhibitors are able to induce apoptosis in ALCL cells, regardless of the presence or absence of an ALK-fusion. Thus, TYK2 is a dependency that is required for ALCL cell survival through activation of MCL1 expression. TYK2 represents an attractive drug target due to its essential enzymatic domain, and TYK2-specific inhibitors show promise as novel targeted inhibitors for ALCL.
Collapse
Affiliation(s)
- Nicole Prutsch
- Clinical Institute of Pathology, Department for Experimental and Laboratory Animal Pathology, Medical University of Vienna, Vienna, Austria
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, USA
- Unit of Laboratory Animal Pathology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Elisabeth Gurnhofer
- Clinical Institute of Pathology, Department for Experimental and Laboratory Animal Pathology, Medical University of Vienna, Vienna, Austria
| | - Tobias Suske
- Clinical Institute of Pathology, Department for Experimental and Laboratory Animal Pathology, Medical University of Vienna, Vienna, Austria
| | - Huan Chang Liang
- Clinical Institute of Pathology, Department for Experimental and Laboratory Animal Pathology, Medical University of Vienna, Vienna, Austria
| | - Michaela Schlederer
- Clinical Institute of Pathology, Department for Experimental and Laboratory Animal Pathology, Medical University of Vienna, Vienna, Austria
| | - Simone Roos
- Unit of Laboratory Animal Pathology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Lawren C Wu
- Department of Oncology, Amgen Discovery Research, South San Francisco, CA, 94080, USA
| | | | | | - Christoph Kornauth
- Clinical Institute of Pathology, Medical University of Vienna, Vienna, Austria
| | - Dario A Leone
- Clinical Institute of Pathology, Department for Experimental and Laboratory Animal Pathology, Medical University of Vienna, Vienna, Austria
| | - Jasmin Svinka
- Institute of Cancer Research, Medical University of Vienna & Comprehensive Cancer Center (CCC), Vienna, Austria
| | - Robert Eferl
- Institute of Cancer Research, Medical University of Vienna & Comprehensive Cancer Center (CCC), Vienna, Austria
| | - Tanja Limberger
- Clinical Institute of Pathology, Department for Experimental and Laboratory Animal Pathology, Medical University of Vienna, Vienna, Austria
| | - Astrid Aufinger
- Clinical Institute of Pathology, Department for Experimental and Laboratory Animal Pathology, Medical University of Vienna, Vienna, Austria
| | - Nitesh Shirsath
- Department of Dermatology and Venereology, Medical University of Graz, Graz, Austria
| | - Peter Wolf
- Department of Dermatology and Venereology, Medical University of Graz, Graz, Austria
| | - Thomas Hielscher
- Division of Biostatistics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Christina Sternberg
- Unit of Laboratory Animal Pathology, University of Veterinary Medicine Vienna, Vienna, Austria
- Department of Molecular Biology, Cancer Cluster Salzburg, Faculty of Natural Sciences, Paris Lodron University, Salzburg, Austria
- Department of Biochemistry, Christian-Albrechts-University Kiel, Kiel, Germany
| | - Fritz Aberger
- Department of Molecular Biology, Cancer Cluster Salzburg, Faculty of Natural Sciences, Paris Lodron University, Salzburg, Austria
| | | | - Dagmar Stoiber
- Ludwig Boltzmann Institute for Cancer Research (LBI-CR), Vienna, Austria
- Institute of Pharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Birgit Strobl
- Institute of Animal Breeding and Genetics, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Ulrich Jäger
- Department of Medicine I, Clinical Division of Hematology and Hemostaseology and Comprehensive Cancer Center (CCC), Medical University of Vienna, Vienna, Austria
| | - Philipp B Staber
- Department of Medicine I, Clinical Division of Hematology and Hemostaseology and Comprehensive Cancer Center (CCC), Medical University of Vienna, Vienna, Austria
| | - Florian Grebien
- Ludwig Boltzmann Institute for Cancer Research (LBI-CR), Vienna, Austria
| | - Richard Moriggl
- Ludwig Boltzmann Institute for Cancer Research (LBI-CR), Vienna, Austria
- Institute of Animal Breeding and Genetics, University of Veterinary Medicine Vienna, Vienna, Austria
- Medical University of Vienna, Vienna, Austria
| | - Mathias Müller
- Institute of Animal Breeding and Genetics, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Giorgio G Inghirami
- Pathology and Laboratory Medicine, Weill Cornell Medical College, New York, NYC, USA
- European Research Initiative for ALK related malignancies (www.erialcl.net), Vienna, Austria
| | - Takaomi Sanda
- Cancer Science Institute of Singapore, National University of Singapore, 117599, Singapore, Singapore
| | - A Thomas Look
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, USA
| | - Suzanne D Turner
- European Research Initiative for ALK related malignancies (www.erialcl.net), Vienna, Austria
- Division of Cellular and Molecular Pathology, Department of Pathology, University of Cambridge, Cambridge, UK
| | - Lukas Kenner
- Clinical Institute of Pathology, Department for Experimental and Laboratory Animal Pathology, Medical University of Vienna, Vienna, Austria.
- Unit of Laboratory Animal Pathology, University of Veterinary Medicine Vienna, Vienna, Austria.
- Ludwig Boltzmann Institute for Cancer Research (LBI-CR), Vienna, Austria.
- European Research Initiative for ALK related malignancies (www.erialcl.net), Vienna, Austria.
- CBMed Core Lab2, Medical University of Vienna, Vienna, Austria.
| | - Olaf Merkel
- Clinical Institute of Pathology, Department for Experimental and Laboratory Animal Pathology, Medical University of Vienna, Vienna, Austria.
- European Research Initiative for ALK related malignancies (www.erialcl.net), Vienna, Austria.
| |
Collapse
|
19
|
Schleussner N, Merkel O, Costanza M, Liang HC, Hummel F, Romagnani C, Durek P, Anagnostopoulos I, Hummel M, Jöhrens K, Niedobitek A, Griffin PR, Piva R, Sczakiel HL, Woessmann W, Damm-Welk C, Hinze C, Stoiber D, Gillissen B, Turner SD, Kaergel E, von Hoff L, Grau M, Lenz G, Dörken B, Scheidereit C, Kenner L, Janz M, Mathas S. The AP-1-BATF and -BATF3 module is essential for growth, survival and TH17/ILC3 skewing of anaplastic large cell lymphoma. Leukemia 2018; 32:1994-2007. [PMID: 29588546 PMCID: PMC6127090 DOI: 10.1038/s41375-018-0045-9] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2017] [Revised: 12/20/2017] [Accepted: 01/08/2018] [Indexed: 01/26/2023]
Abstract
Transcription factor AP-1 is constitutively activated and IRF4 drives growth and survival in ALK+ and ALK- anaplastic large cell lymphoma (ALCL). Here we demonstrate high-level BATF and BATF3 expression in ALCL. Both BATFs bind classical AP-1 motifs and interact with in ALCL deregulated AP-1 factors. Together with IRF4, they co-occupy AP-1-IRF composite elements, differentiating ALCL from non-ALCL. Gene-specific inactivation of BATFs, or global AP-1 inhibition results in ALCL growth retardation and/or cell death in vitro and in vivo. Furthermore, the AP-1-BATF module establishes TH17/group 3 innate lymphoid cells (ILC3)-associated gene expression in ALCL cells, including marker genes such as AHR, IL17F, IL22, IL26, IL23R and RORγt. Elevated IL-17A and IL-17F levels were detected in a subset of children and adolescents with ALK+ ALCL. Furthermore, a comprehensive analysis of primary lymphoma data confirms TH17-, and in particular ILC3-skewing in ALCL compared with PTCL. Finally, pharmacological inhibition of RORC as single treatment leads to cell death in ALCL cell lines and, in combination with the ALK inhibitor crizotinib, enforces death induction in ALK+ ALCL. Our data highlight the crucial role of AP-1/BATFs in ALCL and lead to the concept that some ALCL might originate from ILC3.
Collapse
Affiliation(s)
- Nikolai Schleussner
- Max-Delbrück-Center for Molecular Medicine, 13125, Berlin, Germany
- Hematology, Oncology, and Tumor Immunology, Charité-Universitätsmedizin Berlin, 12200, Berlin, Germany
| | - Olaf Merkel
- Institute of Clinical Pathology, Medical University of Vienna, Vienna, Austria
- European Research Initiative on ALK-Related Malignancies (ERIA), Cambridge, UK
| | - Mariantonia Costanza
- Max-Delbrück-Center for Molecular Medicine, 13125, Berlin, Germany
- Hematology, Oncology, and Tumor Immunology, Charité-Universitätsmedizin Berlin, 12200, Berlin, Germany
- European Research Initiative on ALK-Related Malignancies (ERIA), Cambridge, UK
| | - Huan-Chang Liang
- Institute of Clinical Pathology, Medical University of Vienna, Vienna, Austria
- European Research Initiative on ALK-Related Malignancies (ERIA), Cambridge, UK
| | - Franziska Hummel
- Max-Delbrück-Center for Molecular Medicine, 13125, Berlin, Germany
- Hematology, Oncology, and Tumor Immunology, Charité-Universitätsmedizin Berlin, 12200, Berlin, Germany
| | - Chiara Romagnani
- German Rheumatism Research Centre, German Rheumatism Research Centre (DRFZ), A Leibniz Institute, 10117, Berlin, Germany
- Medical Department I, Charité-Universitätsmedizin Berlin, 12200, Berlin, Germany
| | - Pawel Durek
- German Rheumatism Research Centre, German Rheumatism Research Centre (DRFZ), A Leibniz Institute, 10117, Berlin, Germany
| | | | - Michael Hummel
- Institute of Pathology, Charité-Universitätsmedizin Berlin, 10117, Berlin, Germany
- German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), 69120, Heidelberg, Germany
| | - Korinna Jöhrens
- Institute of Pathology, Charité-Universitätsmedizin Berlin, 10117, Berlin, Germany
| | - Antonia Niedobitek
- Max-Delbrück-Center for Molecular Medicine, 13125, Berlin, Germany
- Hematology, Oncology, and Tumor Immunology, Charité-Universitätsmedizin Berlin, 12200, Berlin, Germany
| | | | - Roberto Piva
- Department of Molecular Biotechnology and Health Sciences, Center for Experimental Research and Medical Studies, University of Torino, Torino, Italy
| | - Henrike L Sczakiel
- Max-Delbrück-Center for Molecular Medicine, 13125, Berlin, Germany
- Hematology, Oncology, and Tumor Immunology, Charité-Universitätsmedizin Berlin, 12200, Berlin, Germany
| | - Wilhelm Woessmann
- European Research Initiative on ALK-Related Malignancies (ERIA), Cambridge, UK
- NHL-BFM Study Centre and Department of Paediatric Haematology and Oncology, Justus-Liebig-University, Giessen, Germany
| | - Christine Damm-Welk
- European Research Initiative on ALK-Related Malignancies (ERIA), Cambridge, UK
- NHL-BFM Study Centre and Department of Paediatric Haematology and Oncology, Justus-Liebig-University, Giessen, Germany
| | - Christian Hinze
- Max-Delbrück-Center for Molecular Medicine, 13125, Berlin, Germany
- Department of Nephrology, Charité-Universitätsmedizin Berlin, 12200, Berlin, Germany
| | - Dagmar Stoiber
- Institute of Pharmacology, Center for Physiology and Pharmacology, Medical University Vienna, Vienna, Austria
- Ludwig Boltzmann Institute for Cancer Research (LBI-CR), Vienna, Austria
| | - Bernd Gillissen
- Hematology, Oncology, and Tumor Immunology, Charité-Universitätsmedizin Berlin, 12200, Berlin, Germany
| | - Suzanne D Turner
- European Research Initiative on ALK-Related Malignancies (ERIA), Cambridge, UK
- Department of Pathology, University of Cambridge, Cambridge, CB21QP, UK
| | - Eva Kaergel
- Max-Delbrück-Center for Molecular Medicine, 13125, Berlin, Germany
| | - Linda von Hoff
- Max-Delbrück-Center for Molecular Medicine, 13125, Berlin, Germany
| | - Michael Grau
- Department of Medicine A, Albert-Schweitzer-Campus 1, University Hospital Münster, 48149, Münster, Germany
- Cluster of Excellence EXC 1003, Cells in Motion, 48149, Münster, Germany
| | - Georg Lenz
- Department of Medicine A, Albert-Schweitzer-Campus 1, University Hospital Münster, 48149, Münster, Germany
- Cluster of Excellence EXC 1003, Cells in Motion, 48149, Münster, Germany
| | - Bernd Dörken
- Max-Delbrück-Center for Molecular Medicine, 13125, Berlin, Germany
- Hematology, Oncology, and Tumor Immunology, Charité-Universitätsmedizin Berlin, 12200, Berlin, Germany
- German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), 69120, Heidelberg, Germany
| | | | - Lukas Kenner
- Institute of Clinical Pathology, Medical University of Vienna, Vienna, Austria.
- European Research Initiative on ALK-Related Malignancies (ERIA), Cambridge, UK.
- Ludwig Boltzmann Institute for Cancer Research (LBI-CR), Vienna, Austria.
- University of Veterinary Medicine, Vienna, Austria.
- CBmed, Center for Biomarker Research in Medicine, 8010, Graz, Austria.
| | - Martin Janz
- Max-Delbrück-Center for Molecular Medicine, 13125, Berlin, Germany
- Hematology, Oncology, and Tumor Immunology, Charité-Universitätsmedizin Berlin, 12200, Berlin, Germany
- Experimental and Clinical Research Center, a joint cooperation of Max-Delbrück-Center for Molecular Medicine and Charité - Universitätsmedizin Berlin, 13125, Berlin, Germany
| | - Stephan Mathas
- Max-Delbrück-Center for Molecular Medicine, 13125, Berlin, Germany.
- Hematology, Oncology, and Tumor Immunology, Charité-Universitätsmedizin Berlin, 12200, Berlin, Germany.
- European Research Initiative on ALK-Related Malignancies (ERIA), Cambridge, UK.
- German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), 69120, Heidelberg, Germany.
- Experimental and Clinical Research Center, a joint cooperation of Max-Delbrück-Center for Molecular Medicine and Charité - Universitätsmedizin Berlin, 13125, Berlin, Germany.
| |
Collapse
|
20
|
Abstract
IL-22 is a critical cytokine in modulating tissue responses during inflammation. IL-22 is upregulated in many chronic inflammatory diseases, making IL-22 biology a potentially rewarding therapeutic target. However, this is complicated by the dual-natured role of IL-22 in inflammation, as the cytokine can be protective or inflammatory depending on the disease model. Although scientific interest in IL-22 has increased considerably in the past 10 y, there is still much we do not know about the environmental, cellular, and molecular factors that regulate the production and function of this cytokine. A better understanding of IL-22 biology will allow us to develop new or improved therapeutics for treating chronic inflammatory diseases. In this article, I will highlight some of the outstanding questions in IL-22 biology.
Collapse
Affiliation(s)
- Lauren A Zenewicz
- Department of Microbiology and Immunology, College of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104
| |
Collapse
|
21
|
Yang JM, Jang JY, Jeon YK, Paik JH. Clinicopathologic implication of microRNA-197 in diffuse large B cell lymphoma. J Transl Med 2018; 16:162. [PMID: 29890998 PMCID: PMC5996506 DOI: 10.1186/s12967-018-1537-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Accepted: 06/06/2018] [Indexed: 02/08/2023] Open
Abstract
Background Diffuse large B cell lymphoma (DLBCL) contains heterogeneous subtypes with various molecular dysregulation at the gene, protein and microRNA levels. Compared with the GCB subtype, the non-germinal center B-like (non-GCB)/activated B cell-like (ABC) subtype exhibits frequent progression despite standard immunochemotherapy. We aimed to investigate the effects of miR-197 on the progression and chemosensitivity of DLBCL with respect to the GCB and non-GCB/ABC subtypes. Methods To screen distinctively expressed microRNAs, microRNA expression patterns were analyzed in 10 DLBCL cases by microarray chip assays. Using quantitative real-time polymerase chain reaction (qRT-PCR), associations between miR-197 expression levels and clinicopathologic variables were investigated in 51 DLBCL tissue samples. The effects of miR-197 on doxorubicin chemosensitivity were investigated using the OCI-Ly1 and SUDHL9 cell lines. Results MicroRNA expression profiling by hierarchical clustering revealed that miR-197 was one of the distinctively expressed microRNAs in DLBCL. Quantitative analysis using qRT-PCR revealed that miR-197 levels were not correlated with clinicopathologic variables, including the international prognostic index, but low miR-197 levels were significantly associated with lymphoma progression defined by refractoriness, relapse or death in the rituximab plus cyclophosphamide, doxorubicin, vincristine, and prednisone (R-CHOP)-treated subgroup (n = 43; p = 0.004). Among the three molecular groups, i.e., the GCB, non-GCB/miR-197low and non-GCB/miR-197high groups, progression was most frequently observed in the non-GCB/miR-197low group in the full cohort (p = 0.013) and the R-CHOP cohort (p = 0.008). In survival analysis, low miR-197 levels were independently predictive of shorter progression-free survival in the R-CHOP cohort (p = 0.031; HR = 27.9) and the non-GCB subgroup (p = 0.037; HR = 21.5) but not in the GCB subgroup. Using SUDHL9 (ABC type) and OCI-Ly1 (GCB type) cells, the effects of doxorubicin on reducing cell viability were enhanced by miR-197 transfection. In apoptosis assays, miR-197 transfection enhanced doxorubicin-induced apoptosis in SUDHL9 cells but not in OCI-Ly1 cells, suggesting a chemosensitizing effect of miR-197 in ABC DLBCL. Conclusions These results suggest the role of miR-197 as a biomarker with potential therapeutic implications. Electronic supplementary material The online version of this article (10.1186/s12967-018-1537-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Jeong Mi Yang
- Department of Pathology, Seoul National University Bundang Hospital, 300 Gumi-dong, Bundang-gu, Seongnam, Gyeonggi, South Korea
| | | | - Yoon Kyung Jeon
- Department of Pathology, Seoul National University Hospital, Seoul, South Korea.,Department of Pathology, Seoul National University College of Medicine, Seoul, South Korea
| | - Jin Ho Paik
- Department of Pathology, Seoul National University Bundang Hospital, 300 Gumi-dong, Bundang-gu, Seongnam, Gyeonggi, South Korea. .,Department of Pathology, Seoul National University College of Medicine, Seoul, South Korea.
| |
Collapse
|
22
|
Novel insights into the pathogenesis of T-cell lymphomas. Blood 2018; 131:2320-2330. [DOI: 10.1182/blood-2017-11-764357] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Accepted: 04/09/2018] [Indexed: 12/11/2022] Open
Abstract
Abstract
T-cell lymphomas are a heterogeneous group of rare malignancies with overlapping clinical, immunologic, and histologic features. Recent advances in our understanding of T-cell differentiation based on gene expression profiling, next-generation sequencing, and transgenic mouse modeling studies have better elucidated the pathogenetic mechanisms underlying the diverse biology of T-cell lymphomas. These studies show that although genetic alterations in epigenetic modifiers are implicated in all subtypes of T-cell lymphomas, specific subtypes demonstrate enrichment for particular recurrent alterations targeting specific genes. In this regard, RHOA and TET2 alterations are prevalent in nodal T-cell lymphomas, particularly angioimmunoblastic T-cell lymphomas, peripheral T-cell lymphomas (PTCLs) not otherwise specified, and nodal PTCLs with T-follicular helper phenotype. JAK-STAT signaling pathways are mutationally activated in many extranodal T-cell lymphomas, such as natural killer/T-cell and hepatosplenic T-cell lymphomas. The functional significance of many of these genetic alterations is becoming better understood. Altogether these advances will continue to refine diagnostic criteria, improve prognostication, and identify novel therapeutic targets, resulting in improved outcomes for patient with T-cell lymphomas.
Collapse
|
23
|
Hernandez P, Gronke K, Diefenbach A. A catch-22: Interleukin-22 and cancer. Eur J Immunol 2018; 48:15-31. [PMID: 29178520 DOI: 10.1002/eji.201747183] [Citation(s) in RCA: 79] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2017] [Revised: 10/31/2017] [Accepted: 11/23/2017] [Indexed: 12/17/2022]
Abstract
Barrier surfaces of multicellular organisms are in constant contact with the environment and infractions to the integrity of epithelial surfaces is likely a frequent event. Interestingly, components of the immune system, that can be activated by environmental compounds such as the microbiota or nutrients, are interspersed among epithelial cells or directly underlie the epithelium. It is now appreciated that immune cells continuously receive and integrate signals from the environment. Curiously, such continuous reception of stimulation does not normally trigger an inflammatory response but mediators produced by immune cells in response to such signals seem to rather promote barrier integrity and repair. The molecular mediators involved in this process are poorly understood. In recent years, the cytokine interleukin-22, produced mainly by group 3 innate lymphoid cells (ILCs), has been studied as a paradigm for how immune cells can control various aspects of epithelial cell function because expression of its receptor is restricted to non-hematopoietic cells. We will summarize here the diverse roles of IL-22 for the malignant transformation of epithelial cells, for tumor growth, wound healing and tissue repair. Furthermore, we will discuss IL-22 as a potential therapeutic target.
Collapse
Affiliation(s)
- Pedro Hernandez
- Institute of Microbiology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
- Macrophages et Développement de l'Immunité, Institut Pasteur, Paris Cedex 15, France
- Max-Planck-Institute for Immunobiology und Epigenetics, Freiburg, Germany
| | - Konrad Gronke
- Institute of Microbiology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
- Max-Planck-Institute for Immunobiology und Epigenetics, Freiburg, Germany
- Institute of Medical Microbiology and Hygiene and Research Centre Immunology, University of Mainz Medical Centre, Mainz, Germany
| | - Andreas Diefenbach
- Institute of Microbiology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| |
Collapse
|
24
|
Ikeda S, Kitadate A, Ito M, Abe F, Nara M, Watanabe A, Takahashi N, Miyagaki T, Sugaya M, Tagawa H. Disruption of CCL20-CCR6 interaction inhibits metastasis of advanced cutaneous T-cell lymphoma. Oncotarget 2017; 7:13563-74. [PMID: 26789110 PMCID: PMC4924661 DOI: 10.18632/oncotarget.6916] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2015] [Accepted: 12/31/2015] [Indexed: 12/19/2022] Open
Abstract
We recently demonstrated that upregulation of a chemokine receptor CCR6 and its ligand CCL20 led to metastasis of advanced cutaneous T-cell lymphoma (CTCL) cells, suggesting the involvement of CCL20-CCR6 interaction in initiating CTCL cell metastasis. In this study, we determined whether this interaction is functional in metastatic CTCL cells. We first demonstrated increased STAT3 expression during the progression of primary CTCL. STAT3 was spontaneously activated and mediated the transcription of CCL20 in CTCL cell lines. Next, to determine whether the transient knockdown of STAT3, CCL20, or CCR6 or treatment with neutralizing antibody against CCL20 (neutralizing CCL20 antibody) could reduce the migration ability of CTCL cells, we conducted an in vitro migration assay. All treatments reduced the nutrition-dependent migration activity of CTCL cells. Notably, treatment with neutralizing CCL20 antibody reduced the migration ability of the cells without decreasing the expression of CCL20 and CCR6. This demonstrated that the CCL20-CCR6 interaction is actually functional in metastatic CTCL cells. Finally, to examine the in vivo effect of neutralizing CCL20 antibody, we used NOD/Shi-scid IL-2γnul mice inoculated with CTCL cells. These mice were expected to die due to metastasis of CTCL cells into multiple organs. However, administration of neutralizing CCL20 antibody significantly prolonged the survival of the xenografted mice. These findings suggested that automatic activation of the STAT3/CCL20/CCR6 cascade was involved in CTCL lymphomagenesis and that disruption of CCL20-CCR6 interaction could be a key therapeutic strategy against advanced CTCL.
Collapse
Affiliation(s)
- Sho Ikeda
- Department of Hematology, Nephrology, and Rheumatology, Akita University Graduate School of Medicine, Akita, Japan
| | - Akihiro Kitadate
- Department of Hematology, Nephrology, and Rheumatology, Akita University Graduate School of Medicine, Akita, Japan
| | - Mitsugu Ito
- Department of Hematology, Nephrology, and Rheumatology, Akita University Graduate School of Medicine, Akita, Japan
| | - Fumito Abe
- Department of Hematology, Nephrology, and Rheumatology, Akita University Graduate School of Medicine, Akita, Japan
| | - Miho Nara
- Department of Hematology, Nephrology, and Rheumatology, Akita University Graduate School of Medicine, Akita, Japan
| | - Atsushi Watanabe
- Department of Hematology, Nephrology, and Rheumatology, Akita University Graduate School of Medicine, Akita, Japan
| | - Naoto Takahashi
- Department of Hematology, Nephrology, and Rheumatology, Akita University Graduate School of Medicine, Akita, Japan
| | | | - Makoto Sugaya
- Department of Dermatology, University of Tokyo, Tokyo, Japan
| | - Hiroyuki Tagawa
- Department of Hematology, Nephrology, and Rheumatology, Akita University Graduate School of Medicine, Akita, Japan
| |
Collapse
|
25
|
Knörr F, Damm-Welk C, Ruf S, Singh VK, Zimmermann M, Reiter A, Woessmann W. Blood cytokine concentrations in pediatric patients with anaplastic lymphoma kinase-positive anaplastic large cell lymphoma. Haematologica 2017; 103:477-485. [PMID: 29242300 PMCID: PMC5830391 DOI: 10.3324/haematol.2017.177972] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2017] [Accepted: 12/07/2017] [Indexed: 01/16/2023] Open
Abstract
Patients with anaplastic lymphoma kinase-positive anaplastic large cell lymphoma often present with B-symptoms or hemophagocytosis and generate an anti-tumor immune response. Specific serum cytokine levels or profiles may reflect the tumor burden, non-specific immune stimulation by the tumor or differences in the strength of the patients’ anti-lymphoma immunity. We systematically correlated pretreatment concentrations of 25 cytokines with clinical and biological characteristics in a well-characterized cohort of 119 uniformly treated pediatric patients with anaplastic large cell lymphoma. Fifteen patients with anaplastic large cell lymphoma in remission and 11 patients with low-stage B-cell lymphoma served as controls. Concentrations of interleukin-9, interleukin-10, interleukin-17a, hepatocyte growth factor, soluble interleukin-2 receptor, and soluble CD30 were significantly higher in initial sera of patients than in the sera of subjects from both control groups, indicating an anaplastic large cell lymphoma-type cytokine signature. The levels of interleukin-6, interferon-γ, interferon γ-induced protein, and soluble interleukin-2 receptor correlated with the stage, initial general condition, minimal disseminated disease, anaplastic lymphoma kinase-antibody titers, and the risk of relapse among patients with anaplastic lymphoma kinase-positive anaplastic large cell lymphoma. Only interleukin-6 showed an independent prognostic value in multivariate analyses. Pretreatment cytokine profiles in patients with anaplastic large cell lymphoma reflect a tumor signature as well as tumor burden and also differences in the strength of the patients’ immune response.
Collapse
Affiliation(s)
- Fabian Knörr
- NHL-BFM Study Center, Department of Pediatric Hematology and Oncology, Justus-Liebig University, Giessen
| | - Christine Damm-Welk
- NHL-BFM Study Center, Department of Pediatric Hematology and Oncology, Justus-Liebig University, Giessen
| | - Stephanie Ruf
- NHL-BFM Study Center, Department of Pediatric Hematology and Oncology, Justus-Liebig University, Giessen
| | - Vijay Kumar Singh
- NHL-BFM Study Center, Department of Pediatric Hematology and Oncology, Justus-Liebig University, Giessen
| | - Martin Zimmermann
- Department of Pediatric Hematology and Oncology, Children's Hospital, Hannover Medical School, Germany
| | - Alfred Reiter
- NHL-BFM Study Center, Department of Pediatric Hematology and Oncology, Justus-Liebig University, Giessen
| | - Wilhelm Woessmann
- NHL-BFM Study Center, Department of Pediatric Hematology and Oncology, Justus-Liebig University, Giessen
| |
Collapse
|
26
|
Victor AR, Nalin AP, Dong W, McClory S, Wei M, Mao C, Kladney RD, Youssef Y, Chan WK, Briercheck EL, Hughes T, Scoville SD, Pitarresi JR, Chen C, Manz S, Wu LC, Zhang J, Ostrowski MC, Freud AG, Leone GW, Caligiuri MA, Yu J. IL-18 Drives ILC3 Proliferation and Promotes IL-22 Production via NF-κB. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2017; 199:2333-2342. [PMID: 28842466 PMCID: PMC5624342 DOI: 10.4049/jimmunol.1601554] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Accepted: 07/27/2017] [Indexed: 12/13/2022]
Abstract
Group 3 innate lymphoid cells (ILC3s) are important regulators of the immune system, maintaining homeostasis in the presence of commensal bacteria, but activating immune defenses in response to microbial pathogens. ILC3s are a robust source of IL-22, a cytokine critical for stimulating the antimicrobial response. We sought to identify cytokines that can promote proliferation and induce or maintain IL-22 production by ILC3s and determine a molecular mechanism for this process. We identified IL-18 as a cytokine that cooperates with an ILC3 survival factor, IL-15, to induce proliferation of human ILC3s, as well as induce and maintain IL-22 production. To determine a mechanism of action, we examined the NF-κB pathway, which is activated by IL-18 signaling. We found that the NF-κB complex signaling component, p65, binds to the proximal region of the IL22 promoter and promotes transcriptional activity. Finally, we observed that CD11c+ dendritic cells expressing IL-18 are found in close proximity to ILC3s in human tonsils in situ. Therefore, we identify a new mechanism by which human ILC3s proliferate and produce IL-22, and identify NF-κB as a potential therapeutic target to be considered in pathologic states characterized by overproduction of IL-18 and/or IL-22.
Collapse
Affiliation(s)
- Aaron R Victor
- Medical Scientist Training Program, Ohio State University, Columbus, OH 43210
| | - Ansel P Nalin
- Medical Scientist Training Program, Ohio State University, Columbus, OH 43210
| | - Wenjuan Dong
- The James Cancer Hospital and Solove Research Institute, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210
| | - Susan McClory
- Medical Scientist Training Program, Ohio State University, Columbus, OH 43210
- The James Cancer Hospital and Solove Research Institute, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210
| | - Min Wei
- The James Cancer Hospital and Solove Research Institute, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210
| | - Charlene Mao
- The James Cancer Hospital and Solove Research Institute, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210
| | - Raleigh D Kladney
- The James Cancer Hospital and Solove Research Institute, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210
- Department of Molecular Virology, Immunology, and Medical Genetics, The Ohio State University, Columbus, OH 43210
- Department of Molecular Genetics, College of Biological Sciences, The Ohio State University, Columbus, OH 43210
| | - Youssef Youssef
- Department of Pathology, The Ohio State University, Columbus, OH 43210
| | - Wing Keung Chan
- The James Cancer Hospital and Solove Research Institute, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210
| | - Edward L Briercheck
- Medical Scientist Training Program, Ohio State University, Columbus, OH 43210
- The James Cancer Hospital and Solove Research Institute, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210
| | - Tiffany Hughes
- The James Cancer Hospital and Solove Research Institute, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210
| | - Steven D Scoville
- Medical Scientist Training Program, Ohio State University, Columbus, OH 43210
- The James Cancer Hospital and Solove Research Institute, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210
| | - Jason R Pitarresi
- The James Cancer Hospital and Solove Research Institute, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210
- Department of Molecular Virology, Immunology, and Medical Genetics, The Ohio State University, Columbus, OH 43210
| | - Charlie Chen
- The James Cancer Hospital and Solove Research Institute, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210
| | - Sarah Manz
- The James Cancer Hospital and Solove Research Institute, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210
| | - Lai-Chu Wu
- The James Cancer Hospital and Solove Research Institute, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210
| | - Jianying Zhang
- Center for Biostatistics, Department of Bioinformatics, The Ohio State University, Columbus, OH 43210; and
| | - Michael C Ostrowski
- The James Cancer Hospital and Solove Research Institute, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210
- Department of Molecular Virology, Immunology, and Medical Genetics, The Ohio State University, Columbus, OH 43210
| | - Aharon G Freud
- The James Cancer Hospital and Solove Research Institute, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210
- Department of Pathology, The Ohio State University, Columbus, OH 43210
| | - Gustavo W Leone
- The James Cancer Hospital and Solove Research Institute, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210
- Department of Molecular Virology, Immunology, and Medical Genetics, The Ohio State University, Columbus, OH 43210
- Department of Molecular Genetics, College of Biological Sciences, The Ohio State University, Columbus, OH 43210
| | - Michael A Caligiuri
- The James Cancer Hospital and Solove Research Institute, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210;
- Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, OH 43210
| | - Jianhua Yu
- The James Cancer Hospital and Solove Research Institute, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210;
- Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, OH 43210
| |
Collapse
|
27
|
Hoareau-Aveilla C, Meggetto F. Crosstalk between microRNA and DNA Methylation Offers Potential Biomarkers and Targeted Therapies in ALK-Positive Lymphomas. Cancers (Basel) 2017; 9:cancers9080100. [PMID: 28771164 PMCID: PMC5575603 DOI: 10.3390/cancers9080100] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Revised: 07/04/2017] [Accepted: 07/28/2017] [Indexed: 12/13/2022] Open
Abstract
The discovery of microRNA (miRNA) has provided new and powerful tools for studying the mechanism, diagnosis and treatment of human cancers. The down-regulation of tumor suppressive miRNA by hypermethylation of CpG island (CpG is shorthand for 5′-C-phosphate-G-3′, that is, cytosine and guanine separated by only one phosphate) is emerging as a common hallmark of cancer and appears to be involved in drug resistance. This review discusses the role of miRNA and DNA methylation in drug resistance mechanisms and highlights their potential as anti-cancer therapies in Anaplastic Lymphoma Kinase (ALK)-positive lymphomas. These are a sub-type of non-Hodgkin’s lymphomas that predominantly affect children and young adults and are characterized by the expression of the nucleophosmin (NPM)/ALK chimeric oncoprotein. Dysregulation of miRNA expression and regulation has been shown to affect several signaling pathways in ALK carcinogenesis and control tumor growth, both in cell lines and mouse models. These data suggest that the modulation of DNA methylation and/or the expression of these miRNA could serve as new biomarkers and have potential therapeutic applications for ALK-positive malignancies.
Collapse
Affiliation(s)
- Coralie Hoareau-Aveilla
- Inserm, UMR1037 CRCT, F-31000 Toulouse, France.
- Université Toulouse III-Paul Sabatier, UMR1037 CRCT, F-31000 Toulouse, France.
- CNRS, ERL5294 CRCT, F-31000 Toulouse, France.
- Laboratoire d'Excellence Toulouse Cancer-TOUCAN, F-31024 Toulouse, France.
| | - Fabienne Meggetto
- Inserm, UMR1037 CRCT, F-31000 Toulouse, France.
- Université Toulouse III-Paul Sabatier, UMR1037 CRCT, F-31000 Toulouse, France.
- CNRS, ERL5294 CRCT, F-31000 Toulouse, France.
- Laboratoire d'Excellence Toulouse Cancer-TOUCAN, F-31024 Toulouse, France.
| |
Collapse
|
28
|
Ciccia F, Guggino G, Rizzo A, Bombardieri M, Raimondo S, Carubbi F, Cannizzaro A, Sireci G, Dieli F, Campisi G, Giacomelli R, Cipriani P, De Leo G, Alessandro R, Triolo G. Interleukin (IL)-22 receptor 1 is over-expressed in primary Sjogren's syndrome and Sjögren-associated non-Hodgkin lymphomas and is regulated by IL-18. Clin Exp Immunol 2015; 181:219-29. [PMID: 25880879 PMCID: PMC4516437 DOI: 10.1111/cei.12643] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2014] [Revised: 04/11/2015] [Accepted: 04/11/2015] [Indexed: 12/19/2022] Open
Abstract
The aim of this study was to elucidate more clearly the role of interleukin (IL)-18 in modulating the IL-22 pathway in primary Sjögren's syndrome (pSS) patients and in pSS-associated lymphomas. Minor salivary glands (MSGs) from patients with pSS and non-specific chronic sialoadenitis (nSCS), parotid glands biopsies from non-Hodgkin lymphomas (NHL) developed in pSS patients, were evaluated for IL-18, IL-22, IL-22 receptor 1 (IL-22R1), IL-22 binding protein (IL-22BP) and signal transducer and activator of transcription-3 (STAT-3) expression. MSGs IL-22R1-expressing cells were characterized by confocal microscopy and flow cytometry in pSS, nSCS and healthy controls . The effect of recombinant IL-18 and IL-22 on peripheral blood mononuclear cells (PBMCs) from pSS and nSCS was studied by flow cytometry and reverse transcription-polymerase chain reaction (RT-PCR). MSGs of pSS and NHL were characterized by an imbalance between IL-22 and IL-22BP protein expression, with IL-18 and IL-22BP being expressed in a mutually exclusive manner and IL-18 and IL-22R1 being correlated directly. Aberrant expression of IL-22R1, induced by IL-18, was observed only among tissue and circulating myeloid cells of pSS patients and macrophages of NHL tissues of pSS patients, but not nSCS. IL-22R1 expression on PBMC of pSS was functional, as its stimulation with recombinant IL-22 significantly up-regulated the expression of STAT-3, IL-17 and IL-22. An IL-18-dependent aberrant expression of IL-22R1 on cells of haematopoietic origin seems to be a specific immunological signature of patients with pSS and pSS-associated lymphomas.
Collapse
Affiliation(s)
- F Ciccia
- Dipartimento Biomedico di Medicina Interna e Specialistica, Sezione di ReumatologiaPalermo, Italy
| | - G Guggino
- Dipartimento Biomedico di Medicina Interna e Specialistica, Sezione di ReumatologiaPalermo, Italy
- Dipartimento di Biopatologia e Biotecnologie Mediche e Forensi, Università di PalermoPalermo, Italy
| | - A Rizzo
- Azienda Ospedaliera Ospedali riuniti Villa Sofia-Cervello, Anatomia PatologicaPalermo, Italy
| | - M Bombardieri
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Queen Mary University LondonUK
| | - S Raimondo
- Dipartimento di Biopatologia e Biotecnologie Mediche e Forensi, Università di PalermoPalermo, Italy
| | - F Carubbi
- Dipartimento di Scienze Cliniche Applicate e Biotecnologiche, Sezione di Reumatologia, Università di L’AquilaItaly
| | - A Cannizzaro
- Azienda Ospedaliera Ospedali riuniti Villa Sofia-Cervello, Anatomia PatologicaPalermo, Italy
| | - G Sireci
- Dipartimento di Biopatologia e Biotecnologie Mediche e Forensi, Università di PalermoPalermo, Italy
| | - F Dieli
- Dipartimento di Biopatologia e Biotecnologie Mediche e Forensi, Università di PalermoPalermo, Italy
| | - G Campisi
- Dipartimento di discipline Chirurgiche, Università di PalermoItaly
| | - R Giacomelli
- Dipartimento di Scienze Cliniche Applicate e Biotecnologiche, Sezione di Reumatologia, Università di L’AquilaItaly
| | - Paola Cipriani
- Dipartimento di Scienze Cliniche Applicate e Biotecnologiche, Sezione di Reumatologia, Università di L’AquilaItaly
| | - G De Leo
- Dipartimento di Biopatologia e Biotecnologie Mediche e Forensi, Università di PalermoPalermo, Italy
| | - R Alessandro
- Dipartimento di Biopatologia e Biotecnologie Mediche e Forensi, Università di PalermoPalermo, Italy
| | - G Triolo
- Dipartimento Biomedico di Medicina Interna e Specialistica, Sezione di ReumatologiaPalermo, Italy
| |
Collapse
|
29
|
Lu T, Liu Y, Li P, Yu S, Huang X, Ma D, Ji C. Decreased circulating Th22 and Th17 cells in patients with aplastic anemia. Clin Chim Acta 2015; 450:90-6. [PMID: 26238188 DOI: 10.1016/j.cca.2015.07.031] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2015] [Revised: 07/08/2015] [Accepted: 07/30/2015] [Indexed: 01/31/2023]
Abstract
BACKGROUND Aplastic anemia (AA) is an immune-mediated disorder and mainly related to active destruction of hematopoietic cells by effector T lymphocytes. T helper (Th) 22 cells characterized as a novel subset of CD4+ T cells participate in the pathogenesis of autoimmune and hematological diseases. However, the role of Th22 subset in AA remains unknown. METHODS 31 untreated AA patients and 30 healthy controls were included in this study. The percentages of Th22, Th17 and pure Th17 cells in peripheral blood were detected by flow cytometry. ELISA to measure interleukin (IL)-22 and IL-17A plasma levels and qRT-PCR for the mRNA levels of Th22 and Th17 related molecules were performed. RESULTS The proportions of Th22, pure Th17, Th17 cells and plasma levels of IL-22 were significantly lower in untreated AA patients than those in normal controls. A positive correlation was found between Th22 and pure Th17 cells in AA. Moreover, percentages of Th22 cells correlated positively with reticulocyte counts and percentages. In addition, STAT3/STAT5 mRNA expression ratio was elevated in AA patients. CONCLUSION Together, our results showed Th22 cells correlating with clinical characteristics of AA patients, indicating a possible role of Th22 immune response in the pathogenesis and therapeutic intervention of AA.
Collapse
Affiliation(s)
- Ting Lu
- Department of Hematology, Qilu Hospital, Shandong University, Jinan 250012, PR China
| | - Yan Liu
- Department of Hematology, Qilu Hospital, Shandong University, Jinan 250012, PR China
| | - Peng Li
- Department of Hematology, Qilu Hospital, Shandong University, Jinan 250012, PR China
| | - Shuang Yu
- Department of Hematology, Qilu Hospital, Shandong University, Jinan 250012, PR China
| | - Xiaoyang Huang
- Department of Paediatrics, Qilu Hospital, Shandong University, Jinan 250012, PR China
| | - Daoxin Ma
- Department of Hematology, Qilu Hospital, Shandong University, Jinan 250012, PR China.
| | - Chunyan Ji
- Department of Hematology, Qilu Hospital, Shandong University, Jinan 250012, PR China.
| |
Collapse
|
30
|
Yong X, Tang B, Li BS, Xie R, Hu CJ, Luo G, Qin Y, Dong H, Yang SM. Helicobacter pylori virulence factor CagA promotes tumorigenesis of gastric cancer via multiple signaling pathways. Cell Commun Signal 2015; 13:30. [PMID: 26160167 PMCID: PMC4702319 DOI: 10.1186/s12964-015-0111-0] [Citation(s) in RCA: 139] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2015] [Accepted: 07/03/2015] [Indexed: 12/12/2022] Open
Abstract
Helicobacter pylori (H. pylori) infection is strongly associated with the development of gastric diseases but also with several extragastric diseases. The clinical outcomes caused by H. pylori infection are considered to be associated with a complex combination of host susceptibility, environmental factors and bacterial isolates. Infections involving H. pylori strains that possess the virulence factor CagA have a worse clinical outcome than those involving CagA-negative strains. It is remarkable that CagA-positive H. pylori increase the risk for gastric cancer over the risk associated with H. pylori infection alone. CagA behaves as a bacterial oncoprotein playing a key role in H. pylori-induced gastric cancer. Activation of oncogenic signaling pathways and inactivation of tumor suppressor pathways are two crucial events in the development of gastric cancer. CagA shows the ability to affect the expression or function of vital protein in oncogenic or tumor suppressor signaling pathways via several molecular mechanisms, such as direct binding or interaction, phosphorylation of vital signaling proteins and methylation of tumor suppressor genes. As a result, CagA continuously dysregulates of these signaling pathways and promotes tumorigenesis. Recent research has enriched our understanding of how CagA effects on these signaling pathways. This review summarizes the results of the most relevant studies, discusses the complex molecular mechanism involved and attempts to delineate the entire signaling pathway.
Collapse
Affiliation(s)
- Xin Yong
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, P.R. China.
| | - Bo Tang
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, P.R. China.
| | - Bo-Sheng Li
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, P.R. China.
| | - Rui Xie
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, P.R. China.
| | - Chang-Jiang Hu
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, P.R. China.
| | - Gang Luo
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, P.R. China.
| | - Yong Qin
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, P.R. China.
| | - Hui Dong
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, P.R. China.
| | - Shi-Ming Yang
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, P.R. China.
| |
Collapse
|
31
|
Azizi G, Rastegar Pouyani M, Navabi SS, Yazdani R, Kiaee F, Mirshafiey A. The Newly Identified T Helper 22 Cells Lodge in Leukemia. Int J Hematol Oncol Stem Cell Res 2015; 9:143-54. [PMID: 26261700 PMCID: PMC4529682] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2014] [Accepted: 05/17/2015] [Indexed: 11/28/2022] Open
Abstract
Leukemia is a hematological tumor in which the malignant myeloid or lymphoid subsets play a pivotal role. Newly identified T helper cell 22 (Th22) is a subset of CD4(+) T cells with distinguished gene expression, function and specific properties apart from other known CD4(+) T cell subsets.Th22 cells are characterized by production of a distinct profile of effector cytokines, including interleukin (IL)-22, IL-13, and tumor necrosis factor-α (TNF-α). The levels of Th22 and cytokine IL-22 are increased and positively related to inflammatory and autoimmune disorders. Recently, several studies have reported the changes in frequency and function of Th22 in acute leukemic disorders as AML and ALL. This review discusses the role of Th22 and its cytokine IL-22 in the immunopathogenesis of leukemic disease.
Collapse
Affiliation(s)
- Gholamreza Azizi
- Imam Hassan Mojtaba Hospital, Alborz University of Medical Sciences, Karaj, Iran,Research Center for Immunodeficiencies, Children’s Medical Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohsen Rastegar Pouyani
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Shadi sadat Navabi
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Reza Yazdani
- Research Center for Immunodeficiencies, Children’s Medical Center, Tehran University of Medical Sciences, Tehran, Iran,Department of Immunology, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Fatemeh Kiaee
- Research Center for Immunodeficiencies, Children’s Medical Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Abbas Mirshafiey
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
32
|
Merkel O, Hamacher F, Griessl R, Grabner L, Schiefer AI, Prutsch N, Baer C, Egger G, Schlederer M, Krenn PW, Hartmann TN, Simonitsch-Klupp I, Plass C, Staber PB, Moriggl R, Turner SD, Greil R, Kenner L. Oncogenic role of miR-155 in anaplastic large cell lymphoma lacking the t(2;5) translocation. J Pathol 2015; 236:445-56. [PMID: 25820993 PMCID: PMC4557053 DOI: 10.1002/path.4539] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2014] [Revised: 03/11/2015] [Accepted: 03/14/2015] [Indexed: 12/20/2022]
Abstract
Anaplastic large cell lymphoma (ALCL) is a rare, aggressive, non-Hodgkin's lymphoma that is characterized by CD30 expression and disease onset in young patients. About half of ALCL patients bear the t(2;5)(p23;q35) translocation, which results in the formation of the nucleophosmin-anaplastic lymphoma tyrosine kinase (NPM–ALK) fusion protein (ALCL ALK+). However, little is known about the molecular features and tumour drivers in ALK-negative ALCL (ALCL ALK−), which is characterized by a worse prognosis. We found that ALCL ALK−, in contrast to ALCL ALK+, lymphomas display high miR-155 expression. Consistent with this, we observed an inverse correlation between miR-155 promoter methylation and miR-155 expression in ALCL. However, no direct effect of the ALK kinase on miR-155 levels was observed. Ago2 immunoprecipitation revealed miR-155 as the most abundant miRNA, and enrichment of target mRNAs C/EBPβ and SOCS1. To investigate its function, we over-expressed miR-155 in ALCL ALK+ cell lines and demonstrated reduced levels of C/EBPβ and SOCS1. In murine engraftment models of ALCL ALK−, we showed that anti-miR-155 mimics are able to reduce tumour growth. This goes hand-in-hand with increased levels of cleaved caspase-3 and high SOCS1 in these tumours, which leads to suppression of STAT3 signalling. Moreover, miR-155 induces IL-22 expression and suppresses the C/EBPβ target IL-8. These data suggest that miR-155 can act as a tumour driver in ALCL ALK− and blocking miR-155 could be therapeutically relevant. Original miRNA array data are to be found in the supplementary material (Table S1). © 2015 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Olaf Merkel
- Department of Translational Oncology, National Centre for Tumour Diseases (NCT), German Cancer Research Centre (DKFZ), Heidelberg, Germany.,Department of Clinical Pathology, Medical University Vienna, Austria.,European Research Initiative on ALK Related Malignancies (www.erialcl.net)
| | - Frank Hamacher
- Laboratory for Immunological and Molecular Cancer Research, Third Medical Department, Oncologic Centre, Paracelsus Medical University, Salzburg, Austria
| | - Robert Griessl
- Laboratory for Immunological and Molecular Cancer Research, Third Medical Department, Oncologic Centre, Paracelsus Medical University, Salzburg, Austria
| | - Lisa Grabner
- Department of Clinical Pathology, Medical University Vienna, Austria
| | - Ana-Iris Schiefer
- Department of Clinical Pathology, Medical University Vienna, Austria
| | - Nicole Prutsch
- Department of Clinical Pathology, Medical University Vienna, Austria
| | - Constance Baer
- Department of Epigenomics and Cancer Risk Factors, German Cancer Research Centre (DKFZ), Heidelberg, Germany
| | - Gerda Egger
- Department of Clinical Pathology, Medical University Vienna, Austria.,European Research Initiative on ALK Related Malignancies (www.erialcl.net)
| | - Michaela Schlederer
- Department of Clinical Pathology, Medical University Vienna, Austria.,Ludwig Boltzmann Institute for Cancer Research, Vienna, Austria
| | - Peter William Krenn
- Laboratory for Immunological and Molecular Cancer Research, Third Medical Department, Oncologic Centre, Paracelsus Medical University, Salzburg, Austria
| | - Tanja Nicole Hartmann
- Laboratory for Immunological and Molecular Cancer Research, Third Medical Department, Oncologic Centre, Paracelsus Medical University, Salzburg, Austria
| | | | - Christoph Plass
- Department of Epigenomics and Cancer Risk Factors, German Cancer Research Centre (DKFZ), Heidelberg, Germany
| | - Philipp Bernhard Staber
- Division of Hematology and Hemostaseology, Comprehensive Cancer Centre Vienna, Medical University of Vienna, 1090, Vienna, Austria
| | - Richard Moriggl
- Ludwig Boltzmann Institute for Cancer Research, Vienna, Austria.,Institute of Animal Breeding and Genetics, University of Veterinary Medicine Vienna and Medical University of Vienna, Austria
| | - Suzanne D Turner
- Division of Molecular Histopathology, Department of Pathology, University of Cambridge, UK.,European Research Initiative on ALK Related Malignancies (www.erialcl.net)
| | - Richard Greil
- Laboratory for Immunological and Molecular Cancer Research, Third Medical Department, Oncologic Centre, Paracelsus Medical University, Salzburg, Austria
| | - Lukas Kenner
- Department of Clinical Pathology, Medical University Vienna, Austria.,Ludwig Boltzmann Institute for Cancer Research, Vienna, Austria.,Unit of Pathology of Laboratory Animals, University of Veterinary Medicine Vienna, Austria.,European Research Initiative on ALK Related Malignancies (www.erialcl.net)
| |
Collapse
|
33
|
Tsirakis G, Pappa CA, Kolovou A, Kokonozaki M, Neonakis I, Alexandrakis MG. Clinical significance of interleukin-22 in multiple myeloma. Hematology 2015; 20:143-7. [PMID: 25055724 DOI: 10.1179/1607845414y.0000000182] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
OBJECTIVE Interleukin-22 (IL-22) is a cytokine participating in many aspects of inflammation. Multiple myeloma (MM) is a malignant disease of plasma cells with characteristic immune deregulation. We estimated serum levels of IL-22 in MM patients, both in activity and remission, in order to apprehend its possible participation in MM biology. METHODS We measured serum levels of IL-22 along with beta-2 microglobulin (B2M), paraprotein, and interleukin-1beta (IL-1beta), as well as degree of bone marrow infiltration, in 51 patients with active MM and in 22 of them in remission. RESULTS We found that IL-22 was higher in active MM patients, compared to both controls and patients in remission, and also in patients in remission compared to controls. Moreover, IL-22 was increasing in parallel with the disease stage and also correlated with B2M, IL1-beta, and degree of infiltration. DISCUSSION We suggest that the elevated levels of IL-22 in active MM patients, in parallel with disease activity, and in positive correlation with IL-1beta, may represent the inflammatory element of the disease. This increased occurrence of IL-22 may enhance myeloma proliferation and growth, and moreover, may participate in the mechanisms of immune deregulation.
Collapse
|
34
|
Di Lullo G, Marcatti M, Heltai S, Brunetto E, Tresoldi C, Bondanza A, Bonini C, Ponzoni M, Tonon G, Ciceri F, Bordignon C, Protti MP. Th22 cells increase in poor prognosis multiple myeloma and promote tumor cell growth and survival. Oncoimmunology 2015; 4:e1005460. [PMID: 26155400 PMCID: PMC4485827 DOI: 10.1080/2162402x.2015.1005460] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Revised: 12/30/2014] [Accepted: 12/30/2014] [Indexed: 12/19/2022] Open
Abstract
There is increased production of plasmacytoid dendritic cells (pDCs) in the bone marrow (BM) of multiple myeloma (MM) patients and these favor Th22 cell differentiation. Here, we found that the frequency of interleukin (IL)-22+IL-17-IL-13+ T cells is significantly increased in peripheral blood (PB) and BM of stage III and relapsed/refractory MM patients compared with healthy donors and patients with asymptomatic or stage I/II disease. Th22 cells cloned from the BM of MM patients were CCR6+CXCR4+CCR4+CCR10- and produced IL-22 and IL-13 but not IL-17. Furthermore, polyfunctional Th22-Th2 and Th22-Th1 clones were identified based on the co-expression of additional chemokine receptors and cytokines (CRTh2 or CXCR3 and IL-5 or interferon gamma [IFNγ], respectively). A fraction of MM cell lines and primary tumors aberrantly expressed the IL-22RA1 and IL-22 induced STAT-3 phosphorylation, cell growth, and resistance to drug-induced cell death in MM cells. IL-13 treatment of normal BM mesenchymal stromal cells (MSCs) induced STAT-6 phosphorylation, adhesion molecule upregulation, and increased IL-6 production and significantly favored MM cell growth compared with untreated BM MSCs. Collectively, our data show that increased frequency of IL-22+IL-17-IL-13+ T cells correlates with poor prognosis in MM through IL-22 and IL-13 protumor activity and suggest that interference with IL-22 and IL-13 signaling pathways could be exploited for therapeutic intervention.
Collapse
Key Words
- Ab, antibody; BM, bone marrow; BMMCs, bone marrow mononuclear cells; DCs, dendritic cells; Dx, dexamethasone; ICS, intracellular cytokine staining; IFN, interferon; IL, interleukin; ISS, International Staging System; LCL, Epstein–Barr virus-transformed B lymphoblastoid cell line; Ln, lenalidomide; MGUS, monoclonal gammopathy of undetermined clinical significance; MM, multiple myeloma; MSC, mesenchymal stromal cell; PB, peripheral blood; PBMCs, peripheral blood mononuclear cells; pDCs, plasmacytoid dendritic cells; SMM, smoldering multiple myeloma; Th, T helper; TNF, tumor necrosis factor; Treg, regulatory T cells; WB, Western blot
- CD4+ T helper lymphocytes
- IL-22RA1
- Th22 cells
- bone marrow mesenchymal stromal cells
- bone marrow microenvironment
- interleukin-13
- interleukin-22
- multiple myeloma
Collapse
Affiliation(s)
- Giulia Di Lullo
- Tumor Immunology Unit; IRCCS San Raffaele Scientific Institute ; Milan, Italy ; Division of Immunology, Transplantation and Infectious Diseases; IRCCS San Raffaele Scientific Institute ; Milan, Italy
| | - Magda Marcatti
- Hematology and Bone Marrow Transplantation Unit; IRCCS San Raffaele Scientific Institute ; Milan, Italy
| | - Silvia Heltai
- Tumor Immunology Unit; IRCCS San Raffaele Scientific Institute ; Milan, Italy ; Division of Immunology, Transplantation and Infectious Diseases; IRCCS San Raffaele Scientific Institute ; Milan, Italy
| | - Emanuela Brunetto
- Tumor Immunology Unit; IRCCS San Raffaele Scientific Institute ; Milan, Italy ; Division of Immunology, Transplantation and Infectious Diseases; IRCCS San Raffaele Scientific Institute ; Milan, Italy
| | - Cristina Tresoldi
- Hematology and Bone Marrow Transplantation Unit; IRCCS San Raffaele Scientific Institute ; Milan, Italy
| | - Attilio Bondanza
- Division of Immunology, Transplantation and Infectious Diseases; IRCCS San Raffaele Scientific Institute ; Milan, Italy ; Leukenia Immunotherapy Group; IRCCS San Raffaele Scientific Institute ; Milan, Italy
| | - Chiara Bonini
- Division of Immunology, Transplantation and Infectious Diseases; IRCCS San Raffaele Scientific Institute ; Milan, Italy ; Experimental Hematology Unit; IRCCS San Raffaele Scientific Institute ; Milan, Italy
| | - Maurilio Ponzoni
- Pathology Unit; IRCCS San Raffaele Scientific Institute ; Milan, Italy ; Division of Molecular Oncology; IRCCS San Raffaele Scientific Institute ; Milan, Italy
| | - Giovanni Tonon
- Division of Molecular Oncology; IRCCS San Raffaele Scientific Institute ; Milan, Italy ; Functional Genomics of Cancer Unit; IRCCS San Raffaele Scientific Institute ; Milan, Italy
| | - Fabio Ciceri
- Hematology and Bone Marrow Transplantation Unit; IRCCS San Raffaele Scientific Institute ; Milan, Italy
| | - Claudio Bordignon
- MolMed SpA ; Milan, Italy ; Vita-Salute San Raffaele University ; Milan, Italy
| | - Maria Pia Protti
- Tumor Immunology Unit; IRCCS San Raffaele Scientific Institute ; Milan, Italy ; Division of Immunology, Transplantation and Infectious Diseases; IRCCS San Raffaele Scientific Institute ; Milan, Italy
| |
Collapse
|
35
|
Liao F, Hsu YC, Kuo SH, Yang YC, Chen JP, Hsu PN, Lin CW, Chen LT, Cheng AL, Fann CSJ, Lin JT, Wu MS. Genetic polymorphisms and tissue expression of interleukin-22 associated with risk and therapeutic response of gastric mucosa-associated lymphoid tissue lymphoma. Blood Cancer J 2014; 4:eXX. [PMID: 25303370 PMCID: PMC4220648 DOI: 10.1038/bcj.2014.70] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2014] [Revised: 08/22/2014] [Accepted: 08/29/2014] [Indexed: 02/07/2023] Open
Abstract
Chronic Helicobacter pylori-stimulated immune reactions determine the pathogenesis of gastric mucosa-associated lymphoid tissue (MALT) lymphoma. We aimed to explore the genetic predisposition to this lymphoma and its clinical implication. A total of 68 patients and 140 unrelated controls were genotyped for 84 single-nucleotide polymorphisms in genes encoding cytokines, chemokines and related receptors that play important roles in T cell-mediated gastrointestinal immunity. Five genotypes in IL-22, namely CC at rs1179246, CC at rs2227485, AA at rs4913428, AA at rs1026788 and TT at rs7314777, were associated with disease susceptibility. The former four genotypes resided in the same linkage disequilibrium block (r(2)=0.99) that conferred an approximately threefold higher risk. In vitro experiments demonstrated that co-culturing peripheral mononuclear cells or CD4(+) T cells with H. pylori stimulated the secretion of interleukin-22 (IL-22), and that IL-22 induced the expression of antimicrobial proteins, RegIIIα and lipocalin-2, in gastric epithelial cells. Furthermore, patients with gastric tissue expressing IL-22 were more likely to respond to H. pylori eradication (14/22 vs 4/19, P<0.006). We conclude that susceptibility of gastric MALT lymphoma is influenced by genetic polymorphisms in IL-22, the product of which is involved in mucosal immunity against H. pylori and associated with tumor response to H. pylori eradication.
Collapse
MESH Headings
- CD4-Positive T-Lymphocytes/metabolism
- Cell Line, Tumor
- Female
- Gene Expression Regulation, Neoplastic
- Genetic Predisposition to Disease
- Helicobacter Infections/genetics
- Helicobacter Infections/metabolism
- Helicobacter Infections/therapy
- Helicobacter pylori
- Humans
- Interleukins/biosynthesis
- Interleukins/genetics
- Lymphoma, B-Cell, Marginal Zone/genetics
- Lymphoma, B-Cell, Marginal Zone/metabolism
- Lymphoma, B-Cell, Marginal Zone/microbiology
- Lymphoma, B-Cell, Marginal Zone/therapy
- Male
- Neoplasm Proteins/biosynthesis
- Neoplasm Proteins/genetics
- Polymorphism, Single Nucleotide
- Stomach Neoplasms/genetics
- Stomach Neoplasms/metabolism
- Stomach Neoplasms/microbiology
- Stomach Neoplasms/therapy
- Interleukin-22
Collapse
Affiliation(s)
- F Liao
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Y-C Hsu
- Department of Internal Medicine, E-Da Hospital/I-Shou University, Kaohsiung, Taiwan
- Center for Database Research, E-Da Hospital/I-Shou University, Kaohsiung, Taiwan
| | - S-H Kuo
- Department of Oncology, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
| | - Y-C Yang
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - J-P Chen
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - P-N Hsu
- Department of Internal Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
| | - C-W Lin
- Department of Pathology, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
| | - L-T Chen
- National Institute of Cancer Research, National Health Research Institute, Tainan, Taiwan
| | - A-L Cheng
- Department of Oncology, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
| | - C S J Fann
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - J-T Lin
- Department of Internal Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
| | - M-S Wu
- Department of Internal Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
| |
Collapse
|
36
|
Ji Y, Yang X, Li J, Lu Z, Li X, Yu J, Li N. IL-22 promotes the migration and invasion of gastric cancer cells via IL-22R1/AKT/MMP-9 signaling. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2014; 7:3694-703. [PMID: 25120745 PMCID: PMC4128980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 04/13/2014] [Accepted: 04/28/2014] [Indexed: 06/03/2023]
Abstract
IL-22, one important inflammatory cytokine of the IL-10 family, exerts its functions via IL-22 receptor that is composed of IL-22R1 and IL-10R2 subunits. Although IL-22 expression is reported to be elevated in many cancers, and increased IL-22 expression correlates with tumor progression and poor prognosis, little is known about the role of IL-22 in gastric cancer. In our study, we found that IL-22 stimulation promoted the migration and invasion of SGC-7901 cells. Furthermore, IL-22 increased AKT activation and MMP-9 production in a time- and dose-dependent manner, while knockdown of IL-22R1 attenuated the effect of IL-22 on gastric cancer cells. In addition, blocking of AKT activation suppressed the expression and secretion of MMP-9. Taken together, this present study suggests that IL-22 stimulation enhances the migration and invasion of gastric cancer cells by regulating IL-22R1/AKT/MMP-9 signaling axis.
Collapse
Affiliation(s)
- Yinghua Ji
- Department of Oncology, The First Affiliated Hospital of Xinxiang Medical UniversityXinxiang, 453000, China
| | - Xiaoyu Yang
- Department of Pathology, Xinxiang Medical UniversityXinxiang, 453003, China
| | - Jinsong Li
- Department of Pathology, The First Affiliated Hospital of Xinxiang Medical UniversityXinxiang, 453000, China
| | - Zhihong Lu
- Department of Oncology, The First Affiliated Hospital of Xinxiang Medical UniversityXinxiang, 453000, China
| | - Xiaorui Li
- Department of Oncology, The First Affiliated Hospital of Xinxiang Medical UniversityXinxiang, 453000, China
| | - Jian Yu
- Department of Pathology, The First Affiliated Hospital of Xinxiang Medical UniversityXinxiang, 453000, China
| | - Na Li
- Department of Pathology, Xinxiang Medical UniversityXinxiang, 453003, China
| |
Collapse
|
37
|
Lim C, Savan R. The role of the IL-22/IL-22R1 axis in cancer. Cytokine Growth Factor Rev 2014; 25:257-71. [PMID: 24856143 DOI: 10.1016/j.cytogfr.2014.04.005] [Citation(s) in RCA: 120] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2014] [Accepted: 04/29/2014] [Indexed: 12/18/2022]
Abstract
Interleukin-22 (IL-22) is an IL-10 family cytokine produced by T cells and innate lymphoid cells. The IL-22 signaling pathway orchestrates mucosal immune defense and tissue regeneration through pleiotropic effects including pro-survival signaling, cell migration, dysplasia and angiogenesis. While these functions can prevent initial establishment of tumors, they can also be hijacked by aggressive cancers to enhance tumor growth and metastasis. Thus, the role of the IL-22/IL-22R1 axis in cancer is complex and context-specific. Evidence of IL-22 involvement manifests as dysregulation of IL-22 expression and signaling in patients with many common cancers including those of the gut, skin, lung and liver. Unlike other cancer-associated cytokines, IL-22 has restricted tissue specificity as its unique receptor IL-22R1 is exclusively expressed on epithelial and tissue cells, but not immune cells. This makes it an attractive target for therapy as there is potential achieve anti-tumor immunity with fewer side effects. This review summarizes current findings on functions of IL-22 in association with general mechanisms for tumorigenesis as well as specific contributions to particular cancers, and ponders how best to approach further research in the field.
Collapse
Affiliation(s)
- Chrissie Lim
- Department of Immunology, University of Washington, Seattle, WA, USA
| | - Ram Savan
- Department of Immunology, University of Washington, Seattle, WA, USA.
| |
Collapse
|
38
|
Ciccia F, Guggino G, Giardina A, Ferrante A, Carrubbi F, Giacomelli R, Triolo G. The role of innate and lymphoid IL-22-producing cells in the immunopathology of primary Sjögren's syndrome. Expert Rev Clin Immunol 2014; 10:533-41. [PMID: 24490899 DOI: 10.1586/1744666x.2014.884461] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
In primary Sjögren's syndrome (pSS) a complex of interconnections between epithelial barrier, innate and adaptive immunity occurs. IL-22 is a pleiotropic cytokine that in pSS may be placed at the intersection of the adaptive and innate branches of immunity. Some evidence suggests that, in pSS, IL-22 may play a prominent pro-inflammatory role driving the early phase of tissue and systemic inflammation and participating in the self-perpetuation of disease. Despite contradictory data in literature about the role of NK cells in pSS, recent data also suggest an important contribution of this subset of cells of the innate immune system in the development and perpetuation of inflammation. Here, we discuss the role of IL-22 in the pathogenesis of pSS and in epithelial barrier function.
Collapse
Affiliation(s)
- Francesco Ciccia
- Dipartimento Biomedico di Medicina Interna e Specialistica, Università degli Studi di Palermo Palermo, Italy
| | | | | | | | | | | | | |
Collapse
|
39
|
Justa S, Zhou X, Sarkar S. Endogenous IL-22 plays a dual role in arthritis: regulation of established arthritis via IFN-γ responses. PLoS One 2014; 9:e93279. [PMID: 24676270 PMCID: PMC3968131 DOI: 10.1371/journal.pone.0093279] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2013] [Accepted: 03/03/2014] [Indexed: 01/15/2023] Open
Abstract
OBJECTIVE IL-22 is elevated in patients with inflammatory arthritis and correlates with disease activity. IL-22 deficient mice have reduced incidence of arthritis. Recombinant IL-22 restrains progression of arthritis via increase in IL-10 responses when administered prior to onset of arthritis. These findings imply a possible dual role of IL-22 in inflammatory arthritis depending on the phase of arthritis. Experiments outlined here were designed to elucidate the contribution of endogenous IL-22 before and after the onset of arthritis. METHODS Collagen induced arthritis (CIA) was induced in DBA1 or IFN-γ deficient mice following immunization with collagen and complete Freund's adjuvant. Anti-IL-22 antibody or isotype control were administered prior to or after onset of arthritis and disease progression assessed by clinical scoring and histopathology. IL-22, IL-17 and IFN-γ responses were measured by ELISA and flowcytometry. Anti-collagen antibody responses were analyzed by ELISA. Expression of IL-22R1 in CD4+ cells was elucidated by flowcytometry and real time PCR. RESULTS Collagen specific IL-22 responses were expanded during arthritis and IL-22 producing cells were discrete from IL-17 or IFN-γ producing cells. Neutralization of IL-22 after onset of arthritis resulted in significant increase in Th1 responses and significantly reduced severity of arthritis. CD4+ cells from arthritic mice showed increased surface expression of IL-22R1. In vitro, CD4+T cells cultured with antigen presenting cells in the presence or absence of IL-22 suppressed or induced IFN-γ, respectively. The protective effect of anti-IL-22 was reversed in IFN-γ deficient mice. Moreover, administration of anti-IL-22 prior to onset of arthritis augmented arthritis severity. CONCLUSION We show for the first time that IL-22 plays a dual role: protective prior to the onset of arthritis and pathogenic after onset of arthritis. The pathogenic effect of IL-22 is dependent on suppression of IFN-γ responses. IL-17 responses remained unchanged with the administration of anti-IL22 antibody. IL-22R1 is upregulated on CD4+T cells during arthritis and regulates IFN-γ in T cells.
Collapse
Affiliation(s)
- Shivali Justa
- Section of Rheumatology, Department of Medicine, and the Arizona Arthritis Center, University of Arizona, Tucson, Arizona, United States of America
| | - Xiaoqun Zhou
- Section of Rheumatology, Department of Medicine, and the Arizona Arthritis Center, University of Arizona, Tucson, Arizona, United States of America
| | - Sujata Sarkar
- Section of Rheumatology, Department of Medicine, and the Arizona Arthritis Center, University of Arizona, Tucson, Arizona, United States of America
| |
Collapse
|
40
|
Murga-Zamalloa C, Lim MS. ALK-driven tumors and targeted therapy: focus on crizotinib. PHARMACOGENOMICS & PERSONALIZED MEDICINE 2014; 7:87-94. [PMID: 24715763 PMCID: PMC3977456 DOI: 10.2147/pgpm.s37504] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Receptor tyrosine kinases have emerged as promising therapeutic targets for a diverse set of tumors. Overactivation of the tyrosine kinase anaplastic lymphoma kinase (ALK) has been reported in several types of malignancies such as anaplastic large cell lymphoma, inflammatory myofibroblastic tumor, neuroblastoma, and non-small-cell lung carcinoma. Further characterization of the molecular role of ALK has revealed an oncogenic signaling signature that results in tumor dependence on ALK. ALK-positive tumors display a different behavior than their ALK-negative counterparts; however, the specific role of ALK in some of these tumors remains to be elucidated. Although more studies are required to establish selective targeting of ALK as a definitive therapeutic option, initial trials have shown extraordinary results in the majority of cases.
Collapse
Affiliation(s)
| | - Megan S Lim
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
41
|
Ito M, Teshima K, Ikeda S, Kitadate A, Watanabe A, Nara M, Yamashita J, Ohshima K, Sawada K, Tagawa H. MicroRNA-150 inhibits tumor invasion and metastasis by targeting the chemokine receptor CCR6, in advanced cutaneous T-cell lymphoma. Blood 2014; 123:1499-511. [PMID: 24385540 DOI: 10.1182/blood-2013-09-527739] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
In this study, we show that microRNA-150 (miR-150) is significantly downregulated in advanced cutaneous T-cell lymphoma (CTCL), and that this downregulation is strongly associated with tumor invasion/metastasis. Inoculation of CTCL cell lines into nonobese diabetic/Shi-scid interleukin 2γ (IL-2γ) null mice led to CTCL cell migration to multiple organs; however, prior transfection of the cells with miR-150 substantially reduced the invasion/metastasis by directly downregulating CCR6, a specific receptor for the chemokine CCL20. We also found that IL-22 and its specific receptor subunit, IL22RA1, were aberrantly overexpressed in advanced CTCL, and that production of IL-22 and CCL20 was increased in cultured CTCL cells. IL22RA1 knockdown specifically reduced CCL20 production in CTCL cells, suggesting that IL-22 upregulation may activate the production of CCL20 and its binding to CCR6, thereby enhancing the multidirectional migration potential of CTCL cells. CTCL cells also exhibited nutrition- and CCL20-dependent chemotaxis, which were inhibited by miR-150 transfection or CCR6 knockdown. From these findings, we conclude that, in the presence of continuous CCR6 upregulation accompanied by miR-150 downregulation, IL-22 activation leads to continuous CCL20-CCR6 interaction in CTCL cells and, in turn, autocrine metastasis to distal organs. This suggests miR-150, CCL20, and CCR6 could be key targets for the treatment of advanced CTCL.
Collapse
MESH Headings
- Animals
- Autocrine Communication
- Cell Line, Tumor
- Chemokine CCL20/genetics
- Chemokine CCL20/metabolism
- Chemotaxis/genetics
- Cluster Analysis
- Disease Models, Animal
- Gene Expression
- Gene Expression Regulation, Neoplastic
- Gene Knockdown Techniques
- Genes, Tumor Suppressor
- Humans
- Interleukins/genetics
- Interleukins/metabolism
- Lymphoma, T-Cell, Cutaneous/genetics
- Lymphoma, T-Cell, Cutaneous/mortality
- Lymphoma, T-Cell, Cutaneous/pathology
- Mice
- MicroRNAs/genetics
- Neoplasm Invasiveness/genetics
- Neoplasm Metastasis/genetics
- Neoplasm Staging
- RNA Interference
- Receptors, CCR6/genetics
- Transfection
- Xenograft Model Antitumor Assays
- Interleukin-22
Collapse
Affiliation(s)
- Mitsugu Ito
- Department of Hematology, Nephrology, and Rheumatology, Akita University Graduate School of Medicine, Akita, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Karabiyik A, Peck AB, Nguyen CQ. The important role of T cells and receptor expression in Sjögren's syndrome. Scand J Immunol 2013; 78:157-66. [PMID: 23679844 DOI: 10.1111/sji.12079] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2013] [Accepted: 05/16/2013] [Indexed: 12/17/2022]
Abstract
Sjögren's syndrome (SjS), an autoimmune disease characterized by exocrine gland dysfunction leading to dry mouth and dry eye diseases, is typified by progressive leucocyte infiltrations of the salivary and lacrimal glands. Histologically, these leucocyte infiltrations generally establish periductal aggregates, referred to as lymphocytic foci (LF), which occasionally appear as germinal centre (GC)-like structures. The formation and organization of these LF suggest an important and dynamic role for helper T cells (TH), specifically TH1, TH2 and the recently discovered TH17, in development and onset of clinical SjS, considered a B cell-mediated hypersensitivity type 2 disease. Despite an ever-increasing focus on identifying the underlying aetiology of SjS, defining factors that initiate this autoimmune disease remain a mystery. Thus, determining interactions between infiltrating TH cells and exocrine gland tissue (auto-)antigens represents a fertile research endeavour. This review discusses pathological functions of TH cells in SjS, the current status of TH cell receptor gene rearrangements associated with human and mouse models of SjS and potential future prospects for identifying receptor-autoantigen interactions.
Collapse
Affiliation(s)
- A Karabiyik
- Department of Pathology and Infectious Diseases, College of Veterinary Medicine, University of Florida, Gainesville, FL, USA
| | | | | |
Collapse
|
43
|
Zhang S, Fujita H, Mitsui H, Yanofsky VR, Fuentes-Duculan J, Pettersen JS, Suárez-Fariñas M, Gonzalez J, Wang CQF, Krueger JG, Felsen D, Carucci JA. Increased Tc22 and Treg/CD8 ratio contribute to aggressive growth of transplant associated squamous cell carcinoma. PLoS One 2013; 8:e62154. [PMID: 23667456 PMCID: PMC3646982 DOI: 10.1371/journal.pone.0062154] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2012] [Accepted: 03/18/2013] [Indexed: 12/25/2022] Open
Abstract
Immune suppressed organ transplant recipients suffer increased morbidity and mortality from primary cutaneous SCC. We studied tumor microenvironment in transplant-associated SCC (TSCC), immune-competent SCC and normal skin by IHC, IF and RT-PCR on surgical discard. We determined T cell polarization in TSCC and SCC by intracellular cytokine staining of T cell crawl outs from human skin explants. We studied the effects of IL-22, an inducer of keratinocyte proliferation, on SCC proliferation in vitro. SCC and TSCC are both associated with significantly higher numbers of CD3(+) and CD8(+) T cells compared to normal skin. TSCC showed a higher proportion of Foxp3(+) T regs to CD8(+) T cells compared to SCC and a lower percentage of IFN-γ producing CD4(+) T cells. TSCC, however, had a higher percentage of IL-22 producing CD8(+) T cells compared to SCC. TSCC showed more diffuse Ki67 and IL-22 receptor (IL-22R) expression by IHC. IL-22 induced SCC proliferation in vitro despite serum starvation. Diminished cytotoxic T cell function in TSCC due to decreased CD8/T-reg ratio may permit tumor progression. Increased IL-22 and IL-22R expression could accelerate tumor growth in transplant patients. IL-22 may be an attractive candidate for targeted therapy of SCC without endangering allograft survival.
Collapse
MESH Headings
- CD3 Complex/metabolism
- CD8-Positive T-Lymphocytes/cytology
- CD8-Positive T-Lymphocytes/drug effects
- CD8-Positive T-Lymphocytes/immunology
- Carcinoma, Squamous Cell/etiology
- Carcinoma, Squamous Cell/immunology
- Carcinoma, Squamous Cell/metabolism
- Carcinoma, Squamous Cell/pathology
- Cell Count
- Cell Line, Tumor
- Cell Proliferation/drug effects
- Forkhead Transcription Factors/metabolism
- Gene Expression Regulation, Neoplastic/drug effects
- Gene Expression Regulation, Neoplastic/immunology
- Humans
- Immunocompetence/immunology
- Interleukins/pharmacology
- Organ Transplantation/adverse effects
- Phosphoproteins/metabolism
- Receptors, Interleukin/metabolism
- STAT3 Transcription Factor/metabolism
- Skin/cytology
- Skin/immunology
- Skin/pathology
- Skin Neoplasms/immunology
- Skin Neoplasms/pathology
- T-Lymphocyte Subsets/cytology
- T-Lymphocyte Subsets/drug effects
- T-Lymphocyte Subsets/immunology
- T-Lymphocytes, Regulatory/cytology
- T-Lymphocytes, Regulatory/drug effects
- T-Lymphocytes, Regulatory/immunology
- Th1 Cells/cytology
- Th1 Cells/drug effects
- Th1 Cells/immunology
- Tumor Microenvironment/drug effects
- Tumor Microenvironment/immunology
- Up-Regulation/drug effects
- Up-Regulation/immunology
- Interleukin-22
Collapse
Affiliation(s)
- Shali Zhang
- Ronald O. Perelman Department of Dermatology, New York University Langone Medical Center, New York, New York, United States of America
| | - Hideki Fujita
- Laboratory for Investigative Dermatology, Rockefeller University, New York, New York, United States of America
| | - Hiroshi Mitsui
- Laboratory for Investigative Dermatology, Rockefeller University, New York, New York, United States of America
| | - Valerie R. Yanofsky
- Albert Einstein College of Medicine, Bronx, New York, United States of America
| | - Judilyn Fuentes-Duculan
- Laboratory for Investigative Dermatology, Rockefeller University, New York, New York, United States of America
| | - Julia S. Pettersen
- Department of Dermatology, Weill Cornell Medical College, New York, New York, United States of America
| | - Mayte Suárez-Fariñas
- Laboratory for Investigative Dermatology, Rockefeller University, New York, New York, United States of America
| | - Juana Gonzalez
- Translational Immunomonitoring Resource Center, The Rockefeller University, New York, New York, United States of America
| | - Claire Q. F. Wang
- Laboratory for Investigative Dermatology, Rockefeller University, New York, New York, United States of America
| | - James G. Krueger
- Laboratory for Investigative Dermatology, Rockefeller University, New York, New York, United States of America
| | - Diane Felsen
- Institute for Pediatric Urology, Weill Cornell Medical College, New York, New York, United States of America
| | - John A. Carucci
- Ronald O. Perelman Department of Dermatology, New York University Langone Medical Center, New York, New York, United States of America
| |
Collapse
|
44
|
Lai R, Ingham RJ. The pathobiology of the oncogenic tyrosine kinase NPM-ALK: a brief update. Ther Adv Hematol 2013; 4:119-31. [PMID: 23610619 DOI: 10.1177/2040620712471553] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Extensive research has been carried out in the past two decades to study the pathobiology of nucleophosmin-anaplastic lymphoma kinase (NPM-ALK), which is an oncogenic fusion protein found exclusively in a specific type of T-cell lymphoid malignancy, namely ALK-positive anaplastic large cell lymphoma. Results from these studies have provided highly useful insights into the mechanisms by which a constitutively tyrosine kinase, such as NPM-ALK, promotes tumorigenesis. Several previous publications have comprehensively summarized the advances in this field. In this review, we provide readers with a brief update on specific areas of NPM-ALK pathobiology. In the first part, the NPM-ALK/signal transducer and activator of transcription 3 (STAT3) signaling axis is discussed, with an emphasis on the existence of multiple biochemical defects that have been shown to amplify the oncogenic effects of this signaling axis. Specifically, findings regarding JAK3, SHP1 and the stimulatory effects of several cytokines including interleukin (IL)-9, IL-21 and IL-22 are summarized. New concepts stemming from recent observations regarding the functional interactions among the NPM-ALK/STAT3 axis, β catenin and glycogen synthase kinase 3β will be postulated. Lastly, new mechanisms by which the NPM-ALK/STAT3 axis promotes tumorigenesis, such as its modulations of Twist1, hypoxia-induced factor 1α, CD274, will be described. In the second part, we summarize recent data generated by mass spectrometry studies of NPM-ALK, and use MSH2 and heat shock proteins as examples to illustrate the use of mass spectrometry data in stimulating new research in this field. In the third part, the evolving field of microRNA in the context of NPM-ALK biology is discussed.
Collapse
Affiliation(s)
- Raymond Lai
- Department of Laboratory Medicine and Pathology, Cross Cancer Institute and University of Alberta, Rm 2338, Cross Cancer Institute, 11560 University Avenue, Edmonton, Alberta, Canada T6G 1Z2
| | | |
Collapse
|
45
|
Huang Y, de Leval L, Gaulard P. Molecular underpinning of extranodal NK/T-cell lymphoma. Best Pract Res Clin Haematol 2013; 26:57-74. [DOI: 10.1016/j.beha.2013.04.006] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
|
46
|
Shao LL, Zhang L, Hou Y, Yu S, Liu XG, Huang XY, Sun YX, Tian T, He N, Ma DX, Peng J, Hou M. Th22 cells as well as Th17 cells expand differentially in patients with early-stage and late-stage myelodysplastic syndrome. PLoS One 2012; 7:e51339. [PMID: 23236476 PMCID: PMC3517399 DOI: 10.1371/journal.pone.0051339] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2012] [Accepted: 11/07/2012] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Immunological mechanisms are increasingly recognized in the progression of myelodysplastic syndrome (MDS). Early-stage MDS (E-MDS) is characterized by autoimmune-mediated myelosuppression whereas late-stage MDS (L-MDS) involves immune evasion, giving dysplastic cells growth potential to progress into acute myeloid leukemia. T-helper (Th) 22 is involved in the pathogenesis of inflammatory autoimmunity and tumorigenesis. The roles of Th22 cells in the pathophysiology of E-MDS and L-MDS remain unsettled. DESIGN AND METHODS We studied 37 MDS patients (E-MDS, n = 17; L-MDS, n = 20) and 20 healthy controls to characterize their peripheral blood (PB), as well as 25 MDS patients and 10 healthy controls to characterize their bone marrow(BM). The expression of Interleukin-22 (IL-22), IL-17 or interferon gamma (IFN-γ) was examined in E-MDS, L-MDS patients and controls by flow cytometry. The mRNA expression levels of RAR-related orphan receptor C (RORC), IL-6, tumor necrosis factor alpha (TNF-α) and IL-23 in peripheral blood mononuclear cells (PBMCs) were determined by real-time quantitative polymerase chain reaction. The levels of IL-22 and IL-17 both in PB and BM plasma were examined by enzyme-linked immunosorbent assay. RESULTS In E-MDS, peripheral Th17 cells were significantly elevated and correlated with peripheral Th22 cells compared with healthy controls and L-MDS. Significantly higher levels of peripheral Th22 expansion, mRNA expression of IL-6, TNF-α and lower level of RORC mRNA expression were observed in L-MDS compared with E-MDS. No statistical difference was found in IL-23 mRNA expression or plasma IL-22, IL-17 levels among E-MDS, L-MDS and controls. CONCLUSIONS Our data demonstrated that L-MDS cohort had increased frequencies of peripheral Th22 cells and higher mRNA expression levels of IL-6 and TNF-α, indicating that Th22 cells along with Th17 cells or not are involved in the dynamic immune responses of MDS.
Collapse
Affiliation(s)
- Lin-lin Shao
- Department of Hematology, Qilu Hospital, Shandong University, Jinan, China
| | - Lei Zhang
- Department of Orthopedics, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, China
| | - Yu Hou
- Medical College, Shandong University, Jinan, China
| | - Shuang Yu
- Department of Hematology, Qilu Hospital, Shandong University, Jinan, China
| | - Xin-guang Liu
- Department of Hematology, Qilu Hospital, Shandong University, Jinan, China
| | - Xiao-yang Huang
- Department of Pediatrics, Qilu Hospital, Shandong University, Jinan, China
| | - Yuan-xin Sun
- Department of Hematology, Qilu Hospital, Shandong University, Jinan, China
| | - Tian Tian
- Department of Hematology, Qilu Hospital, Shandong University, Jinan, China
| | - Na He
- Department of Hematology, Qilu Hospital, Shandong University, Jinan, China
| | - Dao-xin Ma
- Department of Hematology, Qilu Hospital, Shandong University, Jinan, China
| | - Jun Peng
- Department of Hematology, Qilu Hospital, Shandong University, Jinan, China
| | - Ming Hou
- Department of Hematology, Qilu Hospital, Shandong University, Jinan, China
| |
Collapse
|
47
|
Kasahara S, Tsurumi H, Shibata Y, Matsumoto T, Nakamura N, Nakamura H, Kanemura N, Goto N, Hara T, Moriwaki H. Cerebriform variant type of T cell prolymphocytic leukemia with complex karyotype including an additional segment at 1p36.1. Int J Hematol 2012; 96:674-8. [PMID: 23054642 DOI: 10.1007/s12185-012-1176-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2011] [Revised: 09/04/2012] [Accepted: 09/04/2012] [Indexed: 11/29/2022]
Abstract
We describe two patients with T cell prolymphocytic leukemia (T-PLL) who exhibited the same complex karyotype, including an additional segment at 1p36.1. One presented with secondary progression following an initial stable clinical course, and the other with typically progressive disease. Features of the cerebriform variant were identified in the peripheral blood of both patients. Aggressive symptoms, such as lymphocytosis, lymphadenopathy, pleural effusion, cutaneous involvement and hepatosplenomegaly, developed during the progressive phases. Levels of serum soluble interleukin 2 receptor increased when symptoms worsened. These patients did not have the karyotypic 14q11 abnormality and trisomy 8q that are features of non-Japanese patients. The prognoses of these patients were poor; one survived for 2 months and the other survived for 10 months after progression. A chromosomal abnormality may occur in other types of aggressive T-PLL, particularly when extramedullary infiltration is a feature.
Collapse
Affiliation(s)
- Senji Kasahara
- First Department of Internal Medicine, Gifu University School of Medicine, 1-1 Yanagido, Gifu 501-1194, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Kong Q, Wu W, Yang F, Liu Y, Xue Y, Gao M, Lai W, Pan X, Yan Y, Pang Y, Deng Y. Increased expressions of IL-22 and Th22 cells in the coxsackievirus B3-Induced mice acute viral myocarditis. Virol J 2012; 9:232. [PMID: 23050732 PMCID: PMC3544697 DOI: 10.1186/1743-422x-9-232] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2012] [Accepted: 09/30/2012] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Recently, a new subset of T helper (Th) cell that predominantly secret cytokine interleukin-22 (IL-22) is identified, termed Th22 cells. The Th22 subset has been demonstrated to be involved in immunity and tissue inflammation. However, the existence of Th22 cells and role of IL-22 in acute viral myocarditis (AVMC) remain unknown. METHODS BALB/c mice were intraperitoneally (i.p) infected with CVB3 for establishing AVMC models. Control mice were treated with phosphate-buffered saline (PBS) i.p. On day 14 post injection, frequencies of splenic Th22 cells were determined, productions of IL-22 and expressions of IL-22R (IL-22 receptor) were measured. To further investigate the effects of IL-22, AVMC mice treated with Anti-IL-22 neutralizing antibody were explored. The severity of AVMC were monitored; the frequencies of Th22 cells, the expressions of IL-22 and IL-22R were investigated; in addition to IFN-γ, inflammatory cytokines IL-17, TNF-α, IL-6 as well as IL-1β, were evaluated. Cardiac viral replication were detected. RESULTS Compared with control group, significant elevations of circulating Th22 cells and IL-22, cardiac protein and mRNA of IL-22, and IL-22R1 were demonstrated in AVMC group. Treatment of AVMC mice with Anti-IL-22 Ab exacerbated the severity of viral myocarditis, verified by lower survival rate, higher HW/BW ratios and cardiac pathological scores. Anti-IL-22 Ab decreased the frequencies of Th22 cells and the levels of IL-22, and increased the expressions of cardiac IL-22R1. Up-regulations of IL-17, IL-6 and TNF-α, down-regulations of IFN-γ proteins and gene expressions in the plasma and myocardium, were observed in Anti-IL-22 Ab group. Furthermore, neutralization of IL-22 significantly promoted cardiac viral replication. CONCLUSIONS Our data indicate that the increased frequencies of IL-22-producing Th22 cells may play an important role in the pathogenesis of CVB3-induced mice AVMC, IL-22 may act as an myocardium-protective cytokine via the IL-22-IL-22R pathway, and suggest that targeting the Th22 cell and IL-22-IL-22R pathway could provide new therapeutic modalities for the treatment of CVB3-induced AVMC.
Collapse
Affiliation(s)
- Qing Kong
- Department of Cardiology, First Affiliated Hospital of Guangxi Medical University, Guangxi Cardiovascular Institute, Nanning, 530021, China
| | - Weifeng Wu
- Department of Cardiology, First Affiliated Hospital of Guangxi Medical University, Guangxi Cardiovascular Institute, Nanning, 530021, China
| | - Fan Yang
- Department of Cardiology, First Affiliated Hospital of Guangxi Medical University, Guangxi Cardiovascular Institute, Nanning, 530021, China
| | - Yanli Liu
- Department of Cardiology, First Affiliated Hospital of Guangxi Medical University, Guangxi Cardiovascular Institute, Nanning, 530021, China
| | - Yimin Xue
- Department of Cardiology, First Affiliated Hospital of Guangxi Medical University, Guangxi Cardiovascular Institute, Nanning, 530021, China
| | - Mengsha Gao
- Department of Cardiology, First Affiliated Hospital of Guangxi Medical University, Guangxi Cardiovascular Institute, Nanning, 530021, China
| | - Wenyin Lai
- Department of Cardiology, First Affiliated Hospital of Guangxi Medical University, Guangxi Cardiovascular Institute, Nanning, 530021, China
| | - Xiaofen Pan
- Department of Cardiology, First Affiliated Hospital of Guangxi Medical University, Guangxi Cardiovascular Institute, Nanning, 530021, China
| | - Yuluan Yan
- Department of Cardiology, First Affiliated Hospital of Guangxi Medical University, Guangxi Cardiovascular Institute, Nanning, 530021, China
| | - Yu Pang
- Department of Cardiology, First Affiliated Hospital of Guangxi Medical University, Guangxi Cardiovascular Institute, Nanning, 530021, China
| | - Yuanhua Deng
- Department of Cardiology, First Affiliated Hospital of Guangxi Medical University, Guangxi Cardiovascular Institute, Nanning, 530021, China
| |
Collapse
|
49
|
Niv-Spector L, Shpilman M, Levi-Bober M, Katz M, Varol C, Elinav E, Gertler A. Preparation and characterization of mouse IL-22 and its four single-amino-acid muteins that act as IL-22 receptor-1 antagonists. Protein Eng Des Sel 2012; 25:397-404. [PMID: 22691704 DOI: 10.1093/protein/gzs030] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Recombinant mouse interleukin 22 (mIL-22) and its variants encoding four muteins (Y51A, N54A, R55A and E117A) were expressed in Escherichia coli, refolded and purified to homogeneity as monomeric proteins by one-step ion-exchange chromatography. The binding of IL-22 and its four muteins to immobilized mIL-22 receptor α1 extracellular domain (mIL-22 Rα1-ECD) exhibited similar affinity, indicating that the single-amino-acid mutations do not affect its binding properties. Similarly, no differences were found in binding to IL-22 binding protein expressed on the surface of yeast cells, although the affinity of all five proteins to the binding protein was higher than that to IL-22 Rα1-ECD. In an in vitro bioassay, recombinant mIL-22 stimulated signal transducer and activator of transcription-3 phosphorylation in HepG2 cells, whereas the four muteins were completely (Y51A) or almost completely (N54A, R55A and E117A) devoid of this agonistic activity. Furthermore, the agonistic activity of mIL-22 could be inhibited in a dose-dependent manner by the four muteins with almost identical efficiency. mIL-22 and its Y51A mutein were pegylated by methoxy polyethylene glycol-propionylaldehyde-20 kDa, yielding a mixture of mono (75-80%) and double (20-25%) pegylated proteins. The pegylated proteins showed lower affinity (50 and 25%) toward immobilized mIL-22 Rα1-ECD than their non-pegylated analogs. Wild-type pegylated IL-22 exhibited 5- to 10-fold lower activity in the HepG2 bioassay than its non-pegylated counterpart. Preparation of recombinant mIL-22 antagonists provides new tools for the study of IL-22 activity and of eventual therapeutic means for attenuating its negative effects.
Collapse
Affiliation(s)
- Leonora Niv-Spector
- The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University, Rehovot 76100, Israel
| | | | | | | | | | | | | |
Collapse
|
50
|
NPM-ALK: The Prototypic Member of a Family of Oncogenic Fusion Tyrosine Kinases. JOURNAL OF SIGNAL TRANSDUCTION 2012; 2012:123253. [PMID: 22852078 PMCID: PMC3407651 DOI: 10.1155/2012/123253] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/21/2012] [Accepted: 04/28/2012] [Indexed: 02/07/2023]
Abstract
Anaplastic lymphoma kinase (ALK) was first identified in 1994 with the discovery that the gene encoding for this kinase was involved in the t(2;5)(p23;q35) chromosomal translocation observed in a subset of anaplastic large cell lymphoma (ALCL). The NPM-ALK fusion protein generated by this translocation is a constitutively active tyrosine kinase, and much research has focused on characterizing the signalling pathways and cellular activities this oncoprotein regulates in ALCL. We now know about the existence of nearly 20 distinct ALK translocation partners, and the fusion proteins resulting from these translocations play a critical role in the pathogenesis of a variety of cancers including subsets of large B-cell lymphomas, nonsmall cell lung carcinomas, and inflammatory myofibroblastic tumours. Moreover, the inhibition of ALK has been shown to be an effective treatment strategy in some of these malignancies. In this paper we will highlight malignancies where ALK translocations have been identified and discuss why ALK fusion proteins are constitutively active tyrosine kinases. Finally, using ALCL as an example, we will examine three key signalling pathways activated by NPM-ALK that contribute to proliferation and survival in ALCL.
Collapse
|