1
|
Gao H, Nepovimova E, Adam V, Heger Z, Valko M, Wu Q, Kuca K. Age-associated changes in innate and adaptive immunity: role of the gut microbiota. Front Immunol 2024; 15:1421062. [PMID: 39351234 PMCID: PMC11439693 DOI: 10.3389/fimmu.2024.1421062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2024] [Accepted: 08/26/2024] [Indexed: 10/04/2024] Open
Abstract
Aging is generally regarded as an irreversible process, and its intricate relationship with the immune system has garnered significant attention due to its profound implications for the health and well-being of the aging population. As people age, a multitude of alterations occur within the immune system, affecting both innate and adaptive immunity. In the realm of innate immunity, aging brings about changes in the number and function of various immune cells, including neutrophils, monocytes, and macrophages. Additionally, certain immune pathways, like the cGAS-STING, become activated. These alterations can potentially result in telomere damage, the disruption of cytokine signaling, and impaired recognition of pathogens. The adaptive immune system, too, undergoes a myriad of changes as age advances. These include shifts in the number, frequency, subtype, and function of T cells and B cells. Furthermore, the human gut microbiota undergoes dynamic changes as a part of the aging process. Notably, the interplay between immune changes and gut microbiota highlights the gut's role in modulating immune responses and maintaining immune homeostasis. The gut microbiota of centenarians exhibits characteristics akin to those found in young individuals, setting it apart from the microbiota observed in typical elderly individuals. This review delves into the current understanding of how aging impacts the immune system and suggests potential strategies for reversing aging through interventions in immune factors.
Collapse
Affiliation(s)
- Haoyu Gao
- College of Life Science, Yangtze University, Jingzhou, China
| | - Eugenie Nepovimova
- Department of Chemistry, Faculty of Science, University of Hradec Králové, Hradec Králové, Czechia
| | - Vojtech Adam
- Department of Chemistry and Biochemistry, Mendel University in Brno, Brno, Czechia
| | - Zbynek Heger
- Department of Chemistry and Biochemistry, Mendel University in Brno, Brno, Czechia
| | - Marian Valko
- Faculty of Chemical and Food Technology, Slovak University of Technology, Bratislava, Slovakia
| | - Qinghua Wu
- College of Life Science, Yangtze University, Jingzhou, China
- Department of Chemistry, Faculty of Science, University of Hradec Králové, Hradec Králové, Czechia
| | - Kamil Kuca
- Department of Chemistry, Faculty of Science, University of Hradec Králové, Hradec Králové, Czechia
- Andalusian Research Institute in Data Science and Computational Intelligence (DaSCI), University of Granada, Granada, Spain
| |
Collapse
|
2
|
Amann EM, Gowdavally S, Tsamadou C, Platzbecker U, Sala E, Wagner-Drouet E, Valerius T, Kröger N, Wulf G, Einsele H, Thurner L, Schaefer-Eckart K, Freitag S, Casper J, Dürholt M, Kaufmann M, Hertenstein B, Klein S, Ringhoffer M, Frank S, Saal T, Schmid-Möglich A, Neuchel C, Schrezenmeier H, Mytilineos J, Fürst D. The impact of MICB mismatches in unrelated haematopoietic stem cell transplantation. HLA 2024; 103:e15584. [PMID: 38932717 DOI: 10.1111/tan.15584] [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: 10/19/2023] [Revised: 05/28/2024] [Accepted: 06/14/2024] [Indexed: 06/28/2024]
Abstract
MICA polymorphisms have been associated with increased incidence of acute GvHD and adverse outcome in allogeneic haematopoietic stem cell transplantation (HSCT). MICB is another expressed member of MHC class I-related chain genes and its impact on HSCT outcome is yet to be fully defined. We typed a large cohort of patients and donors for MICB polymorphisms and investigated the impact of MICB matching on outcome after unrelated HSCT. 69.2% of the patients were 10/10 human leukocyte antigen (HLA) matched and 30.8% were 9/10 HLA matched. MICB typing was performed using a short amplicon-based NGS typing assay on the Illumina MiSeq platform. Differences in proteins were considered as mismatches. MICA polymorphisms were identified as possible confounder and were therefore included as parameter in the multivariate analyses. Due to the strong linkage disequilibrium with the classical HLA-genes, sub-stratification for HLA matching status was necessary, and no effect of MICB mismatches was seen in the 10/10 HLA matched group when compared to the MICB matched cases. However, in the 9/10 HLA matched group, MICB mismatched cases showed significantly worse disease free survival (DFS), GvHD and relapse free survival (GRFS) compared to the MICB matched cases (DFS: HR 1.24, p = 0.011; GRFS: HR 1.26, p = 0.002). MICA mismatches had no impact on any outcome parameter. According to our findings, effects previously attributed to MICA differences may have been confounded by MICB polymorphisms. We show that MICB differences contribute a small but relevant effect in 9/10 HLA-matched transplantations, which in turn highlights the possible usefulness of MICB typing in donor selection among similarly suitable 9/10 matched donors, especially when HLA-B mismatches have to be accepted.
Collapse
Affiliation(s)
- Elisa Maria Amann
- Institute of Clinical Transfusion Medicine and Immunogenetics Ulm, German Red Cross Blood Transfusion Service, Baden Wuerttemberg - Hessen, Ulm, and University Hospital Ulm, Ulm, Germany
- Institute of Transfusion Medicine, University of Ulm, Ulm, Germany
| | - Sowmya Gowdavally
- Institute of Clinical Transfusion Medicine and Immunogenetics Ulm, German Red Cross Blood Transfusion Service, Baden Wuerttemberg - Hessen, Ulm, and University Hospital Ulm, Ulm, Germany
| | - Chrysanthi Tsamadou
- Institute of Clinical Transfusion Medicine and Immunogenetics Ulm, German Red Cross Blood Transfusion Service, Baden Wuerttemberg - Hessen, Ulm, and University Hospital Ulm, Ulm, Germany
- Institute of Transfusion Medicine, University of Ulm, Ulm, Germany
| | - Uwe Platzbecker
- Department of Hematology/Oncology, University of Leipzig, Leipzig, Germany
| | - Elisa Sala
- Department of Internal Medicine III, University of Ulm, Ulm, Germany
| | - Eva Wagner-Drouet
- Department of Medicine III, Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Thomas Valerius
- Section for Stem Cell Transplantation and Immunotherapy, Department of Medicine II, Christian Albrechts University, Kiel, Germany
| | - Nicolaus Kröger
- Department of Stem Cell Transplantation, University Hospital Hamburg Eppendorf, Hamburg, Germany
| | - Gerald Wulf
- Department of Hematology/Oncology, Georg-August-University Göttingen, Göttingen, Germany
| | - Hermann Einsele
- Department of Internal Medicine II, University Hospital Würzburg, Würzburg, Germany
| | - Lorenz Thurner
- Department Internal Medicine I, Universitätsklinikum des Saarlandes, Homburg, Germany
| | | | - Sebastian Freitag
- Department of Medicine III, Hematology/Oncology/Palliative Care, Rostock University Medical Center, Rostock, Germany
| | - Jochen Casper
- Department of Oncology and Hematology, Klinikum Oldenburg, University Clinic, Oldenburg, Germany
| | - Mareike Dürholt
- Hematology/Oncology, Evangelic Clinic Essen-Werden, Essen, Germany
| | - Martin Kaufmann
- 2nd Department of Internal Medicine, Oncology and Hematology, Robert Bosch Hospital Stuttgart, Stuttgart, Germany
| | | | - Stefan Klein
- Universitätsmedizin Mannheim, Med. Klinik III, Mannheim, Germany
| | - Mark Ringhoffer
- Medizinische Klinik III, Städtisches Klinikum Karlsruhe, Karlsruhe, Germany
| | - Sandra Frank
- DRST - German Registry for Stem Cell Transplantation, Ulm, Germany
| | - Teresa Saal
- Institute of Clinical Transfusion Medicine and Immunogenetics Ulm, German Red Cross Blood Transfusion Service, Baden Wuerttemberg - Hessen, Ulm, and University Hospital Ulm, Ulm, Germany
- Institute of Transfusion Medicine, University of Ulm, Ulm, Germany
| | - Amelie Schmid-Möglich
- Institute of Clinical Transfusion Medicine and Immunogenetics Ulm, German Red Cross Blood Transfusion Service, Baden Wuerttemberg - Hessen, Ulm, and University Hospital Ulm, Ulm, Germany
- Institute of Transfusion Medicine, University of Ulm, Ulm, Germany
| | - Christine Neuchel
- Institute of Clinical Transfusion Medicine and Immunogenetics Ulm, German Red Cross Blood Transfusion Service, Baden Wuerttemberg - Hessen, Ulm, and University Hospital Ulm, Ulm, Germany
- Institute of Transfusion Medicine, University of Ulm, Ulm, Germany
| | - Hubert Schrezenmeier
- Institute of Clinical Transfusion Medicine and Immunogenetics Ulm, German Red Cross Blood Transfusion Service, Baden Wuerttemberg - Hessen, Ulm, and University Hospital Ulm, Ulm, Germany
- Institute of Transfusion Medicine, University of Ulm, Ulm, Germany
| | - Joannis Mytilineos
- DRST - German Registry for Stem Cell Transplantation, Ulm, Germany
- ZKRD - Zentrales Knochenmarkspender-Register für Deutschland, German National Bone Marrow Donor Registry, Ulm, Germany
| | - Daniel Fürst
- Institute of Clinical Transfusion Medicine and Immunogenetics Ulm, German Red Cross Blood Transfusion Service, Baden Wuerttemberg - Hessen, Ulm, and University Hospital Ulm, Ulm, Germany
- Institute of Transfusion Medicine, University of Ulm, Ulm, Germany
| |
Collapse
|
3
|
Hovhannisyan L, Riether C, Aebersold DM, Medová M, Zimmer Y. CAR T cell-based immunotherapy and radiation therapy: potential, promises and risks. Mol Cancer 2023; 22:82. [PMID: 37173782 PMCID: PMC10176707 DOI: 10.1186/s12943-023-01775-1] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Accepted: 04/11/2023] [Indexed: 05/15/2023] Open
Abstract
CAR T cell-based therapies have revolutionized the treatment of hematological malignancies such as leukemia and lymphoma within the last years. In contrast to the success in hematological cancers, the treatment of solid tumors with CAR T cells is still a major challenge in the field and attempts to overcome these hurdles have not been successful yet. Radiation therapy is used for management of various malignancies for decades and its therapeutic role ranges from local therapy to a priming agent in cancer immunotherapy. Combinations of radiation with immune checkpoint inhibitors have already proven successful in clinical trials. Therefore, a combination of radiation therapy may have the potential to overcome the current limitations of CAR T cell therapy in solid tumor entities. So far, only limited research was conducted in the area of CAR T cells and radiation. In this review we will discuss the potential and risks of such a combination in the treatment of cancer patients.
Collapse
Affiliation(s)
- Lusine Hovhannisyan
- Department of Radiation Oncology, Inselspital, Bern University Hospital, Freiburgstrasse 8, Bern, 3008, Switzerland
- Department for Biomedical Research, Radiation Oncology, University of Bern, Murtenstrasse 35, Bern, 3008, Switzerland
- Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, 3010, Switzerland
| | - Carsten Riether
- Department of Medical Oncology, Inselspital, University Hospital and University of Bern, Bern, 3010, Switzerland
| | - Daniel M Aebersold
- Department of Radiation Oncology, Inselspital, Bern University Hospital, Freiburgstrasse 8, Bern, 3008, Switzerland
- Department for Biomedical Research, Radiation Oncology, University of Bern, Murtenstrasse 35, Bern, 3008, Switzerland
| | - Michaela Medová
- Department of Radiation Oncology, Inselspital, Bern University Hospital, Freiburgstrasse 8, Bern, 3008, Switzerland
- Department for Biomedical Research, Radiation Oncology, University of Bern, Murtenstrasse 35, Bern, 3008, Switzerland
| | - Yitzhak Zimmer
- Department of Radiation Oncology, Inselspital, Bern University Hospital, Freiburgstrasse 8, Bern, 3008, Switzerland.
- Department for Biomedical Research, Radiation Oncology, University of Bern, Murtenstrasse 35, Bern, 3008, Switzerland.
| |
Collapse
|
4
|
Iacob S, Cicinnati V, Kabar I, Hüsing-Kabar A, Radtke A, Iacob R, Baba H, Schmidt HH, Paul A, Beckebaum S. Prediction of late allograft dysfunction following liver transplantation by immunological blood biomarkers. Transpl Immunol 2021; 69:101448. [PMID: 34391882 DOI: 10.1016/j.trim.2021.101448] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2021] [Revised: 08/09/2021] [Accepted: 08/10/2021] [Indexed: 02/09/2023]
Abstract
BACKGROUND An accelerated course of hepatic fibrosis may occur in liver transplantation (LT) patients despite normal or slightly abnormal liver blood tests. AIM To identify screening tools based on blood biomarkers to predict late allograft dysfunction in LT recipients. METHODS 174 LT recipients were enrolled. Liver biopsy, liver functional tests, cytokine quantitation in serum, as well as soluble MHC class I polypeptide-related sequence A and B (sMICA/sMICB) and soluble UL16 binding protein 2 (sULBP2) were performed. RESULTS Patients with late graft dysfunction had a significantly higher donor age, lower albumin level, higher alanine (ALT) and aspartate aminotransferase (AST), gamma-glutamyl transpeptidase (GGT), total bilirubin and alkaline phosphatase (ALP), higher sMICA, sULBP2, higher interleukin (IL) 6, interferon γ and lower IL10 in serum as compared to recipients without allograft dysfunction. In order to provide a better statistical accuracy for discriminating 5-year allograft dysfunction from other less progressive subtype of allograft injury, we established a predictive model, based on 7 parameters (serum ALP, ALT, AST, GGT, sMICA, IL6 and albumin) which provided an Area Under the Receiver Operating Characteristics (AUROC) curve of 0.905. CONCLUSIONS Blood-based biomarkers can significantly improve prediction of late liver allograft outcome in LT patients. The new developed score comprising serum parameters, with an excellent AUROC, can be reliably used for diagnosing late allograft dysfunction in transplanted patients.
Collapse
Affiliation(s)
- Speranta Iacob
- Center for Digestive Diseases and Liver Transplantation, Fundeni Clinical Institute, Bucharest, Romania; Carol Davila University of Medicine and Pharmacy, Bucharest, Romania.
| | - Vito Cicinnati
- Department of Medicine B for Gastroenterology and Hepatology, University Hospital Muenster, 48149 Muenster, Germany
| | - Iyad Kabar
- Department of Medicine B for Gastroenterology and Hepatology, University Hospital Muenster, 48149 Muenster, Germany
| | - Anna Hüsing-Kabar
- Department of Medicine B for Gastroenterology and Hepatology, University Hospital Muenster, 48149 Muenster, Germany
| | - Arnold Radtke
- Department of General, Visceral and Transplant Surgery, Comprehensive Cancer Center, University of Tübingen, 72076 Tübingen, Germany
| | - Razvan Iacob
- Center for Digestive Diseases and Liver Transplantation, Fundeni Clinical Institute, Bucharest, Romania; Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
| | - Hideo Baba
- Institute for Pathology, University Hospital Essen, 45147 Essen, Germany
| | - Hartmut H Schmidt
- Department of Medicine B for Gastroenterology and Hepatology, University Hospital Muenster, 48149 Muenster, Germany
| | - Andreas Paul
- Department of General, Visceral and Transplantation Surgery, University Hospital Essen, 45147 Essen, Germany
| | - Susanne Beckebaum
- Department of Medicine B for Gastroenterology and Hepatology, University Hospital Muenster, 48149 Muenster, Germany
| |
Collapse
|
5
|
Constanzo J, Faget J, Ursino C, Badie C, Pouget JP. Radiation-Induced Immunity and Toxicities: The Versatility of the cGAS-STING Pathway. Front Immunol 2021; 12:680503. [PMID: 34079557 PMCID: PMC8165314 DOI: 10.3389/fimmu.2021.680503] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Accepted: 04/26/2021] [Indexed: 12/20/2022] Open
Abstract
In the past decade, radiation therapy (RT) entered the era of personalized medicine, following the striking improvements in radiation delivery and treatment planning optimization, and in the understanding of the cancer response, including the immunological response. The next challenge is to identify the optimal radiation regimen(s) to induce a clinically relevant anti-tumor immunity response. Organs at risks and the tumor microenvironment (e.g. endothelial cells, macrophages and fibroblasts) often limit the radiation regimen effects due to adverse toxicities. Here, we reviewed how RT can modulate the immune response involved in the tumor control and side effects associated with inflammatory processes. Moreover, we discussed the versatile roles of tumor microenvironment components during RT, how the innate immune sensing of RT-induced genotoxicity, through the cGAS-STING pathway, might link the anti-tumor immune response, radiation-induced necrosis and radiation-induced fibrosis, and how a better understanding of the switch between favorable and deleterious events might help to define innovative approaches to increase RT benefits in patients with cancer.
Collapse
Affiliation(s)
- Julie Constanzo
- IRCM, Institut de Recherche en Cancérologie de Montpellier, INSERM U1194, Université de Montpellier, Institut régional du Cancer de Montpellier, Montpellier, France
| | - Julien Faget
- IRCM, Institut de Recherche en Cancérologie de Montpellier, INSERM U1194, Université de Montpellier, Institut régional du Cancer de Montpellier, Montpellier, France
| | - Chiara Ursino
- IRCM, Institut de Recherche en Cancérologie de Montpellier, INSERM U1194, Université de Montpellier, Institut régional du Cancer de Montpellier, Montpellier, France
| | - Christophe Badie
- Cancer Mechanisms and Biomarkers Group, Radiation Effects Department, Centre for Radiation, Chemical & Environmental Hazards Public Health England Chilton, Didcot, United Kingdom
| | - Jean-Pierre Pouget
- IRCM, Institut de Recherche en Cancérologie de Montpellier, INSERM U1194, Université de Montpellier, Institut régional du Cancer de Montpellier, Montpellier, France
| |
Collapse
|
6
|
Gomez Hernandez MP, Starman EE, Davis AB, Harishchandra Hikkaduwa Withanage M, Zeng E, Lieberman SM, Brogden KA, Lanzel EA. A distinguishing profile of chemokines, cytokines, and biomarkers in the saliva of children with Sjögren's syndrome. Rheumatology (Oxford) 2021; 60:4765-4777. [PMID: 33512494 PMCID: PMC8487313 DOI: 10.1093/rheumatology/keab098] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 01/17/2021] [Indexed: 01/08/2023] Open
Abstract
Objective SS is an autoimmune disease most commonly diagnosed in adults but can occur in children. Our objective was to assess the presence of chemokines, cytokines and biomarkers (CCBMs) in saliva from these children that were associated with lymphocyte and mononuclear cell functions. Methods Saliva was collected from 11 children diagnosed with SS prior to age 18 years and 16 normal healthy children. A total of 105 CCBMs were detected in multiplex microparticle-based immunoassays. ANOVA and t test (0.05 level) were used to detect differences. Ingenuity Pathway Analysis (IPA) was used to assess whether elevated CCBMs were in annotations associated with immune system diseases and select leukocyte activities and functions. Machine learning methods were used to evaluate the predictive power of these CCBMs for SS and were measured by receiver operating characteristic (ROC) curve and area under curve (AUC). Results Of the 105 CCBMs detected, 43 (40.9%) differed in children with SS from those in healthy study controls (P < 0.05) and could differentiate the two groups (P < 0.05). Elevated CCBMs in IPA annotations were associated with autoimmune diseases and with leukocyte chemotaxis, migration, proliferation, and regulation of T cell activation. The best AUC value in ROC analysis was 0.93, indicating that there are small numbers of CCBMs that may be useful for diagnosis of SS. Conclusion While 35 of these 43 CCBMs have been previously reported in SS, 8 CCBMs had not. Additional studies focusing on these CCBMs may provide further insight into disease pathogenesis and may contribute to diagnosis of SS in children.
Collapse
Affiliation(s)
| | - Emily E Starman
- Iowa Institute for Oral Health Research, College of Dentistry, University of Iowa, Iowa City, IA USA
| | - Andrew B Davis
- Department of Otolaryngology, College of Medicine, University of Iowa, Iowa City, IA USA
| | | | - Erliang Zeng
- Division of Biostatistics and Computational Biology, College of Dentistry, University of Iowa, Iowa City, IA USA
| | - Scott M Lieberman
- Stead Family Department of Pediatrics-Division of Rheumatology, Allergy and Immunology, Carver College of Medicine, University of Iowa, Iowa City, IA USA
| | - Kim A Brogden
- Iowa Institute for Oral Health Research, College of Dentistry, University of Iowa, Iowa City, IA USA
| | - Emily A Lanzel
- Department of Oral Pathology, Radiology and Medicine, College of Dentistry, University of Iowa, Iowa City, IA USA
| |
Collapse
|
7
|
Ni M, Wang L, Yang M, Neuber B, Sellner L, Hückelhoven-Krauss A, Schubert ML, Luft T, Hegenbart U, Schönland S, Wuchter P, Chen BA, Eckstein V, Krüger W, Yerushalmi R, Beider K, Nagler A, Müller-Tidow C, Dreger P, Schmitt M, Schmitt A. Shaping of CD56 bri Natural Killer Cells in Patients With Steroid-Refractory/Resistant Acute Graft-vs.-Host Disease via Extracorporeal Photopheresis. Front Immunol 2019; 10:547. [PMID: 30949182 PMCID: PMC6436423 DOI: 10.3389/fimmu.2019.00547] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Accepted: 02/28/2019] [Indexed: 11/24/2022] Open
Abstract
CD56bri natural killer (NK) cells play an important role in the pathogenesis of graft-vs. -host disease (GVHD) and immune defense in the early period after allogeneic hematopoietic stem cell transplantation. Extracorporeal photopheresis (ECP) as an immunomodulating therapy has been widely used for GVHD treatment. However, the mechanism of action of ECP still remains to be elucidated, particularly the influence of ECP on NK cells. Thirty-four patients with steroid-refractory/resistant acute GVHD (aGVHD) ≥ °II and moderate to severe chronic GVHD (cGVHD) received ECP therapy. Patient samples obtained during intensive and long-term treatment were analyzed. Immunomonitoring with respect to cell phenotype and function was performed on rested peripheral blood mononuclear cells (PBMCs) using multiparametric flow cytometry. NK activity in terms of cytokine release was analyzed by intracellular cytokine staining after co-culture with K562 cells. Moreover, the proliferative capacity of NK cells, CD4+, and CD8+ T cells was determined by carboxyfluorescein succinimidyl ester (CFSE) staining. Clinically, 75% of aGVHD and 78% of cGVHD patients responded to ECP therapy. Moreover, our data show that aGVHD, cGVHD patients and healthy donors (HDs) present distinct NK patterns: aGVHD patients have a higher frequency of CD56bri NK subsets with stronger NKG2D and CD62L expression, while CD56−CD16+ NK cells with higher expression of CD57 and CD11b stand out as a signature population for cGVHD. ECP therapy could significantly decrease CD56briCD16− NK cells with shifting the quality from a cytotoxic to a regulatory pattern and additionally mature CD56dim NK cells via upregulation of CD57 in complete responding aGVHD patients. Moreover, ECP could keep the anti-viral and anti-leukemic effects intact via maintaining specialized anti-viral/leukemic CD57+NKG2C+CD56dim NK cells as well as remaining the quality and quantity of cytokine release by NK cells. The proliferative capacity of effector cells remained constant over ECP therapy. In conclusion, ECP represents an attractive option to treat GVHD without compromising anti-viral/leukemic effects. Shaping of CD56bri NK cell compartment by downregulating the cytotoxic subset while upregulating the regulatory subset contributes to the mechanisms of ECP therapy in aGVHD.
Collapse
Affiliation(s)
- Ming Ni
- Department of Internal Medicine V, University Clinic Heidelberg, Heidelberg, Germany.,Department of Hematology, the Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Lei Wang
- Department of Internal Medicine V, University Clinic Heidelberg, Heidelberg, Germany
| | - Mingya Yang
- Department of Internal Medicine V, University Clinic Heidelberg, Heidelberg, Germany
| | - Brigitte Neuber
- Department of Internal Medicine V, University Clinic Heidelberg, Heidelberg, Germany
| | - Leopold Sellner
- Department of Internal Medicine V, University Clinic Heidelberg, Heidelberg, Germany
| | | | - Maria-Luisa Schubert
- Department of Internal Medicine V, University Clinic Heidelberg, Heidelberg, Germany
| | - Thomas Luft
- Department of Internal Medicine V, University Clinic Heidelberg, Heidelberg, Germany
| | - Ute Hegenbart
- Department of Internal Medicine V, University Clinic Heidelberg, Heidelberg, Germany
| | - Stefan Schönland
- Department of Internal Medicine V, University Clinic Heidelberg, Heidelberg, Germany
| | - Patrick Wuchter
- German Red Cross Blood Service Baden-Wuerttemberg-Hessen, Institute of Transfusion Medicine and Immunology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Bao-An Chen
- Department of Hematology, Zhongda Hospital, Southeast University, Nanjing, China
| | - Volker Eckstein
- Department of Internal Medicine V, University Clinic Heidelberg, Heidelberg, Germany
| | - William Krüger
- Department of Internal Medicine C, Hematology, Oncology, Stem Cell Transplantation, Palliative Care, University Clinic Greifswald, Greifswald, Germany
| | - Ronit Yerushalmi
- Hematology Division, Chaim Sheba Medical Center, Tel Hashomer, Ramat Gan, Israel
| | - Katia Beider
- Hematology Division, Chaim Sheba Medical Center, Tel Hashomer, Ramat Gan, Israel
| | - Arnon Nagler
- Hematology Division, Chaim Sheba Medical Center, Tel Hashomer, Ramat Gan, Israel
| | - Carsten Müller-Tidow
- Department of Internal Medicine V, University Clinic Heidelberg, Heidelberg, Germany
| | - Peter Dreger
- Department of Internal Medicine V, University Clinic Heidelberg, Heidelberg, Germany
| | - Michael Schmitt
- Department of Internal Medicine V, University Clinic Heidelberg, Heidelberg, Germany
| | - Anita Schmitt
- Department of Internal Medicine V, University Clinic Heidelberg, Heidelberg, Germany
| |
Collapse
|
8
|
Wu D, Zhang J, Qian T, Dai Y, Mashaghi A, Xu J, Hong J. IFN-γ Regulates the Expression of MICA in Human Corneal Epithelium Through miRNA4448 and NFκB. Front Immunol 2018; 9:1530. [PMID: 30013574 PMCID: PMC6036181 DOI: 10.3389/fimmu.2018.01530] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Accepted: 06/20/2018] [Indexed: 12/14/2022] Open
Abstract
Purpose Major histocompatibility complex class I-related chain A (MICA), a non-classical major histocompatibility complex molecule, can stimulate or co-stimulate CD8+ T cells or natural killer (nk) cells, thus affecting cornea allograft survival. This study investigated IFN-γ regulation of MICA expression levels in human corneal epithelium by miRNA4448. Methods MICA expression levels in human corneal epithelial cells (HCECs) stimulated with IFN-γ were detected by qRT-PCR and an enzyme-linked immunosorbent assay, and differential miRNA expression levels were measured. qRT-PCR, Western blotting, and immunofluorescence staining revealed nuclear factor kappa B (NFκB)/P65 expression in IFN-γ-treated and miRNA4448-overexpressed HCECs. A luciferase reporter assay was used to predict the interaction between NFκB and MICA. Additionally, HCECs were transfected with MICA plasmid or treated with IFN-γ and NKG2D-mAb and cocultured with NK cells and CD8+ T cells. Cell apoptosis was measured using Annexin V/PI staining. qRT-PCR detected the expression of anti-apoptosis factor Survivin and apoptosis factor Caspase 3 in MICA-transfected and IFN-γ-treated HCECs after co-culturing with NK cells and CD8+ T cells. Results IFN-γ (500 ng/ml, 24 h) upregulated MICA expression in HCECs in vitro. Among six differentially expressed microRNAs, miRNA4448 levels decreased the most after IFN-γ treatment. The overexpression of miRNA4448 decreased MICA expression. miRNA4448 downregulated NFκB/P65 expression in IFN-γ-induced HCEC, and it was determined that NFκB/P65 directly targeted MICA by binding to the promotor region. A coculture with NK cells and CD8+ T cells demonstrated that MICA overexpression enhanced HCEC apoptosis, which could be inhibited by NKG2D-mAb. Simultaneously, Survivin mRNA expression decreased and Caspase3 mRNA expression increased upon the interaction between MICA and NK (CD8+ T) cells in HCECs. Conclusion IFN-γ enhances the expression of MICA in HCECs by modulating miRNA4448 and NFκB/P65 levels, thereby contributing to HCEC apoptosis induced by NK and CD8+ T cells. This discovery may lead to new insights into the pathogenesis of corneal allograft rejection.
Collapse
Affiliation(s)
- Dan Wu
- Department of Ophthalmology, Eye, Ear, Nose, and Throat Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jing Zhang
- Department of Ophthalmology, Eye, Ear, Nose, and Throat Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Tingting Qian
- Department of Immunology and Biotherapy Research Center, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yiqin Dai
- Department of Ophthalmology, Eye, Ear, Nose, and Throat Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Alireza Mashaghi
- Leiden Academic Centre for Drug Research, Faculty of Mathematics and Natural Sciences, Leiden University, Leiden, Netherlands
| | - Jianjiang Xu
- Department of Ophthalmology, Eye, Ear, Nose, and Throat Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jiaxu Hong
- Department of Ophthalmology, Eye, Ear, Nose, and Throat Hospital, Shanghai Medical College, Fudan University, Shanghai, China.,Leiden Academic Centre for Drug Research, Faculty of Mathematics and Natural Sciences, Leiden University, Leiden, Netherlands.,Department of Ophthalmology, The Affiliated Hospital of Guizhou Medical University, Guiyang, China.,Key Laboratory of Myopia, Ministry of Health (Fudan University), Shanghai, China.,Shanghai Key Laboratory of Visual Impairment and Restoration, Fudan University, Shanghai, China
| |
Collapse
|
9
|
Desimio MG, Giuliani E, Doria M. The histone deacetylase inhibitor SAHA simultaneously reactivates HIV-1 from latency and up-regulates NKG2D ligands sensitizing for natural killer cell cytotoxicity. Virology 2017; 510:9-21. [PMID: 28689087 DOI: 10.1016/j.virol.2017.06.033] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Revised: 06/14/2017] [Accepted: 06/29/2017] [Indexed: 01/01/2023]
Abstract
In pilot HIV-1 eradication studies, patients' immune responses were ineffective at killing viral reservoirs reactivated through latency reversing agents (LRAs) like suberoylanilide hydroxamic acid (SAHA). We hypothesized that T cells harboring reactivated HIV-1 express MIC and ULBP ligands for the activating NKG2D receptor of natural killer (NK) cells. Here, we demonstrated that MICA/B and ULBP2 are induced by SAHA on primary T cells harboring reactivated virus. Using latently HIV-1-infected J-Lat 6.3/8.4/9.2 and J1.1 cell lines, we showed that SAHA reverts latency and, simultaneously, up-regulates MICA/B and ULBP2 acting at the transcriptional level and through ATR activation, thus sensitizing T cells with reactivated virus to NKG2D-mediated killing by NK cells. Moreover, IL-2 and IL-15 potently boosted NKG2D expression and cytotoxicity of NK cells against SAHA-reactivated p24+ target cells. Therefore, immunotherapy with cytokines enhancing NKG2D-mediated NK-cell cytotoxicity combined with administration of LRAs up-modulating NKG2D ligands, represents a promising approach towards HIV-1 eradication.
Collapse
Affiliation(s)
| | - Erica Giuliani
- Laboratory of Immunoinfectivology, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Margherita Doria
- Laboratory of Immunoinfectivology, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy.
| |
Collapse
|
10
|
Guo Q, Zhu D, Bu X, Wei X, Li C, Gao D, Wei X, Ma X, Zhao P. Efficient killing of radioresistant breast cancer cells by cytokine-induced killer cells. Tumour Biol 2017; 39:1010428317695961. [PMID: 28349820 DOI: 10.1177/1010428317695961] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Recurrence of breast cancer after radiotherapy may be partly explained by the presence of radioresistant cells. Thus, it would be desirable to develop an effective therapy against radioresistant cells. In this study, we demonstrated the intense antitumor activity of cytokine-induced killer cells against MCF-7 and radioresistant MCF-7 cells, as revealed by cytokine-induced killer-mediated cytotoxicity, tumor cell proliferation, and tumor invasion. Radioresistant MCF-7 cells were more susceptible to cytokine-induced killer cell killing. The stronger cytotoxicity of cytokine-induced killer cells against radioresistant MCF-7 cells was dependent on the expression of major histocompatibility complex class I polypeptide-related sequence A/B on radioresistant MCF-7 cells after exposure of cytokine-induced killer cells to sensitized targets. In addition, we demonstrated that cytokine-induced killer cell treatment sensitized breast cancer cells to chemotherapy via the downregulation of TK1, TYMS, and MDR1. These results indicate that cytokine-induced killer cell treatment in combination with radiotherapy and/or chemotherapy may induce synergistic antitumor activities and represent a novel strategy for breast cancer.
Collapse
Affiliation(s)
- Qingming Guo
- 1 Biotherapy Center, Qingdao Central Hospital, The Second Affiliated Hospital of Qingdao University Medical College, Qingdao, China
| | - Danni Zhu
- 1 Biotherapy Center, Qingdao Central Hospital, The Second Affiliated Hospital of Qingdao University Medical College, Qingdao, China
| | - Xiaocui Bu
- 2 The Affiliated Cardiovascular Hospital of Qingdao University Medical College, Qingdao, China
| | - Xiaofang Wei
- 1 Biotherapy Center, Qingdao Central Hospital, The Second Affiliated Hospital of Qingdao University Medical College, Qingdao, China
| | - Changyou Li
- 1 Biotherapy Center, Qingdao Central Hospital, The Second Affiliated Hospital of Qingdao University Medical College, Qingdao, China
| | - Daiqing Gao
- 1 Biotherapy Center, Qingdao Central Hospital, The Second Affiliated Hospital of Qingdao University Medical College, Qingdao, China
| | - Xiaoqiang Wei
- 3 Department of Gynaecology, Qingdao Central Hospital, The Second Affiliated Hospital of Qingdao University Medical College, Qingdao, China
| | - Xuezhen Ma
- 4 Department of Oncology, Qingdao Central Hospital, The Second Affiliated Hospital of Qingdao University Medical College, Qingdao, China
| | - Peng Zhao
- 1 Biotherapy Center, Qingdao Central Hospital, The Second Affiliated Hospital of Qingdao University Medical College, Qingdao, China
| |
Collapse
|
11
|
Gam R, Shah P, Crossland RE, Norden J, Dickinson AM, Dressel R. Genetic Association of Hematopoietic Stem Cell Transplantation Outcome beyond Histocompatibility Genes. Front Immunol 2017; 8:380. [PMID: 28421078 PMCID: PMC5377073 DOI: 10.3389/fimmu.2017.00380] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2016] [Accepted: 03/16/2017] [Indexed: 12/18/2022] Open
Abstract
The outcome of hematopoietic stem cell transplantation (HSCT) is controlled by genetic factors among which the leukocyte antigen human leukocyte antigen (HLA) matching is most important. In addition, minor histocompatibility antigens and non-HLA gene polymorphisms in genes controlling immune responses are known to contribute to the risks associated with HSCT. Besides single-nucleotide polymorphisms (SNPs) in protein coding genes, SNPs in regulatory elements such as microRNAs (miRNAs) contribute to these genetic risks. However, genetic risks require for their realization the expression of the respective gene or miRNA. Thus, gene and miRNA expression studies may help to identify genes and SNPs that indeed affect the outcome of HSCT. In this review, we summarize gene expression profiling studies that were performed in recent years in both patients and animal models to identify genes regulated during HSCT. We discuss SNP–mRNA–miRNA regulatory networks and their contribution to the risks associated with HSCT in specific examples, including forkheadbox protein 3 and regulatory T cells, the role of the miR-155 and miR-146a regulatory network for graft-versus-host disease, and the function of MICA and its receptor NKG2D for the outcome of HSCT. These examples demonstrate how SNPs affect expression or function of proteins that modulate the alloimmune response and influence the outcome of HSCT. Specific miRNAs targeting these genes and directly affecting expression of mRNAs are identified. It might be valuable in the future to determine SNPs and to analyze miRNA and mRNA expression in parallel in cohorts of HSCT patients to further elucidate genetic risks of HSCT.
Collapse
Affiliation(s)
- Rihab Gam
- Hematological Sciences, Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, UK
| | - Pranali Shah
- Institute of Cellular and Molecular Immunology, University Medical Centre Göttingen, Göttingen, Germany
| | - Rachel E Crossland
- Hematological Sciences, Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, UK
| | - Jean Norden
- Hematological Sciences, Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, UK
| | - Anne M Dickinson
- Hematological Sciences, Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, UK
| | - Ralf Dressel
- Institute of Cellular and Molecular Immunology, University Medical Centre Göttingen, Göttingen, Germany
| |
Collapse
|
12
|
Carapito R, Aouadi I, Ilias W, Bahram S. Natural Killer Group 2, Member D/NKG2D Ligands in Hematopoietic Cell Transplantation. Front Immunol 2017; 8:368. [PMID: 28396673 PMCID: PMC5366881 DOI: 10.3389/fimmu.2017.00368] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Accepted: 03/14/2017] [Indexed: 12/17/2022] Open
Abstract
Natural killer group 2, member D (NKG2D) is an invariant activatory receptor present on subsets of natural killer and T lymphocytes. It stimulates the cytolytic effector response upon engagement of its various stress-induced ligands NKG2D ligands (NKG2DL). Malignant transformation and conditioning treatment prior to hematopoietic cell transplantation (HCT) are stress factors leading to the activation of the NKG2D/NKG2DL signaling in clinical settings. In the context of HCT, NKG2D-bearing cells can kill both tumor and healthy cells expressing NKG2DL. The NKG2D/NKG2DL engagement has therefore a key role in the regulation of one of the most salient issues in allogeneic HCT, i.e., maintaining a balance between graft-vs.-leukemia effect and graft-vs.-host disease. The present review summarizes the current state of our knowledge pertaining to the role of the NKG2D and NKG2DL in HCT.
Collapse
Affiliation(s)
- Raphael Carapito
- ImmunoRhumatologie Moléculaire, INSERM UMR_S1109, LabEx TRANSPLANTEX, Centre de Recherche d'Immunologie et d'Hématologie, Faculté de Médecine, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Université de Strasbourg, Strasbourg, France; Laboratoire International Associé (LIA) INSERM, Strasbourg (France) - Nagano (Japan), Strasbourg, France; Fédération Hospitalo-Universitaire (FHU) OMICARE, Strasbourg, France; Laboratoire Central d'Immunologie, Pôle de Biologie, Nouvel Hôpital Civil, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | - Ismail Aouadi
- ImmunoRhumatologie Moléculaire, INSERM UMR_S1109, LabEx TRANSPLANTEX, Centre de Recherche d'Immunologie et d'Hématologie, Faculté de Médecine, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Université de Strasbourg, Strasbourg, France; Laboratoire International Associé (LIA) INSERM, Strasbourg (France) - Nagano (Japan), Strasbourg, France; Fédération Hospitalo-Universitaire (FHU) OMICARE, Strasbourg, France
| | - Wassila Ilias
- ImmunoRhumatologie Moléculaire, INSERM UMR_S1109, LabEx TRANSPLANTEX, Centre de Recherche d'Immunologie et d'Hématologie, Faculté de Médecine, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Université de Strasbourg, Strasbourg, France; Laboratoire International Associé (LIA) INSERM, Strasbourg (France) - Nagano (Japan), Strasbourg, France; Fédération Hospitalo-Universitaire (FHU) OMICARE, Strasbourg, France
| | - Seiamak Bahram
- ImmunoRhumatologie Moléculaire, INSERM UMR_S1109, LabEx TRANSPLANTEX, Centre de Recherche d'Immunologie et d'Hématologie, Faculté de Médecine, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Université de Strasbourg, Strasbourg, France; Laboratoire International Associé (LIA) INSERM, Strasbourg (France) - Nagano (Japan), Strasbourg, France; Fédération Hospitalo-Universitaire (FHU) OMICARE, Strasbourg, France; Laboratoire Central d'Immunologie, Pôle de Biologie, Nouvel Hôpital Civil, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| |
Collapse
|
13
|
Matching for the nonconventional MHC-I MICA gene significantly reduces the incidence of acute and chronic GVHD. Blood 2016; 128:1979-1986. [PMID: 27549307 DOI: 10.1182/blood-2016-05-719070] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Accepted: 08/14/2016] [Indexed: 01/12/2023] Open
Abstract
Graft-versus-host disease (GVHD) is among the most challenging complications in unrelated donor hematopoietic cell transplantation (HCT). The highly polymorphic MHC class I chain-related gene A, MICA, encodes a stress-induced glycoprotein expressed primarily on epithelia. MICA interacts with the invariant activating receptor NKG2D, expressed by cytotoxic lymphocytes, and is located in the MHC, next to HLA-B Hence, MICA has the requisite attributes of a bona fide transplantation antigen. Using high-resolution sequence-based genotyping of MICA, we retrospectively analyzed the clinical effect of MICA mismatches in a multicenter cohort of 922 unrelated donor HLA-A, HLA-B, HLA-C, HLA-DRB1, and HLA-DQB1 10/10 allele-matched HCT pairs. Among the 922 pairs, 113 (12.3%) were mismatched in MICA MICA mismatches were significantly associated with an increased incidence of grade III-IV acute GVHD (hazard ratio [HR], 1.83; 95% confidence interval [CI], 1.50-2.23; P < .001), chronic GVHD (HR, 1.50; 95% CI, 1.45-1.55; P < .001), and nonelapse mortality (HR, 1.35; 95% CI, 1.24-1.46; P < .001). The increased risk for GVHD was mirrored by a lower risk for relapse (HR, 0.50; 95% CI, 0.43-0.59; P < .001), indicating a possible graft-versus-leukemia effect. In conclusion, when possible, selecting a MICA-matched donor significantly influences key clinical outcomes of HCT in which a marked reduction of GVHD is paramount. The tight linkage disequilibrium between MICA and HLA-B renders identifying a MICA-matched donor readily feasible in clinical practice.
Collapse
|
14
|
Isernhagen A, Malzahn D, Viktorova E, Elsner L, Monecke S, von Bonin F, Kilisch M, Wermuth JM, Walther N, Balavarca Y, Stahl-Hennig C, Engelke M, Walter L, Bickeböller H, Kube D, Wulf G, Dressel R. The MICA-129 dimorphism affects NKG2D signaling and outcome of hematopoietic stem cell transplantation. EMBO Mol Med 2016; 7:1480-502. [PMID: 26483398 PMCID: PMC4644379 DOI: 10.15252/emmm.201505246] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The MHC class I chain-related molecule A (MICA) is a highly polymorphic ligand for the activating natural killer (NK)-cell receptor NKG2D. A single nucleotide polymorphism causes a valine to methionine exchange at position 129. Presence of a MICA-129Met allele in patients (n = 452) undergoing hematopoietic stem cell transplantation (HSCT) increased the chance of overall survival (hazard ratio [HR] = 0.77, P = 0.0445) and reduced the risk to die due to acute graft-versus-host disease (aGVHD) (odds ratio [OR] = 0.57, P = 0.0400) although homozygous carriers had an increased risk to experience this complication (OR = 1.92, P = 0.0371). Overall survival of MICA-129Val/Val genotype carriers was improved when treated with anti-thymocyte globulin (HR = 0.54, P = 0.0166). Functionally, the MICA-129Met isoform was characterized by stronger NKG2D signaling, triggering more NK-cell cytotoxicity and interferon-γ release, and faster co-stimulation of CD8+ T cells. The MICA-129Met variant also induced a faster and stronger down-regulation of NKG2D on NK and CD8+ T cells than the MICA-129Val isoform. The reduced cell surface expression of NKG2D in response to engagement by MICA-129Met variants appeared to reduce the severity of aGVHD.
Collapse
Affiliation(s)
- Antje Isernhagen
- Institute of Cellular and Molecular Immunology, University Medical Center Göttingen, Göttingen, Germany
| | - Dörthe Malzahn
- Institute of Genetic Epidemiology, University Medical Center Göttingen, Göttingen, Germany
| | - Elena Viktorova
- Institute of Genetic Epidemiology, University Medical Center Göttingen, Göttingen, Germany
| | - Leslie Elsner
- Institute of Cellular and Molecular Immunology, University Medical Center Göttingen, Göttingen, Germany
| | - Sebastian Monecke
- Institute of Cellular and Molecular Immunology, University Medical Center Göttingen, Göttingen, Germany
| | - Frederike von Bonin
- Department of Hematology and Medical Oncology, University Medical Center Göttingen, Göttingen, Germany
| | - Markus Kilisch
- Institute of Molecular Biology, University Medical Center Göttingen, Göttingen, Germany
| | - Janne Marieke Wermuth
- Department of Hematology and Medical Oncology, University Medical Center Göttingen, Göttingen, Germany
| | - Neele Walther
- Department of Hematology and Medical Oncology, University Medical Center Göttingen, Göttingen, Germany
| | - Yesilda Balavarca
- Institute of Genetic Epidemiology, University Medical Center Göttingen, Göttingen, Germany
| | | | - Michael Engelke
- Institute of Cellular and Molecular Immunology, University Medical Center Göttingen, Göttingen, Germany
| | - Lutz Walter
- Primate Genetics Laboratory, German Primate Center, Göttingen, Germany
| | - Heike Bickeböller
- Institute of Genetic Epidemiology, University Medical Center Göttingen, Göttingen, Germany
| | - Dieter Kube
- Department of Hematology and Medical Oncology, University Medical Center Göttingen, Göttingen, Germany
| | - Gerald Wulf
- Department of Hematology and Medical Oncology, University Medical Center Göttingen, Göttingen, Germany
| | - Ralf Dressel
- Institute of Cellular and Molecular Immunology, University Medical Center Göttingen, Göttingen, Germany
| |
Collapse
|
15
|
Gimondi S, Dugo M, Vendramin A, Bermema A, Biancon G, Cavané A, Corradini P, Carniti C. Circulating miRNA panel for prediction of acute graft-versus-host disease in lymphoma patients undergoing matched unrelated hematopoietic stem cell transplantation. Exp Hematol 2016; 44:624-634.e1. [DOI: 10.1016/j.exphem.2016.03.005] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2015] [Revised: 03/08/2016] [Accepted: 03/14/2016] [Indexed: 01/01/2023]
|
16
|
Abstract
Human and mouse NKG2D ligands (NKG2DLs) are absent or only poorly expressed by most normal cells but are upregulated by cell stress, hence, alerting the immune system in case of malignancy or infection. Although these ligands are numerous and highly variable (at genetic, genomic, structural, and biochemical levels), they all belong to the major histocompatibility complex class I gene superfamily and bind to a single, invariant, receptor: NKG2D. NKG2D (CD314) is an activating receptor expressed on NK cells and subsets of T cells that have a key role in the recognition and lysis of infected and tumor cells. Here, we review the molecular diversity of NKG2DLs, discuss the increasing appreciation of their roles in a variety of medical conditions, and propose several explanations for the evolutionary force(s) that seem to drive the multiplicity and diversity of NKG2DLs while maintaining their interaction with a single invariant receptor.
Collapse
Affiliation(s)
- Raphael Carapito
- ImmunoRhumatologie Moléculaire, INSERM UMR_S1109, LabEx TRANSPLANTEX, Centre de Recherche d'Immunologie et d'Hématologie, Faculté de Médecine, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Université de Strasbourg, Strasbourg, France.,Laboratoire International Associé (LIA) INSERM, Strasbourg (France) - Nagano (Japan), Strasbourg, France.,Fédération Hospitalo-Universitaire (FHU) OMICARE, Strasbourg, France
| | - Seiamak Bahram
- ImmunoRhumatologie Moléculaire, INSERM UMR_S1109, LabEx TRANSPLANTEX, Centre de Recherche d'Immunologie et d'Hématologie, Faculté de Médecine, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Université de Strasbourg, Strasbourg, France.,Laboratoire International Associé (LIA) INSERM, Strasbourg (France) - Nagano (Japan), Strasbourg, France.,Fédération Hospitalo-Universitaire (FHU) OMICARE, Strasbourg, France.,Laboratoire Central d'Immunologie, Pôle de Biologie, Nouvel Hôpital Civil, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| |
Collapse
|
17
|
Vanpouille-Box C, Pilones KA, Wennerberg E, Formenti SC, Demaria S. In situ vaccination by radiotherapy to improve responses to anti-CTLA-4 treatment. Vaccine 2015; 33:7415-7422. [PMID: 26148880 PMCID: PMC4684480 DOI: 10.1016/j.vaccine.2015.05.105] [Citation(s) in RCA: 136] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2014] [Revised: 05/20/2015] [Accepted: 05/28/2015] [Indexed: 12/14/2022]
Abstract
Targeting immune checkpoint receptors has emerged as an effective strategy to induce immune-mediated cancer regression in the subset of patients who have significant pre-existing anti-tumor immunity. For the remainder, effective anti tumor responses may require vaccination. Radiotherapy, traditionally used to achieve local tumor control, has acquired a new role, that of a partner for immunotherapy. Ionizing radiation has pro-inflammatory effects that facilitate tumor rejection. Radiation alters the tumor to enhance the concentration of effector T cells via induction of chemokines, cytokines and adhesion molecules. In parallel, radiation can induce an immunogenic death of cancer cells, promoting cross-presentation of tumor-derived antigens by dendritic cells to T cells. Newly generated anti-tumor immune responses have been demonstrated post-radiation in both murine models and occasional patients, supporting the hypothesis that the irradiated tumor can become an in situ vaccine. It is in this role, that radiation can be applied to induce anti-tumor T cells in lymphocyte-poor tumors, and possibly benefit patients who would otherwise fail to respond to immune checkpoint inhibitors. This review summarizes preclinical and clinical data demonstrating that radiation acts in concert with antibodies targeting the immune checkpoint cytotoxic T-lymphocyte antigen-4 (CTLA-4), to induce therapeutically effective anti-tumor T cell responses in tumors otherwise non responsive to anti-CTLA-4 therapy.
Collapse
Affiliation(s)
- Claire Vanpouille-Box
- Department of Pathology, New York University School of Medicine, and NYU Cancer Institute, New York, NY 10016, USA
| | - Karsten A Pilones
- Department of Pathology, New York University School of Medicine, and NYU Cancer Institute, New York, NY 10016, USA
| | - Erik Wennerberg
- Department of Pathology, New York University School of Medicine, and NYU Cancer Institute, New York, NY 10016, USA
| | - Silvia C Formenti
- Department of Radiation Oncology, New York University School of Medicine, and NYU Cancer Institute, New York, NY 10016, USA
| | - Sandra Demaria
- Department of Pathology, New York University School of Medicine, and NYU Cancer Institute, New York, NY 10016, USA; Department of Radiation Oncology, New York University School of Medicine, and NYU Cancer Institute, New York, NY 10016, USA.
| |
Collapse
|
18
|
Muro M, López-Hernández R, Mrowiec A. Immunogenetic biomarkers in inflammatory bowel diseases: Role of the IBD3 region. World J Gastroenterol 2014; 20:15037-15048. [PMID: 25386052 PMCID: PMC4223237 DOI: 10.3748/wjg.v20.i41.15037] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2013] [Revised: 12/19/2013] [Accepted: 04/16/2014] [Indexed: 02/06/2023] Open
Abstract
Many studies have demonstrated the linkage between the IBD3 region (6p21.1-23), an area which encompasses the famous human leukocyte antigen (HLA) complex, and Crohn’s disease (CD) or ulcerative colitis (UC). IBD3 is the only region that meets genome-wide significance, and provides stronger evidence of the linkage than 16p13.1-16q12.2 (IBD1), the locus that contains the susceptibility gene CARD15. However, despite these findings, IBD3 susceptibility genes remain elusive and unclear due to the strong linkage disequilibrium, extensive polymorphism, and high gene density that characterize this area and also due to varying allele frequencies in populations around the world. This area presents an extremely high abundance of genes, including the classical and non-classical major histocompatibility complex (MHC) class I and II genes, and other genes, namely MHC class III genes tumor necrosis factor (TNF)-α and -β, and Hsp, whose proteins play key functions in immunological processes. To date, it is not clear which genes within the MHC family contribute to the IBD pathogenesis, although certain HLA alleles have been associated with IBD. Recent insights into the biological function of other genes encoded within the IBD3 region, such as the MHC class I chain-related (MIC) genes, have led investigators to a more comprehensive exploration of this region. MHC class I chain-related molecule A (MICA) is highly polymorphic and interacts with NKG2D, its receptor on the surface of NK, Tγδ and T CD8+ cells. Increased expression of MICA in intestinal epithelial cells and increased expression of NKG2D in CD4+ T cells (lamina propria) in patients with CD have also been reported. MICA alleles have also been associated with IBD, and a variation at amino acid position 129 of the α2-heavy chain domain seems to categorize MICA alleles into strong and weak binders of NKG2D receptor, thereby influencing the effector cells’ function. In this regard, a relevant role of MICA-129-Val/Met single nucleotide polymorphism has recently been implicated in the pathogenesis of IBD. TNF-α and -β also play an important role in inflammatory response. In fact, IBD is commonly treated with TNF-α inhibitors. Additionally, polymorphisms of TNF-α gene are known to affect the gene expression level and particular TNF-α genotypes may influence the response of IBD patients treated with TNF-α inhibitors.
Collapse
|
19
|
Radiation-induced autophagy potentiates immunotherapy of cancer via up-regulation of mannose 6-phosphate receptor on tumor cells in mice. Cancer Immunol Immunother 2014; 63:1009-21. [PMID: 24943275 DOI: 10.1007/s00262-014-1573-4] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2014] [Accepted: 06/10/2014] [Indexed: 12/13/2022]
Abstract
There is a significant body of evidence demonstrating that radiation therapy (XRT) enhances the effect of immune therapy. However, the precise mechanisms by which XRT potentiates the immunotherapy of cancer remain elusive. Here, we report that XRT potentiates the effect of immune therapy via induction of autophagy and resultant trafficking of mannose-6-phopsphate receptor (MPR) to the cell surface. Irradiation of different tumor cells caused substantial up-regulation of MPR on the cell surface in vitro and in vivo. Down-regulation of MPR in tumor cells with shRNA completely abrogated the combined effect of XRT and immunotherapy (CTLA4 antibody) in B16F10-bearing mice without changes in the tumor-specific responses of T cells. Radiation-induced MPR up-regulation was the result of redistribution of the receptor to the cell surface. This effect was caused by autophagy with redirection of MPR to autophagosomes in a clathrin-dependent manner. In autophagosomes, MPR lost its natural ligands, which resulted in subsequent trafficking of empty receptor(s) back to the surface. Together, our data demonstrated a novel mechanism by which XRT can enhance the effect of immunotherapy and the molecular mechanism of this process.
Collapse
|
20
|
Dayanc BE, Bansal S, Gure AO, Gollnick SO, Repasky EA. Enhanced sensitivity of colon tumour cells to natural killer cell cytotoxicity after mild thermal stress is regulated through HSF1-mediated expression of MICA. Int J Hyperthermia 2014; 29:480-90. [PMID: 23902341 DOI: 10.3109/02656736.2013.821526] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
PURPOSE Previously we showed that mild thermal stress increased natural killer (NK) cell-mediated tumour cytotoxicity and that this could be blocked by anti-NKG2D or anti-MICA (major histolocompatability complex (MHC) class I related chain A) antibodies. Here, we investigated the role of the transcription factor heat shock factor 1 (HSF1) in thermal regulation of MICA expression in tumour cells in vitro and in vivo. MATERIALS AND METHODS Hyperthermia experiments were conducted in vitro and in mice using a target temperature of 39.5 °C. Apoptotic cells and NK cells in situ were visualised by use of the TUNEL assay or expression of NKp46 respectively. Using Colo205 cells, HSF1 message was blocked utilising siRNA while luciferase reporter assays were used to measure the activity of the MICA promoter in vitro. Cell surface MICA was measured by flow cytometry. RESULTS Following whole body hyperthermia (WBH), tumour tissues showed an increase in NK cells and apoptosis. Mild thermal stress resulted in a transient increase in surface MICA and enhanced NK cytotoxicity of the Colo205 colon cancer cell line. Silencing (mRNA) HSF1 expression in Colo205 cells prevented the thermal enhancement of MICA message and surface protein levels, with partial loss of thermally enhanced NK cytotoxicity. Mutations of the HSF1 binding site on the MICA promoter implicated HSF1 in the thermal enhancement of MICA. Some, but not all, patient-derived colon tumour derived xenografts also exhibited an enhanced MICA message expression after WBH. CONCLUSIONS Up-regulation of MICA expression in Colo205 cells and enhanced sensitivity to NK cell killing following mild thermal stress is dependent upon HSF1.
Collapse
Affiliation(s)
- Baris E Dayanc
- Department of Molecular Biology and Genetics, Inonu University, Malatya, Turkey
| | | | | | | | | |
Collapse
|
21
|
Jarduli LR, Sell AM, Reis PG, Sippert EÂ, Ayo CM, Mazini PS, Alves HV, Teixeira JJV, Visentainer JEL. Role of HLA, KIR, MICA, and cytokines genes in leprosy. BIOMED RESEARCH INTERNATIONAL 2013; 2013:989837. [PMID: 23936864 PMCID: PMC3722889 DOI: 10.1155/2013/989837] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 03/20/2013] [Revised: 05/16/2013] [Accepted: 06/05/2013] [Indexed: 01/08/2023]
Abstract
Many genes including HLA, KIR, and MICA genes, as well as polymorphisms in cytokines have been investigated for their role in infectious disease. HLA alleles may influence not only susceptibility or resistance to leprosy, but also the course of the disease. Some combinations of HLA and KIR may result in negative as well as positive interactions between NK cells and infected host cells with M. leprae, resulting in activation or inhibition of NK cells and, consequently, in death of bacillus. In addition, studies have demonstrated the influence of MICA genes in the pathogenesis of leprosy. Specifically, they may play a role in the interaction between NK cells and infected cells. Finally, pro- and anti-inflammatory cytokines have been influencing the clinical course of leprosy. Data from a wide variety of sources support the existence of genetic factors influencing the leprosy pathogenesis. These sources include twin studies, segregation analyses, family-based linkage and association studies, candidate gene association studies, and, most recently, genome-wide association studies (GWAS). The purpose of this brief review was to highlight the importance of some immune response genes and their correlation with the clinical forms of leprosy, as well as their implications for disease resistance and susceptibility.
Collapse
Affiliation(s)
- Luciana Ribeiro Jarduli
- Program of Biosciences Applied to Pharmacy, Department of Clinical Analysis and Biomedicine, Maringa State University, Avenida Colombo 5790, 87020-900 Maringá, PR, Brazil
| | - Ana Maria Sell
- Basic Health Sciences Department, Maringa State University, Avenida Colombo 5790, 87020-900 Maringá, PR, Brazil
| | - Pâmela Guimarães Reis
- Program of Biosciences Applied to Pharmacy, Department of Clinical Analysis and Biomedicine, Maringa State University, Avenida Colombo 5790, 87020-900 Maringá, PR, Brazil
| | - Emília Ângela Sippert
- Program of Biosciences Applied to Pharmacy, Department of Clinical Analysis and Biomedicine, Maringa State University, Avenida Colombo 5790, 87020-900 Maringá, PR, Brazil
| | - Christiane Maria Ayo
- Program of Biosciences Applied to Pharmacy, Department of Clinical Analysis and Biomedicine, Maringa State University, Avenida Colombo 5790, 87020-900 Maringá, PR, Brazil
| | - Priscila Saamara Mazini
- Program of Biosciences Applied to Pharmacy, Department of Clinical Analysis and Biomedicine, Maringa State University, Avenida Colombo 5790, 87020-900 Maringá, PR, Brazil
| | - Hugo Vicentin Alves
- Program of Biosciences Applied to Pharmacy, Department of Clinical Analysis and Biomedicine, Maringa State University, Avenida Colombo 5790, 87020-900 Maringá, PR, Brazil
| | - Jorge Juarez Vieira Teixeira
- Program of Biosciences Applied to Pharmacy, Department of Clinical Analysis and Biomedicine, Maringa State University, Avenida Colombo 5790, 87020-900 Maringá, PR, Brazil
| | | |
Collapse
|
22
|
Lam RA, Chwee JY, Le Bert N, Sauer M, Pogge von Strandmann E, Gasser S. Regulation of self-ligands for activating natural killer cell receptors. Ann Med 2013; 45:384-94. [PMID: 23701136 DOI: 10.3109/07853890.2013.792495] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Natural killer (NK) cells are able to lyse infected and tumor cells while sparing healthy cells. Recognition of diseased cells by NK cells is governed by several activating and inhibitory receptors. We review numerous pathways that have been implicated in the regulation of self-ligands for activating receptors, including NKG2D, DNAM-1, LFA-1, NKp30, NKp44, NKp46, NKp65, and NKp80 found on NK cells and some T cells. Understanding how the regulation of self-encoded ligand expression is regulated may provide novel avenues for future therapeutic approaches to infections and cancer.
Collapse
Affiliation(s)
- Runyi A Lam
- Immunology Programme, Centre for Life Sciences, Department of Microbiology, National University of Singapore 117456, Singapore
| | | | | | | | | | | |
Collapse
|
23
|
The uncertain role of unmodified mesenchymal stem cells in tumor progression: what master switch? Stem Cell Res Ther 2013; 4:22. [PMID: 23510751 PMCID: PMC3707017 DOI: 10.1186/scrt170] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are emerging as promising gene vectors for cancer therapy because of their unique characteristics, including the ease of their expansion and genetic modification and their remarkable tumor-tropic properties. However, there remains a concern that MSCs may promote cancer progression. Surprisingly, there are conflicting reports within the literature describing both the promotion and inhibition of cancer progression by MSCs. The reasons for this discrepancy are still unknown. The surface markers, differentiation ability, and tumorigenic roles of MSCs, as well as their effect on immunoregulation, produce heterogeneity. In this review, we describe the heterogeneity of MSCs by the species from which they are derived, the methodology for their isolation and the context of their interactions with cancer cells. The conflicting roles of MSCs in tumor progression may be attributable to the bimodal effect of unmodified MSCs on immunoregulation. MSCs have been reported to suppress T-cell function and inhibit graft-versus-host disease (GVHD). On the other hand, MSCs elicit the graft-versus-tumor (GVT) effect in some cases. Selective allodepletion may be used to dissociate GVHD from the GVT effect. Understanding the conditions that balance GVHD and the GVT effect of MSCs may be crucial to advance cancer therapy research with respect to MSCs.
Collapse
|
24
|
Fernández-Sánchez A, Baragaño Raneros A, Carvajal Palao R, Sanz AB, Ortiz A, Ortega F, Suárez-Álvarez B, López-Larrea C. DNA demethylation and histone H3K9 acetylation determine the active transcription of the NKG2D gene in human CD8+ T and NK cells. Epigenetics 2012; 8:66-78. [PMID: 23235109 DOI: 10.4161/epi.23115] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
The human activating receptor NKG2D is mainly expressed by NK, NKT, γδ T and CD8(+) T cells and, under certain conditions, by CD4(+) T cells. This receptor recognizes a diverse family of ligands (MICA, MICB and ULBPs 1-6) leading to the activation of effector cells and triggering the lysis of target cells. The NKG2D receptor-ligand system plays an important role in the immune response to infections, tumors, transplanted graft and autoantigens. Elucidation of the regulatory mechanisms of NKG2D is therefore essential for therapeutic purposes. In this study, we speculate whether epigenetic mechanisms, such as DNA methylation and histone acetylation, participate in NKG2D gene regulation in T lymphocytes and NK cells. DNA methylation in the NKG2D gene was observed in CD4(+) T lymphocytes and T cell lines (Jurkat and HUT78), while this gene was unmethylated in NKG2D-positive cells (CD8(+) T lymphocytes, NK cells and NKL cell line) and associated with high levels of histone H3 lysine 9 acetylation (H3K9Ac). Treatment with the histone acetyltransferase (HAT) inhibitor curcumin reduces H3K9Ac levels in the NKG2D gene, downregulates NKG2D transcription and leads to a marked reduction in the lytic capacity of NKG2D-mediated NKL cells. These findings suggest that differential NKG2D expression in the different cell subsets is regulated by epigenetic mechanisms and that its modulation by epigenetic treatments might provide a new strategy for treating several pathologies.
Collapse
|
25
|
van Esch EMG, Welters MJP, Jordanova ES, Trimbos JBMZ, van der Burg SH, van Poelgeest MIE. Treatment failure in patients with HPV 16-induced vulvar intraepithelial neoplasia: understanding different clinical responses to immunotherapy. Expert Rev Vaccines 2012; 11:821-40. [PMID: 22913259 DOI: 10.1586/erv.12.56] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Failure of the immune system to launch a strong and effective immune response to high-risk HPV is related to viral persistence and the development of anogenital (pre)malignant lesions such as vulvar intraepithelial neoplasia (VIN). Different forms of immunotherapy, aimed at overcoming the inertia of the immune system, have been developed and met with clinical success. Unfortunately these, in principal successful, therapeutic approaches also fail to induce clinical responses in a substantial number of cases. In this review, the authors summarize the traits of the immune response to HPV in healthy individuals and in patients with HPV-induced neoplasia. The potential mechanisms involved in the escape of HPV-induced lesions from the immune system indicate gaps in our knowledge. Finally, the interaction between the immune system and VIN is discussed with a special focus on the different forms of immunotherapy applied to treat VIN and the potential causes of therapy failure. The authors conclude that there are a number of pre-existing conditions that determine the patients' responsiveness to immunotherapy. An immunotherapeutic strategy in which different aspects of immune failure are attacked by complementary approaches, will improve the clinical response rate.
Collapse
Affiliation(s)
- Edith M G van Esch
- Department of Gynecology, Leiden University Medical Center, PO Box 9600, 2300 RC, Leiden, The Netherlands
| | | | | | | | | | | |
Collapse
|
26
|
Hervieu A, Rébé C, Végran F, Chalmin F, Bruchard M, Vabres P, Apetoh L, Ghiringhelli F, Mignot G. Dacarbazine-mediated upregulation of NKG2D ligands on tumor cells activates NK and CD8 T cells and restrains melanoma growth. J Invest Dermatol 2012; 133:499-508. [PMID: 22951720 DOI: 10.1038/jid.2012.273] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Dacarbazine (DTIC) is a cytotoxic drug widely used for melanoma treatment. However, the putative contribution of anticancer immune responses in the efficacy of DTIC has not been evaluated. By testing how DTIC affects host immune responses to cancer in a mouse model of melanoma, we unexpectedly found that both natural killer (NK) and CD8(+) T cells were indispensable for DTIC therapeutic effect. Although DTIC did not directly affect immune cells, it triggered the upregulation of NKG2D ligands on tumor cells, leading to NK cell activation and IFNγ secretion in mice and humans. NK cell-derived IFNγ subsequently favored upregulation of major histocompatibility complex class I molecules on tumor cells, rendering them sensitive to cytotoxic CD8(+) T cells. Accordingly, DTIC markedly enhanced cytotoxic T lymphocyte antigen 4 inhibition efficacy in vivo in an NK-dependent manner. These results underscore the immunogenic properties of DTIC and provide a rationale to combine DTIC with immunotherapeutic agents that relieve immunosuppression in vivo.
Collapse
|
27
|
Kahraman A, Fingas CD, Syn WK, Gerken G, Canbay A. Role of stress-induced NKG2D ligands in liver diseases. Liver Int 2012; 32:370-82. [PMID: 22097967 DOI: 10.1111/j.1478-3231.2011.02608.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2011] [Accepted: 06/15/2011] [Indexed: 12/24/2022]
Abstract
Cell death by apoptosis is a prominent feature in a variety of liver diseases. It is likely that apoptosis is the initial cellular response to hepatocyte and biliary injury, which then leads to the initiation of cellular and cytokine cascades culminating in hepatocyte death with subsequent fibrosis and cirrhosis. This sequence of events is of paramount clinical importance. Recently, soluble forms of the major histocompatibility complex class I-related chains A and closely related B (MIC A and B) were reported to be increased in patients with a variety of liver diseases. MIC A and B are cell surface glycoproteins that function as indicators for cellular stress and thus activate circulating cytotoxic natural killer (NK) cells. The interaction between MIC A and B with their cognate receptor natural killer group 2 member D (NKG2D) culminates in enhanced liver cell death, which is mediated in part by apoptotic mechanisms. The present overview focuses on the role of the stress-induced NKG2D ligands MIC A and B in diverse liver diseases. Critical insights into these complex relations may help to promote rationally based therapies in liver diseases. Importantly, we hope that this overview will help to stimulate further studies into mechanisms by which stress ligands mediate cell death and its sequale.
Collapse
Affiliation(s)
- Alisan Kahraman
- University Clinic Essen, University of Duisburg-Essen, Essen, Germany
| | | | | | | | | |
Collapse
|
28
|
Cheon S, Lee JH, Park S, Bang SI, Lee WJ, Yoon DY, Yoon SS, Kim T, Min H, Cho BJ, Lee HJ, Lee KW, Jeong SH, Park H, Cho D. Overexpression of IL-32alpha increases natural killer cell-mediated killing through up-regulation of Fas and UL16-binding protein 2 (ULBP2) expression in human chronic myeloid leukemia cells. J Biol Chem 2011; 286:12049-55. [PMID: 21321117 DOI: 10.1074/jbc.m110.159756] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
IL-32 was recently identified as a proinflammatory cytokine that is induced by IL-18 in natural killer (NK) cells and is highly correlated with inflammatory disorders. However, the relationship between IL-32 and tumor progression is still unknown. In this study, we investigated whether overexpression of IL-32 affects susceptibility of chronic myeloid leukemia (CML) cells to NK cells. Interestingly, IL-32α-overexpressing CML cell lines, K562, Kcl22, and BV173, showed higher NK cell-mediated killing. Flow cytometry analysis revealed that overexpression of IL-32α induced increased expression of Fas and UL16-binding protein 2 (ULBP2) in CML cells. The direct relationship between overexpression of surface molecules by IL-32α and increased NK cell-mediated killing was confirmed by Fas or ULBP2 siRNA transfection. IL-32α-induced Fas and ULBP2 expression are regulated p38 MAPK. In addition, the transcription factor Ets1 plays a key role in ULBP2 specific expression by IL-32α overexpression in ULBP family members. Taken together, these data show that IL-32α stimulates Fas and ULBP2 expression via activation of p38 MAPK, which increases NK susceptibility of CML cells. Enhanced NK cell susceptibility of CML cells by IL-32α overexpression may improve the efficiency of NK cell-based immunotherapy.
Collapse
Affiliation(s)
- Soyoung Cheon
- Department of Life Science, Sookmyung Women's University, and Konkuk University Hospital, Seoul 140-742, Republic of Korea
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
López-Hernández R, Valdés M, Lucas D, Campillo JA, Martínez-Garcia P, Salama H, López M, Salgado G, Botella C, Minguela A, Miras M, Alvarez-López MR, Carballo F, Muro M. Association analysis of MICA gene polymorphism and MICA-129 dimorphism with inflammatory bowel disease susceptibility in a Spanish population. Hum Immunol 2010; 71:512-4. [PMID: 20152875 DOI: 10.1016/j.humimm.2010.02.003] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2009] [Revised: 01/19/2010] [Accepted: 02/01/2010] [Indexed: 12/12/2022]
Abstract
MICA is located at 46 kb centromeric of HLA-B, is highly polymorphic and interactions with NKG2D, its receptor on the surface of NK, Tgammadelta, and T CD8 lymphocytes. A variation at amino acid position 129 of the alpha2-heavy chain domain seems to categorize MICA alleles into strong and weak binder of NKG2D receptor, and thereby to influence effector cell function. Our aim was to study allele polymorphism of MICA and the functionally relevant dimorphism (129val/met) of MICA gene in inflammatory bowel disease (IBD) patients in our population. DNA was obtained from IBD patients (n = 88) and unrelated healthy Murcians (n = 154) and used to MICA genotyping using polymerase chain reaction-sequence-specific oligonucleotides. We did not find statistical differences in the distribution of MICA alleles between the IBD and control groups. However, we found a higher frequency of MICA-129met/met and a lower frequency of MICA-129val/met genotypes in IBD patients (mainly in ulcerative colitis) than in controls (pc = 0.02). These preliminary data could suggest a relevant role of MICA-129-val/met SNP (weak/strong binders of NKG2D receptor) in the pathogenesis of IBD.
Collapse
|
30
|
Donor-recipient mismatches in MHC class I chain-related gene A in unrelated donor transplantation lead to increased incidence of acute graft-versus-host disease. Blood 2009; 114:2884-7. [PMID: 19654407 DOI: 10.1182/blood-2009-05-223172] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The polymorphic products of major histocompatibility complex class I-related chain A (MICA) genes are important in solid organ transplantation rejection. MICA expression is limited to gut epithelium and may play a role in triggering acute graft-versus-host disease (aGVHD). A total of 236 recipients of unrelated donor transplantation were studied. Donor-recipient human leukocyte antigen (HLA) match was 10/10 human leukocyte antigen (HLA-A, -B, -C, -DRB1, -DQB1) in 73% and MICA mismatch in 8.4%. Because of physical vicinity of the loci, MICA mismatch was significantly associated with mismatch at HLA-B and HLA-C. A higher rate of grade II-IV aGVHD was seen in MICA-mismatched patients (80% vs 40%, P = .003) irrespective of degree of HLA matching (HLA 10/10 match: 75% vs 39%, P = .02) and HLA any mismatch (83% vs 46%, P = .003). The rate of grade II-IV gastrointestinal aGVHD was also higher in MICA-mismatched patients (35% vs 17%, P = .05). We conclude that MICA may represent novel a transplantation antigen recognized by human allogeneic T cells. This study was registered at ClinicalTrials.gov (Identifier NCT00506922).
Collapse
|
31
|
Meresse B, Cerf-Bensussan N. Innate T cell responses in human gut. Semin Immunol 2009; 21:121-9. [PMID: 19231234 DOI: 10.1016/j.smim.2009.01.002] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2008] [Accepted: 01/20/2009] [Indexed: 01/27/2023]
Abstract
One arm of the gut-associated immune system is represented by a vast collection of T lymphocytes which participate in the subtle interplay between innate and adaptive immune mechanisms and maintain homeostasis at the main body external surface. Mounting data are providing exciting new insight into the innate-like mechanisms which enable intestinal T cells to rapidly sense local conditions and which broaden the spectrum of their functions and regulation at this strategic location. Herein we discuss how innate-like T cell recognition by unconventional T cell subsets and expression of innate NK receptors might modulate immune T cell responses in the human normal or diseased intestine.
Collapse
Affiliation(s)
- Bertrand Meresse
- INSERM U793, Université Paris Descartes, Medical School, 156 rue de Vaugirard, 75737 Paris Cedex 15, France.
| | | |
Collapse
|
32
|
Oncostatin M enhances the antiviral effects of type I interferon and activates immunostimulatory functions in liver epithelial cells. J Virol 2009; 83:3298-311. [PMID: 19158240 DOI: 10.1128/jvi.02167-08] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Oncostatin M (OSM) is released together with type I interferon (IFN) by activated dendritic cells, suggesting a concerted action of these cytokines in the biological response against infection. We found that OSM increases the antiviral effect of IFN-alpha in Huh7 hepatoma cells infected with hepatitis A or hepatitis C virus and synergizes with IFN-alpha in the induction of antiviral genes. The combination of OSM and IFN-alpha led to upregulation of both STAT1 and STAT3 together with intense and prolonged activation of STAT1, STAT3, and Jak1. OSM with or without IFN-alpha also activated p38 mitogen-activated protein kinase, which is known to enhance transcription of IFN-alpha-inducible genes. Interestingly, OSM combined with IFN-alpha strongly induced immunoproteasome genes and other genes involved in antigen processing and presentation. Moreover, OSM, alone or in combination with IFN-alpha, upregulated relevant innate immunity molecules and increased the expression of intracellular adhesion molecule 1 and interleukin-15 receptor alpha (IL-15Ralpha) in liver cells. Hepatoma cells transfected with a plasmid encoding a viral antigen were able to activate effector T cells when pretreated with IFN-alpha plus OSM but not with each cytokine separately. Also, OSM, more than IFN-alpha, augmented the ability of Huh7 cells to transpresent IL-15 to responding lymphocytes and increased the immunostimulatory activity of liver epithelial cells by presenting a short viral peptide to sensitized cytotoxic T cells. In conclusion, OSM enhances the antiviral effects of type I interferon and cooperates with it in the induction of adaptive immune responses to pathogens. These findings may have therapeutic implications.
Collapse
|
33
|
Lucas D, Campillo JA, López-Hernández R, Martínez-García P, López-Sánchez M, Botella C, Salgado G, Minguela A, Alvarez-López MR, Muro M. Allelic diversity of MICA gene and MICA/HLA-B haplotypic variation in a population of the Murcia region in southeastern Spain. Hum Immunol 2008; 69:655-60. [PMID: 18718856 DOI: 10.1016/j.humimm.2008.07.011] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2008] [Revised: 07/13/2008] [Accepted: 07/18/2008] [Indexed: 12/12/2022]
Abstract
Major histocompatibility complex class I-related chain A (MICA) is located at 46 kb centromeric of HLA-B. It is highly polymorphic and interacts with NKG2D, its receptor on the surface of NK, Tgammadelta and T CD8 lymphocytes. Data on MICA polymorphism in different populations are still limited. Our aim was to establish allelic diversity of MICA gene and linkage disequilibrium with HLA-B in our population. DNA was obtained from 154 unrelated healthy individuals from the Murcia region in southeastern Spain. HLA-B genotyping was performed using polymerase chain reaction (PCR)-sequence-specific oligonucleotide probes and allele-specific PCR-sequence-specific primers, and MICA genotyping by using PCR-sequence-specific oligonucleotide probes. A total of 19 MICA alleles were detected on this study. MICA*008 was the most frequent allele (25.3%), followed by MICA*002 (16.1%), MICA*004 (14.9%), MICA*001 (7.8%), MICA*009 and MICA*016 (7.1%), and MICA*010 (4.6%). Eleven alleles had frequencies of <1%. In the haplotype analysis, MICA*008-B*0702 was found to be the most common, followed by MICA*004-B*4403 and MICA*001-B*1801, MICA*002-B*3501, MICA*008-B*4402, MICA*004-B*4901, MICA*008-B*0801, and MICA*002-B*3801. The frequency of MICA*010-B*1501, MICA*008-B*1302, MICA*015-B*4501, and MICA*008-B*4001 was remarkable inasmuch as these two last haplotypes have not been reported in Spanish population. Indeed, MICA*016 linked to B*1402 has also not been reported in the literature. In conclusion, the allelic diversity in our population is similar to other Caucasian populations; however we found a series of less frequent alleles, in addition to as-yet-undescribed haplotypic associations in other populations of Caucasian origin.
Collapse
Affiliation(s)
- Daniel Lucas
- Immunology Service, University Hospital Virgen de la Arrixaca, Murcia, Spain
| | | | | | | | | | | | | | | | | | | |
Collapse
|