1
|
Bojmar L, Zambirinis CP, Hernandez JM, Chakraborty J, Shaashua L, Kim J, Johnson KE, Hanna S, Askan G, Burman J, Ravichandran H, Zheng J, Jolissaint JS, Srouji R, Song Y, Choubey A, Kim HS, Cioffi M, van Beek E, Sigel C, Jessurun J, Velasco Riestra P, Blomstrand H, Jönsson C, Jönsson A, Lauritzen P, Buehring W, Ararso Y, Hernandez D, Vinagolu-Baur JP, Friedman M, Glidden C, Firmenich L, Lieberman G, Mejia DL, Nasar N, Mutvei AP, Paul DM, Bram Y, Costa-Silva B, Basturk O, Boudreau N, Zhang H, Matei IR, Hoshino A, Kelsen D, Sagi I, Scherz A, Scherz-Shouval R, Yarden Y, Oren M, Egeblad M, Lewis JS, Keshari K, Grandgenett PM, Hollingsworth MA, Rajasekhar VK, Healey JH, Björnsson B, Simeone DM, Tuveson DA, Iacobuzio-Donahue CA, Bromberg J, Vincent CT, O'Reilly EM, DeMatteo RP, Balachandran VP, D'Angelica MI, Kingham TP, Allen PJ, Simpson AL, Elemento O, Sandström P, Schwartz RE, Jarnagin WR, Lyden D. Multi-parametric atlas of the pre-metastatic liver for prediction of metastatic outcome in early-stage pancreatic cancer. Nat Med 2024; 30:2170-2180. [PMID: 38942992 DOI: 10.1038/s41591-024-03075-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Accepted: 05/17/2024] [Indexed: 06/30/2024]
Abstract
Metastasis occurs frequently after resection of pancreatic cancer (PaC). In this study, we hypothesized that multi-parametric analysis of pre-metastatic liver biopsies would classify patients according to their metastatic risk, timing and organ site. Liver biopsies obtained during pancreatectomy from 49 patients with localized PaC and 19 control patients with non-cancerous pancreatic lesions were analyzed, combining metabolomic, tissue and single-cell transcriptomics and multiplex imaging approaches. Patients were followed prospectively (median 3 years) and classified into four recurrence groups; early (<6 months after resection) or late (>6 months after resection) liver metastasis (LiM); extrahepatic metastasis (EHM); and disease-free survivors (no evidence of disease (NED)). Overall, PaC livers exhibited signs of augmented inflammation compared to controls. Enrichment of neutrophil extracellular traps (NETs), Ki-67 upregulation and decreased liver creatine significantly distinguished those with future metastasis from NED. Patients with future LiM were characterized by scant T cell lobular infiltration, less steatosis and higher levels of citrullinated H3 compared to patients who developed EHM, who had overexpression of interferon target genes (MX1 and NR1D1) and an increase of CD11B+ natural killer (NK) cells. Upregulation of sortilin-1 and prominent NETs, together with the lack of T cells and a reduction in CD11B+ NK cells, differentiated patients with early-onset LiM from those with late-onset LiM. Liver profiles of NED closely resembled those of controls. Using the above parameters, a machine-learning-based model was developed that successfully predicted the metastatic outcome at the time of surgery with 78% accuracy. Therefore, multi-parametric profiling of liver biopsies at the time of PaC diagnosis may determine metastatic risk and organotropism and guide clinical stratification for optimal treatment selection.
Collapse
Affiliation(s)
- Linda Bojmar
- Departments of Pediatrics and Cell and Developmental Biology, Children's Cancer and Blood Foundation Laboratories, Drukier Institute for Children's Health, Meyer Cancer Center Weill Cornell Medicine, New York, NY, USA
- Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Constantinos P Zambirinis
- Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
- Hepatopancreatobiliary Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Division of Surgical Oncology, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, USA
| | - Jonathan M Hernandez
- Hepatopancreatobiliary Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Thoracic and Gastrointestinal Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Jayasree Chakraborty
- Hepatopancreatobiliary Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- David M. Rubenstein Center for Pancreatic Cancer Research, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Lee Shaashua
- Departments of Pediatrics and Cell and Developmental Biology, Children's Cancer and Blood Foundation Laboratories, Drukier Institute for Children's Health, Meyer Cancer Center Weill Cornell Medicine, New York, NY, USA
| | - Junbum Kim
- Department of Physiology and Biophysics, Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY, USA
| | - Kofi Ennu Johnson
- Departments of Pediatrics and Cell and Developmental Biology, Children's Cancer and Blood Foundation Laboratories, Drukier Institute for Children's Health, Meyer Cancer Center Weill Cornell Medicine, New York, NY, USA
| | - Samer Hanna
- Departments of Pediatrics and Cell and Developmental Biology, Children's Cancer and Blood Foundation Laboratories, Drukier Institute for Children's Health, Meyer Cancer Center Weill Cornell Medicine, New York, NY, USA
| | - Gokce Askan
- David M. Rubenstein Center for Pancreatic Cancer Research, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jonas Burman
- Departments of Pediatrics and Cell and Developmental Biology, Children's Cancer and Blood Foundation Laboratories, Drukier Institute for Children's Health, Meyer Cancer Center Weill Cornell Medicine, New York, NY, USA
- Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Hiranmayi Ravichandran
- Department of Physiology and Biophysics, Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY, USA
- Caryl and Israel Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Jian Zheng
- Hepatopancreatobiliary Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Joshua S Jolissaint
- Departments of Pediatrics and Cell and Developmental Biology, Children's Cancer and Blood Foundation Laboratories, Drukier Institute for Children's Health, Meyer Cancer Center Weill Cornell Medicine, New York, NY, USA
- Hepatopancreatobiliary Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Rami Srouji
- Departments of Pediatrics and Cell and Developmental Biology, Children's Cancer and Blood Foundation Laboratories, Drukier Institute for Children's Health, Meyer Cancer Center Weill Cornell Medicine, New York, NY, USA
- Hepatopancreatobiliary Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Yi Song
- Hepatopancreatobiliary Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ankur Choubey
- Hepatopancreatobiliary Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Han Sang Kim
- Departments of Pediatrics and Cell and Developmental Biology, Children's Cancer and Blood Foundation Laboratories, Drukier Institute for Children's Health, Meyer Cancer Center Weill Cornell Medicine, New York, NY, USA
| | - Michele Cioffi
- Departments of Pediatrics and Cell and Developmental Biology, Children's Cancer and Blood Foundation Laboratories, Drukier Institute for Children's Health, Meyer Cancer Center Weill Cornell Medicine, New York, NY, USA
| | - Elke van Beek
- Hepatopancreatobiliary Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Carlie Sigel
- David M. Rubenstein Center for Pancreatic Cancer Research, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jose Jessurun
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA
| | | | - Hakon Blomstrand
- Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
- Department of Clinical Pathology, Linköping University, Linköping, Sweden
| | - Carolin Jönsson
- Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Anette Jönsson
- Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Pernille Lauritzen
- Departments of Pediatrics and Cell and Developmental Biology, Children's Cancer and Blood Foundation Laboratories, Drukier Institute for Children's Health, Meyer Cancer Center Weill Cornell Medicine, New York, NY, USA
| | - Weston Buehring
- Departments of Pediatrics and Cell and Developmental Biology, Children's Cancer and Blood Foundation Laboratories, Drukier Institute for Children's Health, Meyer Cancer Center Weill Cornell Medicine, New York, NY, USA
| | - Yonathan Ararso
- Departments of Pediatrics and Cell and Developmental Biology, Children's Cancer and Blood Foundation Laboratories, Drukier Institute for Children's Health, Meyer Cancer Center Weill Cornell Medicine, New York, NY, USA
| | - Dylanne Hernandez
- Departments of Pediatrics and Cell and Developmental Biology, Children's Cancer and Blood Foundation Laboratories, Drukier Institute for Children's Health, Meyer Cancer Center Weill Cornell Medicine, New York, NY, USA
| | - Jessica P Vinagolu-Baur
- Departments of Pediatrics and Cell and Developmental Biology, Children's Cancer and Blood Foundation Laboratories, Drukier Institute for Children's Health, Meyer Cancer Center Weill Cornell Medicine, New York, NY, USA
| | - Madison Friedman
- Departments of Pediatrics and Cell and Developmental Biology, Children's Cancer and Blood Foundation Laboratories, Drukier Institute for Children's Health, Meyer Cancer Center Weill Cornell Medicine, New York, NY, USA
| | - Caroline Glidden
- Departments of Pediatrics and Cell and Developmental Biology, Children's Cancer and Blood Foundation Laboratories, Drukier Institute for Children's Health, Meyer Cancer Center Weill Cornell Medicine, New York, NY, USA
| | - Laetitia Firmenich
- Departments of Pediatrics and Cell and Developmental Biology, Children's Cancer and Blood Foundation Laboratories, Drukier Institute for Children's Health, Meyer Cancer Center Weill Cornell Medicine, New York, NY, USA
| | - Grace Lieberman
- Departments of Pediatrics and Cell and Developmental Biology, Children's Cancer and Blood Foundation Laboratories, Drukier Institute for Children's Health, Meyer Cancer Center Weill Cornell Medicine, New York, NY, USA
| | - Dianna L Mejia
- Departments of Pediatrics and Cell and Developmental Biology, Children's Cancer and Blood Foundation Laboratories, Drukier Institute for Children's Health, Meyer Cancer Center Weill Cornell Medicine, New York, NY, USA
| | - Naaz Nasar
- Hepatopancreatobiliary Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Anders P Mutvei
- Department of Laboratory Medicine, Division of Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Doru M Paul
- Division of Hematology and Medical Oncology, Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Yaron Bram
- Division of Gastroenterology & Hepatology, Weill Cornell Medicine, New York, NY, USA
| | - Bruno Costa-Silva
- Departments of Pediatrics and Cell and Developmental Biology, Children's Cancer and Blood Foundation Laboratories, Drukier Institute for Children's Health, Meyer Cancer Center Weill Cornell Medicine, New York, NY, USA
| | - Olca Basturk
- David M. Rubenstein Center for Pancreatic Cancer Research, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Nancy Boudreau
- Departments of Pediatrics and Cell and Developmental Biology, Children's Cancer and Blood Foundation Laboratories, Drukier Institute for Children's Health, Meyer Cancer Center Weill Cornell Medicine, New York, NY, USA
| | - Haiying Zhang
- Departments of Pediatrics and Cell and Developmental Biology, Children's Cancer and Blood Foundation Laboratories, Drukier Institute for Children's Health, Meyer Cancer Center Weill Cornell Medicine, New York, NY, USA
| | - Irina R Matei
- Departments of Pediatrics and Cell and Developmental Biology, Children's Cancer and Blood Foundation Laboratories, Drukier Institute for Children's Health, Meyer Cancer Center Weill Cornell Medicine, New York, NY, USA
| | - Ayuko Hoshino
- Departments of Pediatrics and Cell and Developmental Biology, Children's Cancer and Blood Foundation Laboratories, Drukier Institute for Children's Health, Meyer Cancer Center Weill Cornell Medicine, New York, NY, USA
| | - David Kelsen
- David M. Rubenstein Center for Pancreatic Cancer Research, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Irit Sagi
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel
| | - Avigdor Scherz
- Department of Plant and Environmental Sciences, Weizmann Institute of Science, Rehovot, Israel
| | - Ruth Scherz-Shouval
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, Israel
| | - Yosef Yarden
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel
| | - Moshe Oren
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Mikala Egeblad
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA
| | - Jason S Lewis
- Radiology and Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Weill Cornell Medical College, New York, NY, USA
| | - Kayvan Keshari
- David M. Rubenstein Center for Pancreatic Cancer Research, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Radiology and Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Paul M Grandgenett
- Eppley Institute for Research in Cancer and Allied Diseases, Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA
| | - Michael A Hollingsworth
- Eppley Institute for Research in Cancer and Allied Diseases, Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA
| | - Vinagolu K Rajasekhar
- Orthopedic Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - John H Healey
- Orthopedic Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Bergthor Björnsson
- Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Diane M Simeone
- Perlmutter Cancer Center, New York University Langone Health, New York, NY, USA
| | | | - Christine A Iacobuzio-Donahue
- David M. Rubenstein Center for Pancreatic Cancer Research, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jaqueline Bromberg
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - C Theresa Vincent
- Department of Laboratory Medicine, Division of Pathology, Karolinska Institutet, Stockholm, Sweden
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY, USA
| | - Eileen M O'Reilly
- David M. Rubenstein Center for Pancreatic Cancer Research, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Division of Hematology and Medical Oncology, Department of Medicine, Weill Cornell Medicine, New York, NY, USA
- Weill Cornell Medical College, New York, NY, USA
| | - Ronald P DeMatteo
- Hepatopancreatobiliary Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Vinod P Balachandran
- Hepatopancreatobiliary Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- David M. Rubenstein Center for Pancreatic Cancer Research, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Immuno-Oncology Service, Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Michael I D'Angelica
- Hepatopancreatobiliary Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- David M. Rubenstein Center for Pancreatic Cancer Research, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - T Peter Kingham
- Hepatopancreatobiliary Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- David M. Rubenstein Center for Pancreatic Cancer Research, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Peter J Allen
- Hepatopancreatobiliary Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Amber L Simpson
- Hepatopancreatobiliary Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Olivier Elemento
- Department of Physiology and Biophysics, Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY, USA
- Caryl and Israel Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Per Sandström
- Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Robert E Schwartz
- Division of Gastroenterology & Hepatology, Weill Cornell Medicine, New York, NY, USA
- Department of Biomedical Engineering, Cornell University, Ithaca, NY, USA
| | - William R Jarnagin
- Hepatopancreatobiliary Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- David M. Rubenstein Center for Pancreatic Cancer Research, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - David Lyden
- Departments of Pediatrics and Cell and Developmental Biology, Children's Cancer and Blood Foundation Laboratories, Drukier Institute for Children's Health, Meyer Cancer Center Weill Cornell Medicine, New York, NY, USA.
- David M. Rubenstein Center for Pancreatic Cancer Research, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| |
Collapse
|
2
|
Jiang H, Jiang J. Balancing act: the complex role of NK cells in immune regulation. Front Immunol 2023; 14:1275028. [PMID: 38022497 PMCID: PMC10652757 DOI: 10.3389/fimmu.2023.1275028] [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: 08/09/2023] [Accepted: 10/20/2023] [Indexed: 12/01/2023] Open
Abstract
Natural killer (NK) cells, as fundamental components of innate immunity, can quickly react to abnormalities within the body. In-depth research has revealed that NK cells possess regulatory functions not only in innate immunity but also in adaptive immunity under various conditions. Multiple aspects of the adaptive immune process are regulated through NK cells. In our review, we have integrated multiple studies to illuminate the regulatory function of NK cells in regulating B cell and T cell responses during adaptive immune processes, focusing on aspects including viral infections and the tumor microenvironment (TME). These insights provide us with many new understandings on how NK cells regulate different phases of the adaptive immune response.
Collapse
Affiliation(s)
- Hongwei Jiang
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
- Jiangsu Engineering Research Center for Tumor Immunotherapy, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
- Institute for Cell Therapy, Soochow University, Changzhou, Jiangsu, China
| | - Jingting Jiang
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
- Jiangsu Engineering Research Center for Tumor Immunotherapy, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
- Institute for Cell Therapy, Soochow University, Changzhou, Jiangsu, China
| |
Collapse
|
3
|
Imaoka Y, Ohira M, Chogahara I, Bekki T, Imaoka K, Sato K, Doskali M, Nakano R, Yano T, Hirata F, Kuroda S, Tahara H, Ide K, Ishiyama K, Kobayashi T, Tanaka Y, Ohdan H. Impact of a new liver immune status index among patients with hepatocellular carcinoma after initial hepatectomy. Ann Gastroenterol Surg 2023; 7:987-996. [PMID: 37927921 PMCID: PMC10623950 DOI: 10.1002/ags3.12702] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Revised: 05/05/2023] [Accepted: 05/14/2023] [Indexed: 11/07/2023] Open
Abstract
Aim The anti-tumor effects of natural killer (NK) cells vary among individuals. Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) expressed on liver NK cells is a marker of anti-tumor cytotoxicity against hepatocellular carcinoma (HCC) in immune cell therapy. This study aimed to develop a liver immune status index (LISI) that predicts low TRAIL expression and validates its ability to predict recurrence after initial hepatectomy for primary HCC. Methods A functional analysis of liver NK cells co-cultured with interleukin-2 for 3 days was performed of 40 liver transplant donors. The LISI, which predicted low TRAIL expression (25% quartile: <33%) in liver NK cells, was calculated using multiple logistic regression analysis. Next, 586 initial hepatectomy cases were analyzed based on the LISI. Results Our model was based on the Fibrosis-4 index+0.1 (odds ratio [OR], 1.33), body mass index (OR, 0.61), and albumin levels+0.1 (OR, 0.54). The area under the receiver operating characteristic curve (AUC) of the LISI for low TRAIL expression was 0.89. Stratification of the recurrence rates (RR) revealed that LISI was an independent predictive factor of RR (moderate risk: hazard ratio, 1.44; high risk: hazard ratio, 3.02). The AUC was similar for the LISI, albumin-indocyanine green evaluation grade, albumin-bilirubin score, and geriatric nutritional risk index for predicting RR. Among the vascular invasion cases, the LISI was more useful than the other indexes. Conclusion Our model facilitates the prediction of RR in high-risk patients by providing LISI to predict the anti-tumor effects of NK cells.
Collapse
Affiliation(s)
- Yuki Imaoka
- Department of Gastroenterological and Transplant SurgeryGraduate School of Biomedical and Health Sciences Hiroshima University, Hiroshima UniversityHiroshimaJapan
| | - Masahiro Ohira
- Department of Gastroenterological and Transplant SurgeryGraduate School of Biomedical and Health Sciences Hiroshima University, Hiroshima UniversityHiroshimaJapan
- Division of Regeneration and Medicine, Medical Center for Translational and Clinical ResearchHiroshima University HospitalHiroshimaJapan
| | - Ichiya Chogahara
- Department of Gastroenterological and Transplant SurgeryGraduate School of Biomedical and Health Sciences Hiroshima University, Hiroshima UniversityHiroshimaJapan
| | - Tomoaki Bekki
- Department of Gastroenterological and Transplant SurgeryGraduate School of Biomedical and Health Sciences Hiroshima University, Hiroshima UniversityHiroshimaJapan
| | - Kouki Imaoka
- Department of Gastroenterological and Transplant SurgeryGraduate School of Biomedical and Health Sciences Hiroshima University, Hiroshima UniversityHiroshimaJapan
| | - Koki Sato
- Department of Gastroenterological and Transplant SurgeryGraduate School of Biomedical and Health Sciences Hiroshima University, Hiroshima UniversityHiroshimaJapan
| | - Marlen Doskali
- Department of Gastroenterological and Transplant SurgeryGraduate School of Biomedical and Health Sciences Hiroshima University, Hiroshima UniversityHiroshimaJapan
| | - Ryosuke Nakano
- Department of Gastroenterological and Transplant SurgeryGraduate School of Biomedical and Health Sciences Hiroshima University, Hiroshima UniversityHiroshimaJapan
| | - Takuya Yano
- Department of Gastroenterological and Transplant SurgeryGraduate School of Biomedical and Health Sciences Hiroshima University, Hiroshima UniversityHiroshimaJapan
| | - Fumihiro Hirata
- Department of Gastroenterological and Transplant SurgeryGraduate School of Biomedical and Health Sciences Hiroshima University, Hiroshima UniversityHiroshimaJapan
| | - Shintaro Kuroda
- Department of Gastroenterological and Transplant SurgeryGraduate School of Biomedical and Health Sciences Hiroshima University, Hiroshima UniversityHiroshimaJapan
| | - Hiroyuki Tahara
- Department of Gastroenterological and Transplant SurgeryGraduate School of Biomedical and Health Sciences Hiroshima University, Hiroshima UniversityHiroshimaJapan
| | - Kentaro Ide
- Department of Gastroenterological and Transplant SurgeryGraduate School of Biomedical and Health Sciences Hiroshima University, Hiroshima UniversityHiroshimaJapan
| | - Kohei Ishiyama
- Department of Gastroenterological and Transplant SurgeryGraduate School of Biomedical and Health Sciences Hiroshima University, Hiroshima UniversityHiroshimaJapan
- Department of Renal Transplant SurgeryAichi Medical University School of MedicineNagakuteJapan
| | - Tsuyoshi Kobayashi
- Department of Gastroenterological and Transplant SurgeryGraduate School of Biomedical and Health Sciences Hiroshima University, Hiroshima UniversityHiroshimaJapan
| | - Yuka Tanaka
- Department of Gastroenterological and Transplant SurgeryGraduate School of Biomedical and Health Sciences Hiroshima University, Hiroshima UniversityHiroshimaJapan
| | - Hideki Ohdan
- Department of Gastroenterological and Transplant SurgeryGraduate School of Biomedical and Health Sciences Hiroshima University, Hiroshima UniversityHiroshimaJapan
| |
Collapse
|
4
|
Fahrner R, Gröger M, Settmacher U, Mosig AS. Functional integration of natural killer cells in a microfluidically perfused liver on-a-chip model. BMC Res Notes 2023; 16:285. [PMID: 37865791 PMCID: PMC10590007 DOI: 10.1186/s13104-023-06575-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: 06/10/2023] [Accepted: 10/13/2023] [Indexed: 10/23/2023] Open
Abstract
OBJECTIVE The liver acts as an innate immunity-dominant organ and natural killer (NK) cells, are the main lymphocyte population in the human liver. NK cells are in close interaction with other immune cells, acting as the first line of defense against pathogens, infections, and injury. A previously developed, three-dimensional, perfused liver-on-a-chip comprised of human cells was used to integrate NK cells, representing pivotal immune cells during liver injury and regeneration. The objective of this study was to integrate functional NK cells in an in vitro model of the human liver and assess utilization of the model for NK cell-dependent studies of liver inflammation. RESULTS NK cells from human blood and liver specimen were isolated by Percoll separation with subsequent magnetic cell separation (MACS), yielding highly purified blood and liver derived NK cells. After stimulation with toll-like-receptor (TLR) agonists (lipopolysaccharides, Pam3CSK4), isolated NK cells showed increased interferon (IFN)-gamma secretion. To study the role of NK cells in a complex hepatic environment, these cells were integrated in the vascular compartment of a microfluidically supported liver-on-a-chip model in close interaction with endothelial and resident macrophages. Successful, functional integration of NK cells was verified by immunofluorescence staining (NKp46), flow cytometry analysis and TLR agonist-dependent secretion of interleukin (IL)-6 and tumor necrosis factor (TNF)-alpha. Lastly, we observed that inflammatory activation of NK cells in the liver-on-a-chip led to a loss of vascular barrier integrity. Overall, our data shows the first successful, functional integration of NK cells in a liver-on-a-chip model that can be utilized to investigate NK cell-dependent effects on liver inflammation in vitro.
Collapse
Affiliation(s)
- René Fahrner
- Department of General, Visceral and Vascular Surgery, Jena University Hospital, 07747, Jena, Germany.
- Department of Vascular Surgery, University Hospital Bern, University of Bern, 3010, Bern, Switzerland.
- Integrated Research and Treatment Center, Center for Sepsis Control and Care (CSCC), Jena University Hospital, 07747, Jena, Germany.
| | - Marko Gröger
- Integrated Research and Treatment Center, Center for Sepsis Control and Care (CSCC), Jena University Hospital, 07747, Jena, Germany
| | - Utz Settmacher
- Department of General, Visceral and Vascular Surgery, Jena University Hospital, 07747, Jena, Germany
| | - Alexander S Mosig
- Integrated Research and Treatment Center, Center for Sepsis Control and Care (CSCC), Jena University Hospital, 07747, Jena, Germany
| |
Collapse
|
5
|
Zou M, Al-Yahya S, Al-Alwan M, BinEssa HA, Khabar KSA, Almohanna F, Assiri AM, Altaweel A, Qattan A, Meyer BF, Alzahrani AS, Shi Y. β-catenin attenuation leads to up-regulation of activating NKG2D ligands and tumor regression in BrafV600E-driven thyroid cancer cells. Front Immunol 2023; 14:1171816. [PMID: 37483610 PMCID: PMC10358762 DOI: 10.3389/fimmu.2023.1171816] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Accepted: 06/20/2023] [Indexed: 07/25/2023] Open
Abstract
Introduction BRAFV600E mutations frequently occur in papillary thyroid cancer (PTC). β-catenin, encoded by CTNNB1, is a key downstream component of the canonical Wnt signaling pathway and is often overexpressed in PTC. BRAFV600E-driven PTC tumors rely on Wnt/β-catenin signaling to sustain growth and progression. Methods In the present study, we investigated the tumorigenicity of thyroid cancer cells derived from BRAFV600E PTC mice following Ctnnb1 ablation (BVE-Ctnnb1null). Results Remarkably, the tumorigenic potential of BVE-Ctnnb1null tumor cells was lost in nude mice. Global gene expression analysis of BVE-Ctnnb1null tumor cells showed up-regulation of NKG2D receptor activating ligands (H60a, H60b, H60c, Raet1a, Raet1b, Raet1c, Raet1d, Raet1e, and Ulbp1) and down-regulation of inhibitory MHC class I molecules H-2L and H-2K2 in BVE-Ctnnb1null tumor cells. In vitro cytotoxicity assay demonstrated that BVE-Ctnnb1wt tumor cells were resistant to NK cell-mediated cytotoxicity, whereas BVE-Ctnnb1null tumor cells were sensitive to NK cell-mediated killing. Furthermore, the overexpression of any one of these NKG2D ligands in the BVE-Ctnnb1wt cell line resulted in a significant reduction of tumor growth in nude mice. Conclusions Our results indicate that active β-catenin signaling inhibits NK cell-mediated immune responses against thyroid cancer cells. Targeting the β-catenin signaling pathway may have significant therapeutic benefits for BRAF-mutant thyroid cancer by not only inhibiting tumor growth but also enhancing host immune surveillance.
Collapse
Affiliation(s)
- Minjing Zou
- Department of Molecular Oncology, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Suhad Al-Yahya
- Department of Molecular Biomedicine, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Monther Al-Alwan
- Department of Stem Cell & Tissue Re-engineering, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia and College of Medicine, Al-Faisal University, Riyadh, Saudi Arabia
| | - Huda A. BinEssa
- Department of Molecular Oncology, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Khalid S. A. Khabar
- Department of Molecular Biomedicine, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Falah Almohanna
- Department of Comparative Medicine, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Abdullah M. Assiri
- Department of Comparative Medicine, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Abdulmohsen Altaweel
- Mawhiba, King Abdulaziz and His Companions Foundation for Giftedness and Creativity, Riyadh, Saudi Arabia
| | - Amal Qattan
- Department of Molecular Oncology, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Brian F. Meyer
- Centre for Genomic Medicine, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Ali S. Alzahrani
- Department of Medicine, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Yufei Shi
- Department of Molecular Oncology, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| |
Collapse
|
6
|
Influenza Vaccination Reduces the Risk of Liver Cancer in Patients with Chronic Kidney Disease: A Nationwide Population-Based Cohort Study. Vaccines (Basel) 2022; 10:vaccines10122008. [PMID: 36560418 PMCID: PMC9784512 DOI: 10.3390/vaccines10122008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 11/19/2022] [Accepted: 11/23/2022] [Indexed: 11/26/2022] Open
Abstract
Previous studies have indicated that influenza vaccination reduces the development of lung cancer. However, the protective effects of influenza vaccination on primary liver cancer in patients with chronic kidney disease (CKD) are unclear. This cohort study identified 12,985 patients aged at least 55 years who had received a diagnosis of CKD between 1 January 2001 and 31 December 2012 from the National Health Insurance Research Database of Taiwan. The patients were classified according to vaccination status. Propensity score matching was used to reduce selection bias. Cox proportional hazards regression analysis was used to evaluate the correlation between influenza vaccination and primary liver cancer in patients with CKD. The prevalence of primary liver cancer was lower in patients with CKD who had received an influenza vaccine (adjusted hazard ratio: 0.45, 95% confidence interval [CI]: 0.35−0.58, p < 0.001). The protective effects were observed regardless of sex, age, and comorbidities. Moreover, dose-dependent protective effects were observed. In the subgroup analysis, where the patients were classified by the number of vaccinations received, the adjusted hazard ratios for 1, 2−3, and ≥4 vaccinations were 0.86 (95% CI: 0.63−1.17), 0.45 (95% CI: 0.31−0.63), and 0.21 (95% CI: 0.14−0.33), respectively. In conclusion, influenza vaccination was associated with a lower incidence of liver cancer in patients with CKD.
Collapse
|
7
|
Wei Y, Bingyu W, Lei Y, Xingxing Y. The antifibrotic role of natural killer cells in liver fibrosis. Exp Biol Med (Maywood) 2022; 247:1235-1243. [PMID: 35475367 PMCID: PMC9379607 DOI: 10.1177/15353702221092672] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Liver fibrosis is the common pathological change of chronic liver diseases characterized by increased deposition of extracellular matrix and reduced matrix degradation. In response to liver injury caused by a variety of pathogenic agents, such as virus and alcohol, hepatic stellate cells (HSCs) are differentiated into myofibroblast-like cells and produce excessive collagens, thus resulting in fibrogenesis. Natural killer (NK) cells are the essential innate immune cells in the liver and generally control fibrosis by killing activated HSCs. This review briefly describes the fibrogenesis process and the phenotypic features of hepatic NK cells. Besides, it focuses on the antifibrotic mechanisms of NK cells and explores the potential of activating NK cells as a therapeutic strategy for the disease.
Collapse
Affiliation(s)
- Yuan Wei
- Department of Hepatology, The First Affiliated Hospital of Hunan University of Traditional Chinese Medicine, Changsha 410000, China
| | - Wang Bingyu
- Department of Gastroenterology, Heilongjiang Academy of Traditional Chinese Medicine, Harbin 150001, China
| | - Yang Lei
- Department of Gastroenterology, Heilongjiang Academy of Traditional Chinese Medicine, Harbin 150001, China
| | - Yuan Xingxing
- Department of Gastroenterology, Heilongjiang Academy of Traditional Chinese Medicine, Harbin 150001, China,Yuan Xingxing.
| |
Collapse
|
8
|
Imaoka Y, Sato K, Ohira M, Imaoka K, Yano T, Nakano R, Tanaka Y, Ohdan H. Acute portal hypertension using portal vein ligation abrogates TRAIL expression of liver-resident NK cells. Hepatol Commun 2022; 6:2551-2564. [PMID: 35726345 PMCID: PMC9426399 DOI: 10.1002/hep4.2017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 03/16/2022] [Accepted: 05/09/2022] [Indexed: 11/29/2022] Open
Abstract
The effects of acute portal hypertension (PHT), which is reported as poor prognostic factors in patients with hepatocellular carcinoma, are not well known on the liver immune system, including natural killer (NK) cells. The aim of this study, therefore, was to investigate how acute PHT influences the functions and characteristics of liver‐resident NK (lr‐NK) cells using an acute PHT mouse model. Acute PHT decreased the number of tumor necrosis factor–related apoptosis‐inducing ligand (TRAIL+) lr‐NK cells by about 20% and attenuated cytotoxic activity against the Hepa1‐6 cell line by about 40%. Among various cytokine, only interleukin‐33 (IL‐33), which inhibits NK activity, significantly increased after portal vein ligation (PVL). Because lr‐NK cells highly expressed ST2/IL‐33R, IL‐33 co‐culture significantly suppressed TRAIL expression on lr‐NK cells by about 50%, and IL‐33 administration markedly decreased TRAIL expression and cytotoxic activity of lr‐NK cells. Furthermore, the TRAIL+ NK cells population was maintained by anti‐IL33 antibody or following portosystemic shunt procedure even after PVL. Finally, we demonstrated that IL‐33 decreased TRAIL expression in lr‐NK cells via AKT–forkhead box O (FoxO) and mitogen‐activated protein kinase (MAPK) signaling. Conclusion: This work demonstrates that PHT suppresses the TRAIL+ lr‐NK cell population and antitumor activities in the liver. Additionally, Akt‐FoxO and MAPK signaling pathways attenuate the TRAIL expression in lt‐NK cells via IL‐33 receptor in mice.
Collapse
Affiliation(s)
- Yuki Imaoka
- Department of Gastroenterological and Transplant Surgery, Graduate School of Biomedical and Health Sciences, Hiroshima University 1-2-3, Hiroshima, Japan
| | - Koki Sato
- Department of Gastroenterological and Transplant Surgery, Graduate School of Biomedical and Health Sciences, Hiroshima University 1-2-3, Hiroshima, Japan
| | - Masahiro Ohira
- Department of Gastroenterological and Transplant Surgery, Graduate School of Biomedical and Health Sciences, Hiroshima University 1-2-3, Hiroshima, Japan.,Medical Center for Translational and Clinical Research Hiroshima University Hospital, Hiroshima, Japan
| | - Kouki Imaoka
- Department of Gastroenterological and Transplant Surgery, Graduate School of Biomedical and Health Sciences, Hiroshima University 1-2-3, Hiroshima, Japan
| | - Takuya Yano
- Department of Surgery, Hiroshima City Hiroshima Citizens Hospital, Hiroshima, Japan
| | - Ryosuke Nakano
- Department of Gastroenterological and Transplant Surgery, Graduate School of Biomedical and Health Sciences, Hiroshima University 1-2-3, Hiroshima, Japan
| | - Yuka Tanaka
- Department of Gastroenterological and Transplant Surgery, Graduate School of Biomedical and Health Sciences, Hiroshima University 1-2-3, Hiroshima, Japan
| | - Hideki Ohdan
- Department of Gastroenterological and Transplant Surgery, Graduate School of Biomedical and Health Sciences, Hiroshima University 1-2-3, Hiroshima, Japan
| |
Collapse
|
9
|
Potential Pathophysiological Mechanisms Underlying Multiple Organ Dysfunction in Cytokine Release Syndrome. Mediators Inflamm 2022; 2022:7137900. [PMID: 35431655 PMCID: PMC9007670 DOI: 10.1155/2022/7137900] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Accepted: 03/16/2022] [Indexed: 12/15/2022] Open
Abstract
In recent decades, many serious respiratory infections have broken out all over the world, including SARS-CoV, MERS, and COVID-19. They are characterized by strong infectivity, rapid disease progression, high mortality, and poor prognosis. Excessive immune system activation results in cytokine hypersecretion, which is an important reason for the aggravation of symptoms, and can spread throughout the body leading to systemic multiple organ dysfunction, namely, cytokine release syndrome (CRS). Although many diseases related to CRS have been identified, the mechanism of CRS is rarely mentioned clearly. This review is intended to clarify the pathogenetic mechanism of CRS in the deterioration of related diseases, describe the important signaling pathways and clinical pathophysiological characteristics of CRS, and provide ideas for further research and development of specific drugs for corresponding targets to treat CRS.
Collapse
|
10
|
Ahmed O, Robinson MW, O'Farrelly C. Inflammatory processes in the liver: divergent roles in homeostasis and pathology. Cell Mol Immunol 2021; 18:1375-1386. [PMID: 33864004 PMCID: PMC8166849 DOI: 10.1038/s41423-021-00639-2] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Accepted: 01/10/2021] [Indexed: 02/07/2023] Open
Abstract
The hepatic immune system is designed to tolerate diverse harmless foreign moieties to maintain homeostasis in the healthy liver. Constant priming and regulation ensure that appropriate immune activation occurs when challenged by pathogens and tissue damage. Failure to accurately discriminate, regulate, or effectively resolve inflammation offsets this balance, jeopardizing overall tissue health resulting from an either overly tolerant or an overactive inflammatory response. Compelling scientific and clinical evidence links dysregulated hepatic immune and inflammatory responses upon sterile injury to several pathological conditions in the liver, particularly nonalcoholic steatohepatitis and ischemia-reperfusion injury. Murine and human studies have described interactions between diverse immune repertoires and nonhematopoietic cell populations in both physiological and pathological activities in the liver, although the molecular mechanisms driving these associations are not clearly understood. Here, we review the dynamic roles of inflammatory mediators in responses to sterile injury in the context of homeostasis and disease, the clinical implications of dysregulated hepatic immune activity and therapeutic developments to regulate liver-specific immunity.
Collapse
Affiliation(s)
- Ola Ahmed
- School of Medicine, Trinity College Dublin, Dublin, Ireland
| | - Mark W Robinson
- Department of Biology, Kathleen Lonsdale Institute for Human Health Research, Maynooth University, Maynooth, Ireland
| | - Cliona O'Farrelly
- School of Medicine, Trinity College Dublin, Dublin, Ireland.
- School of Biochemistry & Immunology, Trinity College Dublin, Dublin 2, Ireland.
| |
Collapse
|
11
|
Araúzo-Bravo MJ, Delic D, Gerovska D, Wunderlich F. Protective Vaccination Reshapes Hepatic Response to Blood-Stage Malaria of Genes Preferentially Expressed by NK Cells. Vaccines (Basel) 2020; 8:vaccines8040677. [PMID: 33202767 PMCID: PMC7712122 DOI: 10.3390/vaccines8040677] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 11/06/2020] [Accepted: 11/11/2020] [Indexed: 02/04/2023] Open
Abstract
The role of natural killer (NK) cells in the liver as first-line post infectionem (p.i.) effectors against blood-stage malaria and their responsiveness to protective vaccination is poorly understood. Here, we investigate the effect of vaccination on NK cell-associated genes induced in the liver by blood-stage malaria of Plasmodium chabaudi. Female Balb/c mice were vaccinated at weeks 3 and 1 before being infected with 106P. chabaudi-parasitized erythrocytes. Genes preferentially expressed by NK cells were investigated in livers of vaccination-protected and non-protected mice on days 0, 1, 4, 8, and 11 p.i. using microarrays, qRT-PCR, and chromosome landscape analysis. Blood-stage malaria induces expression of specific genes in the liver at different phases of infection, i.e., Itga1 in expanding liver-resident NK (lrNK) cells, Itga2 in immigrating conventional NK (cNK) cells; Eomes and Tbx21 encoding transcription factors; Ncr1, Tnfsf10, Prf1, Gzma, Gzmb, Gzmc, Gzmm, and Gzmk encoding cytolytic effectors; natural killer gene complex (NKC)-localized genes encoding the NK cell receptors KLRG1, KLRK1, KLRAs1, 2, 5, 7, KLRD1, KLRC1, KLRC3, as well as the three receptors KLRB1A, KLRB1C, KLRB1F and their potential ligands CLEC2D and CLEC2I. Vaccination enhances this malaria-induced expression of genes, but impairs Gzmm expression, accelerates decline of Tnfsf10 and Clec2d expression, whereas it accelerates increased expression of Clec2i, taking a very similar time course as that of genes encoding plasma membrane proteins of erythroblasts, whose malaria-induced extramedullary generation in the liver is known to be accelerated by vaccination. Collectively, vaccination reshapes the response of the liver NK cell compartment to blood-stage malaria. Particularly, the malaria-induced expansion of lrNK cells peaking on day 4 p.i. is highly significantly (p < 0.0001) reduced by enhanced immigration of peripheral cNK cells, and KLRB1F:CLEC2I interactions between NK cells and erythroid cells facilitate extramedullary erythroblastosis in the liver, thus critically contributing to vaccination-induced survival of otherwise lethal blood-stage malaria of P. chabaudi.
Collapse
Affiliation(s)
- Marcos J. Araúzo-Bravo
- Group of Computational Biology and Systems Biomedicine, Biodonostia Health Research Institute, 20014 San Sebastián, Spain;
- IKERBASQUE, Basque Foundation for Science, 48009 Bilbao, Spain
- TransBioNet Thematic Network of Excellence for Transitional Bioinformatics, Barcelona Supercomputing Center, 08034 Barcelona, Spain
- Correspondence: (M.J.A.-B.); (D.D.); Tel.: +34-943006108 (M.J.A.-B.); +49-735154143839 (D.D.)
| | - Denis Delic
- Boeringer Ingelheim Pharma, 88400 Biberach, Germany
- Correspondence: (M.J.A.-B.); (D.D.); Tel.: +34-943006108 (M.J.A.-B.); +49-735154143839 (D.D.)
| | - Daniela Gerovska
- Group of Computational Biology and Systems Biomedicine, Biodonostia Health Research Institute, 20014 San Sebastián, Spain;
| | - Frank Wunderlich
- Department of Biology, Heinrich-Heine-University, 40225 Düsseldorf, Germany;
| |
Collapse
|
12
|
Ciner AT, Jones K, Muschel RJ, Brodt P. The unique immune microenvironment of liver metastases: Challenges and opportunities. Semin Cancer Biol 2020; 71:143-156. [PMID: 32526354 DOI: 10.1016/j.semcancer.2020.06.003] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 06/04/2020] [Accepted: 06/05/2020] [Indexed: 02/07/2023]
Abstract
Liver metastases from gastrointestinal and non-gastrointestinal malignancies remain a major cause of cancer-related mortality and a major clinical challenge. The liver has unique properties that facilitate metastatic expansion, including a complex immune system that evolved to dampen immunity to neoantigens entering the liver from the gut, through the portal circulation. In this review, we describe the unique microenvironment encountered by cancer cells in the liver, focusing on elements of the innate and adaptive immune response that can act as a double-edge sword, contributing to the elimination of cancer cells on the one hand and promoting their survival and growth, on the other. We discuss this microenvironment in a clinical context, particularly for colorectal carcinoma, and highlight how a better understanding of the role of the microenvironment has spurred an intense effort to develop novel and innovative strategies for targeting liver metastatic disease, some of which are currently being tested in the clinic.
Collapse
Affiliation(s)
- Aaron T Ciner
- Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, United States
| | - Keaton Jones
- Oxford Institute for Radiation Oncology, Department of Surgery, University of Oxford, Oxford, UK
| | - Ruth J Muschel
- Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford, UK
| | - Pnina Brodt
- Departments of Surgery, Medicine and Oncology, McGill University, and the Research Institute of the McGill University Health Center, Montreal, QC, Canada.
| |
Collapse
|
13
|
Key features and homing properties of NK cells in the liver are shaped by activated iNKT cells. Sci Rep 2019; 9:16362. [PMID: 31704965 PMCID: PMC6841958 DOI: 10.1038/s41598-019-52666-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Accepted: 10/16/2019] [Indexed: 11/16/2022] Open
Abstract
The contribution of natural killer (NK) cells to the clearance of hepatic viral infections is well recognized. The recently discovered heterogeneity of NK cell populations renders them interesting targets for immune interventions. Invariant natural killer T (iNKT) cells represent a key interaction partner for hepatic NK cells. The present study addressed whether characteristics of NK cells in the liver can be shaped by targeting iNKT cells. For this, the CD1d-binding pegylated glycolipid αGalCerMPEG was assessed for its ability to modulate the features of NK cells permanently or transiently residing in the liver. In vivo administration resulted in enhanced functionality of educated and highly differentiated CD27+ Mac-1+ NK cells accompanied by an increased proliferation. Improved liver homing was supported by serum-derived and cellular factors. Reduced viral loads in a mCMV infection model confirmed the beneficial effect of NK cells located in the liver upon stimulation with αGalCerMPEG. Thus, targeting iNKT cell-mediated NK cell activation in the liver represents a promising approach for the establishment of liver-directed immune interventions.
Collapse
|
14
|
Natural killer cells involved in tumour immune escape of hepatocellular carcinomar. Int Immunopharmacol 2019; 73:10-16. [PMID: 31078921 DOI: 10.1016/j.intimp.2019.04.057] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 03/21/2019] [Accepted: 04/25/2019] [Indexed: 02/08/2023]
Abstract
Natural killer cells are the first line of host immune surveillance and play major roles in the defence against infection and tumours. Hepatic NK cells exhibit unique phenotypic and functional characteristics compared to circulating and spleen NK cells, such as higher levels of cytolytic activity and cytotoxicity mediators against tumour cells. However, the activities of NK cells may be reversed during tumour progression. Recent studies demonstrated that hepatic NK cells were exhausted in hepatocellular carcinoma (HCC) and exhibited impaired cytolytic activity and decreased production of effector cytokines. The present review discusses current knowledge on the role of exhausted NK cells in promoting HCC development and the mechanisms contributing to tumour immune escape, including an imbalance of activating and inhibitory receptors on NK cells, abnormal receptor-ligand interaction, and cross-talk with immune cells and other stromal cells in the tumour environment. We provide a fundamental basis for further study of innate immunity in tumour progression and serve the purpose of exploring new HCC treatment strategies.
Collapse
|
15
|
Patra T, Ray RB, Ray R. Strategies to Circumvent Host Innate Immune Response by Hepatitis C Virus. Cells 2019; 8:E274. [PMID: 30909456 PMCID: PMC6468774 DOI: 10.3390/cells8030274] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Revised: 03/15/2019] [Accepted: 03/18/2019] [Indexed: 12/13/2022] Open
Abstract
Innate immune responses generate interferons, proinflammatory cytokines, complement activation, and natural killer (NK) cell response. Ultimately, this leads to the induction of a robust virus-specific adaptive immunity. Although the host innate immune system senses and responds to eliminate virus infection, hepatitis C virus (HCV) evades immune attack and establishes persistent infection within the liver. Spontaneous clearance of HCV infection is associated with a prompt induction of innate immunity generated in an infected host. In this review, we have highlighted the current knowledge of our understanding of host⁻HCV interactions, especially for endogenous interferon production, proinflammatory response, NK cell response, and complement activation, which may impair the generation of a strong adaptive immune response for establishment of chronicity. The information may provide novel strategies in augmenting therapeutic intervention against HCV.
Collapse
Affiliation(s)
- Tapas Patra
- Departments of Internal Medicine, Saint Louis University, St. Louis, MO 63104, USA.
| | - Ratna B Ray
- Departments of Pathology, Saint Louis University, St. Louis, MO 63104, USA.
| | - Ranjit Ray
- Departments of Internal Medicine, Saint Louis University, St. Louis, MO 63104, USA.
- Molecular Microbiology & Immunology, Saint Louis University, St. Louis, MO 63104, USA.
| |
Collapse
|
16
|
Castriconi R, Carrega P, Dondero A, Bellora F, Casu B, Regis S, Ferlazzo G, Bottino C. Molecular Mechanisms Directing Migration and Retention of Natural Killer Cells in Human Tissues. Front Immunol 2018; 9:2324. [PMID: 30364222 PMCID: PMC6193061 DOI: 10.3389/fimmu.2018.02324] [Citation(s) in RCA: 83] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Accepted: 09/18/2018] [Indexed: 12/29/2022] Open
Abstract
A large body of data shows that Natural Killer (NK) cells are immune effectors exerting a potent cytolytic activity against tumors and virus infected cells. The discovery and characterization of several inhibitory and activating receptors unveiled most of the mechanisms allowing NK cells to spare healthy cells while selectively attacking abnormal tissues. Nevertheless, the mechanisms ruling NK cell subset recirculation among the different compartments of human body have only lately started to be investigated. This is particularly true for pathological settings such as tumors or infected tissues but also for para-physiological condition like pregnant human uterine mucosa. It is becoming evident that the microenvironment associated to a particular clinical condition can deeply influence the migratory capabilities of NK cells. In this review we describe the main mechanisms and stimuli known to regulate the expression of chemokine receptors and other molecules involved in NK cell homing to either normal or pathological/inflamed tissues, including tumors or organs such as lung and liver. We will also discuss the role played by the chemokine/chemokine receptor axes in the orchestration of physiological events such as NK cell differentiation, lymphoid organ retention/egress and recruitment to decidua during pregnancy.
Collapse
Affiliation(s)
- Roberta Castriconi
- Dipartimento di Medicina Sperimentale, University of Genova, Genova, Italy.,Centro di Eccellenza per la Ricerca Biomedica, University of Genova, Genova, Italy
| | - Paolo Carrega
- Dipartimento di Patologia Umana, University of Messina, Messina, Italy
| | - Alessandra Dondero
- Dipartimento di Medicina Sperimentale, University of Genova, Genova, Italy
| | - Francesca Bellora
- Dipartimento di Medicina Sperimentale, University of Genova, Genova, Italy
| | - Beatrice Casu
- Dipartimento di Medicina Sperimentale, University of Genova, Genova, Italy
| | - Stefano Regis
- Istituto di ricovero e cura a carattere scientifico (IRCCS) Giannina Gaslini, Genova, Italy
| | - Guido Ferlazzo
- Dipartimento di Patologia Umana, University of Messina, Messina, Italy
| | - Cristina Bottino
- Dipartimento di Medicina Sperimentale, University of Genova, Genova, Italy.,Istituto di ricovero e cura a carattere scientifico (IRCCS) Giannina Gaslini, Genova, Italy
| |
Collapse
|
17
|
Li S, Yang D, Peng T, Wu Y, Tian Z, Ni B. Innate lymphoid cell-derived cytokines in autoimmune diseases. J Autoimmun 2017; 83:62-72. [PMID: 28479212 DOI: 10.1016/j.jaut.2017.05.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Revised: 03/31/2017] [Accepted: 05/01/2017] [Indexed: 02/07/2023]
Abstract
The most recently recognized types of immune cells, the innate lymphoid cells (ILCs), have been sub-divided according to respective distinct expression profiles of regulatory factors or/and cytokines. ILCs have also been shown to participate in a variety of beneficial immune responses, including participation in attack against pathogens and mediation of the pre-inflammatory and inflammatory responses through their production of pro-inflammatory cytokines. As such, while the ILCs exert protective effects they may also become detrimental upon dysregulation. Indeed, recent studies of the ILCs have revealed a strong association with the advent and pathogenesis of several common autoimmune diseases, including psoriasis, inflammatory bowel disease (IBD) and multiple sclerosis (MS). Though the ILCs belong to lineage negative cells that are distinctive from the Th cells, the profiles of secreted cytokines from the ILCs overlap with those of the corresponding Th subsets. Nevertheless, considering that the ILCs belong to the innate immune system and the Th cells belong to the adaptive immune system, it is expected that the ILCs should function at the early stage of diseases and the Th cells should exert predominant effects at the late stage of diseases. Therefore, it is intriguing to consider targeting of ILCs for therapy by targeting the corresponding cytokines at the early stage of diseases, with the late stage cytokine targeting mainly influencing the Th cells' function. Here, we review the knowledge to date on the roles of ILCs in various autoimmune diseases and discuss their potential as new therapeutic targets.
Collapse
Affiliation(s)
- Sirui Li
- Department of Pathophysiology and High Altitude Pathology, Third Military Medical University, Chongqing 400038, PR China; Institute of Immunology, PLA, Third Military Medical University, Chongqing 400038, PR China; Battalion 3 of Cadet Brigade, Third Military Medical University, Chongqing 400038, PR China
| | - Di Yang
- Institute of Immunology, PLA, Third Military Medical University, Chongqing 400038, PR China
| | - Tingwei Peng
- Department of Pathophysiology and High Altitude Pathology, Third Military Medical University, Chongqing 400038, PR China; Institute of Immunology, PLA, Third Military Medical University, Chongqing 400038, PR China; Battalion 3 of Cadet Brigade, Third Military Medical University, Chongqing 400038, PR China
| | - Yuzhang Wu
- Institute of Immunology, PLA, Third Military Medical University, Chongqing 400038, PR China.
| | - Zhiqiang Tian
- Institute of Immunology, PLA, Third Military Medical University, Chongqing 400038, PR China.
| | - Bing Ni
- Department of Pathophysiology and High Altitude Pathology, Third Military Medical University, Chongqing 400038, PR China; Institute of Immunology, PLA, Third Military Medical University, Chongqing 400038, PR China.
| |
Collapse
|
18
|
Meyer CE, Key PN, Zhu T, Shabsovich M, Ni A, Tripathy SK. Expression of the inhibitory receptor NKG2A correlates with increased liver and splenic NK cell response to activating receptor engagement. IMMUNITY INFLAMMATION AND DISEASE 2017; 5:177-189. [PMID: 28474506 PMCID: PMC5418142 DOI: 10.1002/iid3.156] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Revised: 01/30/2017] [Accepted: 02/02/2017] [Indexed: 12/16/2022]
Abstract
Introduction Natural killer (NK) cells play a critical role in the innate immune response to viruses and tumors, and comprise a large proportion of the hepatic lymphocyte population. They must remain tolerant to non‐pathogenic antigens while protecting the host from harmful agents. Herein, we investigate how the NK cell response to activation receptor engagement is altered in the liver. Methods In this study, we assess IFN‐γ production and degranulation of splenic NK cells and selected subsets of liver NK cells. Flow cytometry (FCM) was used to asses IFN‐γ production and degranulation following stimulation of the NK cells with plate bound antibodies to activating receptors. Results We show that smaller percentages of hepatic NK cells produce interferon (IFN)–γ and/or degranulate than do splenic NK cells upon stimulation through activating receptors. We also found that smaller percentages of the circulating NK (cNK) cells in the liver produce IFN‐γ and/or degranulate, compared to the liver tissue resident NK (trNK) cells. In addition, IFN‐γ production by liver cNK cells is not increased in IL‐10 deficient mice, suggesting that their hyporesponsiveness is not mediated by the presence of this anti‐inflammatory cytokine in the hepatic microenvironment. On the other hand, liver trNK cells express higher levels of the inhibitory receptor NKG2A than do cNK cells, correlating with their increased IFN‐γ production and degranulation. Conclusions Liver cNK cells’ hyporesponsiveness to stimulation through activating receptors is independent of IL‐10, but correlates with decreased NKG2A expression compared to trNK cells. In addition, we demonstrate that liver NK cells become further hyporesponsive upon continuous engagement of an activating receptor on their cell surface.
Collapse
Affiliation(s)
- Claire E Meyer
- Gastroenterology Division, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Phillip N Key
- Gastroenterology Division, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Toby Zhu
- Gastroenterology Division, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Mark Shabsovich
- Gastroenterology Division, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Ann Ni
- Gastroenterology Division, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Sandeep K Tripathy
- Gastroenterology Division, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| |
Collapse
|
19
|
Tosello-Trampont A, Surette FA, Ewald SE, Hahn YS. Immunoregulatory Role of NK Cells in Tissue Inflammation and Regeneration. Front Immunol 2017; 8:301. [PMID: 28373874 PMCID: PMC5357635 DOI: 10.3389/fimmu.2017.00301] [Citation(s) in RCA: 80] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Accepted: 03/03/2017] [Indexed: 12/17/2022] Open
Abstract
NK cells represent an important first line of defense against viral infection and cancer and are also involved in tissue homeostasis. Studies of NK cell activation in the last decade have revealed that they are able to respond to the inflammatory stimuli evoked by tissue damage and contribute to both progression and resolution of diseases. Exacerbation of the inflammatory response through interactions between immune effector cells facilitates the progression of non-alcoholic fatty liver disease (NAFLD) into steatosis, cirrhosis, and hepatocellular carcinoma (HCC). When hepatic damage is incurred, macrophage activation is crucial for initiating cross talk with neighboring cells present in the liver, including hepatocytes and NK cells, and the importance of this interaction in shaping the immune response in liver disease is increasingly recognized. Inflicted structural damage can be in part regenerated via the process of self-limiting fibrosis, though persistent hepatic damage will lead to chronic fibrosis and loss of tissue organization and function. The cytotoxic activity of NK cells plays an important role in inducing hepatic stellate cell apoptosis and thus curtailing the progression of fibrosis. Alternatively, in some diseases, such as HCC, NK cells may become dysregulated, promoting an immunosuppressive state where tumors are able to escape immune surveillance. This review describes the current understanding of the contributions of NK cells to tissue inflammation and metabolic liver diseases and the ongoing effort to develop therapeutics that target the immunoregulatory function of NK cells.
Collapse
Affiliation(s)
| | - Fionna A Surette
- Beirne B. Carter Center for Immunology Research , Charlottesville, VA , USA
| | - Sarah E Ewald
- Beirne B. Carter Center for Immunology Research, Charlottesville, VA, USA; Department of Microbiology, University of Virginia, Charlottesville, VA, USA
| | - Young S Hahn
- Beirne B. Carter Center for Immunology Research, Charlottesville, VA, USA; Department of Microbiology, University of Virginia, Charlottesville, VA, USA
| |
Collapse
|
20
|
Qian X, Hu C, Han S, Lin Z, Xiao W, Ding Y, Zhang Y, Qian L, Jia X, Zhu G, Gong W. NK1.1 - CD4 + NKG2D + T cells suppress DSS-induced colitis in mice through production of TGF-β. J Cell Mol Med 2017; 21:1431-1444. [PMID: 28224733 PMCID: PMC5487917 DOI: 10.1111/jcmm.13072] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2016] [Accepted: 11/25/2016] [Indexed: 12/23/2022] Open
Abstract
CD4+NKG2D+ T cells are associated with tumour, infection and autoimmune diseases. Some CD4+NKG2D+ T cells secrete IFN‐γ and TNF‐α to promote inflammation, but others produce TGF‐β and FasL to facilitate tumour evasion. Here, murine CD4+NKG2D+ T cells were further classified into NK1.1−CD4+NKG2D+ and NK1.1+CD4+NKG2D+ subpopulations. The frequency of NK1.1−CD4+NKG2D+ cells decreased in inflamed colons, whereas more NK1.1+CD4+NKG2D+ cells infiltrated into colons of mice with DSS‐induced colitis. NK1.1−CD4+NKG2D+ cells expressed TGF‐β and FasL without secreting IFN‐γ, IL‐21 and IL‐17 and displayed no cytotoxicity. The adoptive transfer of NK1.1−CD4+NKG2D+ cells suppressed DSS‐induced colitis largely dependent on TGF‐β. NK1.1−CD4+NKG2D+ cells did not expressed Foxp3, CD223 (LAG‐3) and GITR. The subpopulation was distinct from NK1.1+CD4+NKG2D+ cells in terms of surface markers and RNA transcription. NK1.1−CD4+NKG2D+ cells also differed from Th2 or Th17 cells because the former did not express GATA‐3 and ROR‐γt. Thus, NK1.1−CD4+NKG2D+ cells exhibited immune regulatory functions, and this T cell subset could be developed to suppress inflammation in clinics.
Collapse
Affiliation(s)
- Xingxing Qian
- Department of Immunology, School of Medicine, Yangzhou University, Yangzhou, China
| | - Chunxia Hu
- Department of Immunology, School of Medicine, Yangzhou University, Yangzhou, China
| | - Sen Han
- Department of Immunology, School of Medicine, Yangzhou University, Yangzhou, China
| | - Zhijie Lin
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou, China.,Jiangsu Key Laboratory of Zoonosis, Yangzhou, China
| | - Weiming Xiao
- Department of Gastroenterology, The Second Clinical Medical College, Yangzhou University, Yangzhou, China
| | - Yanbing Ding
- Department of Gastroenterology, The Second Clinical Medical College, Yangzhou University, Yangzhou, China
| | - Yu Zhang
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou, China
| | - Li Qian
- Department of Immunology, School of Medicine, Yangzhou University, Yangzhou, China
| | - Xiaoqing Jia
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou, China
| | - Guoqiang Zhu
- Jiangsu Key Laboratory of Zoonosis, Yangzhou, China.,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| | - Weijuan Gong
- Department of Immunology, School of Medicine, Yangzhou University, Yangzhou, China.,Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou, China.,Jiangsu Key Laboratory of Zoonosis, Yangzhou, China.,Department of Gastroenterology, The Second Clinical Medical College, Yangzhou University, Yangzhou, China.,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| |
Collapse
|
21
|
Wałajtys-Rode E, Dzik JM. Monocyte/Macrophage: NK Cell Cooperation-Old Tools for New Functions. Results Probl Cell Differ 2017; 62:73-145. [PMID: 28455707 DOI: 10.1007/978-3-319-54090-0_5] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Monocyte/macrophage and natural killer (NK) cells are partners from a phylogenetic standpoint of innate immune system development and its evolutionary progressive interaction with adaptive immunity. The equally conservative ways of development and differentiation of both invertebrate hemocytes and vertebrate macrophages are reviewed. Evolutionary conserved molecules occurring in macrophage receptors and effectors have been inherited by vertebrates after their common ancestor with invertebrates. Cytolytic functions of mammalian NK cells, which are rooted in immune cells of invertebrates, although certain NK cell receptors (NKRs) are mammalian new events, are characterized. Broad heterogeneity of macrophage and NK cell phenotypes that depends on surrounding microenvironment conditions and expression profiles of specific receptors and activation mechanisms of both cell types are discussed. The particular tissue specificity of macrophages and NK cells, as well as their plasticity and mechanisms of their polarization to different functional subtypes have been underlined. The chapter summarized studies revealing the specific molecular mechanisms and regulation of NK cells and macrophages that enable their highly specific cross-cooperation. Attention is given to the evolving role of human monocyte/macrophage and NK cell interaction in pathogenesis of hypersensitivity reaction-based disorders, including autoimmunity, as well as in cancer surveillance and progression.
Collapse
Affiliation(s)
- Elżbieta Wałajtys-Rode
- Faculty of Chemistry, Department of Drug Technology and Biotechnology, Warsaw University of Technology, Noakowskiego 3 Str, 00-664, Warsaw, Poland.
| | - Jolanta M Dzik
- Faculty of Agriculture and Biology, Department of Biochemistry, Warsaw University of Life Sciences-SGGW, Warsaw, Poland
| |
Collapse
|
22
|
Tripathi D, Venkatasubramanian S, Cheekatla SS, Paidipally P, Welch E, Tvinnereim AR, Vankayalapati R. A TLR9 agonist promotes IL-22-dependent pancreatic islet allograft survival in type 1 diabetic mice. Nat Commun 2016; 7:13896. [PMID: 27982034 PMCID: PMC5171644 DOI: 10.1038/ncomms13896] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Accepted: 11/08/2016] [Indexed: 12/20/2022] Open
Abstract
Pancreatic islet transplantation is a promising potential cure for type 1 diabetes (T1D). Islet allografts can survive long term in the liver parenchyma. Here we show that liver NK1.1+ cells induce allograft tolerance in a T1D mouse model. The tolerogenic effects of NK1.1+ cells are mediated through IL-22 production, which enhances allograft survival and increases insulin secretion. Increased expression of NKG2A by liver NK1.1+ cells in islet allograft-transplanted mice is involved in the production of IL-22 and in the reduced inflammatory response to allografts. Vaccination of T1D mice with a CpG oligonucleotide TLR9 agonist (ODN 1585) enhances expansion of IL-22-producing CD3-NK1.1+ cells in the liver and prolongs allograft survival. Our study identifies a role for liver NK1.1+ cells, IL-22 and CpG oligonucleotides in the induction of tolerance to islet allografts in the liver parenchyma.
Collapse
Affiliation(s)
- Deepak Tripathi
- Department of Pulmonary Immunology, Center for Biomedical Research, University of Texas Health Science Center at Tyler, Tyler, Texas 75708, USA
| | - Sambasivan Venkatasubramanian
- Department of Pulmonary Immunology, Center for Biomedical Research, University of Texas Health Science Center at Tyler, Tyler, Texas 75708, USA
| | - Satyanarayana S. Cheekatla
- Department of Pulmonary Immunology, Center for Biomedical Research, University of Texas Health Science Center at Tyler, Tyler, Texas 75708, USA
| | - Padmaja Paidipally
- Department of Pulmonary Immunology, Center for Biomedical Research, University of Texas Health Science Center at Tyler, Tyler, Texas 75708, USA
| | - Elwyn Welch
- Department of Pulmonary Immunology, Center for Biomedical Research, University of Texas Health Science Center at Tyler, Tyler, Texas 75708, USA
| | - Amy R. Tvinnereim
- Department of Pulmonary Immunology, Center for Biomedical Research, University of Texas Health Science Center at Tyler, Tyler, Texas 75708, USA
| | - Ramakrishna Vankayalapati
- Department of Pulmonary Immunology, Center for Biomedical Research, University of Texas Health Science Center at Tyler, Tyler, Texas 75708, USA
| |
Collapse
|
23
|
Tseng HC, Kanayama K, Kaur K, Park SH, Park S, Kozlowska A, Sun S, McKenna CE, Nishimura I, Jewett A. Bisphosphonate-induced differential modulation of immune cell function in gingiva and bone marrow in vivo: role in osteoclast-mediated NK cell activation. Oncotarget 2016; 6:20002-25. [PMID: 26343372 PMCID: PMC4652983 DOI: 10.18632/oncotarget.4755] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2015] [Accepted: 07/16/2015] [Indexed: 11/25/2022] Open
Abstract
The aim of this study is to establish osteoclasts as key immune effectors capable of activating the function of Natural Killer (NK) cells, and expanding their numbers, and to determine in vivo and in vitro effect of bisphosphonates (BPs) during NK cell interaction with osteoclasts and on systemic and local immune function. The profiles of 27 cytokines, chemokines and growth factors released from osteoclasts were found to be different from dendritic cells and M1 macrophages but resembling to untreated monocytes and M2 macrophages. Nitrogen-containing BPs Zoledronate (ZOL) and Alendronate (ALN), but not non-nitrogen-containing BPs Etidronate (ETI), triggered increased release of pro-inflammatory mediators from osteoclasts while all three BPs decreased pit formation by osteoclasts. ZOL and ALN mediated significant release of IL-6, TNF-` and IL-1β, whereas they inhibited IL-10 secretion by osteoclasts. Treatment of osteoclasts with ZOL inhibited NK cell mediated cytotoxicity whereas it induced significant secretion of cytokines and chemokines. NK cells lysed osteoclasts much more than their precursor cells monocytes, and this correlated with the decreased expression of MHC class I expression on osteoclasts. Intravenous injection of ZOL in mice induced pro-inflammatory microenvironment in bone marrow and demonstrated significant immune activation. By contrast, tooth extraction wound of gingival tissues exhibited profound immune suppressive microenvironment associated with dysregulated wound healing to the effect of ZOL which could potentially be responsible for the pathogenesis of Osteonecrosis of the Jaw (ONJ). Finally, based on the data obtained in this paper we demonstrate that osteoclasts can be used as targets for the expansion of NK cells with superior function for immunotherapy of cancer.
Collapse
Affiliation(s)
- Han-Ching Tseng
- Division of Oral Biology and Medicine, The Jane and Jerry Weintraub Center for Reconstructive Biotechnology, UCLA School of Dentistry, Los Angeles, CA, USA
| | - Keiichi Kanayama
- Division of Advanced Prosthodontics, The Jane and Jerry Weintraub Center for Reconstructive Biotechnology, UCLA School of Dentistry, Los Angeles, CA, USA.,Department of Periodontology, Asahi University School of Dentistry, Gifu, Japan
| | - Kawaljit Kaur
- Division of Oral Biology and Medicine, The Jane and Jerry Weintraub Center for Reconstructive Biotechnology, UCLA School of Dentistry, Los Angeles, CA, USA
| | - So-Hyun Park
- Division of Oral Biology and Medicine, The Jane and Jerry Weintraub Center for Reconstructive Biotechnology, UCLA School of Dentistry, Los Angeles, CA, USA
| | - Sil Park
- Division of Oral Biology and Medicine, The Jane and Jerry Weintraub Center for Reconstructive Biotechnology, UCLA School of Dentistry, Los Angeles, CA, USA.,Division of Advanced Prosthodontics, The Jane and Jerry Weintraub Center for Reconstructive Biotechnology, UCLA School of Dentistry, Los Angeles, CA, USA
| | - Anna Kozlowska
- Division of Oral Biology and Medicine, The Jane and Jerry Weintraub Center for Reconstructive Biotechnology, UCLA School of Dentistry, Los Angeles, CA, USA.,Department of Tumor Immunology, Poznan University of Medical Sciences, Poznan, Poland
| | - Shuting Sun
- Department of Chemistry, University of Southern California, Los Angeles, CA, USA
| | - Charles E McKenna
- Department of Chemistry, University of Southern California, Los Angeles, CA, USA
| | - Ichiro Nishimura
- Division of Oral Biology and Medicine, The Jane and Jerry Weintraub Center for Reconstructive Biotechnology, UCLA School of Dentistry, Los Angeles, CA, USA.,Division of Advanced Prosthodontics, The Jane and Jerry Weintraub Center for Reconstructive Biotechnology, UCLA School of Dentistry, Los Angeles, CA, USA
| | - Anahid Jewett
- Division of Oral Biology and Medicine, The Jane and Jerry Weintraub Center for Reconstructive Biotechnology, UCLA School of Dentistry, Los Angeles, CA, USA
| |
Collapse
|
24
|
Horst AK, Neumann K, Diehl L, Tiegs G. Modulation of liver tolerance by conventional and nonconventional antigen-presenting cells and regulatory immune cells. Cell Mol Immunol 2016; 13:277-92. [PMID: 27041638 PMCID: PMC4856800 DOI: 10.1038/cmi.2015.112] [Citation(s) in RCA: 176] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2015] [Revised: 12/18/2015] [Accepted: 12/18/2015] [Indexed: 12/11/2022] Open
Abstract
The liver is a tolerogenic organ with exquisite mechanisms of immune regulation that ensure upkeep of local and systemic immune tolerance to self and foreign antigens, but that is also able to mount effective immune responses against pathogens. The immune privilege of liver allografts was recognized first in pigs in spite of major histo-compatibility complex mismatch, and termed the "liver tolerance effect". Furthermore, liver transplants are spontaneously accepted with only low-dose immunosuppression, and induce tolerance for non-hepatic co-transplanted allografts of the same donor. Although this immunotolerogenic environment is favorable in the setting of organ transplantation, it is detrimental in chronic infectious liver diseases like hepatitis B or C, malaria, schistosomiasis or tumorigenesis, leading to pathogen persistence and weak anti-tumor effects. The liver is a primary site of T-cell activation, but it elicits poor or incomplete activation of T cells, leading to their abortive activation, exhaustion, suppression of their effector function and early death. This is exploited by pathogens and can impair pathogen control and clearance or allow tumor growth. Hepatic priming of T cells is mediated by a number of local conventional and nonconventional antigen-presenting cells (APCs), which promote tolerance by immune deviation, induction of T-cell anergy or apoptosis, and generating and expanding regulatory T cells. This review will focus on the communication between classical and nonclassical APCs and lymphocytes in the liver in tolerance induction and will discuss recent insights into the role of innate lymphocytes in this process.
Collapse
Affiliation(s)
- Andrea Kristina Horst
- Institute of Experimental Immunology and Hepatology Center for Experimental Medicine, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, Hamburg D-20246, Germany
| | - Katrin Neumann
- Institute of Experimental Immunology and Hepatology Center for Experimental Medicine, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, Hamburg D-20246, Germany
| | - Linda Diehl
- Institute of Experimental Immunology and Hepatology Center for Experimental Medicine, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, Hamburg D-20246, Germany
| | - Gisa Tiegs
- Institute of Experimental Immunology and Hepatology Center for Experimental Medicine, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, Hamburg D-20246, Germany
| |
Collapse
|
25
|
Chen H, Zhidan W, Xia R, Zhaoxia W, Qing J, Qiang G, Haipeng Y, Hengxiao W. Scorpion venom activates natural killer cells in hepatocellular carcinoma via the NKG2D-MICA pathway. Int Immunopharmacol 2016; 35:307-314. [PMID: 27089390 DOI: 10.1016/j.intimp.2016.03.045] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Revised: 03/25/2016] [Accepted: 03/25/2016] [Indexed: 12/23/2022]
Abstract
Previous studies have demonstrated that polypeptides extracted from scorpion venom (PESV) inhibited cell proliferation in several tumors, however, the effect on dysfunctional and exhausted natural killer cells which contribute to tumor escape from immune surveillance remain to be elucidated. In this study, we determined the effect of PESV on NK infiltration into H22 cells orthotopic transplantation tumors and on the expression of MHC class I chain-related proteins A (MICA) in HepG2 cells. We found that tumor growth in mice was significantly inhibited by PESV and the survival time of tumor-bearing mice treated with PESV was significantly prolonged. Moreover, levels of tumor-infiltrating NK cells, NKG2D protein, perforin and granzyme B mRNA were significantly increased in the group treated with PESV compared with the tumor-bearing control group. In addition, In addition, up-regulation of MICA by PESV enhances the susceptibility of HepG2 cells to NK lysis in vitro. These results indicate that the inhibitory effects of PESV on hepatic carcinoma are likely mediated by up-regulation of NK cell activity via the MICA-NKG2D pathway.
Collapse
Affiliation(s)
- Han Chen
- Institute of Basic Medicine, Shandong Academy of Medical Sciences, Jinan 250062, China; School of Medicine and Life Sciences, University of Jinan-Shandong Academy of Medical Sciences, Jinan 250062, China
| | - Wang Zhidan
- Department of Frontier Health Sciences, Tokyo Metropolitan University, 7-2-10 Higashiogu, Arakawa-ku, Tokyo, Japan
| | - Ren Xia
- Institute of Basic Medicine, Shandong Academy of Medical Sciences, Jinan 250062, China
| | - Wang Zhaoxia
- Institute of Basic Medicine, Shandong Academy of Medical Sciences, Jinan 250062, China
| | - Jia Qing
- Institute of Basic Medicine, Shandong Academy of Medical Sciences, Jinan 250062, China
| | - Guo Qiang
- Institute of Basic Medicine, Shandong Academy of Medical Sciences, Jinan 250062, China
| | - Yin Haipeng
- Institute of Basic Medicine, Shandong Academy of Medical Sciences, Jinan 250062, China
| | - Wang Hengxiao
- Institute of Basic Medicine, Shandong Academy of Medical Sciences, Jinan 250062, China.
| |
Collapse
|
26
|
Tosello-Trampont AC, Krueger P, Narayanan S, Landes SG, Leitinger N, Hahn YS. NKp46(+) natural killer cells attenuate metabolism-induced hepatic fibrosis by regulating macrophage activation in mice. Hepatology 2016; 63:799-812. [PMID: 26662852 PMCID: PMC4764418 DOI: 10.1002/hep.28389] [Citation(s) in RCA: 96] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2015] [Accepted: 12/06/2015] [Indexed: 12/17/2022]
Abstract
UNLABELLED Nonalcoholic steatohepatitis (NASH) affects 3%-5% of the U.S. population, having severe clinical complications to the development of fibrosis and end-stage liver diseases, such as cirrhosis and hepatocellular carcinoma. A critical cause of NASH is chronic systemic inflammation promoted by innate immune cells, such as liver macrophages (Mϕ) and natural killer (NK) cells. However, little is known about how the crosstalk between Mϕ and NK cells contributes to regulate NASH progression to fibrosis. In this report, we demonstrate that NKp46(+) cells play an important role in preventing NASH progression to fibrosis by regulating M1/M2 polarization of liver Mϕ. Using a murine model of NASH, we demonstrate that DX5(+)NKp46(+) NK cells are increased during disease and play a role in polarizing Mϕ toward M1-like phenotypes. This NK's immunoregulatory function depends on the production of interferon-gamma (IFN-γ), but not by granzyme-mediated cytolytic activity. Notably, depletion of NKp46(+) cells promotes the development of fibrosis with increased expression of profibrogenic genes as well as skewed M2 Mϕ phenotypes in hepatic tissues. CONCLUSIONS NK cell-derived IFN-γ may be essential for maintaining a balanced inflammatory environment that promotes tissue integrity and limiting NASH progression to fibrosis.
Collapse
Affiliation(s)
| | - Peter Krueger
- Beirne Carter Center for Immunology Research, University of Virginia, Charlottesville, VA 22908,Department of Microbiology, Immunology and Cancer Biology, University of Virginia, Charlottesville, VA 22908
| | - Sowmya Narayanan
- Beirne Carter Center for Immunology Research, University of Virginia, Charlottesville, VA 22908,Department of Microbiology, Immunology and Cancer Biology, University of Virginia, Charlottesville, VA 22908
| | - Susan G. Landes
- Beirne Carter Center for Immunology Research, University of Virginia, Charlottesville, VA 22908
| | - Norbert Leitinger
- Department of Pharmacology, University of Virginia, Charlottesville, VA 22908
| | - Young S. Hahn
- Beirne Carter Center for Immunology Research, University of Virginia, Charlottesville, VA 22908,Department of Microbiology, Immunology and Cancer Biology, University of Virginia, Charlottesville, VA 22908
| |
Collapse
|
27
|
Souza-Fonseca-Guimaraes F, Young A, Mittal D, Martinet L, Bruedigam C, Takeda K, Andoniou CE, Degli-Esposti MA, Hill GR, Smyth MJ. NK cells require IL-28R for optimal in vivo activity. Proc Natl Acad Sci U S A 2015; 112:E2376-84. [PMID: 25901316 PMCID: PMC4426428 DOI: 10.1073/pnas.1424241112] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Natural killer (NK) cells are naturally circulating innate lymphoid cells that protect against tumor initiation and metastasis and contribute to immunopathology during inflammation. The signals that prime NK cells are not completely understood, and, although the importance of IFN type I is well recognized, the role of type III IFN is comparatively very poorly studied. IL-28R-deficient mice were resistant to LPS and cecal ligation puncture-induced septic shock, and hallmark cytokines in these disease models were dysregulated in the absence of IL-28R. IL-28R-deficient mice were more sensitive to experimental tumor metastasis and carcinogen-induced tumor formation than WT mice, and additional blockade of interferon alpha/beta receptor 1 (IFNAR1), but not IFN-γ, further enhanced metastasis and tumor development. IL-28R-deficient mice were also more susceptible to growth of the NK cell-sensitive lymphoma, RMAs. Specific loss of IL-28R in NK cells transferred into lymphocyte-deficient mice resulted in reduced LPS-induced IFN-γ levels and enhanced tumor metastasis. Therefore, by using IL-28R-deficient mice, which are unable to signal type III IFN-λ, we demonstrate for the first time, to our knowledge, the ability of IFN-λ to directly regulate NK cell effector functions in vivo, alone and in the context of IFN-αβ.
Collapse
Affiliation(s)
- Fernando Souza-Fonseca-Guimaraes
- Immunology in Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute, Herston, QLD 4006, Australia; School of Medicine, University of Queensland, St. Lucia, QLD 4006, Australia
| | - Arabella Young
- Immunology in Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute, Herston, QLD 4006, Australia; School of Medicine, University of Queensland, St. Lucia, QLD 4006, Australia
| | - Deepak Mittal
- Immunology in Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute, Herston, QLD 4006, Australia
| | - Ludovic Martinet
- Immunology in Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute, Herston, QLD 4006, Australia
| | - Claudia Bruedigam
- Translational Leukaemia Research Laboratory, QIMR Berghofer Medical Research Institute, Herston, QLD 4006, Australia
| | - Kazuyoshi Takeda
- Division of Cell Biology, Biomedical Research Center, Graduate School of Medicine, Juntendo University, Bunkyo-ku, Tokyo 113-8421, Japan
| | - Christopher E Andoniou
- Immunology and Virology Program, Centre for Ophthalmology and Visual Science, University of Western Australia, Crawley, WA 6009, Australia; Centre for Experimental Immunology, Lions Eye Institute, Nedlands, WA 6009, Australia
| | - Mariapia A Degli-Esposti
- Immunology and Virology Program, Centre for Ophthalmology and Visual Science, University of Western Australia, Crawley, WA 6009, Australia; Centre for Experimental Immunology, Lions Eye Institute, Nedlands, WA 6009, Australia
| | - Geoffrey R Hill
- Bone Marrow Transplantation Laboratory, QIMR Berghofer Medical Research Institute, Herston, QLD 4006, Australia; and Department of Bone Marrow Transplantation, Royal Brisbane Hospital, Brisbane, QLD 4006, Australia
| | - Mark J Smyth
- Immunology in Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute, Herston, QLD 4006, Australia; School of Medicine, University of Queensland, St. Lucia, QLD 4006, Australia;
| |
Collapse
|
28
|
Yamagiwa S, Sato Y, Ichida T, Setsu T, Tominaga K, Kamimura H, Tsuchiya A, Takamura M, Matsuda Y, Aoyagi Y. Imbalance between CD56+bright and CD56+dim natural killer cell subsets in the liver of patients with recurrent hepatitis C after liver transplantation. Biomed Res 2015; 35:177-84. [PMID: 24942856 DOI: 10.2220/biomedres.35.177] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Progressive liver fibrosis remains a major problem for patients with recurrent chronic hepatitis C(CHC) after liver transplantation (LT). However, the involvement of natural killer (NK) and natural killer T (NKT) cells, which predominate in the liver, in recurrent CHC after LT remains unclear.In the present study, we investigated the status of NK and NKT cells in the liver and peripheral blood obtained from 10 patients with recurrent CHC after LT (LT-C), 15 patients with CHC, and 7 normal donors for living donor LT. CD56+ NK cells were separated into two subsets: CD56+bright subset, which is identified as major NK cytokine producer, and CD56+dim subset, which has greater spontaneous cytotoxicity. We found a significant decrease in the CD56+bright subset in the liver of patients with LT-C compared to patients with CHC (P<0.01) and normal donors (P=0.03). The expression of inhibitory NK cell receptor NKG2A was significantly increased on intrahepatic CD56+bright subset in LT-C patients, and activated CD69+CD56+dim NK cell subset was significantly increased in the liver of LT-C patients. Our results suggest that a significant imbalance between CD56+bright and CD56+dim NK cell subsets in the liver may contribute to the progression of recurrent CHC after LT.
Collapse
Affiliation(s)
- Satoshi Yamagiwa
- Division of Gastroenterology and Hepatology, Niigata University Graduate School of Medical and Dental Sciences, 1-757 Asahimachidori, Chuo-ku, Niigata 951-8510, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Chelakkot-Govindalayathil AL, Mifuji-Moroka R, D'Alessandro-Gabazza CN, Toda M, Matsuda Y, Gil-Bernabe P, Roeen Z, Yasuma T, Yano Y, Gabazza EC, Iwasa M, Takei Y. Protein S exacerbates alcoholic hepatitis by stimulating liver natural killer T cells. J Thromb Haemost 2015; 13:142-54. [PMID: 25399514 DOI: 10.1111/jth.12789] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2014] [Accepted: 11/05/2014] [Indexed: 12/15/2022]
Abstract
BACKGROUND Alcohol consumption is a major cause of liver injury but the mechanisms are not completely understood. Protein S (PS) is an anticoagulant glycoprotein with multiple functions. The role of PS in liver injury is unknown. OBJECTIVES This study investigated the role of PS in acute alcoholic hepatitis. METHODS A mouse overexpressing human PS (hPS-TG) was generated in which acute hepatitis was induced by intraperitoneal injection of ethanol. RESULTS The levels of serum liver enzymes and liver tissue inflammatory cytokines and the degree of hepatic steatosis were significantly increased in hPS-TG mice treated with ethanol compared with ethanol-treated wild type (WT) mice. Cell expansion, activation and inhibition of apoptosis were significantly augmented in natural killer T (NKT) cells from hPS-TG mice compared with WT mice. Liver mononuclear cells from hPS-TG mice express higher levels of inflammatory cytokines than those from WT mice after stimulation with a specific stimulant of NKT cells in vitro. In a co-culture system of hepatocytes and NKT cells, the effects of PS on ethanol-mediated cell injury were suppressed by a CD1d neutralizing antibody. Alcoholic liver injury was significantly improved in mice pre-treated with PS siRNA and anti-protein S antibody compared with control mice. Patients with alcoholic hepatitis showed significantly increased plasma PS levels and enhanced liver expression of PS and CD1d compared with controls. CONCLUSIONS The results of this study suggest that PS exacerbates acute alcoholic hepatitis by inhibiting apoptosis of activated NKT cells.
Collapse
MESH Headings
- Animals
- Antibodies, Neutralizing/pharmacology
- Antigens, CD1d/immunology
- Antigens, CD1d/metabolism
- Apoptosis
- Blood Proteins/genetics
- Blood Proteins/metabolism
- Case-Control Studies
- Cells, Cultured
- Coculture Techniques
- Disease Models, Animal
- Ethanol
- Fatty Liver, Alcoholic/immunology
- Fatty Liver, Alcoholic/metabolism
- Fatty Liver, Alcoholic/pathology
- Hepatitis, Alcoholic/genetics
- Hepatitis, Alcoholic/immunology
- Hepatitis, Alcoholic/metabolism
- Hepatitis, Alcoholic/pathology
- Hepatitis, Alcoholic/prevention & control
- Hepatocytes/immunology
- Hepatocytes/metabolism
- Hepatocytes/pathology
- Humans
- Inflammation Mediators/immunology
- Inflammation Mediators/metabolism
- Liver/immunology
- Liver/metabolism
- Liver/pathology
- Lymphocyte Activation
- Male
- Mice, Inbred C57BL
- Mice, Transgenic
- Natural Killer T-Cells/immunology
- Natural Killer T-Cells/metabolism
- Protein S/genetics
- Protein S/metabolism
- RNAi Therapeutics
- Severity of Illness Index
- Signal Transduction
- Up-Regulation
Collapse
|
30
|
Natural history of chronic hepatitis B virus infection. Med Microbiol Immunol 2014; 204:5-10. [PMID: 25540037 DOI: 10.1007/s00430-014-0369-7] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2014] [Accepted: 10/02/2014] [Indexed: 02/06/2023]
Abstract
Hepatitis B virus infection represents a major global health problem. Currently, there are more than 240 million chronically infected people worldwide. The development of chronic hepatitis B virus-mediated liver disease may lead to liver fibrosis, cirrhosis and eventually hepatocellular carcinoma. Recently, the discovery of the viral entry receptor sodium taurocholate cotransporting polypeptide has facilitated new approaches for a better understanding of viral physiopathology. Hopefully, these novel insights may give rise to the development of more effective antiviral therapy concepts during the next years. In this review, we will discuss the natural history of hepatitis B virus infection including the viral biology, the clinical course of infection and the role of the immune response.
Collapse
|
31
|
Abdel-Latif M, Sakran T, El-Shahawi G, El-Fayoumi H, El-Mallah AM. Immunomodulatory effect of diethylcarbamazine citrate plus filarial excretory-secretory product on rat hepatocarcinogenesis. Int Immunopharmacol 2014; 24:173-181. [PMID: 25499729 DOI: 10.1016/j.intimp.2014.12.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2014] [Revised: 11/06/2014] [Accepted: 12/01/2014] [Indexed: 01/03/2023]
Abstract
Diethylcarbamazine citrate (DEC) had a significance in anti-filarial chemotherapy, while excretory-secretory product (ES) is released from adult filarial females. The target of the current study was to examine the immunomodulatory effect of DEC, Setaria equina ES or a combination of them on rat hepatocellular carcinoma (HCC) induced by diethylnitrosamine (DEN). In vitro effect of combined DEC and ES or ES alone on lipopolysaccharide (LPS)-stimulated rat peripheral blood mononuclear cells (PBMCs) was tested through IFN-γ assay in culture supernatants. In addition, single or repeated doses of DEC, ES or DEC+ES have been applied in white albino rats to test the effect on HCC. Levels of IFN-γ and anti-ES IgG antibodies in rat serum were assayed using ELISA. Hemolytic complement activity (CH50) was determined in serum while the concentration of nitric oxide (NO) was assayed in liver tissue. The infiltration of NK cells as well as the expression of MHC Iproliferating cell nuclear antigen (PCNA), inducible NO synthase (iNOS), Bcl2 and p53 were determined using immunohistochemistry. There was a dose-dependent increase in IFN-γ after in vitro exposure to DEC+ES. Repeated ES doses increased NO concentration (p<0.05) and expression of iNOS but reduced CH50 (p<0.001), while repeated DEC+ES doses could increase anti-ES IgG (p<0.01), IFN-γ level (p<0.05) and NK cell infiltration. The same treatments could also reduce the expression of MHC I expression, PCNA, Bcl2 and p53. This study has shown immunomodulatory and protective effects of DEC+ES repeated doses on rat HCC.
Collapse
Affiliation(s)
- Mahmoud Abdel-Latif
- Zoology Department, Faculty of Science, Beni-Suef University, 62511 Salah Salem Street, Beni-Suef, Egypt.
| | - Thabet Sakran
- Zoology Department, Faculty of Science, Beni-Suef University, 62511 Salah Salem Street, Beni-Suef, Egypt
| | - Gamal El-Shahawi
- Zoology Department, Faculty of Science, Beni-Suef University, 62511 Salah Salem Street, Beni-Suef, Egypt
| | - Hoda El-Fayoumi
- Zoology Department, Faculty of Science, Beni-Suef University, 62511 Salah Salem Street, Beni-Suef, Egypt
| | - Al-Mahy El-Mallah
- Zoology Department, Faculty of Science, Beni-Suef University, 62511 Salah Salem Street, Beni-Suef, Egypt
| |
Collapse
|
32
|
Jouvion G, Brock M, Droin-Bergere S, Ibrahim-Granet O. Duality of liver and kidney lesions after systemic infection of immunosuppressed and immunocompetent mice withAspergillus fumigatus. Virulence 2014; 3:43-50. [DOI: 10.4161/viru.3.1.18654] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
|
33
|
In vivo administration of a JAK3 inhibitor during acute SIV infection leads to significant increases in viral load during chronic infection. PLoS Pathog 2014; 10:e1003929. [PMID: 24603870 PMCID: PMC3946395 DOI: 10.1371/journal.ppat.1003929] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2013] [Accepted: 12/31/2013] [Indexed: 12/30/2022] Open
Abstract
The studies reported herein are the first to document the effect of the in vivo administration of a JAK3 inhibitor for defining the potential role of NK cells during acute SIV infection of a group of 15 rhesus macaques (RM). An additional group of 16 MHC/KIR typed RM was included as controls. The previously optimized in vivo dose regimen (20 mg/kg daily for 35 days) led to a marked depletion of each of the major NK cell subsets both in the blood and gastro-intestinal tissues (GIT) during acute infection. While such depletion had no detectable effects on plasma viral loads during acute infection, there was a significant sustained increase in plasma viral loads during chronic infection. While the potential mechanisms that lead to such increased plasma viral loads during chronic infection remain unclear, several correlates were documented. Thus, during acute infection, the administration of the JAK3 inhibitor besides depleting all NK cell subsets also decreased some CD8+ T cells and inhibited the mobilization of the plasmacytoid dendritic cells in the blood and their localization to the GIT. Of interest is the finding that the administration of the JAK3 inhibitor during acute infection also resulted in the sustained maintenance during chronic infection of a high number of naïve and central memory CD4+ T cells, increases in B cells in the blood, but decreases in the frequencies and function of NKG2a+ NK cells within the GIT and blood, respectively. These data identify a unique role for JAK3 inhibitor sensitive cells, that includes NK cells during acute infection that in concert lead to high viral loads in SIV infected RM during chronic infection without affecting detectable changes in antiviral humoral/cellular responses. Identifying the precise mechanisms by which JAK3 sensitive cells exert their influence is critical with important implications for vaccine design against lentiviruses. In efforts to define the potential role of innate immune effector mechanisms in influencing the course of SIV infection during the acute infection period, our lab utilized the in vivo daily administration of 20 mg/kg orally of a compound called Tofacitinib (a Janus kinase 3 inhibitor) to a group of 15 rhesus macaques starting at day −6 and until day 28 post intravenous SIVmac239 infection. An additional group of 16 similarly SIV infected rhesus macaques served as a placebo control. This drug targets the JAK/STAT pathway that is utilized by cells including the NK cell lineage, a major cell of the innate immune system. The dosage utilized was based on extensive previous PK studies that resulted in a marked depletion of the NK cells. Of interest while such drug administration had no effect on plasma viral loads during acute infection, such drug administration led to significant increases in plasma and gastro-intestinal tissues (GIT) viral loads during chronic infection. A series of phenotypic/functional studies were performed to determine the mechanisms for this delayed effect and the correlates identified. These data are the first to document the effect of JAK-3 inhibitor during acute SIV infection with implications for HIV vaccine design.
Collapse
|
34
|
Hepatitis C virus impairs natural killer cell-mediated augmentation of complement synthesis. J Virol 2013; 88:2564-71. [PMID: 24352446 DOI: 10.1128/jvi.02988-13] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
UNLABELLED Natural killer (NK) cells and the complement system play critical roles in the first line of defense against pathogens. The synthesis of complement components C4 and C3 is transcriptionally downregulated by hepatitis C virus (HCV) core and NS5A proteins, and this negative regulation is apparent in chronically HCV-infected patients. In this study, we have examined the potential contribution of an NK cell line as a model in regulating complement synthesis. Coculture of NK cells (NK3.3) with human hepatoma cells (Huh7.5) expressing HCV core or NS5A protein led to a significant increase in C4 and C3 complement synthesis via enhanced specific transcription factors. Reestablishment of complement protein expression was found to be mediated by direct interaction between NKG2D on NK cells and the hepatocyte protein major histocompatibility complex class I-related chains A and B (MICA/B) and not to be associated with specific cytokine signaling events. On the other hand, C4 and C3 synthesis remained impaired in a coculture of NK cells and Huh7.5 cells infected with cell culture-grown HCV. The association between these two cell types through NKG2D and MICA/B was examined further, with MICA/B expression in HCV-infected hepatocytes found to remain inhibited during coculture. Further experiments revealed that the HCV NS2 and NS5B proteins are responsible for the HCV-associated decrease in MICA/B. These results suggest that HCV disables a key receptor ligand in infected hepatoma cells, thereby inhibiting the ability of infected cells to respond to stimuli from NK cells to positively regulate complement synthesis. IMPORTANCE The complement system contributes to the protection of the host from virus infection. However, the involvement of complement in viral hepatitis has not been well documented. Whether NK cells affect complement component expression in HCV-infected hepatocytes remains unknown. Here, we have shown how HCV subverts the ability of NK cells to positively mediate complement protein expression.
Collapse
|
35
|
Role of natural killer and Gamma-delta T cells in West Nile virus infection. Viruses 2013; 5:2298-310. [PMID: 24061543 PMCID: PMC3798903 DOI: 10.3390/v5092298] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2013] [Revised: 08/30/2013] [Accepted: 09/16/2013] [Indexed: 11/18/2022] Open
Abstract
Natural Killer (NK) cells and Gamma-delta T cells are both innate lymphocytes that respond rapidly and non-specifically to viral infection and other pathogens. They are also known to form a unique link between innate and adaptive immunity. Although they have similar immune features and effector functions, accumulating evidence in mice and humans suggest these two cell types have distinct roles in the control of infection by West Nile virus (WNV), a re-emerging pathogen that has caused fatal encephalitis in North America over the past decade. This review will discuss recent studies on these two cell types in protective immunity and viral pathogenesis during WNV infection.
Collapse
|
36
|
Takahashi Y, Mayne AE, Khowawisetsut L, Pattanapanyasat K, Little D, Villinger F, Ansari AA. In vivo administration of a JAK3 inhibitor to chronically siv infected rhesus macaques leads to NK cell depletion associated with transient modest increase in viral loads. PLoS One 2013; 8:e70992. [PMID: 23923040 PMCID: PMC3724739 DOI: 10.1371/journal.pone.0070992] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2013] [Accepted: 06/30/2013] [Indexed: 01/31/2023] Open
Abstract
Innate immune responses are reasoned to play an important role during both acute and chronic SIV infection and play a deterministic role during the acute stages on the rate of infection and disease progression. NK cells are an integral part of the innate immune system but their role in influencing the course of SIV infection has been a subject of debate. As a means to delineate the effect of NK cells on SIV infection, use was made of a Janus kinase 3 (JAK3) inhibitor that has previously been shown to be effective in the depletion of NK cells in vivo in nonhuman primates (NHP). Extensive safety and in vitro/in vivo PK studies were conducted and an optimal dose that depletes NK cells and NK cell function in vivo identified. Six chronically SIV infected rhesus macaques, 3 with undetectable/low plasma viral loads and 3 with high plasma viral loads were administered a daily oral dose of 10 mg/kg for 35 days. Data obtained showed that, at the dose tested, the major cell lineage affected both in the blood and the GI tissues were the NK cells. Such depletion appeared to be associated with a transient increase in plasma and GI tissue viral loads. Whereas the number of NK cells returned to baseline values in the blood, the GI tissues remained depleted of NK cells for a prolonged period of time. Recent findings show that the JAK3 inhibitor utilized in the studies reported herein has a broader activity than previously reported with dose dependent effects on both JAK2 and JAK1 suggests that it is likely that multiple pathways are affected with the administration of this drug that needs to be taken into account. The findings reported herein are the first studies on the use of a JAK3 inhibitor in lentivirus infected NHP.
Collapse
Affiliation(s)
- Yoshiaki Takahashi
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Ann E. Mayne
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Ladawan Khowawisetsut
- Department of Immunology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Kovit Pattanapanyasat
- Office for Research and Development, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Dawn Little
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Francois Villinger
- Division of Pathology, Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, United States of America
| | - Aftab A. Ansari
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia, United States of America
- * E-mail:
| |
Collapse
|
37
|
Abstract
Organs such as the liver, uterus and lung possess hallmark immunotolerant features, making these organs important for sustaining self-homeostasis. These organs contain a relatively large amount of negative regulatory immune cells, which are believed to take part in the regulation of immune responses. Because natural killer cells constitute a large proportion of all lymphocytes in these organs, increasing attention has been given to the roles that these cells play in maintaining immunotolerance. Here, we review the distribution, differentiation, phenotypic features and functional features of natural killer cells in these immunotolerant organs, in addition to the influence of local microenvironments on these cells and how these factors contribute to organ-specific diseases.
Collapse
|
38
|
Tian Z, Chen Y, Gao B. Natural killer cells in liver disease. Hepatology 2013; 57:1654-62. [PMID: 23111952 PMCID: PMC3573257 DOI: 10.1002/hep.26115] [Citation(s) in RCA: 212] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2012] [Revised: 10/01/2012] [Accepted: 10/12/2012] [Indexed: 12/12/2022]
Abstract
Natural killer (NK) cells are enriched in lymphocytes within the liver and have unique phenotypic features and functional properties, including tumor necrosis factor-related apoptosis-inducing ligand-dependent cytotoxicity and specific cytokine profiles. As a key component of innate immunity in the liver, NK cells perform critical roles in host defense against pathogens and tumors through their natural cytotoxicity and cytokine production, and they also act as regulatory cells by engaging in reciprocal interactions with other types of liver cells through cell-to-cell contact and the production of cytokines. Accumulating evidence from the last decade suggests that NK cells play an important role in controlling viral hepatitis, liver fibrosis, and liver tumorigenesis, but also contribute to the pathogenesis of liver injury and inflammation. The characterization of intrahepatic NK cell functions has not only helped us to better understand the pathogenesis of liver disease, but has also revealed new therapeutic targets for managing this disease.
Collapse
Affiliation(s)
- Zhigang Tian
- Department of Microbiology and Immunology, School of Life Sciences, University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Yongyan Chen
- Department of Microbiology and Immunology, School of Life Sciences, University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Bin Gao
- Laboratory of Liver Diseases, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
39
|
Li N, Puga Yung GL, Pradier A, Toso C, Giostra E, Morard I, Spahr L, Seebach JD. NK cell isolation from liver biopsies: phenotypic and functional analysis of low cell numbers by flow cytometry. Front Immunol 2013; 4:61. [PMID: 23482713 PMCID: PMC3593626 DOI: 10.3389/fimmu.2013.00061] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2012] [Accepted: 02/22/2013] [Indexed: 12/29/2022] Open
Abstract
Natural killer (NK) cells are considered to play a critical role in liver disease. However, the available numbers of intrahepatic lymphocytes (IHL) derived from liver biopsies (LB) for ex vivo analysis of intrahepatic NK cells is very limited; and the isolation method may hamper not only yields and viability, but also phenotype and function of IHL. The aim of the present study was therefore to (1) refine and evaluate the cell yields and viability of a modified isolation protocol from standard size needle LB; and (2) to test the effects of mechanical dissociation and enzymatic tissue digestion, as well as the analysis of very low cell numbers, on the phenotype and function of intrahepatic NK cells. Peripheral blood mononuclear cells (PBMC) and IHL, freshly isolated from the peripheral blood, LB (n = 11) or partial liver resections (n = 5), were used for phenotypic analysis by flow cytometry. NK cell function, i.e., degranulation and cytokine production, was determined by staining of CD107a and intracellular IFN-γ following in vitro stimulation. The mean weight of the LB specimens was 9.1 mg, and a mean number of 7,364 IHL/mg were obtained with a viability of >90%. Exposure of IHL and PBMC to 0.5 mg/ml collagenase IV and 0.02 mg/ml DNase I for 30 min did affect neither the viability, NK cell function, nor the percentages of CD56+, NKp46+, and CD16+ NK cells, whereas the level of CD56 surface expression was reduced. The phenotype of LB-derived NK cells was reliably characterized by acquiring as few as 2,500 IHL per tube for flow cytometry. The functional assay of intrahepatic NK cells was miniaturized by culturing as few as 25,000 IHL in 25 μl (106/ml) using 96-well V-bottom plates with IL-2 and IL-12 overnight, followed by a 4 h stimulation with K562 cells at a NK:K562 ratio of 1:1. In summary, we report reliable phenotypic and functional analyses of small numbers of intrahepatic NK cells isolated from LB specimens providing us with a tool to better address the emerging role of human NK cell immunobiology in liver diseases.
Collapse
Affiliation(s)
- Ning Li
- Division of Clinical Immunology and Allergology, Department of Medical Specialties, University Hospital and Medical Faculty Geneva, Switzerland
| | | | | | | | | | | | | | | |
Collapse
|
40
|
Lipidoid Nanoparticles Containing PD-L1 siRNA Delivered In Vivo Enter Kupffer Cells and Enhance NK and CD8(+) T Cell-mediated Hepatic Antiviral Immunity. MOLECULAR THERAPY. NUCLEIC ACIDS 2013; 2:e72. [PMID: 23423360 PMCID: PMC3586800 DOI: 10.1038/mtna.2012.63] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Effective clinical application of antiviral immunotherapies necessitates enhancing the functional state of natural killer (NK) and CD8+ T cells. An important mechanism for the establishment of viral persistence in the liver is the activation of the PD-1/PD-L1 inhibitory pathway. To examine the role of hepatic myeloid PD-L1 expression during viral infection, we determined the magnitude and quality of antiviral immune responses by administering PD-L1 short-interfering RNA (siRNA) encapsulated in lipidoid nanoparticles (LNP) in mice. Our studies indicate that Kupffer cells (KC) preferentially engulfed PD-L1 LNP within a short period of time and silenced Pdl1 during adenovirus and MCMV infection leading to enhanced NK and CD8+ T cell intrahepatic accumulation, effector function (interferon (IFN)-γ and granzyme B (GrB) production), CD8+ T cell–mediated viral clearance, and memory. Our results demonstrate that PD-L1 knockdown on KCs is central in determining the outcome of liver viral infections, and they represent a new class of gene therapy.
Collapse
|
41
|
Carrega P, Ferlazzo G. Natural killer cell distribution and trafficking in human tissues. Front Immunol 2012; 3:347. [PMID: 23230434 PMCID: PMC3515878 DOI: 10.3389/fimmu.2012.00347] [Citation(s) in RCA: 123] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2012] [Accepted: 11/03/2012] [Indexed: 12/31/2022] Open
Abstract
Few data are available regarding the recirculation of natural killer (NK) cells among human organs. Earlier studies have been often impaired by the use of markers then proved to be either not sufficiently specific for NK cells (e.g., CD57, CD56) or expressed only by subsets of NK cells (e.g., CD16). At the present, available data confirmed that human NK cells populate blood, lymphoid organs, lung, liver, uterus (during pregnancy), and gut. Several studies showed that NK cell homing appears to be subset-specific, as secondary lymphoid organs and probably several solid tissues are preferentially inhabited by CD56brightCD16neg/dull non-cytotoxic NK cells. Similar studies performed in the mouse model showed that lymph node and bone marrow are preferentially populated by CD11bdull NK cells while blood, spleen, and lung by CD27dull NK cells. Therefore, an important topic to be addressed in the human system is the contribution of factors that regulate NK cell tissue homing and egress, such as chemotactic receptors or homeostatic mechanisms. Here, we review the current knowledge on NK cell distribution in peripheral tissues and, based on recent acquisitions, we propose our view regarding the recirculation of NK cells in the human body.
Collapse
|
42
|
Innate immune cells in liver inflammation. Mediators Inflamm 2012; 2012:949157. [PMID: 22933833 PMCID: PMC3425885 DOI: 10.1155/2012/949157] [Citation(s) in RCA: 163] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2011] [Accepted: 06/17/2012] [Indexed: 12/20/2022] Open
Abstract
Innate immune system is the first line of defence against invading pathogens that is critical for the overall survival of the host. Human liver is characterised by a dual blood supply, with 80% of blood entering through the portal vein carrying nutrients and bacterial endotoxin from the gastrointestinal tract. The liver is thus constantly exposed to antigenic loads. Therefore, pathogenic microorganism must be efficiently eliminated whilst harmless antigens derived from the gastrointestinal tract need to be tolerized in the liver. In order to achieve this, the liver innate immune system is equipped with multiple cellular components; monocytes, macrophages, granulocytes, natural killer cells, and dendritic cells which coordinate to exert tolerogenic environment at the same time detect, respond, and eliminate invading pathogens, infected or transformed self to mount immunity. This paper will discuss the innate immune cells that take part in human liver inflammation, and their roles in both resolution of inflammation and tissue repair.
Collapse
|
43
|
Wakabayashi H, Ito T, Fushimi S, Nakashima Y, Itakura J, Qiuying L, Win MM, Cuiming S, Chen C, Sato M, Mino M, Ogino T, Makino H, Yoshimura A, Matsukawa A. Spred-2 deficiency exacerbates acetaminophen-induced hepatotoxicity in mice. Clin Immunol 2012; 144:272-82. [PMID: 22868447 DOI: 10.1016/j.clim.2012.07.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2012] [Revised: 06/30/2012] [Accepted: 07/03/2012] [Indexed: 02/06/2023]
Abstract
MAPKs are involved in acetaminophen (APAP)-hepatotoxicity, but the regulatory mechanism remains unknown. Here, we explored the role of Spred-2 that negatively regulates Ras/ERK pathway in APAP-hepatotoxicity. Spred-2 knockout (KO) mice demonstrated exacerbated liver injury, an event that was associated with increased numbers of CD4(+) T, CD8(+) T and NK cells in the liver compared to the control. Levels of CXCL9/CXCL10 that attract and activate these cells were increased in Spred-2 KO-liver. Kupffer cells isolated from Spred-2 KO mice after APAP challenge expressed higher levels of CXCL9/CXCL10 than those from the control. Upon stimulation with APAP or IFNγ, naïve Kupffer cells from Spred-2 KO mice expressed higher levels of CXCL9/CXCL10. NK cell-depletion attenuated APAP-hepatotoxicity with lowered hepatic IFNγ and decreased numbers of not only NK cells but also CD4(+) T and CD8(+) T cells in the liver. These results suggest that Spred-2 negatively regulates APAP-hepatotoxicity under the control of Kupffer cells and NK cells.
Collapse
Affiliation(s)
- Hiroshi Wakabayashi
- Department of Pathology and Experimental Medicine, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Abstract
Natural killer (NK) cells can be swiftly mobilized by danger signals and are among the earliest arrivals in target organs of disease. However, the role of NK cells in regulating inflammatory responses is far from completely understood in different organs. It is often complex and sometimes paradoxical. The phenotypes and functions of NK cells in the liver, mucosal tissues, uterus, pancreas, joints and brain are influenced by the unique cellular interactions and the local microenvironment within each organ. Hepatic NK cells exhibit an activated phenotype with high levels of cytotoxic effector molecules. These cells have been implicated in promoting liver injury and inhibiting liver fibrosis and regeneration. The liver is also enriched in NK cells with memory-like adaptive immune features. NK cells are detected in healthy lymphoid tissues of the lung, skin and gut, and are recruited to these tissues during infection or inflammation. In the gastrointestinal tract, classical NK cells and a variety of innate lymphoid cells, such as the family of lymphoid tissue-inducer (LTi) cells, are likely to have crucial roles in controlling inflammatory responses. NK cells represent the major lymphocyte subset in the pregnant uterus, with a unique phenotype resembling an early developmental state. Emerging evidence indicates that these cells play a crucial part in mediating the uterine vascular adaptations to pregnancy and promoting the maintenance of healthy pregnancy. In non-obese diabetic (NOD) mice, NK cells are recruited early to the pancreas, become locally activated and then adopt a hyporesponsive phenotype. Although NK cells have a pathogenic role in the natural progression of diabetes in NOD mice, they contribute to diabetes protection induced by complete Freund's adjuvant and to islet allograft tolerance induced by co-stimulatory blockade. NK cells in the inflamed joint uniquely express receptor activator of NF-κB ligand (RANKL) and macrophage colony-stimulating factor (M-CSF), which promote osteoclast differentiation. Although NK cells have a pathogenic role in collagen-induced arthritis in mice, they are also crucial for protection against antibody-induced arthritis mediated by CpG oligonucleotides. Studies in a mouse model of multiple sclerosis have shown that NK cells arrive in the central nervous system (CNS) before pathogenic T cells and have a protective role in the development of CNS inflammation, probably by killing CNS-resident microglia that prime effector T cells. During evolution, different organs might have evolved distinct ways to recruit and influence the effector functions of NK cells. Once we understand these mechanisms, the next challenge will be to exploit this information for harnessing NK cells to develop prophylactic and therapeutic measures against infectious agents, tumours and inflammatory diseases.
Each tissue in our body contains a unique microenvironment that can differentially shape immune reactivity. In this Review article, Shiet al. describe how organ-specific factors influence natural killer cell homing and phenotype, and discuss the local molecular and cellular interactions that determine the protective or pathogenic functions of natural killer cells in the different tissues. Natural killer (NK) cells can be swiftly mobilized by danger signals and are among the earliest arrivals at target organs of disease. However, the role of NK cells in mounting inflammatory responses is often complex and sometimes paradoxical. Here, we examine the divergent phenotypic and functional features of NK cells, as deduced largely from experimental mouse models of pathophysiological responses in the liver, mucosal tissues, uterus, pancreas, joints and brain. Moreover, we discuss how organ-specific factors, the local microenvironment and unique cellular interactions may influence the organ-specific properties of NK cells.
Collapse
|