601
|
Nakagaki BN, Freitas-Lopes MA, Carvalho É, Carvalho-Gontijo R, Castro-Oliveira HM, Rezende RM, Cara DC, Santos MM, Lopes RP, David BA, Menezes GB. Generation of a triple-fluorescent mouse strain allows a dynamic and spatial visualization of different liver phagocytes in vivo. AN ACAD BRAS CIENC 2017; 91:e20170317. [PMID: 29044327 DOI: 10.1590/0001-3765201720170317] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Accepted: 06/28/2017] [Indexed: 12/31/2022] Open
Abstract
Resident and circulating immune cells have been extensively studied due to their almost ubiquitous role in cell biology. Despite their classification under the "immune cell department", it is becoming increasingly clear that these cells are involved in many different non-immune related phenomena, including fetus development, vascular formation, memory, social behavior and many other phenotypes. There is a huge potential in combining high-throughput assays - including flow cytometry and gene analysis - with in vivo imaging. This can improve our knowledge in both basic and clinical cell biology, and accessing the expression of markers that are relevant in the context of both homeostasis and disease conditions might be instrumental. Here we describe how we generated a novel mouse strain that spontaneously express three different fluorescence markers under control of well-studied receptors (CX3CR1, CCR2 and CD11c) that are involved in a plethora of stages of cell ontogenesis, maturation, migration and behavior. Also, we assess the percentage of the expression and co-expression of each marker under homeostasis conditions, and how these cells behave when a local inflammation is induced in the liver applying a cutting-edge technology to image cells by confocal intravital microscopy.
Collapse
Affiliation(s)
- Brenda N Nakagaki
- Center for Gastrointestinal Biology, Departamento de Morfologia, sala N3-140, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Av. Presidente Antônio Carlos, 6627, Pampulha, 31270-901 Belo Horizonte, MG, Brazil
| | - Maria A Freitas-Lopes
- Center for Gastrointestinal Biology, Departamento de Morfologia, sala N3-140, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Av. Presidente Antônio Carlos, 6627, Pampulha, 31270-901 Belo Horizonte, MG, Brazil
| | - Érika Carvalho
- Center for Gastrointestinal Biology, Departamento de Morfologia, sala N3-140, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Av. Presidente Antônio Carlos, 6627, Pampulha, 31270-901 Belo Horizonte, MG, Brazil
| | - Raquel Carvalho-Gontijo
- Center for Gastrointestinal Biology, Departamento de Morfologia, sala N3-140, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Av. Presidente Antônio Carlos, 6627, Pampulha, 31270-901 Belo Horizonte, MG, Brazil
| | - Hortência M Castro-Oliveira
- Center for Gastrointestinal Biology, Departamento de Morfologia, sala N3-140, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Av. Presidente Antônio Carlos, 6627, Pampulha, 31270-901 Belo Horizonte, MG, Brazil
| | - Rafael M Rezende
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, 60 Fenwood Road, 02115, Boston, MA, United States of America
| | - Denise C Cara
- Center for Gastrointestinal Biology, Departamento de Morfologia, sala N3-140, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Av. Presidente Antônio Carlos, 6627, Pampulha, 31270-901 Belo Horizonte, MG, Brazil
| | - Mônica M Santos
- Departamento de Biologia Animal, Universidade Federal de Viçosa, Av. Peter Henry Rolfs, s/n, Campus Universitário, 36570-900 Viçosa, MG, Brazil
| | - Rodrigo Pestana Lopes
- BD Biosciences, Rua Alexandre Dumas, 1976, Chácara Santo Antônio, 04717-040 São Paulo, SP, Brazil
| | - Bruna A David
- Center for Gastrointestinal Biology, Departamento de Morfologia, sala N3-140, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Av. Presidente Antônio Carlos, 6627, Pampulha, 31270-901 Belo Horizonte, MG, Brazil.,Departamento de Bioquímica e Imunologia, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo/USP, Av. Bandeirantes, 3900, Monte Alegre, 14049-900 Ribeirão Preto, SP, Brazil
| | - Gustavo B Menezes
- Center for Gastrointestinal Biology, Departamento de Morfologia, sala N3-140, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Av. Presidente Antônio Carlos, 6627, Pampulha, 31270-901 Belo Horizonte, MG, Brazil
| |
Collapse
|
602
|
Kim HY, Park JW. Current immunotherapeutic strategies in hepatocellular carcinoma: recent advances and future directions. Therap Adv Gastroenterol 2017; 10:805-814. [PMID: 29051790 PMCID: PMC5638179 DOI: 10.1177/1756283x17722061] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2017] [Accepted: 06/20/2017] [Indexed: 02/04/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is a common and serious health problem with high mortality. Treatment for HCC remains largely unsatisfactory owing to its high recurrence rates and frequent accompanying cirrhosis. In addition, the unique immune environment of the liver promotes tolerance, which, in conjunction with immune evasion by the disease, makes HCC a less promising target for conventional immunotherapy. However, recent advances in the immunotherapy have led to novel approaches to overcome these obstacles by manipulating and enhancing tumor-specific immune responses against HCC by using various modalities, such as cancer vaccines and immune checkpoint blockade. These treatments have shown both safety and promising outcomes in patients with HCC of various etiologies and tumor stages. Furthermore, combined strategies have been assessed to achieve optimal outcomes, by using immunotherapies with or without conventional treatments. This review briefly covers the background, recent advances, current issues, and future perspectives on immunotherapy in the field of HCC treatment.
Collapse
Affiliation(s)
- Hwi Young Kim
- Department of Internal Medicine, College of Medicine, Ewha Womans University, Seoul, Republic of Korea
| | | |
Collapse
|
603
|
Li D, He W, Liu X, Zheng S, Qi Y, Li H, Mao F, Liu J, Sun Y, Pan L, Du K, Ye K, Li W, Sui J. A potent human neutralizing antibody Fc-dependently reduces established HBV infections. eLife 2017; 6. [PMID: 28949917 PMCID: PMC5614562 DOI: 10.7554/elife.26738] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Accepted: 08/04/2017] [Indexed: 12/11/2022] Open
Abstract
Hepatitis B virus (HBV) infection is a major global health problem. Currently-available therapies are ineffective in curing chronic HBV infection. HBV and its satellite hepatitis D virus (HDV) infect hepatocytes via binding of the preS1 domain of its large envelope protein to sodium taurocholate cotransporting polypeptide (NTCP). Here, we developed novel human monoclonal antibodies that block the engagement of preS1 with NTCP and neutralize HBV and HDV with high potency. One antibody, 2H5-A14, functions at picomolar level and exhibited neutralization-activity-mediated prophylactic effects. It also acts therapeutically by eliciting antibody-Fc-dependent immunological effector functions that impose durable suppression of viral infection in HBV-infected mice, resulting in reductions in the levels of the small envelope antigen and viral DNA, with no emergence of escape mutants. Our results illustrate a novel antibody-Fc-dependent approach for HBV treatment and suggest 2H5-A14 as a novel clinical candidate for HBV prevention and treatment of chronic HBV infection.
Collapse
Affiliation(s)
- Dan Li
- Peking University-Tsinghua University-National Institute of Biological Sciences Joint Graduate Program, School of Life Sciences, Tsinghua University, Beijing, China.,National Institute of Biological Sciences, Beijing, China
| | - Wenhui He
- National Institute of Biological Sciences, Beijing, China
| | - Ximing Liu
- National Institute of Biological Sciences, Beijing, China.,PTN Joint Graduate Program, College of Life Sciences, Peking University, Beijing, China
| | - Sanduo Zheng
- National Institute of Biological Sciences, Beijing, China
| | - Yonghe Qi
- National Institute of Biological Sciences, Beijing, China
| | - Huiyu Li
- National Institute of Biological Sciences, Beijing, China
| | - Fengfeng Mao
- National Institute of Biological Sciences, Beijing, China.,Graduate Program in College of Life Sciences, Beijing Normal University, Beijing, China
| | - Juan Liu
- National Institute of Biological Sciences, Beijing, China
| | - Yinyan Sun
- National Institute of Biological Sciences, Beijing, China
| | - Lijing Pan
- Peking University-Tsinghua University-National Institute of Biological Sciences Joint Graduate Program, School of Life Sciences, Tsinghua University, Beijing, China.,National Institute of Biological Sciences, Beijing, China
| | - Kaixin Du
- National Institute of Biological Sciences, Beijing, China.,Graduate Program in College of Life Sciences, Beijing Normal University, Beijing, China
| | - Keqiong Ye
- National Institute of Biological Sciences, Beijing, China
| | - Wenhui Li
- National Institute of Biological Sciences, Beijing, China
| | - Jianhua Sui
- National Institute of Biological Sciences, Beijing, China
| |
Collapse
|
604
|
Hickey RM, Kulik LM, Nimeiri H, Kalyan A, Kircher S, Desai K, Riaz A, Lewandowski RJ, Salem R. Immuno-oncology and Its Opportunities for Interventional Radiologists: Immune Checkpoint Inhibition and Potential Synergies with Interventional Oncology Procedures. J Vasc Interv Radiol 2017; 28:1487-1494. [PMID: 28912090 DOI: 10.1016/j.jvir.2017.07.018] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Revised: 07/14/2017] [Accepted: 07/15/2017] [Indexed: 12/18/2022] Open
Abstract
Immunotherapy, specifically the use of immune checkpoint inhibitors, offers a new approach to fighting cancer. Although the results of treatment with immune checkpoint inhibition alone have been remarkable for certain cancers, these results are not universal. Preclinical and early clinical studies indicate the potential for synergistic effects when immune checkpoint inhibition is combined with immunogenic local therapies such as ablation and embolization. This review offers an overview of immunology as it relates to immune checkpoint inhibition and the possibilities for synergy when combined with interventional radiology treatments.
Collapse
Affiliation(s)
- Ryan M Hickey
- Department of Radiology, Section of Interventional Radiology, New York University, 560 First Ave., New York, NY 10016.
| | - Laura M Kulik
- Department of Medicine, Division of Hepatology, Northwestern University, Chicago, Illinois
| | - Halla Nimeiri
- Department of Medicine, Division of Hematology and Oncology, Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, Illinois
| | - Aparna Kalyan
- Department of Medicine, Division of Hematology and Oncology, Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, Illinois
| | - Sheetal Kircher
- Department of Medicine, Division of Hematology and Oncology, Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, Illinois
| | - Kush Desai
- Department of Radiology, Section of Interventional Radiology, Northwestern University, Chicago, Illinois
| | - Ahsun Riaz
- Department of Radiology, Section of Interventional Radiology, Northwestern University, Chicago, Illinois
| | - Robert J Lewandowski
- Department of Radiology, Section of Interventional Radiology, Northwestern University, Chicago, Illinois
| | - Riad Salem
- Department of Medicine, Division of Hematology and Oncology, Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, Illinois; Department of Radiology, Section of Interventional Radiology, Northwestern University, Chicago, Illinois
| |
Collapse
|
605
|
Gasco S, Zaragoza P, García-Redondo A, Calvo AC, Osta R. Inflammatory and non-inflammatory monocytes as novel prognostic biomarkers of survival in SOD1G93A mouse model of Amyotrophic Lateral Sclerosis. PLoS One 2017; 12:e0184626. [PMID: 28886177 PMCID: PMC5591000 DOI: 10.1371/journal.pone.0184626] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Accepted: 08/28/2017] [Indexed: 12/13/2022] Open
Abstract
Amyotrophic Lateral Sclerosis (ALS) has lately become a suitable scenario to study the interplay between the hematopoietic system and disease progression. Recent studies in C9orf72 null mice have demonstrated that C9orf72 is necessary for the normal function of myeloid cells. In this study, we aimed to analyze in depth the connection between the hematopoietic system and secondary lymphoid (spleen) and non-lymphoid (liver and skeletal muscle) organs and tissues along the disease progression in the transgenic SOD1G93A mice. Our findings suggested that the inflammatory response due to the neurodegeneration in this animal model affected all three organs and tissues, especially the liver and the skeletal muscle. However, the liver was able to compensate this inflammatory response by means of the action of non-inflammatory monocytes, while in the skeletal muscle inflammatory monocytes prompted a further inflammation process until the terminal state of the animals. Interestingly, in blood, a positive correlation was found between non-inflammatory monocytes and survival of the transgenic SOD1G93A mice, while the contrary (a negative correlation) was found in the case of inflammatory monocytes, supporting their potential role as biomarkers of disease progression and survival in this animal model. These findings could prompt future translational studies in ALS patients, promoting the identification of new reliable biomarkers of disease progression.
Collapse
Affiliation(s)
- Samanta Gasco
- LAGENBIO, Veterinary Faculty of Zaragoza, Instituto Agroalimentario de Aragón (I2A), CITA, Health Research Institute of Aragon (IIS). University of Zaragoza, Zaragoza, Spain
- * E-mail:
| | - Pilar Zaragoza
- LAGENBIO, Veterinary Faculty of Zaragoza, Instituto Agroalimentario de Aragón (I2A), CITA, Health Research Institute of Aragon (IIS). University of Zaragoza, Zaragoza, Spain
| | - Alberto García-Redondo
- Biochemistry Department, CIBERER U-723. Health Research Institute, October 12th Hospital, Madrid, Spain
| | - Ana C. Calvo
- LAGENBIO, Veterinary Faculty of Zaragoza, Instituto Agroalimentario de Aragón (I2A), CITA, Health Research Institute of Aragon (IIS). University of Zaragoza, Zaragoza, Spain
| | - Rosario Osta
- LAGENBIO, Veterinary Faculty of Zaragoza, Instituto Agroalimentario de Aragón (I2A), CITA, Health Research Institute of Aragon (IIS). University of Zaragoza, Zaragoza, Spain
| |
Collapse
|
606
|
Macpherson AJ, Heikenwalder M, Ganal-Vonarburg SC. The Liver at the Nexus of Host-Microbial Interactions. Cell Host Microbe 2017; 20:561-571. [PMID: 27832587 DOI: 10.1016/j.chom.2016.10.016] [Citation(s) in RCA: 81] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The liver receives blood from the intestine, from the spleen, and directly from the heart and holds a vital position in vertebrate physiology. It plays a role in intermediary metabolism, bile secretion, maintaining blood sterility, serum homeostasis, xenobiotic detoxification, and immunological activity. This article provides our perspective on the liver as a nexus in establishing and maintaining host microbial mutualism. We discuss the role of the liver not only in sanitizing the blood stream from penetrant live microbes, but also in metabolizing xenobiotics that are synthesized or modified by intestinal microbes, and how microbiota modify the signaling potential of bile acids. The combination of bile acids as hormones and the metabolic control from pervasive effects of other absorbed microbial molecules powerfully shape hepatic metabolism. In addition, intestinal microbial metabolites can be sensed by liver-resident immune cells, which may disturb liver homeostasis, leading to fibrosis and liver cancer.
Collapse
Affiliation(s)
- Andrew J Macpherson
- Maurice Müller Laboratories (DKF), Universitätsklinik für Viszerale Chirurgie und Medizin Inselspital, Murtenstrasse 35, University of Bern, 3010 Bern, Switzerland.
| | - Mathias Heikenwalder
- Division of Chronic Inflammation and Cancer, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Stephanie C Ganal-Vonarburg
- Maurice Müller Laboratories (DKF), Universitätsklinik für Viszerale Chirurgie und Medizin Inselspital, Murtenstrasse 35, University of Bern, 3010 Bern, Switzerland.
| |
Collapse
|
607
|
Yu J, Chen Y, Wu Y, Ye L, Lian Z, Wei H, Sun R, Tian Z. The differential organogenesis and functionality of two liver-draining lymph nodes in mice. J Autoimmun 2017; 84:109-121. [PMID: 28886898 DOI: 10.1016/j.jaut.2017.08.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Revised: 08/17/2017] [Accepted: 08/17/2017] [Indexed: 12/13/2022]
Abstract
The liver is an immunological organ. However, fundamental knowledge concerning liver-draining lymph nodes (LNs), which have been newly identified in mice as the portal and celiac LNs, is still lacking. Here, we revealed that the portal LN and celiac LN drain liver lymph through different lymphatic vessels. Although both the portal LN and celiac LN possess typical structures, they have different cell compositions. Interestingly, these two LNs form at different times during fetal development. Moreover, the organogenesis of the celiac LN, but not the portal LN, is controlled by the transcription factor NFIL3. Furthermore, the portal LN and celiac LN also perform different functions. The celiac LN is the predominant site of liver antiviral immune responses, whereas the portal LN functions in the in situ induction of dietary antigen-specific regulatory T cells. In conclusion, the portal LN and celiac LN are two independent liver-draining LNs with different organogenesis histories and separate functions in maintaining immune homeostasis in the liver.
Collapse
Affiliation(s)
- Jiali Yu
- Hefei National Laboratory for Physical Sciences at Microscale, The Key Laboratory of Innate Immunity and Chronic Disease (Chinese Academy of Science), Institute of Immunology, School of Life Sciences and Medical Center, University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Yongyan Chen
- Hefei National Laboratory for Physical Sciences at Microscale, The Key Laboratory of Innate Immunity and Chronic Disease (Chinese Academy of Science), Institute of Immunology, School of Life Sciences and Medical Center, University of Science and Technology of China, Hefei, Anhui 230027, China; Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310003, China
| | - Yuzhang Wu
- Institute of Immunology, Third Military Medical University, Chongqing, 400038, China.
| | - Lilin Ye
- Institute of Immunology, Third Military Medical University, Chongqing, 400038, China
| | - Zhexiong Lian
- Hefei National Laboratory for Physical Sciences at Microscale, The Key Laboratory of Innate Immunity and Chronic Disease (Chinese Academy of Science), Institute of Immunology, School of Life Sciences and Medical Center, University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Haiming Wei
- Hefei National Laboratory for Physical Sciences at Microscale, The Key Laboratory of Innate Immunity and Chronic Disease (Chinese Academy of Science), Institute of Immunology, School of Life Sciences and Medical Center, University of Science and Technology of China, Hefei, Anhui 230027, China; Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310003, China
| | - Rui Sun
- Hefei National Laboratory for Physical Sciences at Microscale, The Key Laboratory of Innate Immunity and Chronic Disease (Chinese Academy of Science), Institute of Immunology, School of Life Sciences and Medical Center, University of Science and Technology of China, Hefei, Anhui 230027, China; Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310003, China
| | - Zhigang Tian
- Hefei National Laboratory for Physical Sciences at Microscale, The Key Laboratory of Innate Immunity and Chronic Disease (Chinese Academy of Science), Institute of Immunology, School of Life Sciences and Medical Center, University of Science and Technology of China, Hefei, Anhui 230027, China; Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310003, China.
| |
Collapse
|
608
|
Gilboa D, Haim-Ohana Y, Deshet-Unger N, Ben-Califa N, Hiram-Bab S, Reuveni D, Zigmond E, Gassmann M, Gabet Y, Varol C, Neumann D. Erythropoietin enhances Kupffer cell number and activity in the challenged liver. Sci Rep 2017; 7:10379. [PMID: 28871174 PMCID: PMC5583293 DOI: 10.1038/s41598-017-11082-7] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Accepted: 08/15/2017] [Indexed: 02/07/2023] Open
Abstract
Erythropoietin (EPO) is the main hormone driving mammalian erythropoiesis, with activity mediated via the surface receptor, EPO-R, on erythroid progenitor cells. Recombinant human EPO is currently used clinically for the treatment of anemia in patients with end-stage renal disease, and in certain cancer patients suffering from anemia induced either by the tumor itself or by chemotherapy. EPO-R expression is also detected in non-erythroid cells, including macrophages present in the peritoneum, spleen, and bone marrow (BM). Here we demonstrate that Kupffer cells (KCs) - the liver-resident macrophages - are EPO targets. We show that, in vitro, EPO initiated intracellular signalling and enhanced phagocytosis in a rat KC line (RKC-2) and in sorted KCs. Moreover, continuous EPO administration in mice, resulted in an increased number of KCs, up-regulation of liver EPO-R expression and elevated production of the monocyte chemoattractant CCL2, with corresponding egress of Ly6Chi monocytes from the BM. In a model of acute acetaminophen-induced liver injury, EPO administration increased the recruitment of Ly6Chi monocytes and neutrophils to the liver. Taken together, our results reveal a new role for EPO in stimulating KC proliferation and phagocytosis, and in recruiting Ly6Chi monocytes in response to liver injury.
Collapse
Affiliation(s)
- Dafna Gilboa
- Department of Cell and Developmental Biology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Yasmin Haim-Ohana
- Department of Cell and Developmental Biology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Naamit Deshet-Unger
- Department of Cell and Developmental Biology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Nathalie Ben-Califa
- Department of Cell and Developmental Biology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Sahar Hiram-Bab
- Department of Anatomy and Anthropology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Debby Reuveni
- The Research Center for Digestive Tract and Liver Diseases, Sourasky Medical Center and Department of Clinical Microbiology and Immunology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Ehud Zigmond
- The Research Center for Digestive Tract and Liver Diseases, Sourasky Medical Center and Department of Clinical Microbiology and Immunology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Max Gassmann
- Institute for Veterinary Physiology, Vetsuisse Faculty and Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, Zurich, Switzerland
| | - Yankel Gabet
- Department of Anatomy and Anthropology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Chen Varol
- The Research Center for Digestive Tract and Liver Diseases, Sourasky Medical Center and Department of Clinical Microbiology and Immunology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Drorit Neumann
- Department of Cell and Developmental Biology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.
| |
Collapse
|
609
|
Hepatic Immune Microenvironment in Alcoholic and Nonalcoholic Liver Disease. BIOMED RESEARCH INTERNATIONAL 2017; 2017:6862439. [PMID: 28852648 PMCID: PMC5567444 DOI: 10.1155/2017/6862439] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Accepted: 07/06/2017] [Indexed: 01/18/2023]
Abstract
Many types of innate (natural killer cells, natural killer T cells, and Kupffer cells/macrophages) and adaptive (T cells and B cells) immune cells are enriched within the liver and function in liver physiology and pathology. Liver pathology is generally induced by two types of immunologic insults: failure to eliminate antigens derived from the gastrointestinal tract which are important for host defense and an impaired tissue protective tolerance mechanism that helps reduce the negative outcomes of immunopathology. Accumulating evidence from the last several decades suggests that hepatic immune cells play an important role in the pathogenesis of alcoholic and nonalcoholic liver injury and inflammation in humans and mice. Here, we focus on the roles of innate and adaptive immune cells in the development and maintenance of alcoholic liver disease and nonalcoholic fatty liver disease/nonalcoholic steatohepatitis. Additionally, the pathogenesis of liver disease and new therapeutic targets for preventing and treating alcoholic liver disease and nonalcoholic fatty liver disease/nonalcoholic steatohepatitis are discussed.
Collapse
|
610
|
Martrus G, Kautz T, Lunemann S, Richert L, Glau L, Salzberger W, Goebels H, Langeneckert A, Hess L, Poch T, Schramm C, Oldhafer KJ, Koch M, Tolosa E, Nashan B, Altfeld M. Proliferative capacity exhibited by human liver-resident CD49a+CD25+ NK cells. PLoS One 2017; 12:e0182532. [PMID: 28792982 PMCID: PMC5549915 DOI: 10.1371/journal.pone.0182532] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2017] [Accepted: 07/19/2017] [Indexed: 12/30/2022] Open
Abstract
The recruitment and retention of Natural Killer (NK) cells in the liver are thought to play an important role during hepatotropic infections and liver cirrhosis. The aims of this study were to determine differences between liver-derived and peripheral blood-derived NK cells in the context of liver inflammation and cirrhosis. We conducted a prospective dual-center cross-sectional study in patients undergoing liver transplantation or tumor-free liver resections, in which both liver tissue and peripheral blood samples were obtained from each consenting study participants. Intrahepatic lymphocytes and PBMCs were stained, fixed and analyzed by flow cytometry. Our results showed that, within cirrhotic liver samples, intrahepatic NK cells were particularly enriched for CD49a+ NK cells when compared to tumor-free liver resection samples. CD49a+ liver-derived NK cells included populations of cells expressing CD25, CD34 and CXCR3. Moreover, CD49a+CD25+ liver-derived NK cells exhibited high proliferative capacity in vitro in response to low doses of IL-2. Our study identified a specific subset of CD49a+CD25+ NK cells in cirrhotic livers bearing functional features of proliferation.
Collapse
Affiliation(s)
- Glòria Martrus
- Heinrich Pette Institute, Leibniz Institute for Experimental Virology, Hamburg, Germany
| | - Tobias Kautz
- Heinrich Pette Institute, Leibniz Institute for Experimental Virology, Hamburg, Germany
| | - Sebastian Lunemann
- Heinrich Pette Institute, Leibniz Institute for Experimental Virology, Hamburg, Germany
| | - Laura Richert
- Heinrich Pette Institute, Leibniz Institute for Experimental Virology, Hamburg, Germany
- Univ. Bordeaux, Inserm, Bordeaux Population Health Research Center, team SISTM, UMR1219 and Inria, Bordeaux, France
| | - Laura Glau
- Department of Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Wilhelm Salzberger
- Heinrich Pette Institute, Leibniz Institute for Experimental Virology, Hamburg, Germany
| | - Hanna Goebels
- Heinrich Pette Institute, Leibniz Institute for Experimental Virology, Hamburg, Germany
| | - Annika Langeneckert
- Heinrich Pette Institute, Leibniz Institute for Experimental Virology, Hamburg, Germany
| | - Leonard Hess
- Heinrich Pette Institute, Leibniz Institute for Experimental Virology, Hamburg, Germany
| | - Tobias Poch
- First Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Christoph Schramm
- First Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Karl J. Oldhafer
- Department of General & Abdominal Surgery, Asklepios Hospital Barmbek, Semmelweis University of Medicine, Asklepios Campus, Hamburg, Germany
| | - Martina Koch
- Department of Hepatobiliary and Transplant Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Eva Tolosa
- Department of Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Björn Nashan
- Department of Hepatobiliary and Transplant Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Marcus Altfeld
- Heinrich Pette Institute, Leibniz Institute for Experimental Virology, Hamburg, Germany
- * E-mail:
| |
Collapse
|
611
|
Verschoor A, Karsten CM, Broadley SP, Laumonnier Y, Köhl J. Old dogs-new tricks: immunoregulatory properties of C3 and C5 cleavage fragments. Immunol Rev 2017; 274:112-126. [PMID: 27782330 DOI: 10.1111/imr.12473] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The activation of the complement system by canonical and non-canonical mechanisms results in the generation of multiple C3 and C5 cleavage fragments including anaphylatoxins C3a and C5a as well as opsonizing C3b/iC3b. It is now well appreciated that anaphylatoxins not only act as pro-inflammatory mediators but as immunoregulatory molecules that control the activation status of cells and tissue at several levels. Likewise, C3b/iC3b is more than the opsonizing fragment that facilitates engulfment and destruction of targets by phagocytes. In the circulation, it also facilitates the transport and delivery of bacteria and immune complexes to phagocytes, through a process known as immune adherence, with consequences for adaptive immunity. Here, we will discuss non-classical immunoregulatory properties of C3 and C5 cleavage fragments. We highlight the influence of anaphylatoxins on Th2 and Th17 cell development during allergic asthma with a particular emphasis on their role in the modulation of CD11b+ conventional dendritic cells and monocyte-derived dendritic cells. Furthermore, we discuss the control of anaphylatoxin-mediated activation of dendritic cells and allergic effector cells by adaptive immune mechanisms that involve allergen-specific IgG1 antibodies and plasma or regulatory T cell-derived IL-10 production. Finally, we take a fresh look at immune adherence with a particular focus on the development of antibacterial cytotoxic T-cell responses.
Collapse
Affiliation(s)
- Admar Verschoor
- Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany.
| | - Christian M Karsten
- Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany
| | - Steven P Broadley
- Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany
| | - Yves Laumonnier
- Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany
| | - Jörg Köhl
- Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany. .,Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.
| |
Collapse
|
612
|
Tarasenko TN, McGuire PJ. The liver is a metabolic and immunologic organ: A reconsideration of metabolic decompensation due to infection in inborn errors of metabolism (IEM). Mol Genet Metab 2017; 121:283-288. [PMID: 28666653 PMCID: PMC5553615 DOI: 10.1016/j.ymgme.2017.06.010] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2017] [Revised: 06/22/2017] [Accepted: 06/23/2017] [Indexed: 12/30/2022]
Abstract
Metabolic decompensation in inborn errors of metabolism (IEM) is characterized by a rapid deterioration in metabolic status leading to life-threatening biochemical perturbations (e.g. hypoglycemia, hyperammonemia, acidosis, organ failure). Infection is the major cause of metabolic decompensation in patients with IEM. We hypothesized that activation of the immune system during infection leads to further perturbations in end-organ metabolism resulting in increased morbidity. To address this, we established model systems of metabolic decompensation due to infection. Using these systems, we have described the pathologic mechanisms of metabolic decompensation as well as changes in hepatic metabolic reserve associated with infection. First and foremost, our studies have demonstrated that the liver experiences a significant local innate immune response during influenza infection that modulates hepatic metabolism. Based on these findings, we are the first to suggest that the role of the liver as a metabolic and immunologic organ is central in the pathophysiology of metabolic decompensation due to infection in IEM. The dual function of the liver as a major metabolic regulator and a lymphoid organ responsible for immunosurveillance places this organ at risk for hepatotoxicity. Mobilization of hepatic reserve and the regenerative capacity of a healthy liver compensates for this calculated risk. However, activation of the hepatic innate immune system may be deleterious in IEM. Based on this assertion, strategies aimed at modulating the innate immune response may be a viable target for intervention in the treatment of hepatic metabolic decompensation.
Collapse
Affiliation(s)
- Tatyana N Tarasenko
- Metabolism, Infection and Immunity Section, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, United States
| | - Peter J McGuire
- Metabolism, Infection and Immunity Section, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, United States.
| |
Collapse
|
613
|
Chen IL, Huang HC, Wu CT, Ou-Yang MC, Chung MY, Chen CC, Suen JL, Hung CH. Analysis of early-onset bloodstream infection due to Escherichia coli infection in premature babies. Medicine (Baltimore) 2017; 96:e7748. [PMID: 28796061 PMCID: PMC5556227 DOI: 10.1097/md.0000000000007748] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
In early-onset bacteremia among preterm neonates, Escherichia coli (E. coli) is the main pathogen and can cause a high mortality rate. Thus, the predictive factors of mortality and extended-spectrum β-lactamase (ESBL)-producing E. coli in preterm babies with E. coli early-onset bacteremia were reported.We retrospectively reviewed preterm neonates who had E. coli bacteremia occurring within 3 days after birth between 2004 and 2015. Maternal and perinatal information were collected from their medical records and analyzed by comparing the survival and nonsurvival groups, and also the ESBL-producing and non-ESBL-producing E. coli bacteremia groups. Mann-Whitney U test, Fisher exact test, and multivariate Cox proportional-hazard model were used for statistical analysis.A total of 27 preterm babies had E. coli bacteremia. The overall mortality rate was 55.56% (15 deaths). Five babies had ESBL-producing E. coli. The low systolic blood pressure of <48 mm Hg and low absolute neutrophil count of <2318 cells/mm were the most significant factors in predicting mortality. Moreover, the level of serum alanine aminotransferase was significantly lower in the ESBL-producing E. coli group than that in the non-ESBL-producing E. coli group.Therefore, the lower systolic blood pressure and absolute neutrophil count were the risk factors of mortality in preterm babies with early-onset E. coli bacteremia, and alanine aminotransferase could be a significant factor in predicting ESBL-producing E. coli.
Collapse
Affiliation(s)
- I-Lun Chen
- Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Hsin-Chun Huang
- Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan
| | - Chih-Te Wu
- Department of Pediatrics, Chang Gung Memorial Hospital, Linkou, Taiwan
| | - Mei-Chen Ou-Yang
- Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan
| | - Mei-Yung Chung
- Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan
| | - Chih-Cheng Chen
- Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan
| | - Jau-Ling Suen
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chih-Hsing Hung
- Department of Pediatrics, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Pediatrics, Kaohsiung Municipal Hsiao-Kang Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| |
Collapse
|
614
|
Cai C, Zhu X, Li P, Li J, Gong J, Shen W, He K. NLRP3 Deletion Inhibits the Non-alcoholic Steatohepatitis Development and Inflammation in Kupffer Cells Induced by Palmitic Acid. Inflammation 2017; 40:1875-1883. [DOI: 10.1007/s10753-017-0628-z] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|
615
|
Xie DL, Zheng MM, Zheng Y, Gao H, Zhang J, Zhang T, Guo JC, Yang XF, Zhong XP, Lou YL. Vibrio vulnificus induces mTOR activation and inflammatory responses in macrophages. PLoS One 2017; 12:e0181454. [PMID: 28719654 PMCID: PMC5515453 DOI: 10.1371/journal.pone.0181454] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Accepted: 07/01/2017] [Indexed: 12/18/2022] Open
Abstract
Vibrio vulnificus (V. vulnificus), a Gram-negative marine bacterium, can cause life-threatening primary septicemia, especially in patients with liver diseases. How V. vulnificus affects the liver and how it acts on macrophages are not well understood. In this report, we demonstrated that V. vulnificus infection causes a strong inflammatory response, marked expansion of liver-resident macrophages, and liver damage in mice. We demonstrated further that V. vulnificus activates mTOR in macrophages and inhibition of mTOR differentially regulates V. vulnificus induced inflammatory responses, suggesting the possibility of targeting mTOR as a strategy to modulate V. vulnificus induced inflammatory responses.
Collapse
Affiliation(s)
- Dan-Li Xie
- Department of Microbiology and Immunology, School of Laboratory Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, China
- China Ministry of Education Key Lab of Laboratory Medicine, Wenzhou, Zhejiang, China
| | - Meng-Meng Zheng
- Department of Microbiology and Immunology, School of Laboratory Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yi Zheng
- Department of Microbiology and Immunology, School of Laboratory Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, China
- China Ministry of Education Key Lab of Laboratory Medicine, Wenzhou, Zhejiang, China
| | - Hui Gao
- Department of Microbiology and Immunology, School of Laboratory Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jie Zhang
- Department of Clinical Laboratory Medicine, Sichuan Provincial People’s Hospital, Chengdu, Sichuan, China
| | - Ting Zhang
- Department of Laboratory Medicine, Jinshan Hospital of Fudan University, Jinshan, Shanghai, China
| | - Jian-Chun Guo
- Department of Microbiology and Immunology, School of Laboratory Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - X. Frank Yang
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
| | - Xiao-Ping Zhong
- Department of Microbiology and Immunology, School of Laboratory Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, China
- China Ministry of Education Key Lab of Laboratory Medicine, Wenzhou, Zhejiang, China
- Department of Pediatrics, Division of Allergy and Immunology, Duke University Medical Center, Durham, NC, United States of America
- * E-mail: (YLL); (XPZ)
| | - Yong-Liang Lou
- Department of Microbiology and Immunology, School of Laboratory Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, China
- China Ministry of Education Key Lab of Laboratory Medicine, Wenzhou, Zhejiang, China
- * E-mail: (YLL); (XPZ)
| |
Collapse
|
616
|
Neuzillet C, de Mestier L, Rousseau B, Mir O, Hebbar M, Kocher HM, Ruszniewski P, Tournigand C. Unravelling the pharmacologic opportunities and future directions for targeted therapies in gastro-intestinal cancers part 2: Neuroendocrine tumours, hepatocellular carcinoma, and gastro-intestinal stromal tumours. Pharmacol Ther 2017; 181:49-75. [PMID: 28723416 DOI: 10.1016/j.pharmthera.2017.07.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Until the 1990s, cytotoxic chemotherapy has been the cornerstone of medical therapy for gastrointestinal (GI) cancers. Better understanding of the cancer cell molecular biology has led to the therapeutic revolution of targeted therapies, i.e. monoclonal antibodies or small molecule inhibitors directed against proteins that are specifically overexpressed or mutated in cancer cells. These agents, being more specific to cancer cells, were expected to be less toxic than conventional cytotoxic agents. However, their effects have sometimes been disappointing, due to intrinsic or acquired resistance mechanisms, or to an activity restricted to some tumour settings, illustrating the importance of patient selection and early identification of predictive biomarkers of response to these therapies. Targeted agents have provided clinical benefit in many GI cancer types. Particularly, some GI tumours are considered chemoresistant and targeted therapies have offered a new therapeutic base for their management. Hence, somatostatin receptor-directed strategies, sorafenib, and imatinib have revolutioned the management of neuroendocrine tumours (NET), hepatocellular carcinoma (HCC), and gastrointestinal stromal tumours (GIST), respectively, and are now used as first-line treatment in many patients affected by these tumours. However, these agents face problems of resistances and identification of predictive biomarkers from imaging and/or biology. We propose a comprehensive two-part review providing a panoramic approach of the successes and failures of targeted agents in GI cancers to unravel the pharmacologic opportunities and future directions for these agents in GI oncology. In this second part, we will focus on NET, HCC, and GIST, whose treatment relies primarily on targeted therapies.
Collapse
Affiliation(s)
- Cindy Neuzillet
- INSERM UMR1149, Beaujon University Hospital (Assistance Publique-Hôpitaux de Paris, AP-HP), Paris 7 Diderot University, 100 Boulevard du Général Leclerc, 92110 Clichy, France; Department of Medical Oncology, Henri Mondor University Hospital (AP-HP), Paris Est Créteil University (UPEC), 51 Avenue du Maréchal de Lattre de Tassigny, 94010 Créteil, France; Tumour Biology Laboratory, Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, United Kingdom; Barts and The London HPB Centre, The Royal London Hospital, Whitechapel, London E1 1BB, United Kingdom.
| | - Louis de Mestier
- INSERM UMR1149, Beaujon University Hospital (Assistance Publique-Hôpitaux de Paris, AP-HP), Paris 7 Diderot University, 100 Boulevard du Général Leclerc, 92110 Clichy, France; Department of Gastroenterology and Pancreatology, Beaujon University Hospital (AP-HP), Paris 7 Diderot University, 100 Boulevard du Général Leclerc, 92110 Clichy, France
| | - Benoît Rousseau
- Department of Medical Oncology, Henri Mondor University Hospital (AP-HP), Paris Est Créteil University (UPEC), 51 Avenue du Maréchal de Lattre de Tassigny, 94010 Créteil, France; Institut Mondor de Recherche Biomédicale, INSERM UMR955 Team 18, Paris Est Créteil University (UPEC), 51 Avenue du Maréchal de Lattre de Tassigny, 94010 Créteil, France
| | - Olivier Mir
- Department of Cancer Medicine - Sarcoma Group, Department of Early Drug Development (DITEP) - Phase 1 Unit, Gustave Roussy Cancer Campus, University of Paris Sud, 114, Rue Edouard Vaillant, 94800 Villejuif, France
| | - Mohamed Hebbar
- Department of Medical Oncology, Lille University Hospital, 1, Rue Polonovski, 59037 Lille, France
| | - Hemant M Kocher
- Tumour Biology Laboratory, Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, United Kingdom; Barts and The London HPB Centre, The Royal London Hospital, Whitechapel, London E1 1BB, United Kingdom
| | - Philippe Ruszniewski
- INSERM UMR1149, Beaujon University Hospital (Assistance Publique-Hôpitaux de Paris, AP-HP), Paris 7 Diderot University, 100 Boulevard du Général Leclerc, 92110 Clichy, France
| | - Christophe Tournigand
- Department of Medical Oncology, Henri Mondor University Hospital (AP-HP), Paris Est Créteil University (UPEC), 51 Avenue du Maréchal de Lattre de Tassigny, 94010 Créteil, France
| |
Collapse
|
617
|
TLR9-Mediated Conditioning of Liver Environment Is Essential for Successful Intrahepatic Immunotherapy and Effective Memory Recall. Mol Ther 2017; 25:2289-2298. [PMID: 28716576 DOI: 10.1016/j.ymthe.2017.06.018] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Revised: 06/08/2017] [Accepted: 06/18/2017] [Indexed: 01/11/2023] Open
Abstract
Immune defense against hepatotropic viruses such as hepatitis B (HBV) and hepatitis C (HCV) poses a major challenge for therapeutic approaches. Intrahepatic cytotoxic CD8 T cells that are crucial for an immune response against these viruses often become exhausted resulting in chronic infection. We elucidated the T cell response upon therapeutic vaccination in inducible transgenic mouse models in which variable percentages of antigen-expressing hepatocytes can be adjusted, providing mosaic antigen distribution and reflecting the varying viral antigen loads observed in patients. Vaccination-induced endogenous CD8 T cells could eliminate low antigen loads in liver but were functionally impaired if confronted with elevated antigen loads. Strikingly, only by conditioning the liver environment with TLR9 ligand prior and early after peripheral vaccination, successful immunization against high intrahepatic antigen density with its elimination was achieved. Moreover, TLR9 immunomodulation was also indispensable for functional memory recall after high frequency antigen challenge. Together, the results indicate that TLR9-mediated conditioning of liver environment during therapeutic vaccination or antigen reoccurrence is crucial for an efficacious intrahepatic T cell response.
Collapse
|
618
|
Zhang K, Yu J, Yu X, Han Z, Cheng Z, Liu F, Liang P. Clinical and survival outcomes of percutaneous microwave ablation for intrahepatic cholangiocarcinoma. Int J Hyperthermia 2017; 34:292-297. [PMID: 28540822 DOI: 10.1080/02656736.2017.1327678] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
OBJECTIVE To evaluate the outcomes of percutaneous microwave ablation (MWA) and explore the prognostic factors for the survival of patients with intrahepatic cholangiocarcinoma (ICC). METHODS A total of 107 patients (age: mean 58.0 years, range 15-85 years) with 171 ICCs (maximum size ≤5 cm, tumour number per patient ≤3) who underwent MWA for ICC during January 2009 to February 2016 were selected, and their clinical and pathological data were collected and reviewed. The MWA-associated mortality, major complication rate and survival were evaluated. The prognostic factors for survival in patients with ICC were analysed with univariate and multivariate analyses. RESULTS The median follow-up after MWA was 20.1 months (2.8-63.5 months). There was no procedure-associated death. The overall procedure-associated major complication rate was 2.8%. The median PFS after MWA was 8.9 months; PFS rates after 6, 12, 18 and 24 months were 67.4%, 41.5%, 18.2% and 8.7%. The median OS was 28.0 months; OS rates after 1, 3 and 5 years were 93.5%, 39.6% and 7.9%. Child-Pugh class A and less tumour number were identified as factors predictive of prolonged PFS (HR for Child-Pugh class: 2.62, p = 0.001; HR for tumour number: 2.07, p = 0.002) and OS (HR for Child-Pugh class: 4.14, p < 0.001; HR for tumour number: 1.95, p = 0.024). CONCLUSIONS Percutaneous ultrasound-guided MWA is safe and effective for ICC. Child-Pugh class A and less tumour number predict prolonged PFS and OS in patients with ICC treated by MWA.
Collapse
Affiliation(s)
- Kai Zhang
- a Department of Interventional Ultrasound , Chinese PLA General Hospital , Beijing , China
| | - Jie Yu
- a Department of Interventional Ultrasound , Chinese PLA General Hospital , Beijing , China
| | - Xiaoling Yu
- a Department of Interventional Ultrasound , Chinese PLA General Hospital , Beijing , China
| | - Zhiyu Han
- a Department of Interventional Ultrasound , Chinese PLA General Hospital , Beijing , China
| | - Zhigang Cheng
- a Department of Interventional Ultrasound , Chinese PLA General Hospital , Beijing , China
| | - Fangyi Liu
- a Department of Interventional Ultrasound , Chinese PLA General Hospital , Beijing , China
| | - Ping Liang
- a Department of Interventional Ultrasound , Chinese PLA General Hospital , Beijing , China
| |
Collapse
|
619
|
Pseudomonas aeruginosa Exolysin promotes bacterial growth in lungs, alveolar damage and bacterial dissemination. Sci Rep 2017; 7:2120. [PMID: 28522850 PMCID: PMC5437091 DOI: 10.1038/s41598-017-02349-0] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Accepted: 04/10/2017] [Indexed: 11/18/2022] Open
Abstract
Exolysin (ExlA) is a recently-identified pore-forming toxin secreted by a subset of Pseudomonas aeruginosa strains identified worldwide and devoid of Type III secretion system (T3SS), a major virulence factor. Here, we characterized at the ultrastructural level the lesions caused by an ExlA-secreting strain, CLJ1, in mouse infected lungs. CLJ1 induced necrotic lesions in pneumocytes and endothelial cells, resulting in alveolo-vascular barrier breakdown. Ectopic expression of ExlA in an exlA-negative strain induced similar tissue injuries. In addition, ExlA conferred on bacteria the capacity to proliferate in lungs and to disseminate in secondary organs, similar to bacteria possessing a functional T3SS. CLJ1 did not promote a strong neutrophil infiltration in the alveoli, owing to the weak pro-inflammatory cytokine reaction engendered by the strain. However, CLJ1 was rapidly eliminated from the blood in a bacteremia model, suggesting that it can be promptly phagocytosed by immune cells. Together, our study ascribes to ExlA-secreting bacteria the capacity to proliferate in the lung and to damage pulmonary tissues, thereby promoting metastatic infections, in absence of substantial immune response exacerbation.
Collapse
|
620
|
Surewaard BGJ, Kubes P. Measurement of bacterial capture and phagosome maturation of Kupffer cells by intravital microscopy. Methods 2017; 128:12-19. [PMID: 28522327 DOI: 10.1016/j.ymeth.2017.05.004] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2017] [Revised: 05/04/2017] [Accepted: 05/09/2017] [Indexed: 11/29/2022] Open
Abstract
It is central to the field of bacterial pathogenesis to define how bacteria are killed by phagocytic cells. During phagocytosis, the microbe is localized to the phagolysosome where crucial defense mechanisms such as acidification and production of reactive oxygen species (ROS) are initiated. This process has extensively been studied in vitro, however many resident tissue phagocytes will phenotypically change upon isolation from their natural environment. Therefore, interrogation of phagocytosis and phagosomal function of cells in the context of their natural tissue environment enhances our understanding of the biological process in vivo. This article outlines a real-time intravital microscopy protocol that utilizes fluorescent dyes to study the process of phagocytosis, which reveals acidification and oxidation of individual bacteria inside host cells of living animals. The novelty of this technique exists in use of bacteria that are covalently labelled with the fluorescent dyes Oxyburst and pHrodo, which respectively report on oxidation or acidification. Intravital microscopy is applied to visualize the uptake and subsequent oxidation or acidification of reporter bacteria in the organ of interest. Fluorescently labelled antibodies can be used to counter stain for host immune cells such as neutrophils and macrophages, along with reference stains to identify all bacteria. Although these assays were originally developed to assess the uptake and survival ofStaphylococcus aureusin liver resident macrophages (Kupffer cells), this protocol may be adapted to investigate any bacterium-host cell interaction.
Collapse
Affiliation(s)
- Bas G J Surewaard
- Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada; Department of Medical Microbiology, University Medical Centre, Utrecht, Netherlands.
| | - Paul Kubes
- Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
621
|
Yao ZH, Liao WY, Ho CC, Chen KY, Shih JY, Chen JS, Lin ZZ, Lin CC, Chih-Hsin Yang J, Yu CJ. Real-World Data on Prognostic Factors for Overall Survival in EGFR Mutation-Positive Advanced Non-Small Cell Lung Cancer Patients Treated with First-Line Gefitinib. Oncologist 2017; 22:1075-1083. [PMID: 28507206 DOI: 10.1634/theoncologist.2016-0331] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Accepted: 03/13/2017] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND This study aimed to identify independent prognostic factors for overall survival (OS) of patients with advanced non-small cell lung cancer (NSCLC) harboring an activating epidermal growth factor receptor (EGFR) mutation and receiving gefitinib as first-line treatment in real-world practice. MATERIALS AND METHODS We enrolled 226 patients from June 2011 to May 2013. During this period, gefitinib was the only EGFR-tyrosine kinase inhibitor reimbursed by the Bureau of National Health Insurance of Taiwan. RESULTS The median progression-free survival and median OS were 11.9 months (95% confidence interval [CI]: 9.7-14.2) and 26.9 months (21.2-32.5), respectively. The Cox proportional hazards regression model revealed that postoperative recurrence, performance status (Eastern Cooperative Oncology Grade [ECOG] ≥2), smoking index (≥20 pack-years), liver metastasis at initial diagnosis, and chronic hepatitis C virus (HCV) infection were independent prognostic factors for OS (hazard ratio [95% CI] 0.3 [0.11-0.83], p = .02; 2.69 [1.60-4.51], p < .001; 1.92 [1.24-2.97], p = .003; 2.26 [1.34-3.82], p = .002; 3.38 [1.85-7.78], p < .001, respectively). However, brain metastasis (BM) at initial diagnosis or intracranial progression during gefitinib treatment had no impact on OS (1.266 [0.83-1.93], p = .275 and 0.75 [0.48-1.19], p = .211, respectively). CONCLUSION HCV infection, performance status (ECOG ≥2), newly diagnosed advanced NSCLC without prior operation, and liver metastasis predicted poor OS in EGFR mutation-positive advanced NSCLC patients treated with first-line gefitinib; however, neither BM at initial diagnosis nor intracranial progression during gefitinib treatment had an impact on OS. IMPLICATIONS FOR PRACTICE The finding that chronic hepatitis C virus (HCV) infection might predict poor overall survival (OS) in epidermal growth factor receptor mutation-positive advanced non-small cell lung cancer (NSCLC) patients treated with first-line gefitinib may raise awareness of benefit from anti-HCV treatment in this patient population. Brain metastasis in the initial diagnosis or intracranial progression during gefitinib treatment is not a prognostic factor for OS. This study, which enrolled a real-world population of NSCLC patients, including sicker patients who were not eligible for a clinical trial, may have impact on guiding usual clinical practice.
Collapse
MESH Headings
- Adult
- Aged
- Aged, 80 and over
- Antineoplastic Agents/therapeutic use
- Brain Neoplasms/genetics
- Brain Neoplasms/mortality
- Brain Neoplasms/secondary
- Brain Neoplasms/therapy
- Carcinoma, Non-Small-Cell Lung/genetics
- Carcinoma, Non-Small-Cell Lung/mortality
- Carcinoma, Non-Small-Cell Lung/pathology
- Carcinoma, Non-Small-Cell Lung/therapy
- Disease Progression
- Disease-Free Survival
- ErbB Receptors/genetics
- Female
- Gain of Function Mutation
- Gefitinib
- Hepatitis C, Chronic/epidemiology
- Humans
- Liver Neoplasms/genetics
- Liver Neoplasms/mortality
- Liver Neoplasms/secondary
- Liver Neoplasms/therapy
- Lung/pathology
- Lung/surgery
- Lung Neoplasms/genetics
- Lung Neoplasms/mortality
- Lung Neoplasms/pathology
- Lung Neoplasms/therapy
- Male
- Middle Aged
- Neoplasm Recurrence, Local/genetics
- Neoplasm Recurrence, Local/mortality
- Neoplasm Recurrence, Local/pathology
- Neoplasm Recurrence, Local/therapy
- Prognosis
- Quinazolines/therapeutic use
- Retrospective Studies
- Smoking/epidemiology
- Taiwan/epidemiology
Collapse
Affiliation(s)
- Zong-Han Yao
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, National Taiwan University Hospital, Yun-Lin Branch, Yun-Lin, Taiwan
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
| | - Wei-Yu Liao
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
| | - Chao-Chi Ho
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
| | - Kuan-Yu Chen
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
| | - Jin-Yuan Shih
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
| | - Jin-Shing Chen
- Department of Surgery, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
| | - Zhong-Zhe Lin
- Department of Oncology, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
| | - Chia-Chi Lin
- Department of Oncology, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
| | - James Chih-Hsin Yang
- Department of Oncology, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
| | - Chong-Jen Yu
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
| |
Collapse
|
622
|
Kraakman MJ, Lee MK, Al-Sharea A, Dragoljevic D, Barrett TJ, Montenont E, Basu D, Heywood S, Kammoun HL, Flynn M, Whillas A, Hanssen NM, Febbraio MA, Westein E, Fisher EA, Chin-Dusting J, Cooper ME, Berger JS, Goldberg IJ, Nagareddy PR, Murphy AJ. Neutrophil-derived S100 calcium-binding proteins A8/A9 promote reticulated thrombocytosis and atherogenesis in diabetes. J Clin Invest 2017; 127:2133-2147. [PMID: 28504650 DOI: 10.1172/jci92450] [Citation(s) in RCA: 159] [Impact Index Per Article: 19.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Accepted: 02/16/2017] [Indexed: 12/18/2022] Open
Abstract
Platelets play a critical role in atherogenesis and thrombosis-mediated myocardial ischemia, processes that are accelerated in diabetes. Whether hyperglycemia promotes platelet production and whether enhanced platelet production contributes to enhanced atherothrombosis remains unknown. Here we found that in response to hyperglycemia, neutrophil-derived S100 calcium-binding proteins A8/A9 (S100A8/A9) interact with the receptor for advanced glycation end products (RAGE) on hepatic Kupffer cells, resulting in increased production of IL-6, a pleiotropic cytokine that is implicated in inflammatory thrombocytosis. IL-6 acts on hepatocytes to enhance the production of thrombopoietin, which in turn interacts with its cognate receptor c-MPL on megakaryocytes and bone marrow progenitor cells to promote their expansion and proliferation, resulting in reticulated thrombocytosis. Lowering blood glucose using a sodium-glucose cotransporter 2 inhibitor (dapagliflozin), depleting neutrophils or Kupffer cells, or inhibiting S100A8/A9 binding to RAGE (using paquinimod), all reduced diabetes-induced thrombocytosis. Inhibiting S100A8/A9 also decreased atherogenesis in diabetic mice. Finally, we found that patients with type 2 diabetes have reticulated thrombocytosis that correlates with glycated hemoglobin as well as increased plasma S100A8/A9 levels. These studies provide insights into the mechanisms that regulate platelet production and may aid in the development of strategies to improve on current antiplatelet therapies and to reduce cardiovascular disease risk in diabetes.
Collapse
Affiliation(s)
- Michael J Kraakman
- Haematopoiesis and Leukocyte Biology, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Man Ks Lee
- Haematopoiesis and Leukocyte Biology, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Annas Al-Sharea
- Haematopoiesis and Leukocyte Biology, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Dragana Dragoljevic
- Haematopoiesis and Leukocyte Biology, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | | | | | - Debapriya Basu
- Division of Endocrinology, Diabetes and Metabolism, New York University School of Medicine, New York, New York, USA
| | - Sarah Heywood
- Haematopoiesis and Leukocyte Biology, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Helene L Kammoun
- Haematopoiesis and Leukocyte Biology, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Michelle Flynn
- Haematopoiesis and Leukocyte Biology, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Alexandra Whillas
- Haematopoiesis and Leukocyte Biology, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Nordin Mj Hanssen
- Haematopoiesis and Leukocyte Biology, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia.,Department of Internal Medicine, Cardiovascular Research Institute Maastricht (CARIM), School of Cardiovascular Diseases, Maastricht University, Maastricht, Netherlands
| | - Mark A Febbraio
- Cellular and Molecular Metabolism Laboratory, Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia
| | - Erik Westein
- Vascular Biomechanics, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | | | - Jaye Chin-Dusting
- Department of Pharmacology, Monash University, Clayton, Victoria, Australia
| | - Mark E Cooper
- Diabetic Complications, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | | | - Ira J Goldberg
- Division of Endocrinology, Diabetes and Metabolism, New York University School of Medicine, New York, New York, USA
| | - Prabhakara R Nagareddy
- Department of Nutrition Sciences, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Andrew J Murphy
- Haematopoiesis and Leukocyte Biology, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia.,Department of Immunology, Monash University, Melbourne, Victoria, Australia
| |
Collapse
|
623
|
Pearson RM, Casey LM, Hughes KR, Miller SD, Shea LD. In vivo reprogramming of immune cells: Technologies for induction of antigen-specific tolerance. Adv Drug Deliv Rev 2017; 114:240-255. [PMID: 28414079 PMCID: PMC5582017 DOI: 10.1016/j.addr.2017.04.005] [Citation(s) in RCA: 83] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Revised: 04/01/2017] [Accepted: 04/11/2017] [Indexed: 02/07/2023]
Abstract
Technologies that induce antigen-specific immune tolerance by mimicking naturally occurring mechanisms have the potential to revolutionize the treatment of many immune-mediated pathologies such as autoimmunity, allograft rejection, and allergy. The immune system intrinsically has central and peripheral tolerance pathways for eliminating or modulating antigen-specific responses, which are being exploited through emerging technologies. Antigen-specific tolerogenic responses have been achieved through the functional reprogramming of antigen-presenting cells or lymphocytes. Alternatively, immune privileged sites have been mimicked using biomaterial scaffolds to locally suppress immune responses and promote long-term allograft survival. This review describes natural mechanisms of peripheral tolerance induction and the various technologies being developed to achieve antigen-specific immune tolerance in vivo. As currently approved therapies are non-specific and carry significant associated risks, these therapies offer significant progress towards replacing systemic immune suppression with antigen-specific therapies to curb aberrant immune responses.
Collapse
Affiliation(s)
- Ryan M Pearson
- Department of Biomedical Engineering, University of Michigan, 1119 Carl A. Gerstacker Building, 2200 Bonisteel Boulevard, Ann Arbor, MI 48109-2099, USA
| | - Liam M Casey
- Department of Chemical Engineering, University of Michigan, 2300 Hayward Ave., Ann Arbor, MI 48105, USA
| | - Kevin R Hughes
- Department of Biomedical Engineering, University of Michigan, 1119 Carl A. Gerstacker Building, 2200 Bonisteel Boulevard, Ann Arbor, MI 48109-2099, USA
| | - Stephen D Miller
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, 6-713 Tarry Building, 303 E. Chicago Avenue, Chicago, IL 60611, USA; Chemistry of Life Processes Institute (CLP), Northwestern University, Evanston, IL 60208, USA; The Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL 60611, USA.
| | - Lonnie D Shea
- Department of Biomedical Engineering, University of Michigan, 1119 Carl A. Gerstacker Building, 2200 Bonisteel Boulevard, Ann Arbor, MI 48109-2099, USA; Department of Chemical Engineering, University of Michigan, 2300 Hayward Ave., Ann Arbor, MI 48105, USA.
| |
Collapse
|
624
|
The Roles of Carcinoembryonic Antigen in Liver Metastasis and Therapeutic Approaches. Gastroenterol Res Pract 2017; 2017:7521987. [PMID: 28588612 PMCID: PMC5447280 DOI: 10.1155/2017/7521987] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Accepted: 04/16/2017] [Indexed: 12/22/2022] Open
Abstract
Metastasis is a highly complicated and sequential process in which primary cancer spreads to secondary organic sites. Liver is a well-known metastatic organ from colorectal cancer. Carcinoembryonic antigen (CEA) is expressed in most gastrointestinal, breast, and lung cancer cells. Overexpression of CEA is closely associated with liver metastasis, which is the main cause of death from colorectal cancer. CEA is widely used as a diagnostic and prognostic tumor marker in cancer patients. It affects many steps of liver metastasis from colorectal cancer cells. CEA inhibits circulating cancer cell death. CEA also binds to heterogeneous nuclear RNA binding protein M4 (hnRNP M4), a Kupffer cell receptor protein, and activates Kupffer cells to secrete various cytokines that change the microenvironments for the survival of colorectal cancer cells in the liver. CEA also activates cell adhesion-related molecules. The close correlation between CEA and cancer has spurred the exploration of many CEA-targeted approaches as anticancer therapeutics. Understanding the detailed functions and mechanisms of CEA in liver metastasis will provide great opportunities for the improvement of anticancer approaches against colorectal cancers. In this report, the roles of CEA in liver metastasis and CEA-targeting anticancer modalities are reviewed.
Collapse
|
625
|
White JT, Cross EW, Kedl RM. Antigen-inexperienced memory CD8 + T cells: where they come from and why we need them. Nat Rev Immunol 2017; 17:391-400. [PMID: 28480897 DOI: 10.1038/nri.2017.34] [Citation(s) in RCA: 116] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Memory-phenotype CD8+ T cells exist in substantial numbers within hosts that have not been exposed to either foreign antigen or overt lymphopenia. These antigen-inexperienced memory-phenotype T cells can be divided into two major subsets: 'innate memory' T cells and 'virtual memory' T cells. Although these two subsets are nearly indistinguishable by surface markers alone, notable developmental and functional differences exist between the two subsets, which suggests that they represent distinct populations. In this Opinion article, we review the available literature on each subset, highlighting the key differences between these populations. Furthermore, we suggest a unifying model for the categorization of antigen-inexperienced memory-phenotype CD8+ T cells.
Collapse
Affiliation(s)
- Jason T White
- Department of Microbiology and Immunology, The Peter Doherty Institute, University of Melbourne, 792 Elizabeth Street, Melbourne, Victoria 3000, Australia
| | - Eric W Cross
- Department of Immunology and Microbiology, University of Colorado Denver at Anschutz Medical Campus, School of Medicine, Mail Stop 8333, Room P18-8115, 12800 East 19th Avenue, Aurora, Colorado 80045-2537, USA
| | - Ross M Kedl
- Department of Immunology and Microbiology, University of Colorado Denver at Anschutz Medical Campus, School of Medicine, Mail Stop 8333, Room P18-8115, 12800 East 19th Avenue, Aurora, Colorado 80045-2537, USA
| |
Collapse
|
626
|
David BA, Rubino S, Moreira TG, Freitas‐Lopes MA, Araújo AM, Paul NE, Rezende RM, Menezes GB. Isolation and high-dimensional phenotyping of gastrointestinal immune cells. Immunology 2017; 151:56-70. [PMID: 28039862 PMCID: PMC5382328 DOI: 10.1111/imm.12706] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Revised: 12/21/2016] [Accepted: 12/24/2016] [Indexed: 12/25/2022] Open
Abstract
The gastrointestinal immune system plays a pivotal role in the host relationship with food antigens, the homeostatic microbiome and enteric pathogens. Here, we describe how to collect and process liver and intestinal samples to efficiently isolate and analyse resident immune cells. Furthermore, we describe a step-by-step methodology showing how to high-dimensionally immunophenotype resident leucocytes using cytometry by time-of-flight, providing a well-characterized antibody platform that allows the identification of every leucocyte subset simultaneously. This protocol also includes instructions to purify and cultivate primary murine hepatocytes, a powerful tool to assess basic cell biology and toxicology assays. Gut and liver samples from the same mouse can be collected, processed and stained in less than 6 hr. This protocol enables the recovery of several populations of purified and viable immune cells from solid and fibrous organs, preventing unwanted loss of adherent cells during isolation.
Collapse
Affiliation(s)
- Bruna A. David
- Departamento de MorfologiaCenter for Gastrointestinal BiologyInstituto de Ciências BiológicasUniversidade Federal de Minas GeraisBelo HorizonteMinas GeraisBrazil
| | - Stephen Rubino
- Ann Romney Center for Neurologic DiseasesBrigham and Women's HospitalHarvard Medical SchoolBostonMAUSA
| | - Thais G. Moreira
- Ann Romney Center for Neurologic DiseasesBrigham and Women's HospitalHarvard Medical SchoolBostonMAUSA
| | - Maria Alice Freitas‐Lopes
- Departamento de MorfologiaCenter for Gastrointestinal BiologyInstituto de Ciências BiológicasUniversidade Federal de Minas GeraisBelo HorizonteMinas GeraisBrazil
| | - Alan M. Araújo
- Departamento de MorfologiaCenter for Gastrointestinal BiologyInstituto de Ciências BiológicasUniversidade Federal de Minas GeraisBelo HorizonteMinas GeraisBrazil
| | - Nicole E. Paul
- Dana‐Farber Cancer InstituteHarvard Medical SchoolBostonMAUSA
| | - Rafael M. Rezende
- Ann Romney Center for Neurologic DiseasesBrigham and Women's HospitalHarvard Medical SchoolBostonMAUSA
| | - Gustavo B. Menezes
- Departamento de MorfologiaCenter for Gastrointestinal BiologyInstituto de Ciências BiológicasUniversidade Federal de Minas GeraisBelo HorizonteMinas GeraisBrazil
| |
Collapse
|
627
|
Brandl K, Kumar V, Eckmann L. Gut-liver axis at the frontier of host-microbial interactions. Am J Physiol Gastrointest Liver Physiol 2017; 312:G413-G419. [PMID: 28232456 PMCID: PMC5451561 DOI: 10.1152/ajpgi.00361.2016] [Citation(s) in RCA: 127] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Revised: 02/13/2017] [Accepted: 02/20/2017] [Indexed: 01/31/2023]
Abstract
Liver and intestine are tightly linked through the venous system of the portal circulation. Consequently, the liver is the primary recipient of gut-derived products, most prominently dietary nutrients and microbial components. It functions as a secondary "firewall" and protects the body from intestinal pathogens and other microbial products that have crossed the primary barrier of the intestinal tract. Disruption of the intestinal barrier enhances microbial exposure of the liver, which can have detrimental or beneficial effects in the organ depending on the specific circumstances. Conversely, the liver also exerts influence over intestinal microbial communities via secretion of bile acids and IgA antibodies. This mini-review highlights key findings and concepts in the area of host-microbial interactions as pertinent to the bilateral communication between liver and gut and highlights the concept of the gut-liver axis.
Collapse
Affiliation(s)
- Katharina Brandl
- 1Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, California; and
| | - Vipin Kumar
- 2Department of Medicine, School of Medicine, University of California, San Diego, La Jolla, California
| | - Lars Eckmann
- Department of Medicine, School of Medicine, University of California, San Diego, La Jolla, California
| |
Collapse
|
628
|
Taner T, Park WD, Stegall MD. Unique molecular changes in kidney allografts after simultaneous liver-kidney compared with solitary kidney transplantation. Kidney Int 2017; 91:1193-1202. [PMID: 28233612 DOI: 10.1016/j.kint.2016.12.016] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Revised: 12/13/2016] [Accepted: 12/15/2016] [Indexed: 12/13/2022]
Abstract
Kidney allografts transplanted simultaneously with liver allografts from the same donor are known to be immunologically privileged. This is especially evident in recipients with high levels of donor-specific anti-HLA antibodies. Here we investigated the mechanisms of liver's protective impact using gene expression in the kidney allograft. Select solitary kidney transplant or simultaneous liver-kidney transplant recipients were retrospectively reviewed and separated into four groups: 16 cross-match negative kidney transplants, 15 cross-match positive kidney transplants, 12 cross-match negative simultaneous liver-kidney transplants, and nine cross-match-positive simultaneous liver-kidney transplants. Surveillance biopsies of cross-match-positive kidney transplants had increased expression of genes associated with donor-specific antigens, inflammation, and endothelial cell activation compared to cross-match-negative kidney transplants. These changes were not found in cross-match-positive simultaneous liver-kidney transplant biopsies when compared to cross-match-negative simultaneous liver-kidney transplants. In addition, simultaneously transplanting a liver markedly increased renal expression of genes associated with tissue integrity/metabolism, regardless of the cross-match status. While the expression of inflammatory gene sets in cross-match-positive simultaneous liver-kidney transplants was not completely reduced to the level of cross-match-negative kidney transplants, the downstream effects of donor-specific anti-HLA antibodies were blocked. Thus, simultaneous liver-kidney transplants can have a profound impact on the kidney allograft, not only by decreasing inflammation and avoiding endothelial cell activation in cross-match-positive recipients, but also by increasing processes associated with tissue integrity/metabolism by unknown mechanisms.
Collapse
Affiliation(s)
- Timucin Taner
- William J. von Liebig Center for Transplantation and Clinical Regeneration, Mayo Clinic, Rochester, Minnesota, USA.
| | - Walter D Park
- William J. von Liebig Center for Transplantation and Clinical Regeneration, Mayo Clinic, Rochester, Minnesota, USA
| | - Mark D Stegall
- William J. von Liebig Center for Transplantation and Clinical Regeneration, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
629
|
Ma C, Greten TF. Commensal bacteria (ab)use CD8 + T cells to induce insulin resistance. Sci Immunol 2017; 2:2/10/eaan1473. [PMID: 28738021 DOI: 10.1126/sciimmunol.aan1473] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2017] [Accepted: 03/30/2017] [Indexed: 12/28/2022]
Abstract
Type I interferon-dependent expansion of intrahepatic CD8+ T cells promotes systemic insulin resistance in obesity.
Collapse
Affiliation(s)
- Chi Ma
- GI-Malignancy Section, Thoracic and Gastrointestinal Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Tim F Greten
- GI-Malignancy Section, Thoracic and Gastrointestinal Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
630
|
Kang HJ, Lee H, Park EM, Kim JM, Min BH, Park CG. D-dimer level, in association with humoral responses, negatively correlates with survival of porcine islet grafts in non-human primates with immunosuppression. Xenotransplantation 2017; 24. [DOI: 10.1111/xen.12299] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Revised: 02/15/2017] [Accepted: 03/01/2017] [Indexed: 01/09/2023]
Affiliation(s)
- Hee Jung Kang
- Department of Laboratory Medicine; Hallym University College of Medicine; Anyang-si Korea
| | - Haneulnari Lee
- Department of Laboratory Medicine; Hallym University College of Medicine; Anyang-si Korea
| | - Eun Mi Park
- Department of Laboratory Medicine; Hallym University College of Medicine; Anyang-si Korea
| | - Jong-Min Kim
- Xenotransplantation Research Center; Seoul National University College of Medicine; Seoul Korea
| | - Byoung-Hoon Min
- Xenotransplantation Research Center; Seoul National University College of Medicine; Seoul Korea
| | - Chung-Gyu Park
- Xenotransplantation Research Center; Seoul National University College of Medicine; Seoul Korea
- Department of Microbiology and Immunology; Department of Biomedical Sciences; Cancer Research Institute; Institute of Endemic Diseases; Seoul National University College of Medicine; Seoul Korea
| |
Collapse
|
631
|
Tumeh PC, Hellmann MD, Hamid O, Tsai KK, Loo KL, Gubens MA, Rosenblum M, Harview CL, Taube JM, Handley N, Khurana N, Nosrati A, Krummel MF, Tucker A, Sosa EV, Sanchez PJ, Banayan N, Osorio JC, Nguyen-Kim DL, Chang J, Shintaku IP, Boasberg PD, Taylor EJ, Munster PN, Algazi AP, Chmielowski B, Dummer R, Grogan TR, Elashoff D, Hwang J, Goldinger SM, Garon EB, Pierce RH, Daud A. Liver Metastasis and Treatment Outcome with Anti-PD-1 Monoclonal Antibody in Patients with Melanoma and NSCLC. Cancer Immunol Res 2017; 5:417-424. [PMID: 28411193 DOI: 10.1158/2326-6066.cir-16-0325] [Citation(s) in RCA: 432] [Impact Index Per Article: 54.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2016] [Revised: 01/30/2017] [Accepted: 04/07/2017] [Indexed: 12/18/2022]
Abstract
We explored the association between liver metastases, tumor CD8+ T-cell count, and response in patients with melanoma or lung cancer treated with the anti-PD-1 antibody, pembrolizumab. The melanoma discovery cohort was drawn from the phase I Keynote 001 trial, whereas the melanoma validation cohort was drawn from Keynote 002, 006, and EAP trials and the non-small cell lung cancer (NSCLC) cohort from Keynote 001. Liver metastasis was associated with reduced response and shortened progression-free survival [PFS; objective response rate (ORR), 30.6%; median PFS, 5.1 months] compared with patients without liver metastasis (ORR, 56.3%; median PFS, 20.1 months) P ≤ 0.0001, and confirmed in the validation cohort (P = 0.0006). The presence of liver metastasis significantly increased the likelihood of progression (OR, 1.852; P < 0.0001). In a subset of biopsied patients (n = 62), liver metastasis was associated with reduced CD8+ T-cell density at the invasive tumor margin (liver metastasis+ group, n = 547 ± 164.8; liver metastasis- group, n = 1,441 ± 250.7; P < 0.016). A reduced response rate and shortened PFS was also observed in NSCLC patients with liver metastasis [median PFS, 1.8 months; 95% confidence interval (CI), 1.4-2.0], compared with those without liver metastasis (n = 119, median PFS, 4.0 months; 95% CI, 2.1-5.1), P = 0.0094. Thus, liver metastatic patients with melanoma or NSCLC that had been treated with pembrolizumab were associated with reduced responses and PFS, and liver metastases were associated with reduced marginal CD8+ T-cell infiltration, providing a potential mechanism for this outcome. Cancer Immunol Res; 5(5); 417-24. ©2017 AACR.
Collapse
Affiliation(s)
- Paul C Tumeh
- University of California, Los Angeles, Los Angeles, California
| | | | | | - Katy K Tsai
- University of California, San Francisco, San Francisco, California
| | - Kimberly L Loo
- University of California, San Francisco, San Francisco, California
| | - Matthew A Gubens
- University of California, San Francisco, San Francisco, California
| | | | | | | | - Nathan Handley
- University of California, San Francisco, San Francisco, California
| | - Neharika Khurana
- University of California, San Francisco, San Francisco, California
| | - Adi Nosrati
- University of California, San Francisco, San Francisco, California
| | | | - Andrew Tucker
- University of California, Los Angeles, Los Angeles, California
| | - Eduardo V Sosa
- University of California, San Francisco, San Francisco, California
| | | | - Nooriel Banayan
- University of California, Los Angeles, Los Angeles, California
| | - Juan C Osorio
- Memorial Sloan Kettering Cancer Center, New York, New York
| | | | - Jeremy Chang
- University of California, Los Angeles, Los Angeles, California
| | | | | | - Emma J Taylor
- University of California, Los Angeles, Los Angeles, California
| | - Pamela N Munster
- University of California, San Francisco, San Francisco, California
| | - Alain P Algazi
- University of California, San Francisco, San Francisco, California
| | | | | | | | - David Elashoff
- University of California, Los Angeles, Los Angeles, California
| | - Jimmy Hwang
- University of California, San Francisco, San Francisco, California
| | | | - Edward B Garon
- University of California, Los Angeles, Los Angeles, California
| | | | - Adil Daud
- University of California, San Francisco, San Francisco, California.
| |
Collapse
|
632
|
Clavien PA, Muller X, de Oliveira ML, Dutkowski P, Sanchez-Fueyo A. Can immunosuppression be stopped after liver transplantation? Lancet Gastroenterol Hepatol 2017; 2:531-537. [PMID: 28606879 DOI: 10.1016/s2468-1253(16)30208-4] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2016] [Revised: 11/30/2016] [Accepted: 11/30/2016] [Indexed: 12/22/2022]
Abstract
Liver transplantation has improved dramatically over the past three decades, mainly as a result of advances in surgical techniques and management of post-transplant complications. The focus has now turned towards rescuing additional organs in the face of scarce organ supply, or prevention of long-term toxicity associated with immunosuppression. The liver appears to be privileged in terms of immune tolerance, with a low incidence of antibody-mediated rejection, which is in sharp contrast to other solid organ transplants, such as kidney, lung, and heart transplants. However, tolerogenic processes remain poorly understood, and strategies for complete drug withdrawal should be selected carefully to avoid graft rejection. In this Review, we summarise the current understanding of liver-specific immune responses and provide an outlook on future approaches.
Collapse
Affiliation(s)
- Pierre-Alain Clavien
- Department of Surgery and Transplantation, University Hospital Zurich, Zurich, Switzerland.
| | - Xavier Muller
- Department of Surgery and Transplantation, University Hospital Zurich, Zurich, Switzerland
| | - Michelle L de Oliveira
- Department of Surgery and Transplantation, University Hospital Zurich, Zurich, Switzerland
| | - Philipp Dutkowski
- Department of Surgery and Transplantation, University Hospital Zurich, Zurich, Switzerland
| | - Alberto Sanchez-Fueyo
- Institute of Liver Studies, MRC Centre for Transplantation, King's College London, London, UK
| |
Collapse
|
633
|
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: 87] [Impact Index Per Article: 10.9] [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
|
634
|
Fischer J, Silva TE, Soares E Silva PE, Colombo BS, Silva MC, Wildner LM, Bazzo ML, Rateke ECM, Frode TS, Mello SVD, Rosa JS, Dantas-Correa EB, Narciso-Schiavon JL, Schiavon LL. From stable disease to acute-on-chronic liver failure: Circulating cytokines are related to prognosis in different stages of cirrhosis. Cytokine 2017; 91:162-169. [PMID: 28082235 DOI: 10.1016/j.cyto.2016.12.017] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2016] [Revised: 11/24/2016] [Accepted: 12/21/2016] [Indexed: 12/19/2022]
Abstract
INTRODUCTION Although both pro- and anti-inflammatory circulating cytokines are known to be elevated in liver cirrhosis, its clinical significance is not completely recognized. Our aim was to evaluate the prognostic significance of circulating cytokines interleukin (IL)-6, IL-17 and IL-10 in different stages of cirrhosis. METHODS This prospective study included two cohorts: (1) stable cirrhosis attended in the Outpatient Clinic (n=118), and (2) subjects hospitalized for acute decompensation (AD) (n=130). Thirty healthy subjects served as control group. RESULTS Patients with cirrhosis exhibited higher levels of cytokines as compared to controls. In stable cirrhosis, during a median follow-up of 17months, liver-related events occurred in 26 patients. Higher IL-10 levels and Child-Pugh B/C were independently associated with reduced event-free survival. In AD cohort, death after 90days of follow-up occurred in 39 patients and was independently associated with ascites, higher IL-6 and model for end-stage liver disease. IL-6 levels also showed higher AUROC than CRP for predicting bacterial infection in the AD cohort (0.831±0.043vs. 0.763±0.048, respectively). IL-17 decreased at third day of hospitalization only in patients who progressed to death. Higher IL-6 levels were observed in acute-on-chronic liver failure (ACLF) patients even in the absence of bacterial infection whereas IL-10 was higher only in subjects with infection-related ACLF. Higher IL-10 and IL-17 levels were associated with progression to death in ACLF. CONCLUSIONS The pattern of immune response seems to vary according to the phase of cirrhosis and is related to prognosis, from stable disease to ACLF.
Collapse
Affiliation(s)
- Josiane Fischer
- Division of Gastroenterology, Federal University of Santa Catarina, Brazil
| | | | | | | | | | | | - Maria Luiza Bazzo
- Department of Clinical Analysis, Federal University of Santa Catarina, Brazil
| | | | - Tania Silvia Frode
- Department of Clinical Analysis, Federal University of Santa Catarina, Brazil
| | | | - Júlia S Rosa
- Department of Clinical Analysis, Federal University of Santa Catarina, Brazil
| | | | | | | |
Collapse
|
635
|
Du Y, Li N, Yang H, Luo C, Gong Y, Tong C, Gao Y, Lü S, Long M. Mimicking liver sinusoidal structures and functions using a 3D-configured microfluidic chip. LAB ON A CHIP 2017; 17:782-794. [PMID: 28112323 DOI: 10.1039/c6lc01374k] [Citation(s) in RCA: 154] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/18/2023]
Abstract
Physiologically, four major types of hepatic cells - the liver sinusoidal endothelial cells, Kupffer cells, hepatic stellate cells, and hepatocytes - reside inside liver sinusoids and interact with flowing peripheral cells under blood flow. It is hard to mimic an in vivo liver sinusoid due to its complex multiple cell-cell interactions, spatiotemporal construction, and mechanical microenvironment. Here we developed an in vitro liver sinusoid chip by integrating the four types of primary murine hepatic cells into two adjacent fluid channels separated by a porous permeable membrane, replicating liver's key structures and configurations. Each type of cells was identified with its respective markers, and the assembled chip presented the liver-specific unique morphology of fenestration. The flow field in the liver chip was quantitatively analyzed by computational fluid dynamics simulations and particle tracking visualization tests. Intriguingly, co-culture and shear flow enhance albumin secretion independently or cooperatively, while shear flow alone enhances HGF production and CYP450 metabolism. Under lipopolysaccharide (LPS) stimulations, the hepatic cell co-culture facilitated neutrophil recruitment in the liver chip. Thus, this 3D-configured in vitro liver chip integrates the two key factors of shear flow and the four types of primary hepatic cells to replicate key structures, hepatic functions, and primary immune responses and provides a new in vitro model to investigate the short-duration hepatic cellular interactions under a microenvironment mimicking the physiology of a liver.
Collapse
Affiliation(s)
- Yu Du
- Key Laboratory of Microgravity (National Microgravity Laboratory), Center of Biomechanics and Bioengineering, and Beijing Key Laboratory of Engineered Construction and Mechanobiology, Institute of Mechanics, Chinese Academy of Sciences, Beijing 100190, China. and School of Engineering Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Ning Li
- Key Laboratory of Microgravity (National Microgravity Laboratory), Center of Biomechanics and Bioengineering, and Beijing Key Laboratory of Engineered Construction and Mechanobiology, Institute of Mechanics, Chinese Academy of Sciences, Beijing 100190, China. and School of Engineering Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Hao Yang
- Key Laboratory of Microgravity (National Microgravity Laboratory), Center of Biomechanics and Bioengineering, and Beijing Key Laboratory of Engineered Construction and Mechanobiology, Institute of Mechanics, Chinese Academy of Sciences, Beijing 100190, China. and School of Engineering Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Chunhua Luo
- Key Laboratory of Microgravity (National Microgravity Laboratory), Center of Biomechanics and Bioengineering, and Beijing Key Laboratory of Engineered Construction and Mechanobiology, Institute of Mechanics, Chinese Academy of Sciences, Beijing 100190, China. and School of Engineering Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yixin Gong
- Key Laboratory of Microgravity (National Microgravity Laboratory), Center of Biomechanics and Bioengineering, and Beijing Key Laboratory of Engineered Construction and Mechanobiology, Institute of Mechanics, Chinese Academy of Sciences, Beijing 100190, China. and School of Engineering Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Chunfang Tong
- Key Laboratory of Microgravity (National Microgravity Laboratory), Center of Biomechanics and Bioengineering, and Beijing Key Laboratory of Engineered Construction and Mechanobiology, Institute of Mechanics, Chinese Academy of Sciences, Beijing 100190, China. and School of Engineering Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yuxin Gao
- Key Laboratory of Microgravity (National Microgravity Laboratory), Center of Biomechanics and Bioengineering, and Beijing Key Laboratory of Engineered Construction and Mechanobiology, Institute of Mechanics, Chinese Academy of Sciences, Beijing 100190, China. and School of Engineering Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Shouqin Lü
- Key Laboratory of Microgravity (National Microgravity Laboratory), Center of Biomechanics and Bioengineering, and Beijing Key Laboratory of Engineered Construction and Mechanobiology, Institute of Mechanics, Chinese Academy of Sciences, Beijing 100190, China. and School of Engineering Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Mian Long
- Key Laboratory of Microgravity (National Microgravity Laboratory), Center of Biomechanics and Bioengineering, and Beijing Key Laboratory of Engineered Construction and Mechanobiology, Institute of Mechanics, Chinese Academy of Sciences, Beijing 100190, China. and School of Engineering Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
636
|
Krentz T, Allen S. Bacterial translocation in critical illness. J Small Anim Pract 2017; 58:191-198. [PMID: 28186322 DOI: 10.1111/jsap.12626] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2015] [Revised: 10/18/2016] [Accepted: 11/08/2016] [Indexed: 12/19/2022]
Abstract
Bacterial translocation involves the passage of intestinal bacteria to extraintestinal sites and has been shown to increase morbidity and mortality in critical illness. This review outlines the pathophysiology of bacterial translocation, host defence mechanisms, and reviews the evidence for the clinical management of critically ill patients in order to minimise the negative outcomes associated with bacterial translocation.
Collapse
Affiliation(s)
- T Krentz
- Department of Emergency and Critical Care, Massachusetts Veterinary Referral Hospital, Woburn, MA, 01801, USA
| | - S Allen
- Department of Emergency and Critical Care, Massachusetts Veterinary Referral Hospital, Woburn, MA, 01801, USA
| |
Collapse
|
637
|
Abstract
The tumor microenvironment (TME) in the liver plays an important role in primary and metastatic liver tumor formation and tumor growth promotion. Cellular and non-cellular components of the TME significantly influence tumor development, growth, metastatic spread, anti-tumor immunity and response to tumor therapy. The cellular components of the TME in the liver not only consist of infiltrating immune cells, but also of liver-resident cells such as liver sinusoidal endothelial cells (LSEC) and hepatic stellate cells (HSC), which promote tumor growth by negatively regulating tumor-associated immune responses. In this review, we characterize cells of the TME with pro- and anti-tumor function in primary and metastatic liver tumors. Furthermore, we summarize mechanisms that permit growth of hepatic tumors despite the occurrence of spontaneous anti-tumor immune responses and how novel therapeutic approaches targeting the TME could unleash tumor-specific immune responses to improve survival of liver cancer patients.
Collapse
|
638
|
Golbar HM, Izawa T, Bondoc A, Wijesundera KK, Tennakoon AH, Kuwamura M, Yamate J. Attenuation of alpha-naphthylisothiocyanate (ANIT)-induced biliary fibrosis by depletion of hepatic macrophages in rats. ACTA ACUST UNITED AC 2017; 69:221-230. [PMID: 28159300 DOI: 10.1016/j.etp.2017.01.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2016] [Accepted: 01/17/2017] [Indexed: 02/07/2023]
Abstract
Biliary fibrosis is a complex process in which macrophages and myofibroblasts may play central roles. We investigated biliary fibrosis lesions induced in the Glisson's sheath in rats by alpha-naphthylisothiocyanate (ANIT) administration under macrophage depletion. Hepatic macrophages were depleted in F344 rats with liposome-encapsulated clodronate (CLD) (10mL/kg body weight, i.v) followed by bile duct injury with ANIT (75mg/kg body weight, i.p) (ANIT+CLD group). Rats received empty-liposomes (Lipo) followed by ANIT, and served as control (ANIT+Lipo group). In both ANIT+Lipo and ANIT+CLD groups, ANIT-induced bile duct injury with inflammatory cell infiltration was seen on days 1-3, and subsequently reparative fibrosis occurred on days 5 and 7. In comparisons between the two groups, macrophages reacting to CD68, CD163, MHC class II and CD204 were less in numbers in ANIT+CLD group; the most sensitive immunophenotype was of CD163-positive. Furthermore, in ANIT+CLD group interstitial mesenchymal cells/myofibroblasts reacting to vimentin, desmin and α-smooth muscle actin were also less in grades and tended to be delayed in appearance. Interestingly, MCP-1, IFN-γ, IL-10, and TGF-β1 mRNAs were significantly increased mainly on day 2 in ANIT+Lipo group, while the levels of these factors were prominently lower in ANIT+CLD group. Collectively, depletion of hepatic macrophages plays roles in attenuating biliary fibrogenesis by production of inflammatory factors. The present results indicated clearly importance of macrophage functions in the pathogenesis of biliary fibrosis.
Collapse
Affiliation(s)
- Hossain M Golbar
- Laboratory of Veterinary Pathology, Graduate School of Life and Environmental Sciences, Osaka Prefecture University, 1-58 Rinku-Ourai-Kita, Izumisano City, Osaka, 598-8531, Japan; Department of Veterinary and Animal Sciences, University of Rajshahi, Motihar, Rajshahi, 6205, Bangladesh
| | - Takeshi Izawa
- Laboratory of Veterinary Pathology, Graduate School of Life and Environmental Sciences, Osaka Prefecture University, 1-58 Rinku-Ourai-Kita, Izumisano City, Osaka, 598-8531, Japan
| | - Alexandra Bondoc
- Laboratory of Veterinary Pathology, Graduate School of Life and Environmental Sciences, Osaka Prefecture University, 1-58 Rinku-Ourai-Kita, Izumisano City, Osaka, 598-8531, Japan
| | - Kavindra K Wijesundera
- Laboratory of Veterinary Pathology, Faculty of Veterinary Medicine and Animal Science, University of Peradeniya, 20200, Sri Lanka
| | | | - Mitsuru Kuwamura
- Laboratory of Veterinary Pathology, Graduate School of Life and Environmental Sciences, Osaka Prefecture University, 1-58 Rinku-Ourai-Kita, Izumisano City, Osaka, 598-8531, Japan
| | - Jyoji Yamate
- Laboratory of Veterinary Pathology, Graduate School of Life and Environmental Sciences, Osaka Prefecture University, 1-58 Rinku-Ourai-Kita, Izumisano City, Osaka, 598-8531, Japan.
| |
Collapse
|
639
|
Abstract
Sepsis is recognized by the presence of physiologic and laboratory changes that reflect the inflammatory response to infection on cellular and systemic levels. Comorbid conditions, such as cirrhosis, end-stage renal disease, and obesity, alter patients' susceptibility to infection and their response to it once present. Baseline changes in vital signs and chronic medications often mask clues to the severity of illness. The physiologic, hematologic, and biochemical adjustments that accompany pregnancy and the puerperium introduce similar challenges. Emergency providers must remain vigilant for subtle alterations in the expected baseline for these conditions to arrive at appropriate management decisions.
Collapse
Affiliation(s)
- Matthew P Borloz
- Department of Emergency Medicine, Virginia Tech Carilion School of Medicine, One Riverside Circle, Roanoke, VA 24016, USA.
| | - Khalief E Hamden
- Department of Emergency Medicine, Virginia Tech Carilion School of Medicine, One Riverside Circle, Roanoke, VA 24016, USA
| |
Collapse
|
640
|
Beuke K, Schildberg FA, Pinna F, Albrecht U, Liebe R, Bissinger M, Schirmacher P, Dooley S, Bode JG, Knolle PA, Kummer U, Breuhahn K, Sahle S. Quantitative and integrative analysis of paracrine hepatocyte activation by nonparenchymal cells upon lipopolysaccharide induction. FEBS J 2017; 284:796-813. [PMID: 28109179 DOI: 10.1111/febs.14022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Revised: 12/02/2016] [Accepted: 01/18/2017] [Indexed: 12/14/2022]
Abstract
Gut-derived bacterial lipopolysaccharides (LPS) stimulate the secretion of tumour necrosis factor (TNF) from liver macrophages (MCs), liver sinusoidal endothelial cells (LSECs) and hepatic stellate cells (HSCs), which control the acute phase response in hepatocytes through activation of the NF-κB pathway. The individual and cooperative impact of nonparenchymal cells on this clinically relevant response has not been analysed in detail due to technical limitations. To gain an integrative view on this complex inter- and intracellular communication, we combined a multiscale mathematical model with quantitative, time-resolved experimental data of different primary murine liver cell types. We established a computational model for TNF-induced NF-κB signalling in hepatocytes, accurately describing dose-responsiveness for physiologically relevant cytokine concentrations. TNF secretion profiles were quantitatively measured for all nonparenchymal cell types upon LPS stimulation. This novel approach allowed the analysis of individual and collective paracrine TNF-mediated NF-κB induction in hepatocytes, revealing strongest effects of MCs and LSECs on hepatocellular NF-κB signalling. Simulations suggest that both cell types act together to maximize the NF-κB pathway response induced by low LPS concentrations (0.1 and 1 ng/mL). Higher LPS concentrations (≥ 5 ng/mL) induced sufficient TNF levels from MCs or LSECs to induce a strong and nonadjustable pathway response. Importantly, these simulations also revealed that the initial cytokine secretion (1-2 h after stimulation) rather than final TNF level (10 h after stimulation) defines the hepatocellular NF-κB response. This raises the question whether the current experimental standard of single high-dose cytokine administration is suitable to mimic in vivo cytokine exposure. DATABASE The computational models described in this manuscript are available in the JWS database via the following link: https://jjj.bio.vu.nl/database/beuke.
Collapse
Affiliation(s)
- Katharina Beuke
- Department of Modeling of Biological Processes, COS Heidelberg/BIOQUANT, Heidelberg University, Germany
| | - Frank A Schildberg
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA, USA
| | - Federico Pinna
- Institute of Pathology, University Hospital of Heidelberg, Germany
| | - Ute Albrecht
- Clinic for Gastroenterology, Heinrich-Heine-University of Düsseldorf, Germany
| | - Roman Liebe
- Molecular Hepatology, Department of Medicine II, Medical Faculty at Mannheim, Heidelberg University, Germany
| | | | | | - Steven Dooley
- Molecular Hepatology, Department of Medicine II, Medical Faculty at Mannheim, Heidelberg University, Germany
| | - Johannes G Bode
- Clinic for Gastroenterology, Heinrich-Heine-University of Düsseldorf, Germany
| | - Percy A Knolle
- Institute of Molecular Immunology and Experimental Oncology, München Rechts der Isar, Technische Universität München, Germany
| | - Ursula Kummer
- Department of Modeling of Biological Processes, COS Heidelberg/BIOQUANT, Heidelberg University, Germany
| | - Kai Breuhahn
- Institute of Pathology, University Hospital of Heidelberg, Germany
| | - Sven Sahle
- Department of Modeling of Biological Processes, COS Heidelberg/BIOQUANT, Heidelberg University, Germany
| |
Collapse
|
641
|
Abstract
After partial hepatectomy, hepatocytes proliferate to restore mass and function of the liver. Macrophages, natural killer (NK) cells, natural killer T (NKT) cells, dendritic cells (DC), eosinophils, gamma delta T (γδT) cells, and conventional T cells, as well as other subsets of the immune cells residing in the liver control liver regeneration, either through direct interactions with hepatocytes or indirectly by releasing inflammatory cytokines. Here, we review recent progress regarding the immune cells in the liver and their functions during liver regeneration.
Collapse
Affiliation(s)
- Na Li
- College of Veterinary Medicine, Shaanxi Center of Stem Cells Engineering & Technology, Northwest A&F University, Yangling, Shaanxi, China
| | - Jinlian Hua
- College of Veterinary Medicine, Shaanxi Center of Stem Cells Engineering & Technology, Northwest A&F University, Yangling, Shaanxi, China
| |
Collapse
|
642
|
Man K, Kutyavin VI, Chawla A. Tissue Immunometabolism: Development, Physiology, and Pathobiology. Cell Metab 2017; 25:11-26. [PMID: 27693378 PMCID: PMC5226870 DOI: 10.1016/j.cmet.2016.08.016] [Citation(s) in RCA: 87] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Revised: 08/15/2016] [Accepted: 08/24/2016] [Indexed: 02/07/2023]
Abstract
Evolution of metazoans resulted in the specialization of cellular and tissue function. This was accomplished by division of labor, which allowed tissue parenchymal cells to prioritize their core functions while ancillary functions were delegated to tissue accessory cells, such as immune, stromal, and endothelial cells. In metabolic organs, the accessory cells communicate with their clients, the tissue parenchymal cells, to optimize cellular processes, allowing organisms to adapt to changes in their environment. Here, we discuss tissue immunometabolism from this vantage point and use examples from adipose tissues (white, beige, and brown) and liver to outline the general principles by which accessory cells support metabolic homeostasis in parenchymal cells. A corollary of this model is that disruption of communication between client and accessory cells might predispose metabolic organs to the development of disease.
Collapse
Affiliation(s)
- Kevin Man
- Cardiovascular Research Institute, University of California, San Francisco, CA 94143-0795, USA
| | - Vassily I Kutyavin
- Cardiovascular Research Institute, University of California, San Francisco, CA 94143-0795, USA
| | - Ajay Chawla
- Cardiovascular Research Institute, University of California, San Francisco, CA 94143-0795, USA; Departments of Physiology and Medicine, University of California, San Francisco, CA 94143-0795, USA.
| |
Collapse
|
643
|
Li F, Hao X, Chen Y, Bai L, Gao X, Lian Z, Wei H, Sun R, Tian Z. The microbiota maintain homeostasis of liver-resident γδT-17 cells in a lipid antigen/CD1d-dependent manner. Nat Commun 2017; 7:13839. [PMID: 28067223 PMCID: PMC5227332 DOI: 10.1038/ncomms13839] [Citation(s) in RCA: 137] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Accepted: 11/04/2016] [Indexed: 02/08/2023] Open
Abstract
The microbiota control regional immunity using mechanisms such as inducing IL-17A-producing γδ T (γδT-17) cells in various tissues. However, little is known regarding hepatic γδT cells that are constantly stimulated by gut commensal microbes. Here we show hepatic γδT cells are liver-resident cells and predominant producers of IL-17A. The microbiota sustain hepatic γδT-17 cell homeostasis, including activation, survival and proliferation. The global commensal quantity affects the number of liver-resident γδT-17 cells; indeed, E. coli alone can generate γδT-17 cells in a dose-dependent manner. Liver-resident γδT-17 cell homeostasis depends on hepatocyte-expressed CD1d, that present lipid antigen, but not Toll-like receptors or IL-1/IL-23 receptor signalling. Supplementing mice in vivo or loading hepatocytes in vitro with exogenous commensal lipid antigens augments the hepatic γδT-17 cell number. Moreover, the microbiota accelerate nonalcoholic fatty liver disease through hepatic γδT-17 cells. Thus, our work describes a unique liver-resident γδT-17 cell subset maintained by gut commensal microbes through CD1d/lipid antigens. γδ T cells are major producers of IL-17A in response to microbial infection. Here the authors show that a high load of commensal microbes can maintain homeostasis of IL-17A+ γδ T cells in the liver via CD1d antigen presentation, with implications for liver diseases.
Collapse
Affiliation(s)
- Fenglei Li
- Institute of Immunology and the Key Laboratory of Innate Immunity and Chronic Disease (Chinese Academy of Science), School of Life Science and Medical Center, University of Science and Technology of China, Hefei 230027, China
| | - Xiaolei Hao
- Hefei National Laboratory for Physical Sciences at Microscale, Hefei, Anhui 230027, China
| | - Yongyan Chen
- Institute of Immunology and the Key Laboratory of Innate Immunity and Chronic Disease (Chinese Academy of Science), School of Life Science and Medical Center, University of Science and Technology of China, Hefei 230027, China
| | - Li Bai
- Institute of Immunology and the Key Laboratory of Innate Immunity and Chronic Disease (Chinese Academy of Science), School of Life Science and Medical Center, University of Science and Technology of China, Hefei 230027, China
| | - Xiang Gao
- Model Animal Research Center, Nanjing University, Nanjing, Jiangsu 210061, China
| | - Zhexiong Lian
- Institute of Immunology and the Key Laboratory of Innate Immunity and Chronic Disease (Chinese Academy of Science), School of Life Science and Medical Center, University of Science and Technology of China, Hefei 230027, China
| | - Haiming Wei
- Institute of Immunology and the Key Laboratory of Innate Immunity and Chronic Disease (Chinese Academy of Science), School of Life Science and Medical Center, University of Science and Technology of China, Hefei 230027, China.,Hefei National Laboratory for Physical Sciences at Microscale, Hefei, Anhui 230027, China
| | - Rui Sun
- Institute of Immunology and the Key Laboratory of Innate Immunity and Chronic Disease (Chinese Academy of Science), School of Life Science and Medical Center, University of Science and Technology of China, Hefei 230027, China.,Hefei National Laboratory for Physical Sciences at Microscale, Hefei, Anhui 230027, China.,Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, China
| | - Zhigang Tian
- Institute of Immunology and the Key Laboratory of Innate Immunity and Chronic Disease (Chinese Academy of Science), School of Life Science and Medical Center, University of Science and Technology of China, Hefei 230027, China.,Hefei National Laboratory for Physical Sciences at Microscale, Hefei, Anhui 230027, China.,Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, China
| |
Collapse
|
644
|
McCarthy DP, Yap JWT, Harp CT, Song WK, Chen J, Pearson RM, Miller SD, Shea LD. An antigen-encapsulating nanoparticle platform for T H1/17 immune tolerance therapy. NANOMEDICINE : NANOTECHNOLOGY, BIOLOGY, AND MEDICINE 2017; 13:191-200. [PMID: 27720992 PMCID: PMC5237397 DOI: 10.1016/j.nano.2016.09.007] [Citation(s) in RCA: 82] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/08/2016] [Revised: 09/12/2016] [Accepted: 09/20/2016] [Indexed: 01/19/2023]
Abstract
Tolerogenic nanoparticles (NPs) are rapidly being developed as specific immunotherapies to treat autoimmune disease. However, many NP-based therapies conjugate antigen (Ag) directly to the NP posing safety concerns due to antibody binding or require the co-delivery of immunosuppressants to induce tolerance. Here, we developed Ag encapsulated NPs comprised of poly(lactide-co-glycolide) [PLG(Ag)] and investigated the mechanism of action for Ag-specific tolerance induction in an autoimmune model of T helper type 1/17 dysfunction - relapse-remitting experimental autoimmune encephalomyelitis (R-EAE). PLG(Ag) completely abrogated disease induction in an organ specific manner, where the spleen was dispensable for tolerance induction. PLG(Ag) delivered intravenously distributed to the liver, associated with macrophages, and recruited Ag-specific T cells. Furthermore, programmed death ligand 1 (PD-L1) was increased on Ag presenting cells and PD-1 blockade lessened tolerance induction. The robust promotion of tolerance by PLG(Ag) without co-delivery of immunosuppressive drugs, suggests that these NPs effectively deliver antigen to endogenous tolerogenic pathways.
Collapse
Affiliation(s)
- Derrick P McCarthy
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | | | - Christopher T Harp
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - W Kelsey Song
- Department of Chemical and Biological Engineering, Evanston, IL, USA
| | - Jeane Chen
- Department of Chemical and Biological Engineering, Evanston, IL, USA
| | - Ryan M Pearson
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Stephen D Miller
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA; Chemistry of Life Processes Institute (CLP), Northwestern University, Evanston, IL, USA; The Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL, USA.
| | - Lonnie D Shea
- Department of Biomedical Engineering, Evanston, IL, USA; Department of Chemical and Biological Engineering, Evanston, IL, USA; Chemistry of Life Processes Institute (CLP), Northwestern University, Evanston, IL, USA; The Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL, USA; Department of Obstetrics and Gynecology, Northwestern University, Chicago, IL, USA; Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA; Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
645
|
Abstract
Sepsis and septic shock are characterized by life-threatening organ dysfunction caused by a dysregulated host response to infection. The liver has a central role during sepsis, and is essential to the regulation of immune defence during systemic infections by mechanisms such as bacterial clearance, acute-phase protein or cytokine production and metabolic adaptation to inflammation. However, the liver is also a target for sepsis-related injury, including hypoxic hepatitis due to ischaemia and shock, cholestasis due to altered bile metabolism, hepatocellular injury due to drug toxicity or overwhelming inflammation, as well as distinct pathologies such as secondary sclerosing cholangitis in critically ill patients. Hence, hepatic dysfunction substantially impairs the prognosis of sepsis and serves as a powerful independent predictor of mortality in the intensive care unit. Sepsis is particularly problematic in patients with liver cirrhosis (who experience increased bacterial translocation from the gut and impaired microbial defence) as it can trigger acute-on-chronic liver failure - a syndrome with high short-term mortality. Here, we review the importance of the liver as a guardian, modifier and target of sepsis, the factors that contribute to sepsis in patients with liver cirrhosis and new therapeutic strategies.
Collapse
|
646
|
|
647
|
Stephen C, Nicolas I. A Summary of The 6th International Conference on Coagulation in Liver Disease: Discussion, Debate, Deliberations. Ann Hepatol 2017; 16:12-15. [PMID: 28051789 DOI: 10.5604/16652681.1226811] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Affiliation(s)
- Caldwell Stephen
- Division of Gastroenterology & Hepatology, University of Virginia, Charlottesville, Virginia, USA
| | - Intagliata Nicolas
- Division of Gastroenterology & Hepatology, University of Virginia, Charlottesville, Virginia, USA
| |
Collapse
|
648
|
Brown DL. Immunopathology of the Hepatobiliary System. MOLECULAR AND INTEGRATIVE TOXICOLOGY 2017:329-417. [DOI: 10.1007/978-3-319-47385-7_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
649
|
L'Hermitte A, Pham S, Cadoux M, Couty JP. [Non alcoholic-fatty liver disease causes selective CD4 + T lymphocytes loss and promotes hepatocarcinogenesis]. Med Sci (Paris) 2016; 32:1023-1026. [PMID: 28008846 DOI: 10.1051/medsci/20163211021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Affiliation(s)
- Antoine L'Hermitte
- Institut Cochin, Université Paris-Descartes, Sorbonne Paris Cité, CNRS UMR 8104, Inserm U1016, département Reproduction, Développement et Cancer, 22, rue Méchain, 475014 Paris, France
| | - Sandrine Pham
- Institut Cochin, Université Paris-Descartes, Sorbonne Paris Cité, CNRS UMR 8104, Inserm U1016, département Reproduction, Développement et Cancer, 22, rue Méchain, 475014 Paris, France
| | - Mathilde Cadoux
- Institut Cochin, Université Paris-Descartes, Sorbonne Paris Cité, CNRS UMR 8104, Inserm U1016, département Reproduction, Développement et Cancer, 22, rue Méchain, 475014 Paris, France
| | - Jean-Pierre Couty
- Institut Cochin, Université Paris-Descartes, Sorbonne Paris Cité, CNRS UMR 8104, Inserm U1016, département Reproduction, Développement et Cancer, 22, rue Méchain, 475014 Paris, France
| |
Collapse
|
650
|
Sood S, Yu L, Visvanathan K, Angus PW, Gow PJ, Testro AG. Immune function biomarker QuantiFERON-monitor is associated with infection risk in cirrhotic patients. World J Hepatol 2016; 8:1569-1575. [PMID: 28050238 PMCID: PMC5165271 DOI: 10.4254/wjh.v8.i35.1569] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2016] [Revised: 10/06/2016] [Accepted: 10/22/2016] [Indexed: 02/06/2023] Open
Abstract
AIM To investigate whether a novel immune function biomarker QuantiFERON-Monitor (QFM) can identify cirrhotic patients at greatest risk of infection. METHODS Adult cirrhotic patients on the liver transplant waiting list were recruited for this observational cohort study from a tertiary liver transplant referral unit. The immune function biomarker, QFM was performed using the same method as the widely available Quantiferon-gold assay, and measures output in interferon gamma in IU/mL after dual stimulation of the innate and adaptive immune systems. Ninety-one cirrhotic patients were recruited, with 47 (52%) transplanted on the day of their QFM. The remaining 44 (48%) were monitored for infections until transplant, death, or census date of 1st February 2014. RESULTS Cirrhotic patients express a median QFM significantly lower than healthy controls (94.5 IU/mL vs 423 IU/mL), demonstrating that they are severely immunosuppressed. Several factors including model for end stage liver disease, presence of hepatocellular carcinoma, bilirubin, international normalized ratio and haemoglobin were associated with QFM on univariate analysis. Disease aetiology did not appear to impact QFM. On multivariate analysis, only Child-Pugh score and urea were significantly associated with a patient's immune function as objectively measured by QFM. In the 44 patients who were not transplanted immediately after their blood test and could be monitored for subsequent infection risk, 13 (29.5%) experienced a pre-transplant infection a median 20 d (range 2-182) post-test. QFM < 214 IU/mL was associated with HR = 4.1 (P = 0.01) for infection. A very low QFM < 30 IU/mL was significantly associated (P = 0.003) with death in three patients who died while awaiting transplantation (HR = 56.6). CONCLUSION QFM is lower in cirrhotics, allowing objective determinations of an individual's unique level of immune dysfunction. Low QFM was associated with increased susceptibility to infection.
Collapse
Affiliation(s)
- Siddharth Sood
- Siddharth Sood, Department of Gastroenterology and Hepatology, University of Melbourne, Royal Melbourne Hospital, Parkville, VIC 3050, Australia
| | - Lijia Yu
- Siddharth Sood, Department of Gastroenterology and Hepatology, University of Melbourne, Royal Melbourne Hospital, Parkville, VIC 3050, Australia
| | - Kumar Visvanathan
- Siddharth Sood, Department of Gastroenterology and Hepatology, University of Melbourne, Royal Melbourne Hospital, Parkville, VIC 3050, Australia
| | - Peter William Angus
- Siddharth Sood, Department of Gastroenterology and Hepatology, University of Melbourne, Royal Melbourne Hospital, Parkville, VIC 3050, Australia
| | - Paul John Gow
- Siddharth Sood, Department of Gastroenterology and Hepatology, University of Melbourne, Royal Melbourne Hospital, Parkville, VIC 3050, Australia
| | - Adam Gareth Testro
- Siddharth Sood, Department of Gastroenterology and Hepatology, University of Melbourne, Royal Melbourne Hospital, Parkville, VIC 3050, Australia
| |
Collapse
|