101
|
Metelli A, Wu BX, Fugle CW, Rachidi S, Sun S, Zhang Y, Wu J, Tomlinson S, Howe PH, Yang Y, Garrett-Mayer E, Liu B, Li Z. Surface Expression of TGFβ Docking Receptor GARP Promotes Oncogenesis and Immune Tolerance in Breast Cancer. Cancer Res 2016; 76:7106-7117. [PMID: 27913437 DOI: 10.1158/0008-5472.can-16-1456] [Citation(s) in RCA: 84] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Revised: 09/23/2016] [Accepted: 09/30/2016] [Indexed: 12/20/2022]
Abstract
GARP encoded by the Lrrc32 gene is the cell surface docking receptor for latent TGFβ, which is expressed naturally by platelets and regulatory T cells (Treg). Although Lrrc32 is amplified frequently in breast cancer, the expression and relevant functions of GARP in cancer have not been explored. Here, we report that GARP exerts oncogenic effects, promoting immune tolerance by enriching and activating latent TGFβ in the tumor microenvironment. We found that human breast, lung, and colon cancers expressed GARP aberrantly. In genetic studies in normal mammary gland epithelial and carcinoma cells, GARP expression increased TGFβ bioactivity and promoted malignant transformation in immunodeficient mice. In breast carcinoma-bearing mice that were immunocompetent, GARP overexpression promoted Foxp3+ Treg activity, which in turn contributed to enhancing cancer progression and metastasis. Notably, administration of a GARP-specific mAb limited metastasis in an orthotopic model of human breast cancer. Overall, these results define the oncogenic effects of the GARP-TGFβ axis in the tumor microenvironment and suggest mechanisms that might be exploited for diagnostic and therapeutic purposes. Cancer Res; 76(24); 7106-17. ©2016 AACR.
Collapse
Affiliation(s)
- Alessandra Metelli
- Department of Microbiology and Immunology, Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina
| | - Bill X Wu
- Department of Microbiology and Immunology, Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina
| | - Caroline W Fugle
- Department of Microbiology and Immunology, Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina
| | - Saleh Rachidi
- Department of Microbiology and Immunology, Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina
| | - Shaoli Sun
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, South Carolina
| | - Yongliang Zhang
- Department of Microbiology and Immunology, Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina
| | - Jennifer Wu
- Department of Microbiology and Immunology, Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina
| | - Stephen Tomlinson
- Department of Microbiology and Immunology, Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina
| | - Philip H Howe
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, South Carolina
| | - Yi Yang
- Department of Microbiology and Immunology, Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina
| | - Elizabeth Garrett-Mayer
- Department of Public Health Sciences, Medical University of South Carolina, Charleston, South Carolina
| | - Bei Liu
- Department of Microbiology and Immunology, Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina
| | - Zihai Li
- Department of Microbiology and Immunology, Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina.
| |
Collapse
|
102
|
Guo H, Lu L, Wang R, Perez-Gutierrez A, Abdulkerim H, Zahorchak A, Sumpter T, Reimann KA, Thomson A, Ezzelarab M. Impact of Human Mutant TGFβ1/Fc Protein on Memory and Regulatory T Cell Homeostasis Following Lymphodepletion in Nonhuman Primates. Am J Transplant 2016; 16:2994-3006. [PMID: 27217298 PMCID: PMC5121100 DOI: 10.1111/ajt.13883] [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: 11/13/2015] [Revised: 04/29/2016] [Accepted: 05/07/2016] [Indexed: 01/25/2023]
Abstract
Transforming growth factor β1 (TGFβ1) plays a key role in T cell homeostasis and peripheral tolerance. We evaluated the influence of a novel human mutant TGFβ1/Fc (human IgG4 Fc) fusion protein on memory CD4+ and CD8+ T cell (Tmem) responses in vitro and their recovery following antithymocyte globulin (ATG)-mediated lymphodepletion in monkeys. TGFβ1/Fc induced Smad2/3 protein phosphorylation in rhesus and human peripheral blood mononuclear cells and augmented the suppressive effect of rapamycin on rhesus Tmem proliferation after either alloactivation or anti-CD3/CD28 stimulation. In combination with IL-2, the incidence of CD4+ CD25hi Foxp3hi regulatory T cells (Treg) and Treg:Th17 ratios were increased. In lymphodepleted monkeys, whole blood trough levels of infused TGFβ1/Fc were maintained between 2 and 7 μg/mL for 35 days. Following ATG administration, total T cell numbers were reduced markedly. In those given TGFβ1/Fc infusion, CD8+ T cell recovery to predepletion levels was delayed compared to controls. Additionally, numbers of CD4+ CD25hi CD127lo Treg increased at 4-6 weeks after depletion but subsequently declined to predepletion levels by 12 weeks. In all monkeys, CD4+ CD25hi Foxp3hi Treg/CD4+ IL-17+ cell ratios were reduced, particularly after stopping TGFβ1/Fc infusion. Thus, human TGFβ1/Fc infusion may delay Tmem recovery following lymphodepletion in nonhuman primates. Combined (low-dose) IL-2 infusion may be required to improve the Treg:Th17 ratio following lymphodepletion.
Collapse
Affiliation(s)
- H. Guo
- Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - L. Lu
- Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - R. Wang
- MassBiologics, University of Massachusetts Medical School, Boston, MA
| | - A. Perez-Gutierrez
- Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - H.S. Abdulkerim
- MassBiologics, University of Massachusetts Medical School, Boston, MA
| | - A.F. Zahorchak
- Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - T.L. Sumpter
- Department of Dermatology, University of Pittsburgh School of Medicine
| | - K. A. Reimann
- MassBiologics, University of Massachusetts Medical School, Boston, MA
| | - A.W. Thomson
- Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA,Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - M.B. Ezzelarab
- Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA,Corresponding author: Mohamed B. Ezzelarab, Starzl Transplantation Institute, University of Pittsburgh School of Medicine, 200 Lothrop Street, E1558 BST, Pittsburgh, PA 15261,
| |
Collapse
|
103
|
Maspi N, Abdoli A, Ghaffarifar F. Pro- and anti-inflammatory cytokines in cutaneous leishmaniasis: a review. Pathog Glob Health 2016; 110:247-260. [PMID: 27660895 DOI: 10.1080/20477724.2016.1232042] [Citation(s) in RCA: 165] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Cutaneous leishmaniasis (CL) is caused by different species of the genus Leishmania. Pro- and anti-inflammatory cytokines play different roles in resistance/susceptibility and the immunopathogenesis of Leishmania infection. The balance and dynamic changes in cytokines may control or predict clinical outcome. T helper 1 (Th1) inflammatory cytokines (especially interferon-γ, tumor necrosis factor-α and interleukin-12) are the crucial factors in the initiation of protective immunity against L. major infection, whereas T helper 2 cytokines including IL-5, IL-4, and IL-13 facilitate the persistence of parasites by downregulating the Th1 immune response. On the other hand, aggravation of inflammatory reactions leads to collateral tissue damage and formation of ulcer. For this reason, immunity system such as T regulatory cells produce regulatory cytokines such as transforming growth factor-β and IL-10 to inhibit possible injures caused by increased inflammatory responses in infection site. In this article, we review the role of pro- and anti-inflammatory cytokines in the immunoprotection and immunopathology of CL.
Collapse
Affiliation(s)
- Nahid Maspi
- a Faculty of Medical Sciences, Department of Parasitology , Tarbiat Modares University , Tehran , Iran
| | - Amir Abdoli
- a Faculty of Medical Sciences, Department of Parasitology , Tarbiat Modares University , Tehran , Iran
| | - Fathemeh Ghaffarifar
- a Faculty of Medical Sciences, Department of Parasitology , Tarbiat Modares University , Tehran , Iran
| |
Collapse
|
104
|
Abstract
The number of blood-borne chemotherapeutic agents implicated in drug-induced lung toxicity continues to increase, although problems in detection remain. The initiation of drug-induced lung injury can have an immunologic or nonimmunologic basis. If endothelial cells are injured, interstitial pulmonary edema may result. Regardless of the source of injury, the progression of drug-induced lung toxicity is often quite similar, involving (1) parenchymal damage, (2) recruitment of inflammatory cells, and (3) progression of the inflammatory process. If the inflammatory reponse is sufficiently severe and disperse, increased collagen can be deposited in interstitial and intra-alveolar areas. The resulting attenuation of gas exchange can induce dyspnea and possibly death. Recent research suggests mediation of the fibrogenic process via cytokines such as transforming growth factor-β and tumor necrosis factor. Preliminary results demonstrating amelioration of cytokine mediated lung-induced fibrosis in animal models with appropriate antibodies suggest a possible future modality of therapy. Certain amphiphilic drugs are capable of eliciting a more specific form of lung toxicity. This class of drugs can interfere with phospholipid metabolism in pulmonary macrophages. In these cases, phospholipidosis results from phospholipid accumulation. The physiologic sequelae in human phospholipidosis is still uncertain.
Collapse
Affiliation(s)
- Mannfred A. Hollinger
- Chairman Dept. of Medical Pharmacology & Toxicology School of Medicine University of California Davis, CA 95616
| |
Collapse
|
105
|
Abstract
The human body combats infection and promotes wound healing through the remarkable process of inflammation. Inflammation is characterized by the recruitment of stromal cell activity including recruitment of immune cells and induction of angiogenesis. These cellular processes are regulated by a class of soluble molecules called cytokines. Based on function, cell target, and structure, cytokines are subdivided into several classes including: interleukins, chemokines, and lymphokines. While cytokines regulate normal physiological processes, chronic deregulation of cytokine expression and activity contributes to cancer in many ways. Gene polymorphisms of all types of cytokines are associated with risk of disease development. Deregulation RNA and protein expression of interleukins, chemokines, and lymphokines have been detected in many solid tumors and hematopoetic malignancies, correlating with poor patient prognosis. The current body of literature suggests that in some tumor types, interleukins and chemokines work against the human body by signaling to cancer cells and remodeling the local microenvironment to support the growth, survival, and invasion of primary tumors and enhance metastatic colonization. Some lymphokines are downregulated to suppress tumor progression by enhancing cytotoxic T cell activity and inhibiting tumor cell survival. In this review, we will describe the structure/function of several cytokine families and review our current understanding on the roles and mechanisms of cytokines in tumor progression. In addition, we will also discuss strategies for exploiting the expression and activity of cytokines in therapeutic intervention.
Collapse
Affiliation(s)
- M Yao
- University of Kansas Medical Center, Kansas City, KS, United States
| | - G Brummer
- University of Kansas Medical Center, Kansas City, KS, United States
| | - D Acevedo
- University of Kansas Medical Center, Kansas City, KS, United States
| | - N Cheng
- University of Kansas Medical Center, Kansas City, KS, United States.
| |
Collapse
|
106
|
Zhou J, Wang S, Sun K, Chng WJ. The emerging roles of exosomes in leukemogeneis. Oncotarget 2016; 7:50698-50707. [PMID: 27191983 PMCID: PMC5226614 DOI: 10.18632/oncotarget.9333] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2016] [Accepted: 05/05/2016] [Indexed: 12/21/2022] Open
Abstract
Communication between leukemia cells and their environment is essential for the development and progression of leukemia. Exosomes are microvesicles secreted by many types of cells that contain protein and RNA and mediate intercellular communication. The involvement of exosomes has been demonstrated in the crosstalk between leukemic cells, stromal cells and endothelial cells, consequently promoting the survival of leukemic cells, protection of leukemic cells from the cytotoxic effects of chemotherapeutic drugs, angiogenesis and cell migration. At the same time, exosomes can be used for the detection and monitoring of leukemia, with some advantage over current methods of detection and surveillance. As they are involved in immune response towards leukemic cells, exosomes can also potentially be exploited to augment immunotherapy in leukemia. In this review, we first describe the general characteristics of exosomes and biogenesis of exosomes. We then highlight the emerging role of exosomes in different types of leukemia. Finally, the clinical value of exosomes as biomarkers, in vivo drug carriers and novel exosome-based immunotherapy are discussed.
Collapse
Affiliation(s)
- Jianbiao Zhou
- Cancer Science Institute of Singapore, National University of Singapore, Centre for Translational Medicine, Singapore, Republic of Singapore
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Republic of Singapore
| | - Sam Wang
- Faculty of Arts and Sciences, University of Toronto, Toronto, ON, Canada
| | - Kangyun Sun
- Suzhou Municipal Hospital, Affiliate of Nanjing Medical University, Suzhou, Jiangsu Province, PR China
| | - Wee-Joo Chng
- Cancer Science Institute of Singapore, National University of Singapore, Centre for Translational Medicine, Singapore, Republic of Singapore
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Republic of Singapore
- Department of Hematology-Oncology, National University Cancer Institute of Singapore (NCIS), The National University Health System (NUHS), Singapore, Republic of Singapore
| |
Collapse
|
107
|
Affiliation(s)
- Keith I Block
- Block Center for Integrative Cancer Care, Evanston, IL 60201, USA.
| | | | | | | |
Collapse
|
108
|
Revisiting the regulatory roles of the TGF-β family of cytokines. Autoimmun Rev 2016; 15:917-22. [PMID: 27392504 DOI: 10.1016/j.autrev.2016.07.007] [Citation(s) in RCA: 83] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Accepted: 06/13/2016] [Indexed: 02/04/2023]
Abstract
TGF-β family members are multipotent cytokines that are involved in many cellular processes, including cell differentiation, organ development, wound healing and immune regulation. TGF-β has pleiotropic effects on adaptive immunity, especially in the regulation of CD4(+) T cell and B cell responses. Furthermore, identification of CD4(+) T cell subsets that produce TGF-β3 revealed unexpected roles of TGF-β3 in the control of adaptive immunity. In contrast to TGF-β1, which induces extensive fibrosis, TGF-β3 induces non-scarring wound healing and counteracts tissue fibrosis. Recent progress in the understanding of the activation mechanism of TGF-β may enable us to develop novel biologic therapies based on advanced protein engineering.
Collapse
|
109
|
Kuhn C, Weiner HL. Therapeutic anti-CD3 monoclonal antibodies: from bench to bedside. Immunotherapy 2016; 8:889-906. [DOI: 10.2217/imt-2016-0049] [Citation(s) in RCA: 132] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
The induction of tolerance is a major goal of immunotherapy. Investigations over the last 20 years have shown that anti-CD3 monoclonal antibodies (mAbs) effectively treat autoimmune disease in animal models and have also shown promise in clinical trials. Tolerance induction by anti-CD3 mAbs is related to the induction of Tregs that control pathogenic autoimmune responses. Here, we review preclinical and clinical studies in which intravenous or mucosal administration of anti-CD3 mAbs has been employed and provide an outlook on future developments to enhance the efficacy of this promising therapeutic approach.
Collapse
Affiliation(s)
- Chantal Kuhn
- Ann Romney Center for Neurologic Diseases, Brigham & Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Howard L Weiner
- Ann Romney Center for Neurologic Diseases, Brigham & Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
110
|
The Discovery and Early Days of TGF-β: A Historical Perspective. Cold Spring Harb Perspect Biol 2016; 8:cshperspect.a021865. [PMID: 27328871 DOI: 10.1101/cshperspect.a021865] [Citation(s) in RCA: 143] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Transforming growth factors (TGFs) were discovered as activities that were secreted by cancer cells, and later by normal cells, and had the ability to phenotypically and reversibly transform immortalized fibroblasts. TGF-β distinguished itself from TGF-α because it did not bind to the same epidermal growth factor (EGF) receptor as TGF-α and, therefore, acted through different cell-surface receptors and signaling mediators. This review summarizes the discovery of TGF-β, the early developments in its molecular and biological characterization with its many biological activities in different cell and tissue contexts and its roles in disease, the realization that there is a family of secreted TGF-β-related proteins with many differentiation functions in development and activities in normal cell and tissue physiology, and the subsequent identification and characterization of the receptors and effectors that mediate TGF-β family signaling responses.
Collapse
|
111
|
Hamel Y, Mauvais FX, Pham HP, Kratzer R, Marchi C, Barilleau É, Waeckel-Enée E, Arnoux JB, Hartemann A, Cordier C, Mégret J, Rocha B, de Lonlay P, Beltrand J, Six A, Robert JJ, van Endert P. A unique CD8(+) T lymphocyte signature in pediatric type 1 diabetes. J Autoimmun 2016; 73:54-63. [PMID: 27318739 DOI: 10.1016/j.jaut.2016.06.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Revised: 06/01/2016] [Accepted: 06/08/2016] [Indexed: 12/16/2022]
Abstract
Human type 1 diabetes results from a destructive auto-reactive immune response in which CD8(+) T lymphocytes play a critical role. Given the intense ongoing efforts to develop immune intervention to prevent and/or cure the disease, biomarkers suitable for prediction of disease risk and progress, as well as for monitoring of immunotherapy are required. We undertook separate multi-parameter analyses of single naïve and activated/memory CD8(+) T lymphocytes from pediatric and adult patients, with the objective of identifying cellular profiles associated with onset of type 1 diabetes. We observe global perturbations in gene and protein expression and in the abundance of T cell populations characterizing pediatric but not adult patients, relative to age-matched healthy individuals. Pediatric diabetes is associated with a unique population of CD8(+) T lymphocytes co-expressing effector (perforin, granzyme B) and regulatory (transforming growth factor β, interleukin-10 receptor) molecules. This population persists after metabolic normalization and is especially abundant in children with high titers of auto-antibodies to glutamic acid decarboxylase and with elevated HbA1c values. These findings highlight striking differences between pediatric and adult type 1 diabetes, indicate prolonged large-scale perturbations in the CD8(+) T cell compartment in the former, and suggest that CD8(+)CD45RA(-) T cells co-expressing effector and regulatory factors are of interest as biomarkers in pediatric type 1 diabetes.
Collapse
Affiliation(s)
- Yamina Hamel
- Institut National de la Sante et de la Recherche Médicale, Unité 1151, 75015 Paris, France; Université Paris Descartes, Sorbonne Paris Cité, Faculté de médecine, 75015 Paris, France; Centre National de la Recherche Scientifique, UMR8253, 75015 Paris, France
| | - François-Xavier Mauvais
- Institut National de la Sante et de la Recherche Médicale, Unité 1151, 75015 Paris, France; Université Paris Descartes, Sorbonne Paris Cité, Faculté de médecine, 75015 Paris, France; Centre National de la Recherche Scientifique, UMR8253, 75015 Paris, France
| | - Hang-Phuong Pham
- Sorbonne Universités, UPMC Université Paris 6, 75015 Paris, France; Institut National de la Sante et de la Recherche Médicale, UMRS 959, Immunology-Immunopathology-Immunotherapy (i3), 75013 Paris, France
| | - Roland Kratzer
- Institut National de la Sante et de la Recherche Médicale, Unité 1151, 75015 Paris, France; Université Paris Descartes, Sorbonne Paris Cité, Faculté de médecine, 75015 Paris, France; Centre National de la Recherche Scientifique, UMR8253, 75015 Paris, France
| | - Christophe Marchi
- Institut National de la Sante et de la Recherche Médicale, Unité 1151, 75015 Paris, France; Université Paris Descartes, Sorbonne Paris Cité, Faculté de médecine, 75015 Paris, France; Centre National de la Recherche Scientifique, UMR8253, 75015 Paris, France
| | - Émilie Barilleau
- Institut National de la Sante et de la Recherche Médicale, Unité 1151, 75015 Paris, France; Université Paris Descartes, Sorbonne Paris Cité, Faculté de médecine, 75015 Paris, France; Centre National de la Recherche Scientifique, UMR8253, 75015 Paris, France
| | - Emmanuelle Waeckel-Enée
- Institut National de la Sante et de la Recherche Médicale, Unité 1151, 75015 Paris, France; Université Paris Descartes, Sorbonne Paris Cité, Faculté de médecine, 75015 Paris, France; Centre National de la Recherche Scientifique, UMR8253, 75015 Paris, France
| | - Jean-Baptiste Arnoux
- Université Paris Descartes, Sorbonne Paris Cité, Faculté de médecine, 75015 Paris, France; Centre de référence des Maladies Héréditaires du Métabolisme, Hôpital Necker, Assistance Publique-Hôpitaux de Paris, 75015 Paris, France
| | - Agnès Hartemann
- Université Pierre & Marie Curie, IHU ICAN, 75013 Paris, France; Service de Diabétologie, Hôpital de la Pitié-Salpétrière, Assistance Publique-Hôpitaux de Paris, 75013 Paris, France
| | - Corinne Cordier
- Institut National de la Sante et de la Recherche Médicale, US24, 75015 Paris, France; Centre National de la Recherche Scientifique, UMS3633, 75015 Paris, France
| | - Jerome Mégret
- Institut National de la Sante et de la Recherche Médicale, US24, 75015 Paris, France; Centre National de la Recherche Scientifique, UMS3633, 75015 Paris, France
| | - Benedita Rocha
- Institut National de la Sante et de la Recherche Médicale, Unité 1151, 75015 Paris, France; Université Paris Descartes, Sorbonne Paris Cité, Faculté de médecine, 75015 Paris, France; Centre National de la Recherche Scientifique, UMR8253, 75015 Paris, France
| | - Pascale de Lonlay
- Université Paris Descartes, Sorbonne Paris Cité, Faculté de médecine, 75015 Paris, France; Centre de référence des Maladies Héréditaires du Métabolisme, Hôpital Necker, Assistance Publique-Hôpitaux de Paris, 75015 Paris, France; Institut Imagine, Institut National de la Sante et de la Recherche Médicale, Unité 1163, 75015 Paris, France
| | - Jacques Beltrand
- Université Paris Descartes, Sorbonne Paris Cité, Faculté de médecine, 75015 Paris, France; Endocrinologie, Gynécologie et Diabétologie Pédiatrique, Hôpital Necker, Assistance Publique-Hôpitaux de Paris, 75015 Paris, France
| | - Adrien Six
- Sorbonne Universités, UPMC Université Paris 6, 75015 Paris, France; Institut National de la Sante et de la Recherche Médicale, UMRS 959, Immunology-Immunopathology-Immunotherapy (i3), 75013 Paris, France
| | - Jean-Jacques Robert
- Université Paris Descartes, Sorbonne Paris Cité, Faculté de médecine, 75015 Paris, France; Endocrinologie, Gynécologie et Diabétologie Pédiatrique, Hôpital Necker, Assistance Publique-Hôpitaux de Paris, 75015 Paris, France
| | - Peter van Endert
- Institut National de la Sante et de la Recherche Médicale, Unité 1151, 75015 Paris, France; Université Paris Descartes, Sorbonne Paris Cité, Faculté de médecine, 75015 Paris, France; Centre National de la Recherche Scientifique, UMR8253, 75015 Paris, France.
| |
Collapse
|
112
|
Beltra JC, Decaluwe H. Cytokines and persistent viral infections. Cytokine 2016; 82:4-15. [DOI: 10.1016/j.cyto.2016.02.006] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2015] [Revised: 02/11/2016] [Accepted: 02/11/2016] [Indexed: 12/14/2022]
|
113
|
Morikawa M, Derynck R, Miyazono K. TGF-β and the TGF-β Family: Context-Dependent Roles in Cell and Tissue Physiology. Cold Spring Harb Perspect Biol 2016; 8:8/5/a021873. [PMID: 27141051 DOI: 10.1101/cshperspect.a021873] [Citation(s) in RCA: 956] [Impact Index Per Article: 106.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The transforming growth factor-β (TGF-β) is the prototype of the TGF-β family of growth and differentiation factors, which is encoded by 33 genes in mammals and comprises homo- and heterodimers. This review introduces the reader to the TGF-β family with its complexity of names and biological activities. It also introduces TGF-β as the best-studied factor among the TGF-β family proteins, with its diversity of roles in the control of cell proliferation and differentiation, wound healing and immune system, and its key roles in pathology, for example, skeletal diseases, fibrosis, and cancer.
Collapse
Affiliation(s)
- Masato Morikawa
- Ludwig Cancer Research, Science for Life Laboratory, Uppsala University, Biomedical Center, SE-751 24 Uppsala, Sweden
| | - Rik Derynck
- Department of Cell and Tissue Biology, University of California at San Francisco, San Francisco, California 94143
| | - Kohei Miyazono
- Department of Molecular Pathology, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Japan
| |
Collapse
|
114
|
Mitchell BI, Byron MM, Ng RC, Chow DC, Ndhlovu LC, Shikuma CM. Elevation of Non-Classical (CD14+/lowCD16++) Monocytes Is Associated with Increased Albuminuria and Urine TGF-β1 in HIV-Infected Individuals on Stable Antiretroviral Therapy. PLoS One 2016; 11:e0153758. [PMID: 27097224 PMCID: PMC4838224 DOI: 10.1371/journal.pone.0153758] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2015] [Accepted: 04/04/2016] [Indexed: 12/30/2022] Open
Abstract
OBJECTIVE High rates of albuminuria are observed among HIV-infected individuals on stable antiretroviral therapy (ART). Though pro-inflammatory and pro-fibrotic responses are described as components of albuminuria in the general population, it is unclear how these responses are associated to albuminuria in ART-treated chronic HIV. We investigated the relationship of monocyte subsets and urine inflammatory and fibrotic biomarkers to albuminuria in ART-treated HIV-infected participants. DESIGN AND METHODS Cross-sectional analyses were performed on Hawaii Aging with HIV-cardiovascular disease study cohort participants who were required at entry to be ≥40 years old and on ART ≥3 months. Monocyte subpopulations were determined in banked peripheral blood mononuclear cells (PBMC) using multi-parametric flow-cytometry. Entry random urine samples were assessed for albumin-to-creatinine ratios (UACR). Urine samples were measured for inflammatory and fibrotic biomarkers using Luminex technology. RESULTS Among 96 HIV-infected subjects with measured UACR (87% male, 59% Caucasian, and 89% undetectable HIV RNA with median CD4 of 495.5 cells/μL), 18 patients (19%) had albuminuria. Non-classical (CD14low/+CD16++) monocytes were significantly elevated in subjects with albuminuria (p = 0.034) and were correlated to UACR (r = 0.238, p = 0.019). Elevated non-classical monocyte counts were significant predictors of worsening albuminuria, independent of traditional- and ART-associated risk factors (β = 0.539, p = 0.007). Urine TGF-β1 and collagen-IV were significantly higher in albuminuric compared to non-albuminuric participants (TGF-β1; p = 0.039 and collagen-IV; p = 0.042). Urine TGF-β1 was significantly correlated with non-classical monocyte counts (r = 0.464, p = 0.017). CONCLUSION Alterations in monocyte subpopulations and urine pro-fibrotic factors may play a role in kidney dysfunction during chronic HIV infection and warrants further study.
Collapse
Affiliation(s)
- Brooks I. Mitchell
- Department of Tropical Medicine, University of Hawaii, Honolulu, Hawaii, United States of America
- Hawaii Center for AIDS, University of Hawaii, Honolulu, Hawaii, United States of America
- * E-mail:
| | - Mary Margaret Byron
- Department of Tropical Medicine, University of Hawaii, Honolulu, Hawaii, United States of America
| | - Roland C. Ng
- Department of Medicine, University of Hawaii, Honolulu, Hawaii, United States of America
| | - Dominic C. Chow
- Hawaii Center for AIDS, University of Hawaii, Honolulu, Hawaii, United States of America
- Department of Medicine, University of Hawaii, Honolulu, Hawaii, United States of America
| | - Lishomwa C. Ndhlovu
- Department of Tropical Medicine, University of Hawaii, Honolulu, Hawaii, United States of America
- Hawaii Center for AIDS, University of Hawaii, Honolulu, Hawaii, United States of America
| | - Cecilia M. Shikuma
- Department of Tropical Medicine, University of Hawaii, Honolulu, Hawaii, United States of America
- Hawaii Center for AIDS, University of Hawaii, Honolulu, Hawaii, United States of America
- Department of Medicine, University of Hawaii, Honolulu, Hawaii, United States of America
| |
Collapse
|
115
|
Hisamatsu T, Erben U, Kühl AA. The Role of T-Cell Subsets in Chronic Inflammation in Celiac Disease and Inflammatory Bowel Disease Patients: More Common Mechanisms or More Differences? Inflamm Intest Dis 2016; 1:52-62. [PMID: 29922658 DOI: 10.1159/000445133] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Accepted: 03/02/2016] [Indexed: 12/13/2022] Open
Abstract
Background Chronic intestinal inflammation due to noninfectious causes represents a growing health issue all over the world. Celiac disease as well as inflammatory bowel diseases (IBD) like Crohn's disease and ulcerative and microscopic colitis involve uncontrolled T-cell activation and T-cell-mediated damage as common denominators. Therefore, diagnosis and treatment decisions clearly benefit from the knowledge of the intricacies of the systemic and the local T-cell activity. Summary Depending on the cytokine milieu, CD4+ T cells can differentiate into proinflammatory T helper 1 (Th1), anti-inflammatory Th2, antimicrobial Th17, pleiotropic Th9, tissue-instructing Th22 cells, and in the regulatory compartment forkhead box protein 3+ Treg, suppressive Tr1 or Th3 cells. Additionally, follicular Th cells provide B-cell help in antibody class switching; cytotoxic CD8+ T cells target virus-infected or tumor cells. This review discusses our current knowledge on the contribution of defined T-cell subpopulations to establishing and maintaining chronic intestinal inflammation in either of the above entities. It also puts emphasis on the differences in the prevalence of these diseases between Eastern and Western countries. Key Messages In celiac disease, the driving role of T cells in the lamina propria and in the epithelium mainly specific for two defined antigens is well established. Differences in genetics and lifestyle between Western and Eastern countries were instrumental in understanding underlying mechanisms. In IBD, the vast amount of potential antigens and the corresponding antigen-specific T cells makes it unlikely to find universal triggers. Increased mucosal CD4+ regulatory T cells in all four entities fail to control or abrogate local inflammatory processes. Thus, prevailing differences in the functional T-cell subtypes driving chronic intestinal inflammation in celiac disease and IBD at best allow some overlap in the treatment options for either disease.
Collapse
Affiliation(s)
- Tadakazu Hisamatsu
- The Third Department of Internal Medicine, Kyorin University School of Medicine, Mitaka, Tokyo, Japan
| | - Ulrike Erben
- Medical Department (Gastroenterology/Infectious Diseases/Rheumatology), Campus Benjamin Franklin, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Research Center ImmunoSciences, Campus Benjamin Franklin, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Anja A Kühl
- Medical Department (Gastroenterology/Infectious Diseases/Rheumatology), Campus Benjamin Franklin, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Research Center ImmunoSciences, Campus Benjamin Franklin, Charité - Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
116
|
Fridrich S, Hahn SA, Linzmaier M, Felten M, Zwarg J, Lennerz V, Tuettenberg A, Stöcker W. How Soluble GARP Enhances TGFβ Activation. PLoS One 2016; 11:e0153290. [PMID: 27054568 PMCID: PMC4824412 DOI: 10.1371/journal.pone.0153290] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2015] [Accepted: 03/25/2016] [Indexed: 11/18/2022] Open
Abstract
GARP (glycoprotein A repetitions predominant) is a cell surface receptor on regulatory T-lymphocytes, platelets, hepatic stellate cells and certain cancer cells. Its described function is the binding and accommodation of latent TGFβ (transforming growth factor), before the activation and release of the mature cytokine. For regulatory T cells it was shown that a knockdown of GARP or a treatment with blocking antibodies dramatically decreases their immune suppressive capacity. This confirms a fundamental role of GARP in the basic function of regulatory T cells. Prerequisites postulated for physiological GARP function include membrane anchorage of GARP, disulfide bridges between the propeptide of TGFβ and GARP and connection of this propeptide to αvβ6 or αvβ8 integrins of target cells during mechanical TGFβ release. Other studies indicate the existence of soluble GARP complexes and a functionality of soluble GARP alone. In order to clarify the underlying molecular mechanism, we expressed and purified recombinant TGFβ and a soluble variant of GARP. Surprisingly, soluble GARP and TGFβ formed stable non-covalent complexes in addition to disulfide-coupled complexes, depending on the redox conditions of the microenvironment. We also show that soluble GARP alone and the two variants of complexes mediate different levels of TGFβ activity. TGFβ activation is enhanced by the non-covalent GARP-TGFβ complex already at low (nanomolar) concentrations, at which GARP alone does not show any effect. This supports the idea of soluble GARP acting as immune modulator in vivo.
Collapse
Affiliation(s)
- Sven Fridrich
- Cell and Matrix Biology, Institute of Zoology, JGU Mainz, Mainz, Germany
| | | | - Marion Linzmaier
- Cell and Matrix Biology, Institute of Zoology, JGU Mainz, Mainz, Germany
| | - Matthias Felten
- Cell and Matrix Biology, Institute of Zoology, JGU Mainz, Mainz, Germany
| | - Jenny Zwarg
- University Hospital Mainz, 3rd medical center, Mainz, Germany
| | - Volker Lennerz
- University Hospital Mainz, 3rd medical center, Mainz, Germany
| | | | - Walter Stöcker
- Cell and Matrix Biology, Institute of Zoology, JGU Mainz, Mainz, Germany
- * E-mail:
| |
Collapse
|
117
|
Klapal L, Igelhorst BA, Dietzel-Meyer ID. Changes in Neuronal Excitability by Activated Microglia: Differential Na(+) Current Upregulation in Pyramid-Shaped and Bipolar Neurons by TNF-α and IL-18. Front Neurol 2016; 7:44. [PMID: 27065940 PMCID: PMC4812774 DOI: 10.3389/fneur.2016.00044] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Accepted: 03/14/2016] [Indexed: 12/22/2022] Open
Abstract
Microglia are activated during pathological events in the brain and are capable of releasing various types of inflammatory cytokines. Here, we demonstrate that the addition of 5% microglia activated by 1 μg/ml lipopolysaccharides (LPS) to hippocampal cultures upregulates Na+ current densities (INavD) of bipolar as well as pyramid-shaped neurons, thereby increasing their excitability. Deactivation of microglia by the addition of 10 ng/ml transforming growth factor-β (TGF-β) decreases INavD below control levels suggesting that the residual activated microglial cells influence neuronal excitability in control cultures. Preincubation of hippocampal cultures with 10 ng/ml tumor necrosis factor-α (TNF-α), a major cytokine released by activated microglia, upregulated INavD significantly by ~30% in bipolar cells, whereas in pyramid-shaped cells, the upregulation only reached an increase of ~14%. Incubation of the cultures with antibodies against either TNF-receptor 1 or 2 blocked the upregulation of INavD in bipolar cells, whereas in pyramid-shaped cells, increases in INavD were exclusively blocked by antibodies against TNF-receptor 2, suggesting that both cell types respond differently to TNF-α exposure. Since additional cytokines, such as interleukin-18 (IL-18), are released from activated microglia, we tested potential effects of IL-18 on INavD in both cell types. Exposure to 5–10 ng/ml IL-18 for 4 days increased INavD in both pyramid-shaped as well as bipolar neurons, albeit the dose–response curves were shifted to lower concentrations in bipolar cells. Our results suggest that by secretion of cytokines, microglial cells upregulate Na+ current densities in bipolar and pyramid-shaped neurons to some extent differentially. Depending on the exact cytokine composition and concentration released, this could change the balance between the activity of inhibitory bipolar and excitatory pyramid-shaped cells. Since bipolar cells show a larger upregulation of INavD in response to TNF-α as well as respond to smaller concentrations of IL-18, our results offer an explanation for the finding, that in certain conditions of brain inflammations periods of dizziness are followed by epileptic seizures.
Collapse
Affiliation(s)
- Lars Klapal
- Department of Biochemistry II, Ruhr-University Bochum , Bochum , Germany
| | - Birte A Igelhorst
- Department of Biochemistry II, Ruhr-University Bochum , Bochum , Germany
| | | |
Collapse
|
118
|
Effects of Quorum Sensing Systems on Regulatory T Cells in Catheter-Related Pseudomonas aeruginosa Biofilm Infection Rat Models. Mediators Inflamm 2016; 2016:4012912. [PMID: 27069314 PMCID: PMC4812362 DOI: 10.1155/2016/4012912] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2015] [Accepted: 02/23/2016] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Quorum sensing (QS) systems play an important role in modulating biofilm formation. Recent studies have found that the QS molecules had complex effects on the host immune systems. In addition, regulatory T cells (Tregs), known as important negative regulators in the immune system, have been found upregulated in multiple chronic infections. Therefore, the QS systems were hypothesized to be involved in modulating Tregs in biofilm-associated infections. Object. To explore the effects of QS systems on Tregs in catheter-related Pseudomonas aeruginosa biofilm infection rat models. METHOD Catheter-related Pseudomonas aeruginosa biofilm infection rat models were established; the bacterial clearance rates, total cell counts in bronchoalveolar lavage (BAL) fluid, pathological changes of lungs, and the levels of Foxp3, TGF-β1, and IL-10 in PAO1 strain group were examined and compared with the QS-mutant ΔlasRΔrhlR and ΔlasIΔrhlI groups. RESULTS In PAO1 group, the bacterial clearance rates were lower, total cell counts were higher, pathological changes were severer, and the levels of Foxp3, TGF-β1, and IL-10 were significantly higher compared with QS-mutant groups (p < 0.05). No significant difference was observed between the two QS-mutant groups (p > 0.05). CONCLUSION QS systems can trigger host immune system, accompanied with the upregulation of Tregs.
Collapse
|
119
|
Kaesler S, Skabytska Y, Chen KM, Kempf WE, Volz T, Köberle M, Wölbing F, Hein U, Hartung T, Kirschning C, Röcken M, Biedermann T. Staphylococcus aureus-derived lipoteichoic acid induces temporary T-cell paralysis independent of Toll-like receptor 2. J Allergy Clin Immunol 2016; 138:780-790.e6. [PMID: 26949056 DOI: 10.1016/j.jaci.2015.11.043] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2015] [Revised: 11/09/2015] [Accepted: 11/20/2015] [Indexed: 12/19/2022]
Abstract
BACKGROUND The interplay between microbes and surface organs, such as the skin, shapes a complex immune system with several checks and balances. The first-line defense is mediated by innate immune pathways leading to inflammation. In the second phase specific T cells invade the infected organ, amplifying inflammation and defense. Consecutively, termination of inflammation is crucial to avoid chronic inflammation triggered by microbes, such as in patients with atopic dermatitis. OBJECTIVE We aimed to elucidate how the Staphylococcus aureus-derived cell-wall component lipoteichoic acid (LTA) governs the second phase of immune responses when high concentrations of LTA access T cells directly through disrupted skin. METHODS We analyzed the direct exposure of T cells to LTA in vitro. For in vivo analyses, we used fluorescein isothiocyanate contact hypersensitivity and ovalbumin-induced dermatitis as models for TH2-mediated cutaneous inflammation. RESULTS We observed that LTA potently suppressed T-lymphocyte activation in a Toll-like receptor 2-independent manner. LTA-exposed T cells did not proliferate and did not produce cytokines. Importantly, these T cells remained completely viable and were responsive to consecutive activation signals on subsequent removal of LTA. Thus LTA exposure resulted in temporary functional T-cell paralysis. In vivo experiments revealed that T-cell cytokine production and cutaneous recall responses were significantly suppressed by LTA. CONCLUSION We identified a new mechanism through which bacterial compounds directly but temporarily modulate adaptive immune responses.
Collapse
Affiliation(s)
- Susanne Kaesler
- Department of Dermatology, Eberhard Karls University, Liebermeisterstr, Tubingen, Germany; Department of Dermatology and Allergology, Technische Universität München, Munich, Germany
| | - Yuliya Skabytska
- Department of Dermatology, Eberhard Karls University, Liebermeisterstr, Tubingen, Germany
| | - Ko-Ming Chen
- Department of Dermatology, Eberhard Karls University, Liebermeisterstr, Tubingen, Germany; Derma Labor Düsseldorf, Dusseldorf, Germany
| | - Wolfgang E Kempf
- Department of Dermatology, Eberhard Karls University, Liebermeisterstr, Tubingen, Germany; Department of Dermatology and Allergology, Technische Universität München, Munich, Germany
| | - Thomas Volz
- Department of Dermatology, Eberhard Karls University, Liebermeisterstr, Tubingen, Germany; Department of Dermatology and Allergology, Technische Universität München, Munich, Germany
| | - Martin Köberle
- Department of Dermatology, Eberhard Karls University, Liebermeisterstr, Tubingen, Germany; Department of Dermatology and Allergology, Technische Universität München, Munich, Germany
| | - Florian Wölbing
- Department of Dermatology, Eberhard Karls University, Liebermeisterstr, Tubingen, Germany; Department of Dermatology and Allergology, Technische Universität München, Munich, Germany
| | - Ulrike Hein
- Department of Dermatology, Eberhard Karls University, Liebermeisterstr, Tubingen, Germany
| | - Thomas Hartung
- Center for Alternatives to Animal Testing Europe, University of Konstanz, Konstanz, Germany; Johns Hopkins University, Bloomberg School of Public Health, Baltimore, Md
| | - Carsten Kirschning
- Institute of Medical Microbiology, University of Duisburg-Essen, Essen, Germany
| | - Martin Röcken
- Department of Dermatology, Eberhard Karls University, Liebermeisterstr, Tubingen, Germany
| | - Tilo Biedermann
- Department of Dermatology, Eberhard Karls University, Liebermeisterstr, Tubingen, Germany; Department of Dermatology and Allergology, Technische Universität München, Munich, Germany.
| |
Collapse
|
120
|
Cuende J, Liénart S, Dedobbeleer O, van der Woning B, De Boeck G, Stockis J, Huygens C, Colau D, Somja J, Delvenne P, Hannon M, Baron F, Dumoutier L, Renauld JC, De Haard H, Saunders M, Coulie PG, Lucas S. Monoclonal antibodies against GARP/TGF-β1 complexes inhibit the immunosuppressive activity of human regulatory T cells in vivo. Sci Transl Med 2016; 7:284ra56. [PMID: 25904740 DOI: 10.1126/scitranslmed.aaa1983] [Citation(s) in RCA: 122] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Regulatory T cells (Tregs) are essential to prevent autoimmunity, but excessive Treg function contributes to cancer progression by inhibiting antitumor immune responses. Tregs exert contact-dependent inhibition of immune cells through the production of active transforming growth factor-β1 (TGF-β1). On the Treg cell surface, TGF-β1 is in an inactive form bound to membrane protein GARP and then activated by an unknown mechanism. We demonstrate that GARP is involved in this activation mechanism. Two anti-GARP monoclonal antibodies were generated that block the production of active TGF-β1 by human Tregs. These antibodies recognize a conformational epitope that requires amino acids GARP137-139 within GARP/TGF-β1 complexes. A variety of antibodies recognizing other GARP epitopes did not block active TGF-β1 production by Tregs. In a model of xenogeneic graft-versus-host disease in NSG mice, the blocking antibodies inhibited the immunosuppressive activity of human Tregs. These antibodies may serve as therapeutic tools to boost immune responses to infection or cancer via a mechanism of action distinct from that of currently available immunomodulatory antibodies. Used alone or in combination with tumor vaccines or antibodies targeting the CTLA4 or PD1/PD-L1 pathways, blocking anti-GARP antibodies may improve the efficiency of cancer immunotherapy.
Collapse
Affiliation(s)
- Julia Cuende
- de Duve Institute, Université catholique de Louvain, and WELBIO, B1200 Brussels, Belgium
| | - Stéphanie Liénart
- de Duve Institute, Université catholique de Louvain, and WELBIO, B1200 Brussels, Belgium
| | - Olivier Dedobbeleer
- de Duve Institute, Université catholique de Louvain, and WELBIO, B1200 Brussels, Belgium
| | | | - Gitte De Boeck
- arGEN-X BVBA, Technologiepark 30, B9052 Zwijnaarde, Gent, Belgium
| | - Julie Stockis
- de Duve Institute, Université catholique de Louvain, and WELBIO, B1200 Brussels, Belgium
| | - Caroline Huygens
- de Duve Institute, Université catholique de Louvain, and WELBIO, B1200 Brussels, Belgium
| | | | - Joan Somja
- Department of Pathology, University Hospital of Liège, and Interdisciplinary Cluster of Applied Genoproteomics (GIGA), Laboratory of Experimental Pathology, University of Liège, B4000 Liège, Belgium
| | - Philippe Delvenne
- Department of Pathology, University Hospital of Liège, and Interdisciplinary Cluster of Applied Genoproteomics (GIGA), Laboratory of Experimental Pathology, University of Liège, B4000 Liège, Belgium
| | - Muriel Hannon
- Department of Pathology, University Hospital of Liège, and Interdisciplinary Cluster of Applied Genoproteomics (GIGA), Laboratory of Experimental Pathology, University of Liège, B4000 Liège, Belgium
| | - Frédéric Baron
- Department of Pathology, University Hospital of Liège, and Interdisciplinary Cluster of Applied Genoproteomics (GIGA), Laboratory of Experimental Pathology, University of Liège, B4000 Liège, Belgium
| | | | | | - Hans De Haard
- arGEN-X BVBA, Technologiepark 30, B9052 Zwijnaarde, Gent, Belgium
| | - Michael Saunders
- arGEN-X BVBA, Technologiepark 30, B9052 Zwijnaarde, Gent, Belgium
| | - Pierre G Coulie
- de Duve Institute, Université catholique de Louvain, and WELBIO, B1200 Brussels, Belgium
| | - Sophie Lucas
- de Duve Institute, Université catholique de Louvain, and WELBIO, B1200 Brussels, Belgium.
| |
Collapse
|
121
|
Okeke EB, Uzonna JE. In Search of a Cure for Sepsis: Taming the Monster in Critical Care Medicine. J Innate Immun 2016; 8:156-70. [PMID: 26771196 DOI: 10.1159/000442469] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Accepted: 11/16/2015] [Indexed: 12/21/2022] Open
Abstract
In spite of over half a century of research, sepsis still constitutes a major problem in health care delivery. Although advances in research have significantly increased our knowledge of the pathogenesis of sepsis and resulted in better prognosis and improved survival outcome, sepsis still remains a major challenge in modern medicine with an increase in occurrence predicted and a huge socioeconomic burden. It is generally accepted that sepsis is due to an initial hyperinflammatory response. However, numerous efforts aimed at targeting the proinflammatory cytokine network have been largely unsuccessful and the search for novel potential therapeutic targets continues. Recent studies provide compelling evidence that dysregulated anti-inflammatory responses may also contribute to sepsis mortality. Our previous studies on the role of regulatory T cells and phosphoinositide 3-kinases in sepsis highlight immunological approaches that could be explored for sepsis therapy. In this article, we review the current and emerging concepts in sepsis, highlight novel potential therapeutic targets and immunological approaches for sepsis treatment and propose a biphasic treatment approach for management of the condition.
Collapse
Affiliation(s)
- Emeka B Okeke
- Department of Immunology, Faculty of Medicine, University of Manitoba, Winnipeg, Man., Canada
| | | |
Collapse
|
122
|
Ohira S, Inoue T, Iwao K, Takahashi E, Tanihara H. Factors Influencing Aqueous Proinflammatory Cytokines and Growth Factors in Uveitic Glaucoma. PLoS One 2016; 11:e0147080. [PMID: 26771310 PMCID: PMC4714891 DOI: 10.1371/journal.pone.0147080] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2015] [Accepted: 12/27/2015] [Indexed: 12/14/2022] Open
Abstract
PURPOSE To analyze the effects of factors on aqueous humor proinflammatory cytokine and growth factor levels in patients with uveitic glaucoma (UG). METHODS In this cross-sectional study, we enrolled 143 participants: 1) UG patients (n = 39); 2) primary open-angle glaucoma (POAG) patients (n = 36); and 3) cataract surgery patients, as a comparative group (n = 68). Aqueous humor samples were obtained at the start of surgery. Aqueous cytokine levels were determined using a multiplex immunoassay (xMAP and the Human Cytokine/Chemokine Panel I). RESULTS In UG cases, mean interleukin (IL)-6, IL-8, monocyte chemotactic protein (MCP)-1, tumor necrosis factor (TNF)-α, platelet-derived growth factor (PDGF)-AA, PDGF-AB/BB, and VEGF levels were 171.1, 214.5, 2791.7, 3.5, 23.9, 5.4, and 168.9 pg/mL, respectively, and were higher than those in cataract (non-glaucomatous) cases except PDGF. Levels of IL-6, MCP-1, and VEGF were higher in UG cases than in POAG cases. UG cases with a history of phacoemulsification displayed significantly higher levels of IL-6 (P = 0.0164), IL-8 (P = 0.0003), MCP-1 (P = 0.0465), and PDGF-AB/BB (P = 0.0062) compared to the phakic cases. The presence of cells in the anterior chamber was related to higher levels of IL-8 (P = 0.0002), TNF-α (P = 0.0037), and PDGF-AB/BB (P = 0.0009). The level of PDGF-AB/BB was higher in infectious uveitis than in non-infectious uveitis (P = 0.0211). The level of transforming growth factor (TGF)-β2 was negatively correlated with the levels of MCP-1 (adjusted R2 = 0.28, t = -2.45, P = 0.031) and TNF-α (adjusted R2 = 0.27, t = -2.43, P = 0.032). CONCLUSION A history of phacoemulsification, the presence of cells in the anterior chamber, and infectious uveitis were related to aqueous proinflammatory cytokine levels in patients with UG. TGF-β2 might be an anti-inflammatory factor in aqueous humor of UG patients.
Collapse
Affiliation(s)
- Saori Ohira
- Department of Ophthalmology, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Toshihiro Inoue
- Department of Ophthalmology, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
- * E-mail:
| | - Keiichiro Iwao
- Department of Ophthalmology, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Eri Takahashi
- Department of Ophthalmology, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Hidenobu Tanihara
- Department of Ophthalmology, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| |
Collapse
|
123
|
Liu X, Luo D, Yang N. Cytosolic Low Molecular Weight FGF2 Orchestrates RIG-I-Mediated Innate Immune Response. THE JOURNAL OF IMMUNOLOGY 2015; 195:4943-52. [PMID: 26466960 DOI: 10.4049/jimmunol.1501503] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/06/2015] [Accepted: 09/15/2015] [Indexed: 12/21/2022]
Abstract
Fibroblast growth factor (FGF)2,which is one of the 22 members of the FGF family, functions as an extracellular molecule involved in canonical receptor tyrosine kinase signaling. It has been implicated in angiogenesis and the development of the CNS. In this article, we reveal that cytosolic low m.w. isoform (LMW) FGF2 (18 kDa), not its secreted form, plays an unexpected role in the innate immune response. Cytosolic LMW FGF2 directly associated with inactivated RIG-I under physiological conditions, which enhanced RIG-I protein stability, thereby maintaining basal RIG-I levels. However, during RIG-I activation induced by viral RNA, cytosolic FGF2 bound to the caspase recruitment domains of activated RIG-I, which blocked RIG-I-MAVS complex formation. LMW FGF2 deficiency increased type I IFN production, whereas the overexpression of LMW FGF2 exerted the opposite effect. Cytosolic LMW FGF2 functions as a negative regulator in RIG-I-mediated antiviral signaling. This work provides insight into the role of FGF2 in innate immune response.
Collapse
Affiliation(s)
- Xin Liu
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, China; and
| | - Deyan Luo
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, China; and
| | - Ning Yang
- Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| |
Collapse
|
124
|
Herbertz S, Sawyer JS, Stauber AJ, Gueorguieva I, Driscoll KE, Estrem ST, Cleverly AL, Desaiah D, Guba SC, Benhadji KA, Slapak CA, Lahn MM. Clinical development of galunisertib (LY2157299 monohydrate), a small molecule inhibitor of transforming growth factor-beta signaling pathway. Drug Des Devel Ther 2015; 9:4479-99. [PMID: 26309397 PMCID: PMC4539082 DOI: 10.2147/dddt.s86621] [Citation(s) in RCA: 281] [Impact Index Per Article: 28.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Transforming growth factor-beta (TGF-β) signaling regulates a wide range of biological processes. TGF-β plays an important role in tumorigenesis and contributes to the hallmarks of cancer, including tumor proliferation, invasion and metastasis, inflammation, angiogenesis, and escape of immune surveillance. There are several pharmacological approaches to block TGF-β signaling, such as monoclonal antibodies, vaccines, antisense oligonucleotides, and small molecule inhibitors. Galunisertib (LY2157299 monohydrate) is an oral small molecule inhibitor of the TGF-β receptor I kinase that specifically downregulates the phosphorylation of SMAD2, abrogating activation of the canonical pathway. Furthermore, galunisertib has antitumor activity in tumor-bearing animal models such as breast, colon, lung cancers, and hepatocellular carcinoma. Continuous long-term exposure to galunisertib caused cardiac toxicities in animals requiring adoption of a pharmacokinetic/pharmacodynamic-based dosing strategy to allow further development. The use of such a pharmacokinetic/pharmacodynamic model defined a therapeutic window with an appropriate safety profile that enabled the clinical investigation of galunisertib. These efforts resulted in an intermittent dosing regimen (14 days on/14 days off, on a 28-day cycle) of galunisertib for all ongoing trials. Galunisertib is being investigated either as monotherapy or in combination with standard antitumor regimens (including nivolumab) in patients with cancer with high unmet medical needs such as glioblastoma, pancreatic cancer, and hepatocellular carcinoma. The present review summarizes the past and current experiences with different pharmacological treatments that enabled galunisertib to be investigated in patients.
Collapse
Affiliation(s)
| | - J Scott Sawyer
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, USA
| | - Anja J Stauber
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, USA
| | | | - Kyla E Driscoll
- Lilly Research Laboratories, Eli Lilly and Company, New York, NY, USA
| | - Shawn T Estrem
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, USA
| | - Ann L Cleverly
- Lilly Research Laboratories, Eli Lilly and Company, Windlesham, Surrey, UK
| | - Durisala Desaiah
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, USA
| | - Susan C Guba
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, USA
| | - Karim A Benhadji
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, USA
| | | | - Michael M Lahn
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, USA
| |
Collapse
|
125
|
Yang EY, Kronenfeld JP, Gattás-Asfura KM, Bayer AL, Stabler CL. Engineering an "infectious" T(reg) biomimetic through chemoselective tethering of TGF-β1 to PEG brush surfaces. Biomaterials 2015. [PMID: 26197412 DOI: 10.1016/j.biomaterials.2015.07.009] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Modulation of immunological responses to allografts following transplantation is of pivotal importance to improving graft outcome and duration. Of the many approaches, harnessing the dominant tolerance induced by regulatory T cells (Treg) holds tremendous promise. Recent studies have highlighted the unique potency of cell surface-bound TGF-β1 on Treg for promoting infectious tolerance, i.e. to confer suppressive capacity from one cell to another. To mimic this characteristic, TGF-β1 was chemoselectively tethered to inert and viable polymer grafting platforms using Staudinger ligation. We report the synthesis and functional characterization of these engineered TGF-β1 surfaces. Inert beads tethered with TGF-β1 were capable of efficiently converting naïve CD4(+) CD62L(hi) T cells to functional Treg. Concordantly, translation of conjugation scheme from inert surfaces to viable cells also led to efficient generation of functional Treg. Further, the capacity of these platforms to generate antigen-specific Treg was demonstrated. These findings illustrate the unique faculty of tethered TGF-β1 biomaterial platforms to function as an "infectious" Treg and provide a compelling approach for generating tolerogenic microenvironments for allograft transplantation.
Collapse
Affiliation(s)
- E Y Yang
- Diabetes Research Institute, University of Miami, Miami, FL, USA; Department of Biochemistry and Molecular Biology, University of Miami, Miami, FL, USA
| | - J P Kronenfeld
- Diabetes Research Institute, University of Miami, Miami, FL, USA; Department of Medicine, University of Miami, Miami, FL, USA
| | | | - A L Bayer
- Diabetes Research Institute, University of Miami, Miami, FL, USA; Department of Microbiology and Immunology, University of Miami, Miami, FL, USA
| | - C L Stabler
- Diabetes Research Institute, University of Miami, Miami, FL, USA; Department of Biochemistry and Molecular Biology, University of Miami, Miami, FL, USA; Department of Biomedical Engineering, University of Miami, Miami, FL, USA; Department of Surgery, University of Miami, Miami, FL, USA.
| |
Collapse
|
126
|
Ly J, Lagman M, Saing T, Singh MK, Tudela EV, Morris D, Anderson J, Daliva J, Ochoa C, Patel N, Pearce D, Venketaraman V. Liposomal Glutathione Supplementation Restores TH1 Cytokine Response to Mycobacterium tuberculosis Infection in HIV-Infected Individuals. J Interferon Cytokine Res 2015; 35:875-87. [PMID: 26133750 PMCID: PMC4642835 DOI: 10.1089/jir.2014.0210] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Cytokines are signaling biomolecules that serve as key regulators of our immune system. CD4+ T-cells can be grouped into 2 major categories based on their cytokine profile: T-helper 1 (TH1) subset and T-helper 2 (TH2) subset. Protective immunity against HIV infection requires TH1-directed CD4 T-cell responses, mediated by cytokines, such as interleukin-1β (IL-1β), IL-12, interferon-γ (IFN-γ), and tumor necrosis factor-α (TNF-α). Cytokines released by the TH1 subset of CD4 T-cells are considered important for mediating effective immune responses against intracellular pathogens such as Mycobacterium tuberculosis (M. tb). Oxidative stress and redox imbalance that occur during HIV infection often lead to inappropriate immune responses. Glutathione (GSH) is an antioxidant present in nearly all cells and is recognized for its function in maintaining redox homeostasis. Our laboratory previously reported that individuals with HIV infection have lower levels of GSH. In this study, we report a link between lower levels of GSH and dysregulation of TH1- and TH2-associated cytokines in the plasma samples of HIV-positive subjects. Furthermore, we demonstrate that supplementing individuals with HIV infection for 13 weeks with liposomal GSH (lGSH) resulted in a significant increase in the levels of TH1 cytokines, IL-1β, IL-12, IFN-γ, and TNF-α. lGSH supplementation in individuals with HIV infection also resulted in a substantial decrease in the levels of free radicals and immunosuppressive cytokines, IL-10 and TGF-β, relative to those in a placebo-controlled cohort. Finally, we determined the effects of lGSH supplementation in improving the functions of immune cells to control M. tb infection by conducting in vitro assays using peripheral blood mononuclear cells collected from HIV-positive individuals at post-GSH supplementation. Our studies establish a correlation between low levels of GSH and increased susceptibility to M. tb infection through TH2-directed response, which may be relieved with lGSH supplementation enhancing the TH1 response.
Collapse
Affiliation(s)
- Judy Ly
- 1 Graduate College of Biomedical Sciences, Western University of Health Sciences , Pomona, California.,2 Department of Basic Medical Sciences, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences , Pomona, California
| | - Minette Lagman
- 1 Graduate College of Biomedical Sciences, Western University of Health Sciences , Pomona, California.,2 Department of Basic Medical Sciences, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences , Pomona, California
| | - Tommy Saing
- 1 Graduate College of Biomedical Sciences, Western University of Health Sciences , Pomona, California
| | - Manpreet Kaur Singh
- 1 Graduate College of Biomedical Sciences, Western University of Health Sciences , Pomona, California.,2 Department of Basic Medical Sciences, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences , Pomona, California
| | - Enrique Vera Tudela
- 1 Graduate College of Biomedical Sciences, Western University of Health Sciences , Pomona, California.,2 Department of Basic Medical Sciences, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences , Pomona, California
| | - Devin Morris
- 2 Department of Basic Medical Sciences, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences , Pomona, California
| | - Jessica Anderson
- 2 Department of Basic Medical Sciences, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences , Pomona, California
| | - John Daliva
- 2 Department of Basic Medical Sciences, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences , Pomona, California
| | - Cesar Ochoa
- 3 Western Diabetes Institute , Pomona, California
| | | | - Daniel Pearce
- 4 Center for Comparative Effectiveness and Outcomes Research, Loma Linda University , Loma Linda, California
| | - Vishwanath Venketaraman
- 1 Graduate College of Biomedical Sciences, Western University of Health Sciences , Pomona, California.,2 Department of Basic Medical Sciences, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences , Pomona, California
| |
Collapse
|
127
|
Abstract
Importance of chronic fibroproliferative diseases (FDs) including pulmonary fibrosis, chronic kidney diseases, inflammatory bowel disease, and cardiovascular or liver fibrosis is rapidly increasing and they have become a major public health problem. According to some estimates about 45% of all deaths are attributed to FDs in the developed world. Independently of their etiology the common hallmark of FDs is chronic inflammation. Infiltrating immune cells, endothelial, epithelial, and other resident cells of the injured organ release an orchestra of inflammatory mediators, which stimulate the proliferation and excessive extracellular matrix (ECM) production of myofibroblasts, the effector cells of organ fibrosis. Abnormal amount of ECM disturbs the original organ architecture leading to the decline of function. Although our knowledge is rapidly expanding, we still have neither a diagnostic tool to detect nor a drug to specifically target fibrosis. Therefore, there is an urgent need for the more comprehensive understanding of the pathomechanism of fibrosis and development of novel diagnostic and therapeutic strategies. In the present review we provide an overview of the common key mediators of organ fibrosis highlighting the role of interleukin-10 (IL-10) cytokine family members (IL-10, IL-19, IL-20, IL-22, IL-24, and IL-26), which recently came into focus as tissue remodeling-related inflammatory cytokines.
Collapse
|
128
|
ATaenia crassicepsfactor induces apoptosis of spleen CD4+T cells and TFG-β andFoxp3gene expression in mice. J Helminthol 2015; 90:223-31. [DOI: 10.1017/s0022149x1500022x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
AbstractThis study was undertaken to determine whether a parasite substance produces structural pathology in the mouse spleen. A low-molecular-weightTaenia crassicepsmetacestode factor (MF) isolated from the peritoneal fluid of female mice infected withT. crassicepsmetacestodes induced pathological and immunological changes in mouse spleen cellsin vivo.Electron microscopy and confocal microscopy revealed severe changes in the spleen histoarchitecture ofT. crassiceps-infected and MF-treated mice. Apoptotic degenerated spleen cells were observed in the white and red pulps and were more conspicuous in the white pulp of the spleen from theT. crassiceps-infected mice than in that of the MF-treated mice. Flow cytometry analysis revealed that the numbers of spleen CD4+T cells were significantly lower in both experimental groups than in control mice. Theex vivoexpression of transforming growth factor (TGF)-β and factor Foxp3 were significantly higher in splenocytes of the experimental mice than the basal expression observed in the control cells. These findings may have potential applications for a better understanding of the host–parasite relationship in human neurocysticercosis.
Collapse
|
129
|
Ren S, Chai L, Wang C, Li C, Ren Q, Yang L, Wang F, Qiao Z, Li W, He M, Riker AI, Han Y, Yu Q. Human malignant melanoma-derived progestagen-associated endometrial protein immunosuppresses T lymphocytes in vitro. PLoS One 2015; 10:e0119038. [PMID: 25785839 PMCID: PMC4364885 DOI: 10.1371/journal.pone.0119038] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2014] [Accepted: 01/17/2015] [Indexed: 01/04/2023] Open
Abstract
Progestagen-associated endometrial protein (PAEP) is a glycoprotein of the lipocalin family that acts as a negative regulator of T cell receptor-mediated activation. However, the function of tumor-derived PAEP on the human immune system in the tumor microenvironment is unknown. PAEP is highly expressed in intermediate and thick primary melanomas (Breslow’s 2.5mm or greater) and metastatic melanomas, correlating with its expression in daughter cell lines established in vitro. The current study investigates the role of melanoma cell-secreted PAEP protein in regulating T cell function. Upon the enrichment of CD3+, CD4+ and CD8+ T cells from human peripheral blood mononuclear cells, each subset was then mixed with either melanoma-derived PAEP protein or PAEP-poor supernatant of gene-silenced tumor cells. IL-2 and IFN-γ secretion of CD4+ T cells significantly decreased with the addition of PAEP-rich supernatant. And the addition of PAEP-positive cell supernatant to activated lymphocytes significantly inhibited lymphocyte proliferation and cytotoxic T cell activity, while increasing lymphocyte apoptosis. Our result suggests that melanoma cell-secreted PAEP protein immunosuppresses the activation, proliferation and cytotoxicity of T lymphocytes, which might partially explain the mechanism of immune tolerance induced by melanoma cells within the tumor microenvironment.
Collapse
Affiliation(s)
- Suping Ren
- Department of Immunohematology, Beijing Institute of Transfusion Medicine, Beijing, China
- * E-mail: (SR); (QY)
| | - Lina Chai
- Department of Immunohematology, Beijing Institute of Transfusion Medicine, Beijing, China
| | - Chunyan Wang
- Department of Immunohematology, Beijing Institute of Transfusion Medicine, Beijing, China
- Department of poisoning treatment, Beijing 307 hospital, Beijing, China
| | - Changlan Li
- Department of Immunohematology, Beijing Institute of Transfusion Medicine, Beijing, China
| | - Qiquan Ren
- People’s Hospital of Anqiu City, Anqiu, Shandong, China
| | - Lihua Yang
- People’s Hospital of Anqiu City, Anqiu, Shandong, China
| | - Fumei Wang
- People’s Hospital of Anqiu City, Anqiu, Shandong, China
| | - Zhixin Qiao
- Department of Immunohematology, Beijing Institute of Transfusion Medicine, Beijing, China
| | - Weijing Li
- Department of Immunohematology, Beijing Institute of Transfusion Medicine, Beijing, China
| | - Min He
- Department of Immunohematology, Beijing Institute of Transfusion Medicine, Beijing, China
| | - Adam I. Riker
- Advocate Christ Medical Center, Advocate Cancer Institute, Oak Lawn, IL, United States of America
| | - Ying Han
- Department of Immunohematology, Beijing Institute of Transfusion Medicine, Beijing, China
| | - Qun Yu
- Department of Immunohematology, Beijing Institute of Transfusion Medicine, Beijing, China
- * E-mail: (SR); (QY)
| |
Collapse
|
130
|
George J, Lewis MG, Renne R, Mattapallil JJ. Suppression of transforming growth factor β receptor 2 and Smad5 is associated with high levels of microRNA miR-155 in the oral mucosa during chronic simian immunodeficiency virus infection. J Virol 2015; 89:2972-8. [PMID: 25540365 PMCID: PMC4325739 DOI: 10.1128/jvi.03248-14] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2014] [Accepted: 12/15/2014] [Indexed: 12/12/2022] Open
Abstract
Chronic human immunodeficiency virus and simian immunodeficiency virus (HIV and SIV) infections are characterized by mucosal inflammation in the presence of anti-inflammatory cytokines such as transforming growth factor β (TGFβ). The mechanisms for refractiveness to TGFβ are not clear. Here we show that the expression of microRNA miR-155 was significantly upregulated in the oropharyngeal mucosa during chronic SIV infection and was coincident with downregulation of TGFβ receptor 2 (TGFβ-R2) and SMAD5, key TGFβ signaling genes that harbor putative target sites for miR-155. Ectopic expression of miR-155 in vitro was found to significantly downregulate TGFβ-R2 and Smad5 expression, suggesting a role for miR-155 in the suppression of TGFβ-R2 and SMAD5 genes in vivo. The downregulation of TGFβ signaling genes by miR-155 likely contributes to the nonresponsiveness to TGFβ during SIV infection and may inadvertently aid in increased immune activation during HIV and SIV infections.
Collapse
Affiliation(s)
- Jeffy George
- Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
| | | | - Rolf Renne
- University of Florida, Gainesville, Florida, USA
| | | |
Collapse
|
131
|
Sanders YY, Cui Z, Le Saux CJ, Horowitz JC, Rangarajan S, Kurundkar A, Antony VB, Thannickal VJ. SMAD-independent down-regulation of caveolin-1 by TGF-β: effects on proliferation and survival of myofibroblasts. PLoS One 2015; 10:e0116995. [PMID: 25658089 PMCID: PMC4319960 DOI: 10.1371/journal.pone.0116995] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2014] [Accepted: 12/17/2014] [Indexed: 12/30/2022] Open
Abstract
Transforming growth factor-β (TGF-β) mediates growth-inhibitory effects on most target cells via activation of the canonical SMAD signaling pathway. This growth-inhibitory activity may be coupled with cellular differentiation. Our studies demonstrate that TGF-β1 inhibits proliferation of primary, non-transformed human lung fibroblasts in association with the induction of myofibroblast differentiation. Differentiated myofibroblasts maintain the capacity to proliferate in response to exogenous mitogenic stimuli and are resistant to serum deprivation-induced apoptosis. These proliferative and anti-apoptotic properties of myofibroblasts are related, in part, to the down-regulation of caveolin-1 (Cav-1) by TGF-β1. Cav-1 down-regulation is mediated by early activation of p38 MAPK and does not require SMAD signaling. In contrast, myofibroblast differentiation is dependent on activation of the SMAD pathway, but not on p38 MAPK. Thus, combinatorial signaling by TGF-β1 of myofibroblast differentiation and down-regulation of Cav-1 by SMAD and p38 MAPK pathways, respectively, confer proliferative and apoptosis-resistant properties to myofibroblasts. Selective targeting of this SMAD-independent, p38-MAPK/Cav-1-dependent pathway is likely to be effective in the treatment of pathological conditions characterized by TGF-β signaling and myofibroblast activation.
Collapse
Affiliation(s)
- Yan Y. Sanders
- Division of Pulmonary, Allergy and Critical Care, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, 35294, United States of America
| | - Zongbin Cui
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan, 48109, United States of America
| | - Claude Jourdan Le Saux
- Division of Cardiology, Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas, 78229, United States of America
| | - Jeffrey C. Horowitz
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan, 48109, United States of America
| | - Sunad Rangarajan
- Division of Pulmonary, Allergy and Critical Care, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, 35294, United States of America
| | - Ashish Kurundkar
- Division of Pulmonary, Allergy and Critical Care, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, 35294, United States of America
| | - Veena B. Antony
- Division of Pulmonary, Allergy and Critical Care, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, 35294, United States of America
| | - Victor J. Thannickal
- Division of Pulmonary, Allergy and Critical Care, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, 35294, United States of America
- * E-mail:
| |
Collapse
|
132
|
Xiao B, Wang Y, Reinach PS, Ren Y, Li J, Hua S, Lu H, Chen W. Dynamic ocular surface and lacrimal gland changes induced in experimental murine dry eye. PLoS One 2015; 10:e0115333. [PMID: 25590134 PMCID: PMC4295848 DOI: 10.1371/journal.pone.0115333] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2014] [Accepted: 11/21/2014] [Indexed: 01/22/2023] Open
Abstract
Dry eye disease can be a consequence of lacrimal gland insufficiency in Sjögren’s Syndrome or increased tear film evaporation despite normal lacrimal gland function. To determine if there is a correlation between severity effects in these models and underlying pathophysiological responses, we compared the time dependent changes in each of these parameters that occur during a 6 week period. Dry eye was induced in 6-week-old female C57BL/6 mice by exposing them to an Intelligently Controlled Environmental System (ICES). Sixty mice were housed in ICES for 1, 2, 4 and 6 weeks respectively. Twelve were raised in normal environment and received subcutaneous injections of scopolamine hydrobromide (SCOP) 3 times daily for 5 days. Another sixty mice were housed in a normal environment and received no treatment. Corneal fluorescein staining along with corneal MMP-9 and caspase-3 level measurements were performed in parallel with the TUNEL assay. Interleukin-17(IL-17), IL-23, IL-6, IL-1, TNF-α, IFN-γ and TGF-β2 levels were estimated by real-time PCR measurements of conjunctival and lacrimal gland samples (LGs). Immunohistochemistry of excised LGs along with flow cytometry in cervical lymph nodes evaluated immune cell infiltration. Light and transmission electron microscopy studies evaluated LGs cytoarchitectural changes. ICES induced corneal epithelial destruction and apoptosis peaked at 2 weeks and kept stable in the following 4 weeks. In the ICES group, lacrimal gland proinflammatory cytokine level increases were much lower than those in the SCOP group. In accord with the lower proinflammatory cytokine levels, in the ICES group, lacrimal gland cytosolic vesicular density and size exceeded that in the SCOP group. ICES and SCOP induced murine dry eye effects became progressively more severe over a two week period. Subsequently, the disease process stabilized for the next four weeks. ICES induced local effects in the ocular surface, but failed to elicit lacrimal gland inflammation and cytoarchitectural changes, which accounts for less dry eye severity in the ICES model than that in the SCOP model.
Collapse
Affiliation(s)
- Bing Xiao
- School of Ophthalmology and Optometry, Wenzhou Medical University, Zhejiang, China
| | - Yu Wang
- School of Ophthalmology and Optometry, Wenzhou Medical University, Zhejiang, China
| | - Peter S. Reinach
- School of Ophthalmology and Optometry, Wenzhou Medical University, Zhejiang, China
- Department of Biological Sciences, College of Optometry, State University of New York, New York, NY 10036, United States of America
| | - Yueping Ren
- School of Ophthalmology and Optometry, Wenzhou Medical University, Zhejiang, China
| | - Jinyang Li
- School of Ophthalmology and Optometry, Wenzhou Medical University, Zhejiang, China
| | | | - Huihui Lu
- School of Ophthalmology and Optometry, Wenzhou Medical University, Zhejiang, China
| | - Wei Chen
- School of Ophthalmology and Optometry, Wenzhou Medical University, Zhejiang, China
- * E-mail:
| |
Collapse
|
133
|
Shao Y, Cheng Z, Li X, Chernaya V, Wang H, Yang XF. Immunosuppressive/anti-inflammatory cytokines directly and indirectly inhibit endothelial dysfunction--a novel mechanism for maintaining vascular function. J Hematol Oncol 2014; 7:80. [PMID: 25387998 PMCID: PMC4236671 DOI: 10.1186/s13045-014-0080-6] [Citation(s) in RCA: 111] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2014] [Accepted: 10/13/2014] [Indexed: 12/14/2022] Open
Abstract
Endothelial dysfunction is a pathological status of the vascular system, which can be broadly defined as an imbalance between endothelium-dependent vasoconstriction and vasodilation. Endothelial dysfunction is a key event in the progression of many pathological processes including atherosclerosis, type II diabetes and hypertension. Previous reports have demonstrated that pro-inflammatory/immunoeffector cytokines significantly promote endothelial dysfunction while numerous novel anti-inflammatory/immunosuppressive cytokines have recently been identified such as interleukin (IL)-35. However, the effects of anti-inflammatory cytokines on endothelial dysfunction have received much less attention. In this analytical review, we focus on the recent progress attained in characterizing the direct and indirect effects of anti-inflammatory/immunosuppressive cytokines in the inhibition of endothelial dysfunction. Our analyses are not only limited to the importance of endothelial dysfunction in cardiovascular disease progression, but also expand into the molecular mechanisms and pathways underlying the inhibition of endothelial dysfunction by anti-inflammatory/immunosuppressive cytokines. Our review suggests that anti-inflammatory/immunosuppressive cytokines serve as novel therapeutic targets for inhibiting endothelial dysfunction, vascular inflammation and cardio- and cerebro-vascular diseases.
Collapse
Affiliation(s)
- Ying Shao
- Department of Pharmacology, Center for Metabolic Disease Research and Cardiovascular Research Center, Temple University School of Medicine, MERB 1059, 3500 North Broad Street, Philadelphia, PA, 19140, USA.
| | - Zhongjian Cheng
- Department of Pharmacology, Center for Metabolic Disease Research and Cardiovascular Research Center, Temple University School of Medicine, MERB 1059, 3500 North Broad Street, Philadelphia, PA, 19140, USA.
| | - Xinyuan Li
- Department of Pharmacology, Center for Metabolic Disease Research and Cardiovascular Research Center, Temple University School of Medicine, MERB 1059, 3500 North Broad Street, Philadelphia, PA, 19140, USA.
| | - Valeria Chernaya
- Department of Pharmacology, Center for Metabolic Disease Research and Cardiovascular Research Center, Temple University School of Medicine, MERB 1059, 3500 North Broad Street, Philadelphia, PA, 19140, USA.
| | - Hong Wang
- Department of Pharmacology, Center for Metabolic Disease Research and Cardiovascular Research Center, Temple University School of Medicine, MERB 1059, 3500 North Broad Street, Philadelphia, PA, 19140, USA.
| | - Xiao-feng Yang
- Department of Pharmacology, Center for Metabolic Disease Research and Cardiovascular Research Center, Temple University School of Medicine, MERB 1059, 3500 North Broad Street, Philadelphia, PA, 19140, USA. .,Department of Microbiology and Immunology, Temple University School of Medicine, Philadelphia, PA, 19140, USA.
| |
Collapse
|
134
|
Engwerda CR, Ng SS, Bunn PT. The Regulation of CD4(+) T Cell Responses during Protozoan Infections. Front Immunol 2014; 5:498. [PMID: 25352846 PMCID: PMC4195384 DOI: 10.3389/fimmu.2014.00498] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2014] [Accepted: 09/25/2014] [Indexed: 12/20/2022] Open
Abstract
CD4(+) T cells are critical for defense against protozoan parasites. Intracellular protozoan parasite infections generally require the development of a Th1 cell response, characterized by the production of IFNγ and TNF that are critical for the generation of microbicidal molecules by phagocytes, as well as the expression of cytokines and cell surface molecules needed to generate cytolytic CD8(+) T cells that can recognize and kill infected host cells. Over the past 25 years, much has been learnt about the molecular and cellular components necessary for the generation of Th1 cell responses, and it has become clear that these responses need to be tightly controlled to prevent disease. However, our understanding of the immunoregulatory mechanisms activated during infection is still not complete. Furthermore, it is apparent that although these mechanisms are critical to prevent inflammation, they can also promote parasite persistence and development of disease. Here, we review how CD4(+) T cells are controlled during protozoan infections and how these regulatory mechanisms can influence parasite growth and disease outcome.
Collapse
Affiliation(s)
| | - Susanna S. Ng
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
- School of Natural Sciences, Griffith University, Nathan, QLD, Australia
| | - Patrick T. Bunn
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
- Institute of Glycomics, Griffith University, Gold Coast, QLD, Australia
| |
Collapse
|
135
|
Lind A, Koenderman L, Kusters JG, Siersema PD. Squamous tissue lymphocytes in the esophagus of controls and patients with reflux esophagitis and Barrett's esophagus are characterized by a non-inflammatory phenotype. PLoS One 2014; 9:e106261. [PMID: 25170842 PMCID: PMC4149547 DOI: 10.1371/journal.pone.0106261] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2013] [Accepted: 08/03/2014] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND AND OBJECTIVE Reflux esophagitis (RE) is characterized by inflammation of the squamous epithelium (SQ) of the esophagus and may progress to Barrett's esophagus (BE) characterized by intestinal metaplasia. The role of inflammation in this transition has been postulated but lacks experimental evidence. Here, the inflammatory responses in the esophagus of these patients were investigated. PATIENTS AND METHODS Fifty-one esophageal biopsies from with patients BE (n = 19), RE (n = 8) and controls (n = 23) were analyzed. T-cells were analyzed before and after ex vivo expansion (14 days) by multicolor flow cytometric analysis. The following markers were studied: CD3, CD4, CD8 (T-cell markers), Granzyme B (marker of cytotoxicity), CD103 (αE/epithelial integrin) and NKg2a (inhibitory receptor on T-cells and NK-cells). RESULTS Analysis of ex vivo cultures from normal looking SQ from controls, RE patients, and BE patients revealed no significant differences in the number and phenotypes of T-cells. In contrast, tissue from RE was different to normal SQ in four aspects: 1) higher percentages of CD3+ CD4+-cells (72±7% vs 48±6%, p = 0.01) and 2) CD8+ GranzymeB+-cells (53±11% vs 26±4%, p<0.05), while 3) lower percentages of CD4+ CD103+-cells (45±19% vs 80±3%, p = 0.02) and 4) CD8+ NKg2a+-cells (31±12% vs 44±5%). CONCLUSION Despite the fact that both tissues are exposed to the same reflux associated inflammatory triggers, the immune response observed in RE is clearly distinct from that in SQ of BE. The differences in immune responses in BE tissue might contribute to its susceptibility for transformation into intestinal metaplasia.
Collapse
Affiliation(s)
- Alexandra Lind
- Department of Respiratory Medicine, University Medical Center Utrecht, Utrecht, the Netherlands
- Department of Gastroenterology and Hepatology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Leo Koenderman
- Department of Respiratory Medicine, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Johannes G. Kusters
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Peter D. Siersema
- Department of Gastroenterology and Hepatology, University Medical Center Utrecht, Utrecht, the Netherlands
- * E-mail:
| |
Collapse
|
136
|
Tu E, Chia PZC, Chen W. TGFβ in T cell biology and tumor immunity: Angel or devil? Cytokine Growth Factor Rev 2014; 25:423-35. [PMID: 25156420 DOI: 10.1016/j.cytogfr.2014.07.014] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
The evolutionally conserved transforming growth factor β (TGFβ) affects multiple cell types in the immune system by either stimulating or inhibiting their differentiation and function. Studies using transgenic mice with ablation of TGFβ or its receptor have revealed the biological significance of TGFβ signaling in the control of T cells. However, it is now clear that TGFβ is more than an immunosuppressive cytokine. Disruption of TGFβ signaling pathway also leads to impaired generation of certain T cell populations. Therefore, in the normal physiological state, TGFβ actively maintains T cell homeostasis and regulates T cell function. However, in the tumor microenvironment, TGFβ creates an immunosuppressive milieu that inhibits antitumor immunity. Here, we review recent advances in our understanding of the roles of TGFβ in the regulation of T cells and tumor immunity.
Collapse
Affiliation(s)
- Eric Tu
- Mucosal Immunology Section, OPCB, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892, USA
| | - Pei Zhi Cheryl Chia
- Mucosal Immunology Section, OPCB, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892, USA
| | - Wanjun Chen
- Mucosal Immunology Section, OPCB, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
137
|
Tan KKB, Salgado G, Connolly JE, Chan JKY, Lane EB. Characterization of fetal keratinocytes, showing enhanced stem cell-like properties: a potential source of cells for skin reconstruction. Stem Cell Reports 2014; 3:324-38. [PMID: 25254345 PMCID: PMC4175556 DOI: 10.1016/j.stemcr.2014.06.005] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2013] [Revised: 06/05/2014] [Accepted: 06/06/2014] [Indexed: 01/18/2023] Open
Abstract
Epidermal stem cells have been in clinical application as a source of culture-generated grafts. Although applications for such cells are increasing due to aging populations and the greater incidence of diabetes, current keratinocyte grafting technology is limited by immunological barriers and the time needed for culture amplification. We studied the feasibility of using human fetal skin cells for allogeneic transplantation and showed that fetal keratinocytes have faster expansion times, longer telomeres, lower immunogenicity indicators, and greater clonogenicity with more stem cell indicators than adult keratinocytes. The fetal cells did not induce proliferation of T cells in coculture and were able to suppress the proliferation of stimulated T cells. Nevertheless, fetal keratinocytes could stratify normally in vitro. Experimental transplantation of fetal keratinocytes in vivo seeded on an engineered plasma scaffold yielded a well-stratified epidermal architecture and showed stable skin regeneration. These results support the possibility of using fetal skin cells for cell-based therapeutic grafting. Properties of fetal and adult keratinocytes are compared in tissue culture and grafts Fetal skin cells can be engrafted and show stable human-to-mouse skin regeneration Fetal keratinocytes are stem cell rich and need no differentiation before grafting Fetal keratinocytes are able to suppress proliferation of stimulated T cells in vitro
Collapse
Affiliation(s)
- Kenneth K B Tan
- A(∗)STAR Institute of Medical Biology, Immunos, Singapore 138648, Singapore; NUS Graduate School for Integrative Sciences and Engineering, Centre for Life Sciences, Singapore 117597, Singapore
| | - Giorgiana Salgado
- A(∗)STAR Institute of Medical Biology, Immunos, Singapore 138648, Singapore
| | - John E Connolly
- Singapore Immunology Network, A(∗)STAR, Immunos, Singapore 138648, Singapore
| | - Jerry K Y Chan
- Department of Reproductive Medicine, KK Women's and Children's Hospital, Singapore 229899, Singapore; Cancer and Stem Cell Biology Program, Duke-NUS Graduate Medical School, Singapore 169857, Singapore; Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, Singapore 119228, Singapore.
| | - E Birgitte Lane
- A(∗)STAR Institute of Medical Biology, Immunos, Singapore 138648, Singapore.
| |
Collapse
|
138
|
Abstract
Conventional therapy for malignant glioma (MG) fails to specifically eliminate tumor cells, resulting in toxicity that limits therapeutic efficacy. In contrast, antibody-based immunotherapy uses the immune system to eliminate tumor cells with exquisite specificity. Increased understanding of the pathobiology of MG and the profound immunosuppression present among patients with MG has revealed several biologic targets amenable to antibody-based immunotherapy. Novel antibody engineering techniques allow for the production of fully human antibodies or antibody fragments with vastly reduced antigen-binding dissociation constants, increasing safety when used clinically as therapeutics. In this report, we summarize the use of antibody-based immunotherapy for MG. Approaches currently under investigation include the use of antibodies or antibody fragments to: (1) redirect immune effector cells to target tumor mutations, (2) inhibit immunosuppressive signals and thereby stimulate an immunological response against tumor cells, and (3) provide costimulatory signals to evoke immunologic targeting of tumor cells. These approaches demonstrate highly compelling safety and efficacy for the treatment of MG, providing a viable adjunct to current standard-of-care therapy for MG.
Collapse
Affiliation(s)
- Patrick C Gedeon
- Duke Brain Tumor Immunotherapy Program, Division of Neurosurgery, Department of Surgery, Duke University Medical Center, Durham, NC; Department of Biomedical Engineering, Duke University, Durham, NC; The Preston Robert Tisch Brain Tumor Center, Duke University Medical Center, Durham, NC.
| | - Katherine A Riccione
- Duke Brain Tumor Immunotherapy Program, Division of Neurosurgery, Department of Surgery, Duke University Medical Center, Durham, NC; Department of Biomedical Engineering, Duke University, Durham, NC; The Preston Robert Tisch Brain Tumor Center, Duke University Medical Center, Durham, NC
| | - Peter E Fecci
- Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - John H Sampson
- Duke Brain Tumor Immunotherapy Program, Division of Neurosurgery, Department of Surgery, Duke University Medical Center, Durham, NC; Department of Biomedical Engineering, Duke University, Durham, NC; The Preston Robert Tisch Brain Tumor Center, Duke University Medical Center, Durham, NC
| |
Collapse
|
139
|
Li X, Shen L, Tan H. Polymorphisms and plasma level of transforming growth factor-Beta 1 and risk for preeclampsia: a systematic review. PLoS One 2014; 9:e97230. [PMID: 24823830 PMCID: PMC4019528 DOI: 10.1371/journal.pone.0097230] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2014] [Accepted: 04/16/2014] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Transforming growth factor-beta 1 (TGF-β1) is thought to be involved in the pathogenesis of preeclampsia (PE), but the results are inconsistent among studies. This article aims to compile an overview of the studies about the associations of TGF-β 1 polymorphism and plasma level with PE risk and to provide recommendations for future research. METHODS AND RESULTS The databases PubMed, Embase and Web of Science were searched up to December 2013. Five studies investigating the associations of four polymorphisms with the risks of PE were involved. A meta-analysis was conducted for the 869T>C polymorphism and PE risk. The results show that genotype TT of 869T>C polymorphism is a protective factor of PE (pooled odds ratio=0.73, 95% CI: 0.56, 0.95). Eight case-control studies reported the plasma level of TGF-β 1. The substantial heterogeneity among studies may be attributed to the differences in the blood sample processing and the TGF-β 1 analysis kits. The results suggest that plasma TGF-β 1 level in the second trimester was significantly lower in the PE group than in the normal pregnancy group, but was significantly higher in the PE group during the third trimester. CONCLUSIONS The current results support that the TGF-β 1 869 T>C polymorphism was associated with the risk of PE. However, the number of eligible studies is small and more studies are needed to clarify whether this association can be detected on larger sample sizes and different populations. Owing to the heterogeneity between studies, no conclusion on the association between plasma TGF-β 1 level and PE risk can be drawn from this review. Further studies about the TGF-β 1 levels at different stages of pregnancy and the development of TGF-β 1 assay methodology are required to reveal the role of TGF-β 1 in the pathological development of PE.
Collapse
Affiliation(s)
- Xun Li
- Department of Epidemiology and Health Statistics, School of Public Health, Central South University, Changsha, Hunan, China
| | - Lin Shen
- Department of Epidemiology and Health Statistics, School of Public Health, Central South University, Changsha, Hunan, China
| | - Hongzhuan Tan
- Department of Epidemiology and Health Statistics, School of Public Health, Central South University, Changsha, Hunan, China
- * E-mail:
| |
Collapse
|
140
|
Papel del factor de crecimiento transformador-beta (TGF-β) en la fisiopatología de la artritis reumatoide. ACTA ACUST UNITED AC 2014; 10:174-9. [DOI: 10.1016/j.reuma.2014.01.009] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2013] [Revised: 01/11/2014] [Accepted: 01/21/2014] [Indexed: 11/24/2022]
|
141
|
Gonzalo-Gil E, Galindo-Izquierdo M. Role of Transforming Growth Factor-Beta (TGF) Beta in the Physiopathology of Rheumatoid Arthritis. ACTA ACUST UNITED AC 2014. [DOI: 10.1016/j.reumae.2014.01.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
142
|
Li MO. Close bonding: the first report of TGF-β and T cells. THE JOURNAL OF IMMUNOLOGY 2014; 192:2937-8. [PMID: 24659786 DOI: 10.4049/jimmunol.1400405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Affiliation(s)
- Ming O Li
- Immunology Program, Memorial Sloan-Kettering Cancer Center, New York, NY 10065
| |
Collapse
|
143
|
The role(s) of cytokines/chemokines in urinary bladder inflammation and dysfunction. BIOMED RESEARCH INTERNATIONAL 2014; 2014:120525. [PMID: 24738044 PMCID: PMC3971501 DOI: 10.1155/2014/120525] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/13/2013] [Revised: 01/26/2014] [Accepted: 02/05/2014] [Indexed: 12/17/2022]
Abstract
Bladder pain syndrome (BPS)/interstitial cystitis (IC) is a chronic pain syndrome characterized by pain, pressure, or discomfort perceived to be bladder related and with at least one urinary symptom. It was recently concluded that 3.3-7.9 million women (>18 years old) in the United States exhibit BPS/IC symptoms. The impact of BPS/IC on quality of life is enormous and the economic burden is significant. Although the etiology and pathogenesis of BPS/IC are unknown, numerous theories including infection, inflammation, autoimmune disorder, toxic urinary agents, urothelial dysfunction, and neurogenic causes have been proposed. Altered visceral sensations from the urinary bladder (i.e., pain at low or moderate bladder filling) that accompany BPS/IC may be mediated by many factors including changes in the properties of peripheral bladder afferent pathways such that bladder afferent neurons respond in an exaggerated manner to normally innocuous stimuli (allodynia). The goals for this review are to describe chemokine/receptor (CXCL12/CXCR4; CCL2/CCR2) signaling and cytokine/receptor (transforming growth factor (TGF-β)/TGF-β type 1 receptor) signaling that may be valuable LUT targets for pharmacologic therapy to improve urinary bladder function and reduce somatic sensitivity associated with urinary bladder inflammation.
Collapse
|
144
|
Tidball JG, Dorshkind K, Wehling-Henricks M. Shared signaling systems in myeloid cell-mediated muscle regeneration. Development 2014; 141:1184-96. [PMID: 24595286 PMCID: PMC3943178 DOI: 10.1242/dev.098285] [Citation(s) in RCA: 101] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Much of the focus in muscle regeneration has been placed on the identification and delivery of stem cells to promote regenerative capacity. As those efforts have advanced, we have learned that complex features of the microenvironment in which regeneration occurs can determine success or failure. The immune system is an important contributor to that complexity and can determine the extent to which muscle regeneration succeeds. Immune cells of the myeloid lineage play major regulatory roles in tissue regeneration through two general, inductive mechanisms: instructive mechanisms that act directly on muscle cells; and permissive mechanisms that act indirectly to influence regeneration by modulating angiogenesis and fibrosis. In this article, recent discoveries that identify inductive actions of specific populations of myeloid cells on muscle regeneration are presented, with an emphasis on how processes in muscle and myeloid cells are co-regulated.
Collapse
Affiliation(s)
- James G. Tidball
- Molecular, Cellular & Integrative Physiology Program, University of California, Los Angeles, CA 90095-1606, USA
- Department of Integrative Biology and Physiology, University of California, Los Angeles, CA 90095-1606, USA
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at UCLA, University of California, Los Angeles, CA 90095-1732, USA
| | - Kenneth Dorshkind
- Molecular, Cellular & Integrative Physiology Program, University of California, Los Angeles, CA 90095-1606, USA
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at UCLA, University of California, Los Angeles, CA 90095-1732, USA
| | - Michelle Wehling-Henricks
- Department of Integrative Biology and Physiology, University of California, Los Angeles, CA 90095-1606, USA
| |
Collapse
|
145
|
TGF-β upregulates CD70 expression and induces exhaustion of effector memory T cells in B-cell non-Hodgkin's lymphoma. Leukemia 2014; 28:1872-84. [PMID: 24569779 DOI: 10.1038/leu.2014.84] [Citation(s) in RCA: 112] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2013] [Revised: 01/17/2014] [Accepted: 02/03/2014] [Indexed: 12/16/2022]
Abstract
Transforming growth factor beta (TGF-β) has an important role in mediating T-cell suppression in B-cell non-Hodgkin lymphoma (NHL). However, the underlying mechanism responsible for TGF-β-mediated inhibition of effector memory T (Tm) cells is largely unknown. As reported here, we show that exhaustion is a major mechanism by which TGF-β inhibits Tm cells, and TGF-β mediated exhaustion is associated with upregulation of CD70. We found that TGF-β upregulates CD70 expression on effector Tm cells while it preferentially induces Foxp3 expression in naive T cells. CD70 induction by TGF-β is Smad3-dependent and involves IL-2/Stat5 signaling. CD70+ T cells account for TGF-β-induced exhaustion of effector Tm cells. Both TGF-β-induced and preexisting intratumoral CD70+ effector Tm cells from B-cell NHL have an exhausted phenotype and express higher levels of PD-1 and TIM-3 compared with CD70- T cells. Signaling transduction, proliferation and cytokine production are profoundly decreased in these cells, and they are highly susceptible to apoptosis. Clinically, intratumoral CD70-expressing T cells are prevalent in follicular B-cell lymphoma (FL) biopsy specimens, and increased numbers of intratumoral CD70+ T cells correlate with an inferior patient outcome. These findings confirm TGF-β-mediated effector Tm cell exhaustion as an important mechanism of immune suppression in B-cell NHL.
Collapse
|
146
|
Li MO, Flavell RA. TGF-β, T-cell tolerance and immunotherapy of autoimmune diseases and cancer. Expert Rev Clin Immunol 2014; 2:257-65. [DOI: 10.1586/1744666x.2.2.257] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
147
|
Stromnes IM, Schmitt TM, Chapuis AG, Hingorani SR, Greenberg PD. Re-adapting T cells for cancer therapy: from mouse models to clinical trials. Immunol Rev 2014; 257:145-64. [PMID: 24329795 PMCID: PMC4015625 DOI: 10.1111/imr.12141] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Adoptive T-cell therapy involves the isolation, expansion, and reinfusion of T lymphocytes with a defined specificity and function as a means to eradicate cancer. Our research has focused on specifying the requirements for tumor eradication with antigen-specific T cells and T cells transduced to express a defined T-cell receptor (TCR) in mouse models and then translating these strategies to clinical trials. Our design of T-cell-based therapy for cancer has reflected efforts to identify the obstacles that limit sustained effector T-cell activity in mice and humans, design approaches to enhance T-cell persistence, develop methods to increase TCR affinity/T-cell functional avidity, and pursue strategies to overcome tolerance and immunosuppression. With the advent of genetic engineering, a highly functional population of T cells can now be rapidly generated and tailored for the targeted malignancy. Preclinical studies in faithful and informative mouse models, in concert with knowledge gained from analyses of successes and limitations in clinical trials, are shaping how we continue to develop, refine, and broaden the applicability of this approach for cancer therapy.
Collapse
Affiliation(s)
- Ingunn M. Stromnes
- Clinical Research Division, Program in Immunology, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
- Department of Immunology, University of Washington, Seattle, WA, USA
| | - Thomas M. Schmitt
- Clinical Research Division, Program in Immunology, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Aude G. Chapuis
- Clinical Research Division, Program in Immunology, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Sunil R. Hingorani
- Clinical Research Division and Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Philip D. Greenberg
- Clinical Research Division, Program in Immunology, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
- Department of Immunology, University of Washington, Seattle, WA, USA
- Department of Medicine, Division of Medical Oncology, University of Washington School of Medicine, Seattle, WA, USA
| |
Collapse
|
148
|
Lee YH, Schiemann WP. Chemotherapeutic Targeting of the Transforming Growth Factor-β Pathway in Breast Cancers. BREAST CANCER MANAGEMENT 2014; 3:73-85. [PMID: 25904986 DOI: 10.2217/bmt.13.74] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Transforming growth factor (TGF-β) is a multifunctional cytokine that plays essential roles in regulating mammary gland development, morphogenesis, differentiation, and involution. TGF-β also regulates mammary gland homeostasis and prevents its transformation by prohibiting dysregulated cell cycle progression, and by inducing apoptosis; it also creates cell microenvironments that readily inhibit cell migration, invasion, and metastasis. Interestingly, while early-stage mammary tumors remain sensitive to the tumor suppressing activities of TGF-β, late-stage breast cancers become insensitive to the anticancer functions of this cytokine and instead rely upon TGF-β to drive disease and metastatic progression. This switch in TGF-β function is known as the "TGF-β Paradox" and represents the rationale for developing chemotherapies to inactivate the TGF-β pathway and its oncogenic functions in late-stage breast cancers. Here we outline the molecular mechanisms that manifest the "TGF-β Paradox" and discuss the challenges associated with the development and use of anti-TGF-β agents to treat breast cancer patients.
Collapse
Affiliation(s)
- Yong-Hun Lee
- Case Comprehensive Cancer Center, Division of General Medical Sciences-Oncology, Case Western Reserve University, Wolstein Research Building, 2103 Cornell Road Cleveland, OH 44106
| | - William P Schiemann
- Case Comprehensive Cancer Center, Division of General Medical Sciences-Oncology, Case Western Reserve University, Wolstein Research Building, 2103 Cornell Road Cleveland, OH 44106
| |
Collapse
|
149
|
Sang LX, Chang B, Dai C, Gao N, Liu WX, Jiang M. Heat-killed VSL#3 ameliorates dextran sulfate sodium (DSS)-induced acute experimental colitis in rats. Int J Mol Sci 2013; 15:15-28. [PMID: 24451125 PMCID: PMC3907795 DOI: 10.3390/ijms15010015] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2013] [Revised: 12/09/2013] [Accepted: 12/10/2013] [Indexed: 12/12/2022] Open
Abstract
To determine the effects of heat-killed VSL#3 (B. breve, B. longum and B. infantis; L. plantarum, L. bulgaricus, L. casei and L. acidophilus; S. salivarius subsp. thermophilus) therapy in the dextran sulfate sodium (DSS)-induced acute experimental colitis in rats. Acute experimental colitis was induced in rats by 5% DSS and freely drink for seven days. Beginning on Day 8, rats underwent gavage once daily for seven days with heat-killed probiotic VSL#3 (0.6 g/kg/day), colonic damage was evaluated histologically and biochemically seven days after gavage. Expression of inflammatory related mediators (STAT3, P-STAT3) and cytokines (IL-6, IL-23, TGFβ) in colonic tissue were detected. The results revealed that heat-killed and live VSL#3 have identical anti-inflammatory properties by the assessed DAI (disease activity index), colon length, histological tissue and MPO activity. Heat-killed and live VSL#3 results in reduced IL-6, IL-23, TGFβ, STAT3 and P-STAT3 expression in colonic tissue. Heat-killed and live VSL#3 have showed the similar anti-inflammatory activity by inhibiting IL-6/STAT3 pathway in the DSS-induced acute experimental colitis in rats.
Collapse
Affiliation(s)
- Li-Xuan Sang
- Department of Cadre Ward II, First Affiliated Hospital of China Medical University, Shenyang 110001, Liaoning, China.
| | - Bing Chang
- Department of Gastroenterology, First Affiliated Hospital of China Medical University, Shenyang 110001, Liaoning, China.
| | - Cong Dai
- Department of Gastroenterology, First Affiliated Hospital of China Medical University, Shenyang 110001, Liaoning, China.
| | - Nan Gao
- Department of Cadre Ward II, First Affiliated Hospital of China Medical University, Shenyang 110001, Liaoning, China.
| | - Wei-Xin Liu
- Department of Gastroenterology, First Affiliated Hospital of China Medical University, Shenyang 110001, Liaoning, China.
| | - Min Jiang
- Department of Gastroenterology, First Affiliated Hospital of China Medical University, Shenyang 110001, Liaoning, China.
| |
Collapse
|
150
|
Talaat RM, Dondeti MF, El-Shenawy SZ, Khamiss OA. Transforming Growth Factor- β 1 Gene Polymorphism (T29C) in Egyptian Patients with Hepatitis B Virus Infection: A Preliminary Study. HEPATITIS RESEARCH AND TREATMENT 2013; 2013:293274. [PMID: 24455227 PMCID: PMC3878635 DOI: 10.1155/2013/293274] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/07/2013] [Revised: 10/15/2013] [Accepted: 10/29/2013] [Indexed: 12/16/2022]
Abstract
The interindividual variations in the capacity of transforming growth factor- β 1 (TGF- β 1) production have been ascribed to genetic polymorphisms in TGF- β 1 gene. As pathogenesis of HBV has a genetic background, this preliminary study was designed to assess the impact of TGF- β 1 (T29C) on the susceptibility of Egyptians to HBV infection. Genotyping was performed using single stranded polymorphism-polymerase chain reaction (SSP-PCR) in 65 Egyptian hepatitis B patients and 50 healthy controls. TGF- β 1 plasma levels were measured using Enzyme-linked immunosorbent assay (ELISA). The frequency of CC genotype was significantly higher (P < 0.05) in HBV patients compared to controls. On the contrary, TC genotype did not show significant difference in both groups. TT genotype was significantly higher (P < 0.01) in controls than HBV patients. Our current preliminary data revealed that the frequency of the genotypes in the controls were within Hardy-Weinberg equilibrium (HWE) while the patients group was out of HWE (P < 0.01). TGF- β 1 was significantly (r = -0.684; P < 0.001) deceased in the sera of patients as compared to normal subjects. Depending on our preliminary work, CC genotype may act as a host genetic factor in the susceptibility to HBV infection in Egyptians. Taken together, the current data pointed to the importance of polymorphism of TGF- β 1 gene (T29C) in HBV infection.
Collapse
Affiliation(s)
- Roba M. Talaat
- Molecular Biology Department, Genetic Engineering and Biotechnology Research Institute (GEBRI), University of Sadat City, Sadat City 22857, Egypt
| | - Mahmoud F. Dondeti
- Molecular Biology Department, Genetic Engineering and Biotechnology Research Institute (GEBRI), University of Sadat City, Sadat City 22857, Egypt
| | - Soha Z. El-Shenawy
- Biochemistry Department, National Liver Institute (NLI), Menoufiya University, Shebeen El-Kom, Menoufiya 32511, Egypt
| | - Omaima A. Khamiss
- Animal Biotechnology Department, Genetic Engineering and Biotechnology Research Institute (GEBRI), University of Sadat City, Sadat City 22857, Egypt
| |
Collapse
|