1
|
Montgomery A, Tam F, Gursche C, Cheneval C, Besler K, Enns W, Manku S, Rey K, Hanson PJ, Rose-John S, McManus BM, Choy JC. Overlapping and distinct biological effects of IL-6 classic and trans-signaling in vascular endothelial cells. Am J Physiol Cell Physiol 2021; 320:C554-C565. [PMID: 33471622 DOI: 10.1152/ajpcell.00323.2020] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 12/31/2020] [Indexed: 02/08/2023]
Abstract
IL-6 affects tissue protective/reparative and inflammatory properties of vascular endothelial cells (ECs). This cytokine can signal to cells through classic and trans-signaling mechanisms, which are differentiated based on the expression of IL-6 receptor (IL-6R) on the surface of target cells. The biological effects of these IL-6-signaling mechanisms are distinct and have implications for vascular pathologies. We have directly compared IL-6 classic and trans-signaling in ECs. Human ECs expressed IL-6R in culture and in situ in coronary arteries from heart transplants. Stimulation of human ECs with IL-6, to model classic signaling, triggered the activation of phosphatidylinositol 3-kinase (PI3K)-Akt and ERK1/2 signaling pathways, whereas stimulation with IL-6 + sIL-6R, to model trans-signaling, triggered activation of STAT3, PI3K-Akt, and ERK1/2 pathways. IL-6 classic signaling reduced persistent injury of ECs in an allograft model of vascular rejection and inhibited cell death induced by growth factor withdrawal. When inflammatory effects were examined, IL-6 classic signaling did not induce ICAM or CCL2 expression but was sufficient to induce secretion of CXCL8 and support transmigration of neutrophil-like cells. IL-6 trans-signaling induced all inflammatory effects studied. Our findings show that IL-6 classic and trans-signaling have overlapping but distinct properties in controlling EC survival and inflammatory activation. This has implications for understanding the effects of IL-6 receptor-blocking therapies as well as for vascular responses in inflammatory and immune conditions.
Collapse
MESH Headings
- Adult
- Aged
- Animals
- Aorta, Abdominal/drug effects
- Aorta, Abdominal/metabolism
- Aorta, Abdominal/pathology
- Aorta, Abdominal/transplantation
- Cells, Cultured
- Cytokine Receptor gp130/agonists
- Cytokine Receptor gp130/metabolism
- Disease Models, Animal
- Endothelial Cells/drug effects
- Endothelial Cells/metabolism
- Endothelial Cells/pathology
- Endothelial Cells/transplantation
- Female
- Graft Rejection/metabolism
- Graft Rejection/pathology
- Graft Rejection/prevention & control
- Humans
- Inflammation Mediators/metabolism
- Interleukin-6/pharmacology
- Male
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Middle Aged
- Receptors, Interleukin-6/agonists
- Receptors, Interleukin-6/metabolism
- Signal Transduction
- Mice
Collapse
Affiliation(s)
- Ashani Montgomery
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Franklin Tam
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Chris Gursche
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Catherine Cheneval
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Katrina Besler
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Winnie Enns
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Sukhkbir Manku
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Kevin Rey
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Paul J Hanson
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
- Centre for Heart and Lung Innovation, St. Paul's Hospital, Vancouver, British Columbia, Canada
| | - Stefan Rose-John
- Institute of Biochemistry, Christian-Albrechts University Kiel, Kiel, Germany
| | - Bruce M McManus
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
- Centre for Heart and Lung Innovation, St. Paul's Hospital, Vancouver, British Columbia, Canada
| | - Jonathan C Choy
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, British Columbia, Canada
| |
Collapse
|
2
|
Sims NA. The JAK1/STAT3/SOCS3 axis in bone development, physiology, and pathology. Exp Mol Med 2020; 52:1185-1197. [PMID: 32788655 PMCID: PMC8080635 DOI: 10.1038/s12276-020-0445-6] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 04/03/2020] [Accepted: 04/20/2020] [Indexed: 12/15/2022] Open
Abstract
Bone growth and the maintenance of bone structure are controlled by multiple endocrine and paracrine factors, including cytokines expressed locally within the bone microenvironment and those that are elevated, both locally and systemically, under inflammatory conditions. This review focuses on those bone-active cytokines that initiate JAK–STAT signaling, and outlines the discoveries made from studying skeletal defects caused by induced or spontaneous modifications in this pathway. Specifically, this review describes defects in JAK1, STAT3, and SOCS3 signaling in mouse models and in humans, including mutations designed to modify these pathways downstream of the gp130 coreceptor. It is shown that osteoclast formation is generally stimulated indirectly by these pathways through JAK1 and STAT3 actions in inflammatory and other accessory cells, including osteoblasts. In addition, in bone remodeling, osteoblast differentiation is increased secondary to stimulated osteoclast formation through an IL-6-dependent pathway. In growth plate chondrocytes, STAT3 signaling promotes the normal differentiation process that leads to bone lengthening. Within the osteoblast lineage, STAT3 signaling promotes bone formation in normal physiology and in response to mechanical loading through direct signaling in osteocytes. This activity, particularly that of the IL-6/gp130 family of cytokines, must be suppressed by SOCS3 for the normal formation of cortical bone. Maintaining normal bone structure and strength depends on a group of signaling proteins called cytokines that bind to receptor molecules on cell surfaces. Natalie Sims at St. Vincent’s Institute of Medical Research and The University of Melbourne in Australia reviews the role of cytokines in a specific signaling pathway in bone development and disease. Two of the proteins in this pathway respond to cytokine activity, whereas the third inhibits the cytokines’ effects. Studies in mice and humans have identified links between specific bone defects and spontaneous or experimentally induced mutations in the genes that code for the three proteins. The review covers the significance of recent findings to several types of cells that form new bone, degrade bone as part of normal bone turnover, and sustain the structure of bone and cartilage.
Collapse
Affiliation(s)
- Natalie A Sims
- St. Vincent's Institute of Medical Research, and Department of Medicine at St. Vincent's Hospital, The University of Melbourne, Parkville, VIC, Australia.
| |
Collapse
|
3
|
Béziat V, Tavernier SJ, Chen YH, Ma CS, Materna M, Laurence A, Staal J, Aschenbrenner D, Roels L, Worley L, Claes K, Gartner L, Kohn LA, De Bruyne M, Schmitz-Abe K, Charbonnier LM, Keles S, Nammour J, Vladikine N, Maglorius Renkilaraj MRL, Seeleuthner Y, Migaud M, Rosain J, Jeljeli M, Boisson B, Van Braeckel E, Rosenfeld JA, Dai H, Burrage LC, Murdock DR, Lambrecht BN, Avettand-Fenoel V, Vogel TP, Esther CR, Haskologlu S, Dogu F, Ciznar P, Boutboul D, Ouachée-Chardin M, Amourette J, Lebras MN, Gauvain C, Tcherakian C, Ikinciogullari A, Beyaert R, Abel L, Milner JD, Grimbacher B, Couderc LJ, Butte MJ, Freeman AF, Catherinot É, Fieschi C, Chatila TA, Tangye SG, Uhlig HH, Haerynck F, Casanova JL, Puel A. Dominant-negative mutations in human IL6ST underlie hyper-IgE syndrome. J Exp Med 2020; 217:e20191804. [PMID: 32207811 PMCID: PMC7971136 DOI: 10.1084/jem.20191804] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Revised: 01/14/2020] [Accepted: 02/18/2020] [Indexed: 11/29/2022] Open
Abstract
Autosomal dominant hyper-IgE syndrome (AD-HIES) is typically caused by dominant-negative (DN) STAT3 mutations. Patients suffer from cold staphylococcal lesions and mucocutaneous candidiasis, severe allergy, and skeletal abnormalities. We report 12 patients from 8 unrelated kindreds with AD-HIES due to DN IL6ST mutations. We identified seven different truncating mutations, one of which was recurrent. The mutant alleles encode GP130 receptors bearing the transmembrane domain but lacking both the recycling motif and all four STAT3-recruiting tyrosine residues. Upon overexpression, the mutant proteins accumulate at the cell surface and are loss of function and DN for cellular responses to IL-6, IL-11, LIF, and OSM. Moreover, the patients' heterozygous leukocytes and fibroblasts respond poorly to IL-6 and IL-11. Consistently, patients with STAT3 and IL6ST mutations display infectious and allergic manifestations of IL-6R deficiency, and some of the skeletal abnormalities of IL-11R deficiency. DN STAT3 and IL6ST mutations thus appear to underlie clinical phenocopies through impairment of the IL-6 and IL-11 response pathways.
Collapse
Affiliation(s)
- Vivien Béziat
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Institut National de la Santé et de la Recherche Médicale (INSERM) U1163, Paris, France
- University of Paris, Imagine Institute, Paris, France
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY
| | - Simon J. Tavernier
- Primary Immune Deficiency Research Laboratory, Department of Internal Diseases and Pediatrics, Centre for Primary Immunodeficiency Ghent, Jeffrey Modell Diagnosis and Research Centre, Ghent University Hospital, Ghent, Belgium
- VIB-UGent Center for Inflammation Research, Unit of Molecular Signal Transduction in Inflammation, Ghent, Belgium
| | - Yin-Huai Chen
- Translational Gastroenterology Unit, John Radcliffe Hospital, University of Oxford, Oxford, UK
- Department of Paediatrics, University of Oxford, Oxford, UK
| | - Cindy S. Ma
- Immunology Division, Garvan Institute of Medical Research, Darlinghurst, Sydney, New South Wales, Australia
- St. Vincent's Clinical School, UNSW Sydney, Sydney, New South Wales, Australia
| | - Marie Materna
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Institut National de la Santé et de la Recherche Médicale (INSERM) U1163, Paris, France
- University of Paris, Imagine Institute, Paris, France
| | - Arian Laurence
- Translational Gastroenterology Unit, John Radcliffe Hospital, University of Oxford, Oxford, UK
- Department of Paediatrics, University of Oxford, Oxford, UK
| | - Jens Staal
- VIB-UGent Center for Inflammation Research, Unit of Molecular Signal Transduction in Inflammation, Ghent, Belgium
| | - Dominik Aschenbrenner
- Translational Gastroenterology Unit, John Radcliffe Hospital, University of Oxford, Oxford, UK
- Department of Paediatrics, University of Oxford, Oxford, UK
| | - Lisa Roels
- Primary Immune Deficiency Research Laboratory, Department of Internal Diseases and Pediatrics, Centre for Primary Immunodeficiency Ghent, Jeffrey Modell Diagnosis and Research Centre, Ghent University Hospital, Ghent, Belgium
| | - Lisa Worley
- Immunology Division, Garvan Institute of Medical Research, Darlinghurst, Sydney, New South Wales, Australia
- St. Vincent's Clinical School, UNSW Sydney, Sydney, New South Wales, Australia
| | - Kathleen Claes
- Center for Medical Genetics, Ghent University Hospital, Ghent, Belgium
| | - Lisa Gartner
- Translational Gastroenterology Unit, John Radcliffe Hospital, University of Oxford, Oxford, UK
- Department of Paediatrics, University of Oxford, Oxford, UK
| | - Lisa A. Kohn
- Division of Immunology, Allergy, and Rheumatology, Department of Pediatrics, University of California, Los Angeles, Los Angeles, CA
| | - Marieke De Bruyne
- Center for Medical Genetics, Ghent University Hospital, Ghent, Belgium
| | - Klaus Schmitz-Abe
- Division of Newborn Medicine and Neonatal Genomics Program, Boston Children's Hospital, Harvard Medical School, Boston, MA
- Division of Genetics and Genomics, Boston Children's Hospital, Harvard Medical School, Boston, MA
- The Manton Center for Orphan Disease Research, Boston Children's Hospital, Harvard Medical School, Boston, MA
| | - Louis-Marie Charbonnier
- Department of Pediatrics, Harvard Medical School, Boston, MA
- Division of Immunology, Boston Children’s Hospital, Boston, MA
| | - Sevgi Keles
- Necmettin Erbakan University, Meram Medical Faculty, Division of Pediatric Allergy and Immunology, Konya, Turkey
| | - Justine Nammour
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Institut National de la Santé et de la Recherche Médicale (INSERM) U1163, Paris, France
- University of Paris, Imagine Institute, Paris, France
| | - Natasha Vladikine
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Institut National de la Santé et de la Recherche Médicale (INSERM) U1163, Paris, France
- University of Paris, Imagine Institute, Paris, France
| | - Majistor Raj Luxman Maglorius Renkilaraj
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Institut National de la Santé et de la Recherche Médicale (INSERM) U1163, Paris, France
- University of Paris, Imagine Institute, Paris, France
| | - Yoann Seeleuthner
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Institut National de la Santé et de la Recherche Médicale (INSERM) U1163, Paris, France
- University of Paris, Imagine Institute, Paris, France
| | - Mélanie Migaud
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Institut National de la Santé et de la Recherche Médicale (INSERM) U1163, Paris, France
- University of Paris, Imagine Institute, Paris, France
| | - Jérémie Rosain
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Institut National de la Santé et de la Recherche Médicale (INSERM) U1163, Paris, France
- University of Paris, Imagine Institute, Paris, France
| | - Mohamed Jeljeli
- Cochin University Hospital, Biological Immunology Unit, Assistance Publique Hôpitaux de Paris (AP-HP), Paris, France
| | - Bertrand Boisson
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Institut National de la Santé et de la Recherche Médicale (INSERM) U1163, Paris, France
- University of Paris, Imagine Institute, Paris, France
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY
| | - Eva Van Braeckel
- Department of Respiratory Medicine, Ghent University Hospital, Ghent Belgium
| | - Jill A. Rosenfeld
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX
| | - Hongzheng Dai
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX
| | - Lindsay C. Burrage
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX
| | - David R. Murdock
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX
| | - Bart N. Lambrecht
- VIB-UGent Center for Inflammation Research, Unit of Immunoregulation and Mucosal Immunology, Ghent, Belgium
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
| | - Véronique Avettand-Fenoel
- Laboratory of Clinical Microbiology, Virology Unit, Necker Hospital for Sick Children, AP-HP, Paris, France
| | - Tiphanie P. Vogel
- Division of Rheumatology, Department of Pediatrics, Baylor College of Medicine and Texas Children's Hospital, Houston, TX
| | | | - Charles R. Esther
- Pediatric Pulmonology, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Sule Haskologlu
- Division of Pediatric Immunology and Allergy, Ankara University School of Medicine, Sıhhıye, Ankara, Turkey
| | - Figen Dogu
- Division of Pediatric Immunology and Allergy, Ankara University School of Medicine, Sıhhıye, Ankara, Turkey
| | - Peter Ciznar
- Department of Pediatrics, Faculty of Medicine Comenius University and Children's University Hospital, Bratislava, Slovakia
| | - David Boutboul
- Clinical Immunology Department, Saint Louis Hospital, AP-HP de Paris University of Paris, Paris, France
| | - Marie Ouachée-Chardin
- Department of Pediatric Hematology and Immunology, Robert Debré Hospital, AP-HP, Paris, France
| | - Jean Amourette
- Pulmonology Department, Centre Hospitalier d'Arras, Arras, France
| | - Marie-Noëlle Lebras
- Pediatric Pulmonology, Infectious Disease and Internal Medicine Department, AP-HP, Robert Debré Hospital, Paris, France
| | - Clément Gauvain
- Thoracic Oncology Department, Lille University Hospital, Lille, France
| | | | - Aydan Ikinciogullari
- Division of Pediatric Immunology and Allergy, Ankara University School of Medicine, Sıhhıye, Ankara, Turkey
| | - Rudi Beyaert
- VIB-UGent Center for Inflammation Research, Unit of Molecular Signal Transduction in Inflammation, Ghent, Belgium
| | - Laurent Abel
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Institut National de la Santé et de la Recherche Médicale (INSERM) U1163, Paris, France
- University of Paris, Imagine Institute, Paris, France
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY
| | - Joshua D. Milner
- National Institute of Allergy and Infectious Diseases, Bethesda, MD
- Division of Allergy, Immunology and Rheumatology, Department of Pediatrics, Columbia University Irving Medical Center, New York, NY
| | - Bodo Grimbacher
- Institute for Immunodeficiency, Center for Chronic Immunodeficiency, Medical Center, Faculty of Medicine, Albert Ludwig University of Freiburg, Freiburg, Germany
- German Center for Infection Research, Satellite Center Freiburg, Freiburg, Germany
- Centre for Integrative Biological Signaling Studies, Albert Ludwig University, Freiburg, Germany
- RESIST, Cluster of Excellence 2155 to Hanover Medical School, Satellite Center Freiburg, Freiburg, Germany
- Institute of Immunity and Transplantation, Royal Free Hospital, University College London, London, UK
| | - Louis-Jean Couderc
- Hôpital Foch, Pulmonology Department, Suresnes, France
- Simone Veil Faculty of Life Sciences, Versailles-Paris Saclay University, UPRES EA-220, Suresnes, France
| | - Manish J. Butte
- Division of Immunology, Allergy, and Rheumatology, Department of Pediatrics, University of California, Los Angeles, Los Angeles, CA
| | | | | | - Claire Fieschi
- Clinical Immunology Department, Saint Louis Hospital, AP-HP de Paris University of Paris, Paris, France
- INSERM UMR1126, Institut de Recherche Saint-Louis, Université de Paris, Paris, France
| | - Talal A. Chatila
- Department of Pediatrics, Harvard Medical School, Boston, MA
- Division of Immunology, Boston Children’s Hospital, Boston, MA
| | - Stuart G. Tangye
- Immunology Division, Garvan Institute of Medical Research, Darlinghurst, Sydney, New South Wales, Australia
- St. Vincent's Clinical School, UNSW Sydney, Sydney, New South Wales, Australia
| | - Holm H. Uhlig
- Translational Gastroenterology Unit, John Radcliffe Hospital, University of Oxford, Oxford, UK
- Department of Paediatrics, University of Oxford, Oxford, UK
| | - Filomeen Haerynck
- Primary Immune Deficiency Research Laboratory, Department of Internal Diseases and Pediatrics, Centre for Primary Immunodeficiency Ghent, Jeffrey Modell Diagnosis and Research Centre, Ghent University Hospital, Ghent, Belgium
- Department of Internal Medicine and Pediatrics, Division of Pediatric Immunology and Pulmonology, Ghent University Hospital, Ghent, Belgium
| | - Jean-Laurent Casanova
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Institut National de la Santé et de la Recherche Médicale (INSERM) U1163, Paris, France
- University of Paris, Imagine Institute, Paris, France
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY
- Pediatric Hematology-Immunology Unit, Necker Hospital for Sick Children, AP-HP, Paris, France
- Howard Hughes Medical Institute, New York, NY
| | - Anne Puel
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Institut National de la Santé et de la Recherche Médicale (INSERM) U1163, Paris, France
- University of Paris, Imagine Institute, Paris, France
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY
| |
Collapse
|
4
|
Arshad S, Naveed M, Ullia M, Javed K, Butt A, Khawar M, Amjad F. Targeting STAT-3 signaling pathway in cancer for development of novel drugs: Advancements and challenges. Genet Mol Biol 2020; 43:e20180160. [PMID: 32167126 PMCID: PMC7198026 DOI: 10.1590/1678-4685-gmb-2018-0160] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2018] [Accepted: 10/20/2018] [Indexed: 12/25/2022] Open
Abstract
Signal transducers and activators of transcription 3 (STAT-3) is a transcription
factor that regulates the gene expression of several target genes. These factors
are activated by the binding of cytokines and growth factors with STAT-3
specific receptors on cell membrane. Few years ago, STAT-3 was considered an
acute phase response element having several cellular functions such as
inflammation, cell survival, invasion, metastasis and proliferation, genetic
alteration, and angiogenesis. STAT-3 is activated by several types of
inflammatory cytokines, carcinogens, viruses, growth factors, and oncogenes.
Thus, the STAT3 pathway is a potential target for cancer therapeutics. Abnormal
STAT-3 activity in tumor development and cellular transformation can be targeted
by several genomic and pharmacological methodologies. An extensive review of the
literature has been conducted to emphasize the role of STAT-3 as a unique cancer
drug target. This review article discusses in detail the wide range of STAT-3
inhibitors that show antitumor effects both in vitro and
in vivo. Thus, targeting constitutive STAT-3 signaling is a
remarkable therapeutic methodology for tumor progression. Finally, current
limitations, trials and future perspectives of STAT-3 inhibitors are also
critically discussed.
Collapse
Affiliation(s)
- Sundas Arshad
- University of Lahore, Department of Allied Health Sciences, Gujrat Campus, Pakistan
| | - Muhammad Naveed
- University of Central Punjab, Faculty of life sciences, Department of Biotechnology, Lahore, Pakistan
| | - Mahad Ullia
- University of Gujrat, Department of Biochemistry and Biotechnology Sialkot sub Campus, Pakistan
| | - Khadija Javed
- University of Gujrat, Department of Biochemistry and Biotechnology Sialkot sub Campus, Pakistan
| | - Ayesha Butt
- University of Gujrat, Department of Biochemistry and Biotechnology Sialkot sub Campus, Pakistan
| | - Masooma Khawar
- University of Gujrat, Department of Biochemistry and Biotechnology Sialkot sub Campus, Pakistan
| | - Fazeeha Amjad
- University of Gujrat, Department of Biochemistry and Biotechnology Sialkot sub Campus, Pakistan
| |
Collapse
|
5
|
Scherger AK, Al-Maarri M, Maurer HC, Schick M, Maurer S, Öllinger R, Gonzalez-Menendez I, Martella M, Thaler M, Pechloff K, Steiger K, Sander S, Ruland J, Rad R, Quintanilla-Martinez L, Wunderlich FT, Rose-John S, Keller U. Activated gp130 signaling selectively targets B cell differentiation to induce mature lymphoma and plasmacytoma. JCI Insight 2019; 4:128435. [PMID: 31391340 DOI: 10.1172/jci.insight.128435] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Accepted: 07/09/2019] [Indexed: 12/22/2022] Open
Abstract
Aberrant activity of the glycoprotein 130 130/JAK/STAT3 (gp130/JAK/STAT3) signaling axis is a recurrent event in inflammation and cancer. In particular, it is associated with a wide range of hematological malignancies, including multiple myeloma and leukemia. Novel targeted therapies have only been successful for some subtypes of these malignancies, underlining the need for developing robust mouse models to better dissect the role of this pathway in specific tumorigenic processes. Here, we investigated the role of selective gp130/JAK/STAT3 activation by generating a conditional mouse model. This model targeted constitutively active, cell-autonomous gp130 activity to B cells, as well as to the entire hematopoietic system. We found that regardless of the timing of activation in B cells, constitutively active gp130 signaling resulted in the formation specifically of mature B cell lymphomas and plasma cell disorders with full penetrance, only with different latencies, where infiltrating CD138+ cells were a dominant feature in every tumor. Furthermore, constitutively active gp130 signaling in all adult hematopoietic cells also led to the development specifically of largely mature, aggressive B cell cancers, again with a high penetrance of CD138+ tumors. Importantly, gp130 activity abrogated the differentiation block induced by a B cell-targeted Myc transgene and resulted in a complete penetrance of the gp130-associated, CD138+, mature B cell lymphoma phenotype. Thus, gp130 signaling selectively provides a strong growth and differentiation advantage for mature B cells and directs lymphomagenesis specifically toward terminally differentiated B cell cancers.
Collapse
Affiliation(s)
- Anna K Scherger
- Internal Medicine III, Technische Universität München, Munich, Germany
| | - Mona Al-Maarri
- Max Planck Institute for Metabolism Research, Center for Endocrinology, Preventive Medicine and Diabetes, Cologne, Germany
| | | | - Markus Schick
- Internal Medicine III, Technische Universität München, Munich, Germany
| | - Sabine Maurer
- Internal Medicine III, Technische Universität München, Munich, Germany
| | - Rupert Öllinger
- Internal Medicine II.,Center for Translational Cancer Research, and.,Institute of Molecular Oncology and Functional Genomics, Technische Universität München, Munich, Germany
| | | | - Manuela Martella
- Institute of Pathology, Eberhard-Karls-Universität Tübingen, Tübingen, Germany
| | - Markus Thaler
- Institute of Clinical Chemistry and Pathobiochemistry, Technische Universität München, Munich, Germany
| | - Konstanze Pechloff
- Center for Translational Cancer Research, and.,Institute of Clinical Chemistry and Pathobiochemistry, Technische Universität München, Munich, Germany.,German Cancer Consortium and German Cancer Research Center, Heidelberg, Germany
| | - Katja Steiger
- German Cancer Consortium and German Cancer Research Center, Heidelberg, Germany.,Institute of Pathology, Technische Universität München, Munich, Germany
| | - Sandrine Sander
- Adaptive Immunity and Lymphoma, German Cancer Research Center/National Center for Tumor Diseases Heidelberg, Heidelberg, Germany
| | - Jürgen Ruland
- Center for Translational Cancer Research, and.,Institute of Clinical Chemistry and Pathobiochemistry, Technische Universität München, Munich, Germany.,German Cancer Consortium and German Cancer Research Center, Heidelberg, Germany
| | - Roland Rad
- Internal Medicine II.,Center for Translational Cancer Research, and.,Institute of Molecular Oncology and Functional Genomics, Technische Universität München, Munich, Germany.,German Cancer Consortium and German Cancer Research Center, Heidelberg, Germany
| | | | - Frank T Wunderlich
- Max Planck Institute for Metabolism Research, Center for Endocrinology, Preventive Medicine and Diabetes, Cologne, Germany
| | - Stefan Rose-John
- Institute of Biochemistry, Christian-Albrechts-Universität zu Kiel, Kiel, Germany
| | - Ulrich Keller
- Internal Medicine III, Technische Universität München, Munich, Germany.,German Cancer Consortium and German Cancer Research Center, Heidelberg, Germany.,Department of Hematology, Oncology and Tumor Immunology (Campus Benjamin Franklin), Charité - Universitätsmedizin Berlin, Germany
| |
Collapse
|
6
|
Chiodetti AL, Sánchez Vallecillo MF, Dolina JS, Crespo MI, Marin C, Schoenberger SP, Allemandi DA, Palma SD, Pistoresi-Palencia MC, Morón G, Maletto BA. Class-B CpG-ODN Formulated With a Nanostructure Induces Type I Interferons-Dependent and CD4 + T Cell-Independent CD8 + T-Cell Response Against Unconjugated Protein Antigen. Front Immunol 2018; 9:2319. [PMID: 30364187 PMCID: PMC6192457 DOI: 10.3389/fimmu.2018.02319] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Accepted: 09/18/2018] [Indexed: 12/19/2022] Open
Abstract
There is a need for new vaccine adjuvant strategies that offer both vigorous antibody and T-cell mediated protection to combat difficult intracellular pathogens and cancer. To this aim, we formulated class-B synthetic oligodeoxynucleotide containing unmethylated cytosine-guanine motifs (CpG-ODN) with a nanostructure (Coa-ASC16 or coagel) formed by self-assembly of 6-0-ascorbyl palmitate ester. Our previous results demonstrated that mice immunized with ovalbumin (OVA) and CpG-ODN formulated with Coa-ASC16 (OVA/CpG-ODN/Coa-ASC16) elicited strong antibodies (IgG1 and IgG2a) and Th1/Th17 cellular responses without toxic systemic effects. These responses were superior to those induced by a solution of OVA with CpG-ODN or OVA/CpG-ODN formulated with aluminum salts. In this study, we investigated the capacity of this adjuvant strategy (CpG-ODN/Coa-ASC16) to elicit CD8+ T-cell response and some of the underlying cellular and molecular mechanisms involved in adaptive response. We also analyzed whether this adjuvant strategy allows a switch from an immunization scheme of three-doses to one of single-dose. Our results demonstrated that vaccination with OVA/CpG-ODN/Coa-ASC16 elicited an antigen-specific long-lasting humoral response and importantly-high quality CD8+ T-cell immunity with a single-dose immunization. Moreover, Coa-ASC16 promoted co-uptake of OVA and CpG-ODN by dendritic cells. The CD8+ T-cell response induced by OVA/CpG-ODN/Coa-ASC16 was dependent of type I interferons and independent of CD4+ T-cells, and showed polyfunctionality and efficiency against an intracellular pathogen. Furthermore, the cellular and humoral responses elicited by the nanostructured formulation were IL-6-independent. This system provides a simple and inexpensive adjuvant strategy with great potential for future rationally designed vaccines.
Collapse
Affiliation(s)
- Ana L. Chiodetti
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas, Centro de Investigaciones en Bioquímica Clínica e Inmunología, Córdoba, Argentina
| | - María F. Sánchez Vallecillo
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas, Centro de Investigaciones en Bioquímica Clínica e Inmunología, Córdoba, Argentina
| | - Joseph S. Dolina
- Division of Developmental Immunology, La Jolla Institute for Allergy and Immunology, La Jolla, CA, United States
| | - María I. Crespo
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas, Centro de Investigaciones en Bioquímica Clínica e Inmunología, Córdoba, Argentina
| | - Constanza Marin
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas, Centro de Investigaciones en Bioquímica Clínica e Inmunología, Córdoba, Argentina
| | - Stephen P. Schoenberger
- Division of Developmental Immunology, La Jolla Institute for Allergy and Immunology, La Jolla, CA, United States
| | - Daniel A. Allemandi
- Departamento de Farmacia, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas, Unidad de Tecnología Farmacéutica, Córdoba, Argentina
| | - Santiago D. Palma
- Departamento de Farmacia, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas, Unidad de Tecnología Farmacéutica, Córdoba, Argentina
| | - María C. Pistoresi-Palencia
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas, Centro de Investigaciones en Bioquímica Clínica e Inmunología, Córdoba, Argentina
| | - Gabriel Morón
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas, Centro de Investigaciones en Bioquímica Clínica e Inmunología, Córdoba, Argentina
| | - Belkys A. Maletto
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas, Centro de Investigaciones en Bioquímica Clínica e Inmunología, Córdoba, Argentina
| |
Collapse
|
7
|
Annamalai D, Clipstone NA. Prostaglandin F2α Inhibits Adipogenesis Via an Autocrine-Mediated Interleukin-11/Glycoprotein 130/STAT1-Dependent Signaling Cascade. J Cell Biochem 2014; 115:1308-21. [DOI: 10.1002/jcb.24785] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2013] [Accepted: 02/06/2014] [Indexed: 11/09/2022]
Affiliation(s)
- Damodaran Annamalai
- Department of Molecular Pharmacology and Therapeutics; Stritch School of Medicine; Loyola University Chicago; Maywood Illinois 60153
| | - Neil A. Clipstone
- Department of Molecular Pharmacology and Therapeutics; Stritch School of Medicine; Loyola University Chicago; Maywood Illinois 60153
| |
Collapse
|
8
|
David J, Barkema HW, Mortier R, Ghosh S, Guan LL, De Buck J. Gene expression profiling and putative biomarkers of calves 3 months after infection with Mycobacterium avium subspecies paratuberculosis. Vet Immunol Immunopathol 2014; 160:107-17. [PMID: 24841487 DOI: 10.1016/j.vetimm.2014.04.006] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2013] [Revised: 03/31/2014] [Accepted: 04/22/2014] [Indexed: 01/09/2023]
Abstract
Mycobacterium avium subsp. paratuberculosis (MAP) causes Johne's disease (JD), a chronic granulomatous intestinal inflammation of ruminants. Current diagnostic tools lack sensitivity to detect JD early in infection; therefore, alternatives are desired. The objective was to identify potential biomarkers in whole blood of high- and low-dose (LD) MAP-challenged Holstein-Friesian calves 3 months after inoculation. Infected calves were designated MAP-positive using the IFN-γ release assay. Differential expression of transcripts in whole blood was compared between non-infected controls and HD, as well as LD calves, using the Affymetrix(®) GeneChip(®) Bovine Genome Array. Microarray data were analyzed using RMA and PLIER algorithms; 296 transcripts were differentially expressed (17 had ≥ 1.5 fold change). The HD and LD calves had differential gene expression profiles for up to 80% of differentially expressed genes. Pathway analyses using Ingenuity Pathway Analysis (IPA(®)) indicated inhibition of several defence mechanisms, including apoptosis, leukocyte and lymphocyte trafficking, overall repression of gene expression and potentially hydrogen peroxide production in macrophages. Further validation using qPCR verified increased expression of CD46, ICOS, and CEP350, but decreased expression of CTLA4, YARS, and PARVB in infected calves. Additionally, a comparison of seropositive and seronegative infected calves identified transcripts predictive of seroconversion. We concluded that IL6ST/gp130 and CD22 may have important roles in the induction of antibodies against MAP. Putative biomarkers of early MAP infection with roles in immune responses were identified; in addition, the importance of infective dose on biomarkers was determined.
Collapse
Affiliation(s)
- Joel David
- Department of Production Animal Health, University of Calgary, 3330 Hospital Drive, Calgary, Alberta, Canada T2N 4N1.
| | - Herman W Barkema
- Department of Production Animal Health, University of Calgary, 3330 Hospital Drive, Calgary, Alberta, Canada T2N 4N1; Department of Medicine, University of Calgary, Calgary, Alberta, Canada.
| | - Rienske Mortier
- Department of Production Animal Health, University of Calgary, 3330 Hospital Drive, Calgary, Alberta, Canada T2N 4N1.
| | - Subrata Ghosh
- Department of Production Animal Health, University of Calgary, 3330 Hospital Drive, Calgary, Alberta, Canada T2N 4N1; Department of Medicine, University of Calgary, Calgary, Alberta, Canada.
| | - Le Luo Guan
- Department of Agricultural, Food & Nutritional Science, University of Alberta, Edmonton, Alberta, Canada T6G 2P5.
| | - Jeroen De Buck
- Department of Production Animal Health, University of Calgary, 3330 Hospital Drive, Calgary, Alberta, Canada T2N 4N1.
| |
Collapse
|
9
|
Gowane GR, Sharma AK, Sankar M, Narayanan K, Bisht P, Subramaniam S, Pattnaik B. The expression of IL6 and 21 in crossbred calves upregulated by inactivated trivalent FMD vaccine. Anim Biotechnol 2014; 25:108-18. [PMID: 24555796 DOI: 10.1080/10495398.2013.834826] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Foot and mouth disease (FMD) is an economically important disease and a whole-virus inactivated trivalent virus vaccine is the mainstay for controlling the disease in India. The protective humoral immune response to FMD vaccination is a complex, but, tightly regulated process mediated by the interplay of interleukins (IL). Based on the specific role of IL6 and 21 in adaptive immune response, we hypothesized that inactivated trivalent FMD vaccine would stimulate IL6 and 21 expression in the circulating lymphocytes. The expressions of IL6 and 21 were assayed on 0, 28, 60, 90, and 120 d post-vaccination (DPV) by quantitative PCR (qPCR) with simultaneous assessment of FMDV antibody titer by liquid phase blocking ELISA. The results revealed that the peak expression of IL6 and 21 was on DPV 28 which correlated well with the FMDV antibody titer and plummeted to the prevaccination titer level by 60 DPV. As IL21 is the final effector of antibody production as compared to IL6, we investigated the expression of IL21 in calves that had protective titer (>1.8) with the unprotected group (<1.8). Expression of IL21 on 28 DPV was numerically higher in the protected than that of the unprotected group of calves.
Collapse
Affiliation(s)
- G R Gowane
- a Central Sheep & Wool Research Institute , Avikanagar , Rajasthan , India
| | | | | | | | | | | | | |
Collapse
|
10
|
Priming microenvironments dictate cytokine requirements for T helper 17 cell lineage commitment. Immunity 2011; 35:1010-22. [PMID: 22137454 DOI: 10.1016/j.immuni.2011.10.013] [Citation(s) in RCA: 85] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2010] [Revised: 08/29/2011] [Accepted: 10/20/2011] [Indexed: 11/20/2022]
Abstract
Activation of pattern recognition receptors on dendritic cells (DCs) and macrophages leads to secretion of cytokines that control differentiation of CD4(+) T cells. The current understanding is that interleukin-6 (IL-6) in combination with transforming growth factor-β (TGF-β) leads to generation of T helper 17 (Th17) lineage cells. Here, we have discovered that the cytokine requirements for Th17 cell polarization depend on the site of priming. Although IL-6 played a critical role in Th17 cell lineage priming in the skin and mucosal tissues, it was not required for Th17 cell priming in the spleen. In contrast, IL-1 played an irreplaceable role for priming of Th17 lineage cells in all tissues. Importantly, we have demonstrated that IL-6-independent and -dependent pathways of Th17 cell differentiation are guided by DCs residing in various tissues. These results reveal fundamental differences by which the systemic, mucosal, and cutaneous immune systems guide Th17 cell lineage commitment.
Collapse
|
11
|
Stumhofer JS, Tait ED, Quinn WJ, Hosken N, Spudy B, Goenka R, Fielding CA, O'Hara AC, Chen Y, Jones ML, Saris CJM, Rose-John S, Cua DJ, Jones SA, Elloso MM, Grötzinger J, Cancro MP, Levin SD, Hunter CA. A role for IL-27p28 as an antagonist of gp130-mediated signaling. Nat Immunol 2010; 11:1119-26. [PMID: 21057510 PMCID: PMC3059498 DOI: 10.1038/ni.1957] [Citation(s) in RCA: 149] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2010] [Accepted: 10/12/2010] [Indexed: 12/11/2022]
Abstract
The heterodimeric cytokine interleukin 27 (IL-27) signals through the IL-27Rα subunit of its receptor, combined with gp130, a common receptor chain used by several cytokines, including IL-6. Notably, the IL-27 subunits p28 (IL-27p28) and EBI3 are not always expressed together, which suggests that they may have unique functions. Here we show that IL-27p28, independently of EBI3, antagonized cytokine signaling through gp130 and IL-6-mediated production of IL-17 and IL-10. Similarly, the ability to generate antibody responses was dependent on the activity of gp130-signaling cytokines. Mice transgenic for expression of IL-27p28 showed a substantial defect in the formation of germinal centers and antibody production. Thus, IL-27p28, as a natural antagonist of gp130-mediated signaling, may be useful as a therapeutic for managing inflammation mediated by cytokines that signal through gp130.
Collapse
Affiliation(s)
- Jason S Stumhofer
- University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Silver JS, Hunter CA. gp130 at the nexus of inflammation, autoimmunity, and cancer. J Leukoc Biol 2010; 88:1145-56. [PMID: 20610800 DOI: 10.1189/jlb.0410217] [Citation(s) in RCA: 191] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Glycoprotein 130 (gp130) is a shared receptor utilized by several related cytokines, including IL-6, IL-11, IL-27, Leukemia Inhibitory Factor (LIF), Oncostatin M (OSM), Ciliary Neurotrophic Factor (CNTF), Cardiotrophin 1 (CT-1) and Cardiotrophin-like Cytokine (CLC). Gp130 plays critical roles during development and gp130-deficient mice are embryonically lethal. However, the best characterized facet of this receptor and its associated cytokines is the ability to promote or suppress inflammation. The aim of this review is to discuss the role of gp130 in promoting or preventing the development of autoimmunity and cancer, two processes that are associated with aberrant inflammatory responses.
Collapse
Affiliation(s)
- J S Silver
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19143, USA
| | | |
Collapse
|
13
|
Dienz O, Eaton SM, Bond JP, Neveu W, Moquin D, Noubade R, Briso EM, Charland C, Leonard WJ, Ciliberto G, Teuscher C, Haynes L, Rincon M. The induction of antibody production by IL-6 is indirectly mediated by IL-21 produced by CD4+ T cells. ACTA ACUST UNITED AC 2009; 206:69-78. [PMID: 19139170 PMCID: PMC2626667 DOI: 10.1084/jem.20081571] [Citation(s) in RCA: 345] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Abstract
Interleukin (IL) 6 is a proinflammtory cytokine produced by antigen-presenting cells and nonhematopoietic cells in response to external stimuli. It was initially identified as a B cell growth factor and inducer of plasma cell differentiation in vitro and plays an important role in antibody production and class switching in vivo. However, it is not clear whether IL-6 directly affects B cells or acts through other mechanisms. We show that IL-6 is sufficient and necessary to induce IL-21 production by naive and memory CD4+ T cells upon T cell receptor stimulation. IL-21 production by CD4+ T cells is required for IL-6 to promote B cell antibody production in vitro. Moreover, administration of IL-6 with inactive influenza virus enhances virus-specific antibody production, and importantly, this effect is dependent on IL-21. Thus, IL-6 promotes antibody production by promoting the B cell helper capabilities of CD4+ T cells through increased IL-21 production. IL-6 could therefore be a potential coadjuvant to enhance humoral immunity.
Collapse
Affiliation(s)
- Oliver Dienz
- Department of Medicine/Immunobiology Program, University of Vermont, Burlington, VT 05405, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Fornek JL, Tygrett LT, Waldschmidt TJ, Poli V, Rickert RC, Kansas GS. Critical role for Stat3 in T-dependent terminal differentiation of IgG B cells. Blood 2005; 107:1085-91. [PMID: 16223771 PMCID: PMC1895906 DOI: 10.1182/blood-2005-07-2871] [Citation(s) in RCA: 117] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Stat proteins are latent cytoplasmic transcription factors that are crucial in many aspects of mammalian development. In the immune system, Stat3 has distinct roles in T-cell, neutrophil, and macrophage function, but a role for Stat3 in B-cell development, particularly in the terminal differentiation of B cells into antibody-secreting plasma cells, has never been directly tested. In this study, we used the Cre/lox system to generate a mouse strain in which Stat3 was conditionally deleted in the B-cell lineage (Stat3(fl/fl)CD19(Cre/+)). B-cell development, establishment of the peripheral B-cell compartment, and baseline serum antibody levels were unperturbed in Stat3(fl/fl)CD19(Cre/+) mice. Strikingly, Stat3(fl/fl)CD19(Cre/+) mice displayed profound defects in T-dependent (TD) IgG responses, but normal TD IgM, IgE, and IgA responses and T-independent (TI) IgM and IgG3 responses. In addition, germinal center (GC) formation, isotype switching, and generation of memory B cells, including IgG+ memory cells, were all intact in Stat3(fl/fl)CD19(Cre/+) mice, indicating that the requirement for Stat3 was limited to plasma cell differentiation. These results demonstrate a profound yet highly selective role for Stat3 in TD IgG plasma cell differentiation, and therefore represent a unique example of a transcription factor regulating isotype-specific terminal B-cell differentiation.
Collapse
Affiliation(s)
- Jamie L Fornek
- Department of Microbiology-Immunology, Feinberg School of Medicine of Northwestern University, Chicago, IL 60611, USA
| | | | | | | | | | | |
Collapse
|
15
|
Ohtani T, Ishihara K, Atsumi T, Yoshida Y, Nishida K, Narimatsu M, Shirogane T, Hibi M, Hirano T. gp130-mediated signalling as a therapeutic target. ACTA ACUST UNITED AC 2005. [DOI: 10.1517/14728222.4.4.459] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
16
|
Kamimura D, Ishihara K, Hirano T. IL-6 signal transduction and its physiological roles: the signal orchestration model. Rev Physiol Biochem Pharmacol 2004; 149:1-38. [PMID: 12687404 DOI: 10.1007/s10254-003-0012-2] [Citation(s) in RCA: 340] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Interleukin (IL)-6 is a pleiotropic cytokine that not only affects the immune system, but also acts in other biological systems and many physiological events in various organs. In a target cell, IL-6 can simultaneously generate functionally distinct or sometimes contradictory signals through its receptor complex, IL-6Ralpha and gp130. One good illustration is derived from the in vitro observations that IL-6 promotes the growth arrest and differentiation of M1 cells through gp130-mediated STAT3 activation, whereas the Y759/SHP-2-mediated cascade by gp130 stimulation has growth-enhancing effects. The final physiological output can be thought of as a consequence of the orchestration of the diverse signaling pathways generated by a given ligand. This concept, the signal orchestration model, may explain how IL-6 can elicit proinflammatory or anti-inflammatory effects, depending on the in vivo environmental circumstances. Elucidation of the molecular mechanisms underlying this issue is a challenging subject for future research. Intriguingly, recent in vivo studies indicated that the SHP-2-binding site- and YXXQ-mediated pathways through gp130 are not mutually exclusive but affect each other: a mutation at the SHP-2-binding site prolongs STAT3 activation, and a loss of STAT activation by gp130 truncation leads to sustained SHP-2/ERK MAPK phosphorylation. Although IL-6/gp130 signaling is a promising target for drug discovery for many human diseases, the interdependence of each signaling pathway may be an obstacle to the development of a nonpeptide orally active small molecule to inhibit one of these IL-6 signaling cascades, because it would disturb the signal orchestration. In mice, a consequence of the imbalanced signals causes unexpected results such as gastrointestinal disorders, autoimmune diseases, and/or chronic inflammatory proliferative diseases. However, lessons learned from IL-6 KO mice indicate that IL-6 is not essential for vital biological processes, but a significant impact on disease progression in many experimental models for human disorders. Thus, IL-6/gp130 signaling will become a more attractive therapeutic target for human inflammatory diseases when a better understanding of IL-6 signaling, including the identification of the conductor for gp130 signal transduction, is achieved.
Collapse
Affiliation(s)
- D Kamimura
- Department of Molecular Oncology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | | | | |
Collapse
|
17
|
Jego G, Palucka AK, Blanck JP, Chalouni C, Pascual V, Banchereau J. Plasmacytoid dendritic cells induce plasma cell differentiation through type I interferon and interleukin 6. Immunity 2003; 19:225-34. [PMID: 12932356 DOI: 10.1016/s1074-7613(03)00208-5] [Citation(s) in RCA: 795] [Impact Index Per Article: 37.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Dendritic cells (DCs) initiate and control immune responses. Plasmacytoid DCs (pDCs) represent a unique DC subset able to promptly release large amounts of type I interferon (IFN-alphabeta) upon viral encounter. Here we report that depletion of pDCs from human blood mononuclear cells abrogates the secretion of specific and polyclonal IgGs in response to influenza virus. Furthermore, purified pDCs triggered with virus induce CD40-activated B cells to differentiate into plasma cells. Two pDC cytokines act sequentially, with IFN-alphabeta generating non-Ig-secreting plasma blasts and IL-6 inducing their differentiation into Ig-secreting plasma cells. These plasma cells display the high levels of CD38 found on tissue plasma cells. Thus, pDCs are critical for the generation of plasma cells and antibody responses.
Collapse
Affiliation(s)
- Gaetan Jego
- Baylor Institute for Immunology Research, 3434 Live Oak Street, Dallas, Texas 75204, USA
| | | | | | | | | | | |
Collapse
|
18
|
Feldhahn N, Schwering I, Lee S, Wartenberg M, Klein F, Wang H, Zhou G, Wang SM, Rowley JD, Hescheler J, Krönke M, Rajewsky K, Küppers R, Müschen M. Silencing of B cell receptor signals in human naive B cells. J Exp Med 2002; 196:1291-305. [PMID: 12438421 PMCID: PMC2193982 DOI: 10.1084/jem.20020881] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
To identify changes in the regulation of B cell receptor (BCR) signals during the development of human B cells, we generated genome-wide gene expression profiles using the serial analysis of gene expression (SAGE) technique for CD34(+) hematopoietic stem cells (HSCs), pre-B cells, naive, germinal center (GC), and memory B cells. Comparing these SAGE profiles, genes encoding positive regulators of BCR signaling were expressed at consistently lower levels in naive B cells than in all other B cell subsets. Conversely, a large group of inhibitory signaling molecules, mostly belonging to the immunoglobulin superfamily (IgSF), were specifically or predominantly expressed in naive B cells. The quantitative differences observed by SAGE were corroborated by semiquantitative reverse transcription-polymerase chain reaction (RT-PCR) and flow cytometry. In a functional assay, we show that down-regulation of inhibitory IgSF receptors and increased responsiveness to BCR stimulation in memory as compared with naive B cells at least partly results from interleukin (IL)-4 receptor signaling. Conversely, activation or impairment of the inhibitory IgSF receptor LIRB1 affected BCR-dependent Ca(2+) mobilization only in naive but not memory B cells. Thus, LIRB1 and IL-4 may represent components of two nonoverlapping gene expression programs in naive and memory B cells, respectively: in naive B cells, a large group of inhibitory IgSF receptors can elevate the BCR signaling threshold to prevent these cells from premature activation and clonal expansion before GC-dependent affinity maturation. In memory B cells, facilitated responsiveness upon reencounter of the immunizing antigen may result from amplification of BCR signals at virtually all levels of signal transduction.
Collapse
Affiliation(s)
- Niklas Feldhahn
- Institute for Medical Microbiology, Immunology and Hygiene, University of Cologne, 50931 Köln, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Abstract
An estimated 30% of cancer deaths are attributed to cachexia and its consequences. Cachexia (wasting syndrome) is the hypercatabolism of the body's carbon sources, proteins and lipids, for conversion into energy. It is induced by a variety of pathological conditions, including cancer. Among the inflammatory responses to cancer is the synthesis of cytokines, including IL-6 and related cytokines. These cytokines have been found to induce cachexia by altering metabolism of lipids and proteins. IL-6-like cytokines have been found to inhibit lipid biosynthesis by adipocytes, which increased the rate of lipid catabolism. Others have described the atrophy and increased catabolism of muscle protein due to IL-6. A cytokine closely-related to IL-6 is leptin, which plays a major role in lipid metabolism under normal conditions. The role of leptin in pathological conditions such as cancer cachexia has not yet been fully elucidated. Detailed mechanistic information about the induction of cancer cachexia by IL-6-like cytokines requires more research.
Collapse
Affiliation(s)
- B E Barton
- Department of Surgery, University of Medicine & Dentistry of New Jersey, New Jersey Medical School, Newark 07103, USA.
| |
Collapse
|
20
|
Felitsyn N, Kitova EN, Klassen JS. Thermal decomposition of a gaseous multiprotein complex studied by blackbody infrared radiative dissociation. Investigating the origin of the asymmetric dissociation behavior. Anal Chem 2001; 73:4647-61. [PMID: 11605843 DOI: 10.1021/ac0103975] [Citation(s) in RCA: 151] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The blackbody infrared radiative dissociation technique was used to study the thermal decomposition of the gaseous B5 pentamer of the Shiga-like toxin I and its complexes with the Pk trisaccharide and a decavalent Pk-based oligosaccharide ligand (STARFISH, S). Dissociation of the protonated pentamer, (B5 + nH)n+ triple bond B5n+ where n = 11-14, proceeds almost exclusively by the loss of a single subunit (B) with a disproportionately large fraction (30-50%) of the parent ion charge. The degree of charge enrichment of the leaving subunit increases with increasing parent ion charge state. For n = 12-14, a distribution of product ion charge states is observed. The yields of the complementary pairs of product ions are sensitive to the reaction temperature, with higher temperatures favoring greater charge enrichment of the leaving subunit for +13 and +14, and the opposite effect for +12. These results indicate that some of the protons are rapidly exchanged between subunits in the gas phase. Dissociation of B5(14+) x S proceeds exclusively by the loss of one subunit, although the ligand increases the stability of the complex and also reduces the degree of charge enrichment in the ejected monomer. For B5(12+)(Pk)1-3, the loss of neutral Pk competes with loss of a subunit at low temperatures. Linear Arrhenius plots were obtained from the temperature-dependent dissociation rate constants measured for the loss of B from B5n+ and B514+ x S. The magnitude of the Arrhenius parameters is highly dependent on the charge state of the pentamer: Ea = 35 kcal/mol and A = 1,019 s(-1) (+14), 46 kcal/mol and 1,023 S(-1) (+13), 50 kcal/mol and 1026 s(-1) (+12), and 80 kcal/mol and 10(39) (+11). The Ea and A for B5(14+) x S are 59 kcal/mol and 10(30) s(-1), respectively. The reaction pathways leading to greater charge enrichment of the subunit lost from the B5(14+) and B5(13+) ions correspond to higher energy processes, however, these pathways are kinetically preferred at higher temperatures due to their large A factors. A simple electrostatic model, whereby charge enrichment leads to Coulombic repulsion-induced denaturation of the subunits and disruption of the intersubunit interactions, provides an explanation for the magnitude of the Arrhenius parameters and the origin of the asymmetric dissociation behavior of the complexes.
Collapse
Affiliation(s)
- N Felitsyn
- Department of Chemistry, University of Alberta, Edmonton, Canada
| | | | | |
Collapse
|
21
|
Ernst M, Inglese M, Waring P, Campbell IK, Bao S, Clay FJ, Alexander WS, Wicks IP, Tarlinton DM, Novak U, Heath JK, Dunn AR. Defective gp130-mediated signal transducer and activator of transcription (STAT) signaling results in degenerative joint disease, gastrointestinal ulceration, and failure of uterine implantation. J Exp Med 2001; 194:189-203. [PMID: 11457894 PMCID: PMC2193459 DOI: 10.1084/jem.194.2.189] [Citation(s) in RCA: 167] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
The receptor subunit gp130 transduces multiple cell type-specific activities of the leukemia inhibitory factor (LIF)/interleukin (IL)-6 family of cytokines through the signal transducer and activator of transcription (STAT) and src homology 2 domain-bearing protein tyrosine phosphatase (SHP)-2/ras/Erk pathways. To define STAT-dependent physiological responses, we generated mice with a COOH-terminal gp130(DeltaSTAT) "knock-in" mutation which deleted all STAT-binding sites. gp130(DeltaSTAT) mice phenocopyed mice deficient for IL-6 (impaired humoral and mucosal immune and hepatic acute phase responses) and LIF (failure of blastocyst implantation). However, unlike mice with null mutations in any of the components in the gp130 signaling pathway, gp130(DeltaSTAT) mice also displayed gastrointestinal ulceration and a severe joint disease with features of chronic synovitis, cartilaginous metaplasia, and degradation of the articular cartilage. Mitogenic hyperresponsiveness of synovial cells to the LIF/IL-6 family of cyto-kines was caused by sustained gp130-mediated SHP-2/ras/Erk activation due to impaired STAT-mediated induction of suppressor of cytokine signaling (SOCS) proteins which normally limits gp130 signaling. Therefore, the joint pathology in gp130(DeltaSTAT) mice is likely to arise from the disturbance of the otherwise balanced activation of the SHP-2/ras/Erk and STAT signaling cascades emanating from gp130.
Collapse
Affiliation(s)
- M Ernst
- Ludwig Institute for Cancer Research, PO Royal Melbourne Hospital, VIC 3050, Australia.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Uozumi H, Hiroi Y, Zou Y, Takimoto E, Toko H, Niu P, Shimoyama M, Yazaki Y, Nagai R, Komuro I. gp130 plays a critical role in pressure overload-induced cardiac hypertrophy. J Biol Chem 2001; 276:23115-9. [PMID: 11262406 DOI: 10.1074/jbc.m100814200] [Citation(s) in RCA: 67] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
gp130, a common receptor for the interleukin 6 family, plays pivotal roles in growth and survival of cardiac myocytes. In the present study, we examined the role of gp130 in pressure overload-induced cardiac hypertrophy using transgenic (TG) mice, which express a dominant negative mutant of gp130 in the heart under the control of alpha myosin heavy chain promoter. TG mice were apparently healthy and fertile. There were no differences in body weight and heart weight between TG mice and littermate wild type (WT) mice. Pressure overload-induced increases in the heart weight/body weight ratio, ventricular wall thickness, and cross-sectional areas of cardiac myocytes were significantly smaller in TG mice than in WT mice. Northern blot analysis revealed that pressure overload-induced up-regulation of brain natriuretic factor gene and down-regulation of sarcoplasmic reticulum Ca(2+) ATPase 2 gene were attenuated in TG mice. Pressure overload activated ERKs and STAT3 in the heart of WT mice, whereas pressure overload-induced activation of STAT3, but not of ERKs, was suppressed in TG mice. These results suggest that gp130 plays a critical role in pressure overload-induced cardiac hypertrophy possibly through the STAT3 pathway.
Collapse
Affiliation(s)
- H Uozumi
- Department of Cardiovascular Medicine, University of Tokyo Graduate School of Medicine, Tokyo 113-8655, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Abstract
It has been known for many years that the epithelia of the urogenital system derive from mesenchyme. Essential regulators of this conversion have recently been discovered, and cellular changes have been described. However, we do not have a coherent view of how these dramatic changes are integrated, nor do we know the source or identity of extracellular signals that must regulate epithelialization of mesenchymal precursors. The metanephric kidney, Wolffian duct, and the Drosophila midgut are the leading model systems to describe how epithelia derive from mesenchyme.
Collapse
Affiliation(s)
- J Barasch
- Department of Medicine, College of Physicians and Surgeons of Columbia University, New York, New York 10032, USA.
| |
Collapse
|
24
|
Tone E, Kunisada K, Kumanogoh A, Negoro S, Funamoto M, Osugi T, Kishimoto T, Yamauchi-Takihara K. gp130-Dependent signalling pathway is not enhanced in gp130 transgenic heart after LIF stimulation. Cytokine 2000; 12:1512-8. [PMID: 11023666 DOI: 10.1006/cyto.2000.0751] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Activation of gp130 transduces a hypertrophic signal in the heart, but it is not clear whether signalling through gp130 is enhanced when gp130 is overexpressed in vivo. We generated gp130 transgenic mice (TG) and examined the activation of signalling pathways downstream of gp130 in the hearts. The tyrosine phosphorylation of gp130 was enhanced, the phosphorylation of STAT3 and ERK (extracellular signal regulated kinase) 1/2 was increased and induction of the beta-myosin heavy chain (MHC) gene was observed in TG hearts without significant phenotypic changes. Intravenous administration of leukaemia inhibitory factor (LIF) induced tyrosine phosphorylation of STAT3 and ERK 1/2 and expression of c-fos and beta-MHC mRNAs in wild-type littermates' (WT) hearts. However, enhancement of STAT3 and ERK 1/2 phosphorylation or augmented mRNA expressions was not observed in TG hearts after LIF stimulation. Next, STAT-induced STAT inhibitor (SSI) mRNA expression was examined. The expression of SSI-1, SSI-2, and SSI-3 mRNAs was significantly augmented in TG hearts after LIF stimulation. These results indicate that overexpressed gp130 does not always enhance downstream signals in the hearts and suggest that the SSI family plays a role in the regulation of the gp130-dependent signalling pathway in the hearts.
Collapse
Affiliation(s)
- E Tone
- Department of Molecular Medicine, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | | | | | | | | | | | | | | |
Collapse
|
25
|
Karras JG, McKay RA, Lu T, Pych J, Frank DA, Rothstein TL, Monia BP. STAT3 regulates the growth and immunoglobulin production of BCL(1) B cell lymphoma through control of cell cycle progression. Cell Immunol 2000; 202:124-35. [PMID: 10896772 DOI: 10.1006/cimm.2000.1661] [Citation(s) in RCA: 22] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
STAT3 is constitutively phosphorylated on tyrosine(705) in self-renewing, CD5(+) murine B-1 lymphocytes. Nuclear extracts from untreated primary B-1 or CD5(+) BCL(1) B lymphoma cells were found to contain immunoreactive STAT3 protein that binds to a sis-inducible element present in the promoter of the p21(waf1/cip1) tumor suppressor gene and is constitutively phosphorylated on serine(727). To determine the functional significance of constitutive STAT3 activation in B lymphoma cells, a specific STAT3 antisense oligonucleotide was developed and used to examine basal BCL(1) cell growth and IgM production. Abrogating STAT3 expression in BCL(1) cells inhibited their proliferative capacity and induced a corresponding decrease in secretion of IgM. Cell cycle analysis showed a block in progression through G1 in BCL(1) cells treated with the STAT3 antisense oligonucleotide. These results indicate that STAT3 controls cell growth and immunoglobulin secretion by enhancing progression through the G1 phase of the cell cycle in BCL(1) B cell lymphoma.
Collapse
Affiliation(s)
- J G Karras
- Department of Molecular and Cellular Pharmacology, Isis Pharmaceuticals, Inc., 2292 Faraday Avenue, Carlsbad, California 92008, USA
| | | | | | | | | | | | | |
Collapse
|
26
|
Hirano T, Ishihara K, Hibi M. Roles of STAT3 in mediating the cell growth, differentiation and survival signals relayed through the IL-6 family of cytokine receptors. Oncogene 2000; 19:2548-56. [PMID: 10851053 DOI: 10.1038/sj.onc.1203551] [Citation(s) in RCA: 922] [Impact Index Per Article: 38.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Members of the IL-6 cytokine family are involved in a variety of biological responses, including the immune response, inflammation, hematopoiesis, and oncogenesis by regulating cell growth, survival, and differentiation. These cytokines use gp130 as a common receptor subunit. The binding of ligand to gp130 activates the JAK/STAT signal transduction pathway, where STAT3 plays a central role in transmitting the signals from the membrane to the nucleus. STAT3 is essential for gp130-mediated cell survival and G1 to S cell-cycle-transition signals. Both c-myc and pim have been identified as target genes of STAT3 and together can compensate for STAT3 in cell survival and cell-cycle transition. STAT3 is also required for gp130-mediated maintenance of the pluripotential state of proliferating embryonic stem cells and for the gp130-induced macrophage differentiation of M1 cells. Furthermore, STAT3 regulates cell movement, such as leukocyte, epidermal cell, and keratinocyte migration. STAT3 also appears to regulate B cell differentiation into antibody-forming plasma cells. Since the IL-6/gp130/STAT3 signaling pathway is involved in both B cell growth and differentiation into plasma cells it is likely to play a central role in the generation of plasma cell neoplasias. Oncogene (2000).
Collapse
Affiliation(s)
- T Hirano
- Division of Molecular Oncology C-7, Biomedical Research Center, Osaka University Graduate School of Medicine, 2-2 Yamada-oko, Suita, Osaka 565-0871, Japan
| | | | | |
Collapse
|
27
|
Ohtani T, Ishihara K, Atsumi T, Nishida K, Kaneko Y, Miyata T, Itoh S, Narimatsu M, Maeda H, Fukada T, Itoh M, Okano H, Hibi M, Hirano T. Dissection of signaling cascades through gp130 in vivo: reciprocal roles for STAT3- and SHP2-mediated signals in immune responses. Immunity 2000; 12:95-105. [PMID: 10661409 DOI: 10.1016/s1074-7613(00)80162-4] [Citation(s) in RCA: 186] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
We generated a series of knockin mouse lines, in which the cytokine receptor gp130-dependent STAT3 and/or SHP2 signals were disrupted, by replacing the mouse gp130 gene with human gp130 mutant cDNAs. The SHP2 signal-deficient mice (gp130F759/F759 were born normal but displayed splenomegaly and lymphadenopathy and an enhanced acute phase reaction. In contrast, the STAT3 signal-deficient mice (gp130FXQ/FXXQ) died perinatally, like the gp130-deficient mice (gp130D/D). The gp130F759/F759 mice showed prolonged gp130-induced STAT3 activation, indicating a negative regulatory role for SHP2. Th1-type cytokine production and IgG2a and IgG2b production were increased in the gp130F759/F759 mice, while they were decreased in the gp130FXXQ/FXXQ immune system. These results indicate that the balance of positive and negative signals generated through gp130 regulates the immune responses.
Collapse
Affiliation(s)
- T Ohtani
- Division of Molecular Oncology (C7), Biomedical Research Center, Osaka University Graduate School of Medicine, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Saito S, Aikawa R, Shiojima I, Nagai R, Yazaki Y, Komuro I. Endothelin-1 induces expression of fetal genes through the interleukin-6 family of cytokines in cardiac myocytes. FEBS Lett 1999; 456:103-7. [PMID: 10452539 DOI: 10.1016/s0014-5793(99)00936-9] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
We here examined the role of the interleukin-6 (IL-6) family of cytokines in endothelin-1 (ET-1)-induced hypertrophic responses using cultured cardiac myocytes of neonatal rats. ET-1 induced expression of IL-6 and leukemia inhibitory factor (LIF) genes. ET-1-induced LIF gene expression was abolished by inhibition of protein kinase C activity. ET-1 activated the promoter of atrial natriuretic peptide and beta-type myosin heavy chain genes through the tyrosine kinase pathway and IL-6 receptor gp130. These results suggest that the IL-6 family of cytokines mediates ET-1-induced expression of some fetal genes in cardiac myocytes.
Collapse
Affiliation(s)
- S Saito
- Department of Cardiovascular Medicine, University of Tokyo Graduate School of Medicine, Japan
| | | | | | | | | | | |
Collapse
|
29
|
O'Brien CA, Gubrij I, Lin SC, Saylors RL, Manolagas SC. STAT3 activation in stromal/osteoblastic cells is required for induction of the receptor activator of NF-kappaB ligand and stimulation of osteoclastogenesis by gp130-utilizing cytokines or interleukin-1 but not 1,25-dihydroxyvitamin D3 or parathyroid hormone. J Biol Chem 1999; 274:19301-8. [PMID: 10383440 DOI: 10.1074/jbc.274.27.19301] [Citation(s) in RCA: 224] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Interleukin (IL)-6-type cytokines stimulate osteoclastogenesis by activating gp130 in stromal/osteoblastic cells and may mediate some of the osteoclastogenic effects of other cytokines and hormones. To determine whether STAT3 is a downstream effector of gp130 in the osteoclast support function of stromal/osteoblastic cells and whether the gp130/STAT3 pathway is utilized by other osteoclastogenic agents, we conditionally expressed dominant negative (dn)-STAT3 or dn-gp130 in a stromal/osteoblastic cell line (UAMS-32) that supports osteoclast formation. Expression of either dominant negative protein abolished osteoclast formation stimulated by IL-6 + soluble IL-6 receptor, oncostatin M, or IL-1 but not by parathyroid hormone or 1,25-dihydroxyvitamin D3. Because previous studies suggested that IL-6-type cytokines may stimulate osteoclastogenesis by inducing expression of the tumor necrosis factor-related protein, receptor activator of NF-kappaB ligand (RANKL), we conditionally expressed RANKL in UAMS-32 cells and found that this was sufficient to stimulate osteoclastogenesis. Moreover, dn-STAT3 blocked the ability of either IL-6 + soluble IL-6 receptor or oncostatin M to induce RANKL. These results establish that STAT3 is essential for gp130-mediated osteoclast formation and that the target of STAT3 during this process is induction of RANKL. In addition, this study demonstrates that activation of the gp130-STAT3 pathway in stromal/osteoblastic cells mediates the osteoclastogenic effects of IL-1, but not parathyroid hormone or 1, 25-dihydroxyvitamin D3.
Collapse
Affiliation(s)
- C A O'Brien
- Departments of Medicine and Pediatrics, Center for Osteoporosis and Metabolic Bone Diseases, University of Arkansas for Medical Sciences, Little Rock, Arkansas 72205, USA.
| | | | | | | | | |
Collapse
|
30
|
Abstract
Receptors for interleukins, colony stimulating factors, and hormones have a homology in their extracellular regions, characterized by the conserved cysteine residues and the tryptophan-serine-x-tryptophan-serine motif, thus, they are classified to the type 1 cytokine receptor superfamily. Janus tyrosine kinase (JAKs) have been found to be involved in the signal transduction through type I cytokine receptors. JAKs associate with the membrane proximal region in the cytoplasmic domain having box1 and box2, which are conserved among the family, and upon the stimulation JAKs can be aggregated following the receptor dimerization and activated probably by transphosphorylation. JAKs then phosphorylate the receptor and various signal transducing molecules, including STATs (signal transducer and activator of transcriptions) and other SH2-containing adapter molecules. STATs were initially identified as transcription factors containing a SH2 domain and regulating interferons-inducible genes. STATs can be tyrosine phosphorylated by JAKs and form dimer (either hetero- or homo-dimers) to enter the nucleus, resulting in the expression of a set of genes. On the other hand, adapter molecules such as Shc, GRB2, and SHP-2 have been shown to link the cytokine receptors to Ras, followed by the activation of the Raf-MEK-MAP kinase pathway, leading to the activation of various transcription factors in the nucleus. These two signals are generated by different ways upon the stimulation of the receptors and they elicit a variety of biological functions in various cell types. In this review, we will discuss the mechanism by which cytokines activate JAKs, STATs, and a variety of adapter molecules. We further discuss the roles of each signal transduction pathways in the expression of biological activities of cytokines.
Collapse
Affiliation(s)
- M Hibi
- Division of Molecular Oncology, Osaka University Medical School, Japan
| | | |
Collapse
|
31
|
Kopf M, Herren S, Wiles MV, Pepys MB, Kosco-Vilbois MH. Interleukin 6 influences germinal center development and antibody production via a contribution of C3 complement component. J Exp Med 1998; 188:1895-906. [PMID: 9815267 PMCID: PMC2212418 DOI: 10.1084/jem.188.10.1895] [Citation(s) in RCA: 159] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/1997] [Revised: 08/07/1998] [Indexed: 11/04/2022] Open
Abstract
Mice rendered deficient for interleukin (IL) 6 by gene targeting were evaluated for their response to T cell-dependent antigens. Antigen-specific immunoglobulin (Ig)M levels were unaffected whereas all IgG isotypes showed varying degrees of alteration. Germinal center reactions occurred but remained physically smaller in comparison to those in the wild-type mice. This concurred with the observations that molecules involved in initial signaling events leading to germinal center formation were not altered (e.g., B7.2, CD40 and tumor necrosis factor R1). T cell priming was not impaired nor was a gross imbalance of T helper cell (Th) 1 versus Th2 cytokines observed. However, B7.1 molecules, absent from wild-type counterparts, were detected on germinal center B cells isolated from the deficient mice suggesting a modification of costimulatory signaling. A second alteration involved impaired de novo synthesis of C3 both in serum and germinal center cells from IL-6-deficient mice. Indeed, C3 provided an essential stimulatory signal for wild-type germinal center cells as both monoclonal antibodies that interrupted C3-CD21 interactions and sheep anti-mouse C3 antibodies caused a significant decrease in antigen-specific antibody production. In addition, germinal center cells isolated from C3-deficient mice produced a similar defect in isotype production. Low density cells with dendritic morphology were the local source of IL-6 and not the germinal center lymphocytes. Adding IL-6 in vitro to IL-6-deficient germinal center cells stimulated cell cycle progression and increased levels of antibody production. These findings reveal that the germinal center produces and uses molecules of the innate immune system, evolutionarily pirating them in order to optimally generate high affinity antibody responses.
Collapse
Affiliation(s)
- M Kopf
- Basel Institute for Immunology, CH-4005 Basel, Switzerland
| | | | | | | | | |
Collapse
|
32
|
Barton BE. IL-6: insights into novel biological activities. CLINICAL IMMUNOLOGY AND IMMUNOPATHOLOGY 1997; 85:16-20. [PMID: 9325064 DOI: 10.1006/clin.1997.4420] [Citation(s) in RCA: 191] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
IL-6 has many novel activities both within the adaptive immune system and without. It has therapeutic potential in acute inflammation, such as toxic or septic shock, and it is a potential target for cachexia, multiple myeloma, and osteoporosis. Further work on these aspects of IL-6 biology should yield new insight into the possibility of IL-6 both as a therapeutic agent and as a target for antagonists.
Collapse
Affiliation(s)
- B E Barton
- Department of Surgery, UMDNJ-NJMS, Newark 07103, USA.
| |
Collapse
|