1
|
Saini I, Joshi J, Kaur S. Unleashing the role of potential adjuvants in leishmaniasis. Int J Pharm 2025; 669:125077. [PMID: 39675537 DOI: 10.1016/j.ijpharm.2024.125077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 11/26/2024] [Accepted: 12/10/2024] [Indexed: 12/17/2024]
Abstract
Leishmaniasis is amongst one of the most neglected tropical disease, caused by an intracellular protozoan of genus Leishmania. Currently, the most promising strategy to combat leishmaniasis, relies on chemotherapy but the toxicity and increasing resistance of the standard drugs, presses the demand for new alternatives. Immunization is arguably the best strategy for cure because an individual once infected becomes immune to the disease. Yet, there is no efficient vaccine capable of providing enduring immunity against the parasite. Achieving the goal of developing highly efficacious and durable vaccine is limited due to lack of an appropriate adjuvant. Adjuvants are recognized as 'immune potentiators' which redirect or amplify the immune response. A number of adjuvants like alum, MPL-A, CpG ODN, GLA-SE, imiquimod, saponins etc. have been used in combination with various classes of Leishmania antigens. However, only few have reached clinical trials. Thus, the choice of an adjuvant is critically dependent on many factors such as the route of administration, the nature of antigen, formulation, the type of required immune response, their mode of action and the immunization schedule. This review provides an updated status on the types of adjuvants used in leishmaniasis so far and their mechanism of action if known.
Collapse
Affiliation(s)
- Isha Saini
- Parasitology Laboratory, Department of Zoology, Panjab University, Chandigarh, India
| | - Jyoti Joshi
- Goswami Ganesh Dutta Sanatan Dharma College, Sector-32C, Chandigarh, India
| | - Sukhbir Kaur
- Parasitology Laboratory, Department of Zoology, Panjab University, Chandigarh, India.
| |
Collapse
|
2
|
Mutsaers SE, Miles T, Prêle CM, Hoyne GF. Emerging role of immune cells as drivers of pulmonary fibrosis. Pharmacol Ther 2023; 252:108562. [PMID: 37952904 DOI: 10.1016/j.pharmthera.2023.108562] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 11/01/2023] [Accepted: 11/06/2023] [Indexed: 11/14/2023]
Abstract
The pathogenesis of pulmonary fibrosis, including idiopathic pulmonary fibrosis (IPF) and other forms of interstitial lung disease, involves a complex interplay of various factors including host genetics, environmental pollutants, infection, aberrant repair and dysregulated immune responses. Highly variable clinical outcomes of some ILDs, in particular IPF, have made it difficult to identify the precise mechanisms involved in disease pathogenesis and thus the development of a specific cure or treatment to halt and reverse the decline in patient health. With the advent of in-depth molecular diagnostics, it is becoming evident that the pathogenesis of IPF is unlikely to be the same for all patients and therefore will likely require different treatment approaches. Chronic inflammation is a cardinal feature of IPF and is driven by both innate and adaptive immune responses. Inflammatory cells and activated fibroblasts secrete various pro-inflammatory cytokines and chemokines that perpetuate the inflammatory response and contribute to the recruitment and activation of more immune cells and fibroblasts. The balance between pro-inflammatory and regulatory immune cell subsets, as well as the interactions between immune cell types and resident cells within the lung microenvironment, ultimately determines the extent of fibrosis and the potential for resolution. This review examines the role of the innate and adaptive immune responses in pulmonary fibrosis, with an emphasis on IPF. The role of different immune cell types is discussed as well as novel anti-inflammatory and immunotherapy approaches currently in clinical trial or in preclinical development.
Collapse
Affiliation(s)
- Steven E Mutsaers
- Institute for Respiratory Health, The University of Western Australia, Nedlands, WA, Australia.
| | - Tylah Miles
- Institute for Respiratory Health, The University of Western Australia, Nedlands, WA, Australia
| | - Cecilia M Prêle
- Institute for Respiratory Health, The University of Western Australia, Nedlands, WA, Australia; School of Medical, Molecular and Forensic Sciences, Murdoch University, WA, Australia
| | - Gerard F Hoyne
- Institute for Respiratory Health, The University of Western Australia, Nedlands, WA, Australia; The School of Health Sciences and Physiotherapy, University of Notre Dame Australia, Fremantle, WA, Australia
| |
Collapse
|
3
|
Exploring the Role of Staphylococcus aureus in Inflammatory Diseases. Toxins (Basel) 2022; 14:toxins14070464. [PMID: 35878202 PMCID: PMC9318596 DOI: 10.3390/toxins14070464] [Citation(s) in RCA: 65] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Revised: 06/23/2022] [Accepted: 07/01/2022] [Indexed: 02/04/2023] Open
Abstract
Staphylococcus aureus is a very common Gram-positive bacterium, and S. aureus infections play an extremely important role in a variety of diseases. This paper describes the types of virulence factors involved, the inflammatory cells activated, the process of host cell death, and the associated diseases caused by S. aureus. S. aureus can secrete a variety of enterotoxins and other toxins to trigger inflammatory responses and activate inflammatory cells, such as keratinocytes, helper T cells, innate lymphoid cells, macrophages, dendritic cells, mast cells, neutrophils, eosinophils, and basophils. Activated inflammatory cells can express various cytokines and induce an inflammatory response. S. aureus can also induce host cell death through pyroptosis, apoptosis, necroptosis, autophagy, etc. This article discusses S. aureus and MRSA (methicillin-resistant S. aureus) in atopic dermatitis, psoriasis, pulmonary cystic fibrosis, allergic asthma, food poisoning, sarcoidosis, multiple sclerosis, and osteomyelitis. Summarizing the pathogenic mechanism of Staphylococcus aureus provides a basis for the targeted treatment of Staphylococcus aureus infection.
Collapse
|
4
|
Ripszky Totan A, Greabu M, Stanescu-Spinu II, Imre M, Spinu TC, Miricescu D, Ilinca R, Coculescu EC, Badoiu SC, Coculescu BI, Albu C. The Yin and Yang dualistic features of autophagy in thermal burn wound healing. Int J Immunopathol Pharmacol 2022; 36:3946320221125090. [PMID: 36121435 PMCID: PMC9490459 DOI: 10.1177/03946320221125090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/09/2022] Open
Abstract
Burn healing should be regarded as a dynamic process consisting of two main, interrelated phases: (a) the inflammatory phase when neutrophils and monocytes infiltrate the injury site, through localized vasodilation and fluid extravasation, and (b) the proliferative-remodeling phase, which represents a key event in wound healing. In the skin, both canonical autophagy (induced by starvation, oxidative stress, and environmental aggressions) and non-canonical or selective autophagy have evolved to play a discrete, but, essential, “housekeeping” role, for homeostasis, immune tolerance, and survival. Experimental data supporting the pro-survival roles of autophagy, highlighting its Yang, luminous and positive feature of this complex but insufficient explored molecular pathway, have been reported. Autophagic cell death describes an “excessive” degradation of important cellular components that are necessary for normal cell function. This deadly molecular mechanism brings to light the darker, concealed, Yin feature of autophagy. Autophagy seems to perform dual, conflicting roles in the angiogenesis context, revealing once again, its Yin–Yang features. Autophagy with its Yin–Yang features remains the shadow player, able to decide quietly whether the cell survives or dies.
Collapse
Affiliation(s)
- Alexandra Ripszky Totan
- Department of Biochemistry, 367124Carol Davila University of Medicine and Pharmacy, Faculty of Dental Medicine, Romania
| | - Maria Greabu
- Department of Biochemistry, 367124Carol Davila University of Medicine and Pharmacy, Faculty of Dental Medicine, Romania
| | - Iulia-Ioana Stanescu-Spinu
- Department of Biochemistry, 367124Carol Davila University of Medicine and Pharmacy, Faculty of Dental Medicine, Romania
| | - Marina Imre
- Department of Complete Denture, Carol Davila University of Medicine and Pharmacy, Faculty of Dental Medicine, Romania
| | - Tudor-Claudiu Spinu
- Department of Fixed Prosthodontics and Occlusology, Carol Davila University of Medicine and Pharmacy, Faculty of Dental Medicine, Romania
| | - Daniela Miricescu
- Department of Biochemistry, 367124Carol Davila University of Medicine and Pharmacy, Faculty of Dental Medicine, Romania
| | - Radu Ilinca
- Department of Biophysics, Carol Davila University of Medicine and Pharmacy, Faculty of Dental Medicine, Romania
| | - Elena Claudia Coculescu
- Department of Oral Pathology, Carol Davila University of Medicine and Pharmacy, Faculty of Dental Medicine, Romania
| | - Silviu Constantin Badoiu
- Department of Anatomy and Embryology, Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
| | - Bogdan-Ioan Coculescu
- Cantacuzino National Medico-Military Institute for Research and Development, Bucharest, Romania
| | - Crenguta Albu
- Department of Genetics, Carol Davila University of Medicine and Pharmacy, Faculty of Dental Medicine, Bucharest, Romania
| |
Collapse
|
5
|
Oyesola OO, Shanahan MT, Kanke M, Mooney BM, Webb LM, Smita S, Matheson MK, Campioli P, Pham D, Früh SP, McGinty JW, Churchill MJ, Cahoon JL, Sundaravaradan P, Flitter BA, Mouli K, Nadjsombati MS, Kamynina E, Peng SA, Cubitt RL, Gronert K, Lord JD, Rauch I, von Moltke J, Sethupathy P, Tait Wojno ED. PGD2 and CRTH2 counteract Type 2 cytokine-elicited intestinal epithelial responses during helminth infection. J Exp Med 2021; 218:e20202178. [PMID: 34283207 PMCID: PMC8294949 DOI: 10.1084/jem.20202178] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2020] [Revised: 04/28/2021] [Accepted: 06/21/2021] [Indexed: 01/22/2023] Open
Abstract
Type 2 inflammation is associated with epithelial cell responses, including goblet cell hyperplasia, that promote worm expulsion during intestinal helminth infection. How these epithelial responses are regulated remains incompletely understood. Here, we show that mice deficient in the prostaglandin D2 (PGD2) receptor CRTH2 and mice with CRTH2 deficiency only in nonhematopoietic cells exhibited enhanced worm clearance and intestinal goblet cell hyperplasia following infection with the helminth Nippostrongylus brasiliensis. Small intestinal stem, goblet, and tuft cells expressed CRTH2. CRTH2-deficient small intestinal organoids showed enhanced budding and terminal differentiation to the goblet cell lineage. During helminth infection or in organoids, PGD2 and CRTH2 down-regulated intestinal epithelial Il13ra1 expression and reversed Type 2 cytokine-mediated suppression of epithelial cell proliferation and promotion of goblet cell accumulation. These data show that the PGD2-CRTH2 pathway negatively regulates the Type 2 cytokine-driven epithelial program, revealing a mechanism that can temper the highly inflammatory effects of the anti-helminth response.
Collapse
Affiliation(s)
- Oyebola O. Oyesola
- Department of Immunology, University of Washington, Seattle, WA
- Baker Institute for Animal Health and Department of Microbiology and Immunology, Cornell University College of Veterinary Medicine, Ithaca, NY
| | - Michael T. Shanahan
- Department of Biomedical Sciences, Cornell University College of Veterinary Medicine, Ithaca, NY
| | - Matt Kanke
- Department of Biomedical Sciences, Cornell University College of Veterinary Medicine, Ithaca, NY
| | | | - Lauren M. Webb
- Department of Immunology, University of Washington, Seattle, WA
| | - Shuchi Smita
- Department of Immunology, University of Washington, Seattle, WA
| | | | - Pamela Campioli
- Baker Institute for Animal Health and Department of Microbiology and Immunology, Cornell University College of Veterinary Medicine, Ithaca, NY
| | - Duc Pham
- Baker Institute for Animal Health and Department of Microbiology and Immunology, Cornell University College of Veterinary Medicine, Ithaca, NY
| | - Simon P. Früh
- Baker Institute for Animal Health and Department of Microbiology and Immunology, Cornell University College of Veterinary Medicine, Ithaca, NY
| | - John W. McGinty
- Department of Immunology, University of Washington, Seattle, WA
| | - Madeline J. Churchill
- Department of Molecular Microbiology and Immunology, Oregon Health and Science University, Portland, OR
| | | | | | - Becca A. Flitter
- Vision Science Program, School of Optometry, University of California, Berkeley, Berkeley, CA
| | - Karthik Mouli
- Vision Science Program, School of Optometry, University of California, Berkeley, Berkeley, CA
| | | | - Elena Kamynina
- Baker Institute for Animal Health and Department of Microbiology and Immunology, Cornell University College of Veterinary Medicine, Ithaca, NY
| | - Seth A. Peng
- Baker Institute for Animal Health and Department of Microbiology and Immunology, Cornell University College of Veterinary Medicine, Ithaca, NY
| | - Rebecca L. Cubitt
- Baker Institute for Animal Health and Department of Microbiology and Immunology, Cornell University College of Veterinary Medicine, Ithaca, NY
- Department of Biomedical Sciences, Cornell University College of Veterinary Medicine, Ithaca, NY
| | - Karsten Gronert
- Vision Science Program, School of Optometry, University of California, Berkeley, Berkeley, CA
| | - James D. Lord
- Benaroya Research Institute at Virginia Mason Medical Center, Division of Gastroenterology, Seattle, WA
| | - Isabella Rauch
- Department of Molecular Microbiology and Immunology, Oregon Health and Science University, Portland, OR
| | | | - Praveen Sethupathy
- Department of Biomedical Sciences, Cornell University College of Veterinary Medicine, Ithaca, NY
| | | |
Collapse
|
6
|
Florsheim EB, Sullivan ZA, Khoury-Hanold W, Medzhitov R. Food allergy as a biological food quality control system. Cell 2021; 184:1440-1454. [PMID: 33450204 DOI: 10.1016/j.cell.2020.12.007] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 10/21/2020] [Accepted: 12/04/2020] [Indexed: 12/17/2022]
Abstract
Food is simultaneously a source of essential nutrients and a potential source of lethal toxins and pathogens. Consequently, multiple sensory mechanisms evolved to monitor the quality of food based on the presence and relative abundance of beneficial and harmful food substances. These include the olfactory, gustatory, and gut chemosensory systems. Here we argue that, in addition to these systems, allergic immunity plays a role in food quality control by mounting allergic defenses against food antigens associated with noxious substances. Exaggeration of these defenses can result in pathological food allergy.
Collapse
Affiliation(s)
- Esther B Florsheim
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Zuri A Sullivan
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06510, USA
| | - William Khoury-Hanold
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Ruslan Medzhitov
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06510, USA; Howard Hughes Medical Institute, New Haven, CT 06510, USA.
| |
Collapse
|
7
|
Hung LY, Tanaka Y, Herbine K, Pastore C, Singh B, Ferguson A, Vora N, Douglas B, Zullo K, Behrens EM, Li Hui Tan T, Kohanski MA, Bryce P, Lin C, Kambayashi T, Reed DR, Brown BL, Cohen NA, Herbert DR. Cellular context of IL-33 expression dictates impact on anti-helminth immunity. Sci Immunol 2020; 5:5/53/eabc6259. [PMID: 33188058 DOI: 10.1126/sciimmunol.abc6259] [Citation(s) in RCA: 87] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Accepted: 09/28/2020] [Indexed: 12/13/2022]
Abstract
Interleukin-33 (IL-33) is a pleiotropic cytokine that can promote type 2 inflammation but also drives immunoregulation through Foxp3+Treg expansion. How IL-33 is exported from cells to serve this dual role in immunosuppression and inflammation remains unclear. Here, we demonstrate that the biological consequences of IL-33 activity are dictated by its cellular source. Whereas IL-33 derived from epithelial cells stimulates group 2 innate lymphoid cell (ILC2)-driven type 2 immunity and parasite clearance, we report that IL-33 derived from myeloid antigen-presenting cells (APCs) suppresses host-protective inflammatory responses. Conditional deletion of IL-33 in CD11c-expressing cells resulted in lowered numbers of intestinal Foxp3+Treg cells that express the transcription factor GATA3 and the IL-33 receptor ST2, causing elevated IL-5 and IL-13 production and accelerated anti-helminth immunity. We demonstrate that cell-intrinsic IL-33 promoted mouse dendritic cells (DCs) to express the pore-forming protein perforin-2, which may function as a conduit on the plasma membrane facilitating IL-33 export. Lack of perforin-2 in DCs blocked the proliferative expansion of the ST2+Foxp3+Treg subset. We propose that perforin-2 can provide a plasma membrane conduit in DCs that promotes the export of IL-33, contributing to mucosal immunoregulation under steady-state and infectious conditions.
Collapse
Affiliation(s)
- Li-Yin Hung
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Yukinori Tanaka
- Department of Dental Anesthesiology and Pain Management, Tohoku University Hospital, Sendai, Miyagi 980-8574, Japan
| | - Karl Herbine
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Christopher Pastore
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Brenal Singh
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Annabel Ferguson
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Nisha Vora
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Bonnie Douglas
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Kelly Zullo
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Edward M Behrens
- Division of Rheumatology, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Tiffany Li Hui Tan
- Department of Otorhinolaryngology-Head and Neck Surgery, Perelman School of Medicine at The University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Michael A Kohanski
- Department of Otorhinolaryngology-Head and Neck Surgery, Perelman School of Medicine at The University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Paul Bryce
- Immunology and Inflammation Therapeutic Area, Sanofi US, Cambridge, MA 02319, USA
| | - Cailu Lin
- Monell Chemical Senses Center, Philadelphia, PA 19104, USA
| | - Taku Kambayashi
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | | | - Breann L Brown
- Department of Biochemistry, Center for Structural Biology, Vanderbilt University, Nashville, TN 37232, USA
| | - Noam A Cohen
- Department of Otorhinolaryngology-Head and Neck Surgery, Perelman School of Medicine at The University of Pennsylvania, Philadelphia, PA 19104, USA.,Monell Chemical Senses Center, Philadelphia, PA 19104, USA.,Michael J. Crescenz Veterans Affairs Medical Center Surgical Service, Philadelphia, PA 19104, USA
| | - De'Broski R Herbert
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
8
|
Galli SJ, Metz M, Starkl P, Marichal T, Tsai M. Mast cells and IgE in defense against lethality of venoms: Possible "benefit" of allergy[]. ALLERGO JOURNAL INTERNATIONAL 2020; 29:46-62. [PMID: 33224714 PMCID: PMC7673288 DOI: 10.1007/s40629-020-00118-6] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Accepted: 10/20/2019] [Indexed: 01/15/2023]
Abstract
Physicians think of mast cells and IgE primarily in the context of allergic disorders, including fatal anaphylaxis. This 'bad side' of mast cells and IgE is so well accepted that it can be difficult to think of them in other contexts, particularly those in which they may have beneficial functions. However, there is evidence that mast cells and IgE, as well as basophils (circulating granulocytes whose functions partially overlap with those of mast cells), can contribute to host defense as components of adaptive type 2 immune responses to helminths, ticks and certain other parasites. Accordingly, allergies often are conceptualized as "misdirected" type 2 immune responses, in which IgE antibodies are produced against any of a diverse group of apparently harmless antigens, and against components of animal venoms. Indeed, certain unfortunate patients who have become sensitized to venoms develop severe IgE-associated allergic reactions, including fatal anaphylaxis, upon subsequent venom exposure. In this review, we will describe evidence that mast cells can enhance innate resistance, and survival, to challenge with reptile or arthropod venoms during a first exposure to such venoms. We also will discuss findings indicating that, in mice surviving an initial encounter with venom, acquired type 2 immune responses, IgE antibodies, the high affinity IgE receptor (FcεRI), and mast cells can contribute to acquired resistance to the lethal effects of both honeybee venom and Russell's viper venom. These findings support the hypothesis that mast cells and IgE can help protect the host against venoms and perhaps other noxious substances.
Collapse
Affiliation(s)
- Stephen J. Galli
- Department of Pathology and the Sean N. Parker Center for Allergy Research, Stanford University School of Medicine, Stanford, California, 94305; USA
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, California, 94305; USA
| | - Martin Metz
- Department of Dermatology and Allergy, Charité – Universitätsmedizin Berlin, 10117 Berlin, Germany
| | - Philipp Starkl
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, 1090 Vienna, Austria
- Department of Medicine 1, Laboratory of Infection Biology, Medical University of Vienna, 1090 Vienna, Austria
| | - Thomas Marichal
- GIGA-Research and Faculty of Veterinary Medicine, University of Liege, 4000 Liege, Belgium
| | - Mindy Tsai
- Department of Pathology and the Sean N. Parker Center for Allergy Research, Stanford University School of Medicine, Stanford, California, 94305; USA
| |
Collapse
|
9
|
Galli SJ, Metz M, Starkl P, Marichal T, Tsai M. Mast cells and IgE in defense against lethality of venoms: Possible "benefit" of allergy*. ALLERGO JOURNAL 2020. [DOI: 10.1007/s15007-020-0746-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
10
|
Sil P, Wong SW, Martinez J. More Than Skin Deep: Autophagy Is Vital for Skin Barrier Function. Front Immunol 2018; 9:1376. [PMID: 29988591 PMCID: PMC6026682 DOI: 10.3389/fimmu.2018.01376] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Accepted: 06/04/2018] [Indexed: 12/30/2022] Open
Abstract
The skin is a highly organized first line of defense that stretches up to 1.8 m2 and is home to more than a million commensal bacteria. The microenvironment of skin is driven by factors such as pH, temperature, moisture, sebum level, oxidative stress, diet, resident immune cells, and infectious exposure. The skin has a high turnover of cells as it continually bares itself to environmental stresses. Notwithstanding these limitations, it has devised strategies to adapt as a nutrient-scarce site. To perform its protective function efficiently, it relies on mechanisms to continuously remove dead cells without alarming the immune system, actively purging the dying/senescent cells by immunotolerant efferocytosis. Both canonical (starvation-induced, reactive oxygen species, stress, and environmental insults) and non-canonical (selective) autophagy in the skin have evolved to perform astute due-diligence and housekeeping in a quiescent fashion for survival, cellular functioning, homeostasis, and immune tolerance. The autophagic “homeostatic rheostat” works tirelessly to uphold the delicate balance in immunoregulation and tolerance. If this equilibrium is upset, the immune system can wreak havoc and initiate pathogenesis. Out of all the organs, the skin remains under-studied in the context of autophagy. Here, we touch upon some of the salient features of autophagy active in the skin.
Collapse
Affiliation(s)
- Payel Sil
- Immunity, Inflammation, and Disease Laboratory, National Institute of Environmental Health Sciences, Durham, NC, United States
| | - Sing-Wai Wong
- Immunity, Inflammation, and Disease Laboratory, National Institute of Environmental Health Sciences, Durham, NC, United States.,Oral and Craniofacial Biomedicine Curriculum, School of Dentistry, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Jennifer Martinez
- Immunity, Inflammation, and Disease Laboratory, National Institute of Environmental Health Sciences, Durham, NC, United States
| |
Collapse
|
11
|
Bao K, Carr T, Wu J, Barclay W, Jin J, Ciofani M, Reinhardt RL. BATF Modulates the Th2 Locus Control Region and Regulates CD4+ T Cell Fate during Antihelminth Immunity. THE JOURNAL OF IMMUNOLOGY 2016; 197:4371-4381. [PMID: 27798167 DOI: 10.4049/jimmunol.1601371] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/08/2016] [Accepted: 09/27/2016] [Indexed: 11/19/2022]
Abstract
The AP-1 factor basic leucine zipper transcription factor, ATF-like (BATF) is important for CD4+ Th17, Th9, and follicular Th cell development. However, its precise role in Th2 differentiation and function remains unclear, and the requirement for BATF in nonallergic settings of type-2 immunity has not been explored. In this article, we show that, in response to parasitic helminths, Batf-/- mice are unable to generate follicular Th and Th2 cells. As a consequence, they fail to establish productive type-2 immunity during primary and secondary infection. Batf-/- CD4+ T cells do not achieve type-2 cytokine competency, which implies that BATF plays a key role in the regulation of IL-4 and IL-13. In contrast to Th17 and Th9 cell subsets in which BATF binds directly to promoter and enhancer regions to regulate cytokine expression, our results show that BATF is significantly enriched at Rad50 hypersensitivity site (RHS)6 and RHS7 of the locus control region relative to AP-1 sites surrounding type-2 cytokine loci in Th2 cells. Indeed, Batf-/- CD4+ T cells do not obtain permissive epigenetic modifications within the Th2 locus, which were linked to RHS6 and RHS7 function. In sum, these findings reveal BATF as a central modulator of peripheral and humoral hallmarks of type-2 immunity and begin to elucidate a novel mechanism by which it regulates type-2 cytokine production through its modification of the Th2 locus control region.
Collapse
Affiliation(s)
- Katherine Bao
- Department of Immunology, Duke University Medical Center, Durham, NC 27710; and
| | - Tiffany Carr
- Department of Immunology, Duke University Medical Center, Durham, NC 27710; and
| | - Jianxuan Wu
- Department of Immunology, Duke University Medical Center, Durham, NC 27710; and
| | - William Barclay
- Department of Immunology, Duke University Medical Center, Durham, NC 27710; and
| | - Jingxiao Jin
- Department of Immunology, Duke University Medical Center, Durham, NC 27710; and
| | - Maria Ciofani
- Department of Immunology, Duke University Medical Center, Durham, NC 27710; and
| | - R Lee Reinhardt
- Department of Biomedical Research, National Jewish Health, Denver, CO 80206
| |
Collapse
|
12
|
The Microbiome, Timing, and Barrier Function in the Context of Allergic Disease. Immunity 2016; 44:728-38. [PMID: 27096316 DOI: 10.1016/j.immuni.2016.02.002] [Citation(s) in RCA: 106] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2015] [Indexed: 12/14/2022]
Abstract
Allergic disease affects millions. Despite many advances in our understanding of the immune system in the past century, the physiologic underpinning for the existence of allergy remains largely mysterious. Food allergies, in particular, have increased dramatically in recent years, adding a new sense of urgency to unraveling this mystery. The concurrence of significant lifestyle changes in Western societies with increasing disease prevalence implies a causal link. Demographic variables that influence the composition and function of the commensal microbiota early in life seem to be most important. Identifying the evolutionary and physiologic foundations of allergic disease and defining what about our modern environment is responsible for its increased incidence will provide insights critical to the development of new approaches to prevention and treatment.
Collapse
|
13
|
Galli SJ. The Mast Cell-IgE Paradox: From Homeostasis to Anaphylaxis. THE AMERICAN JOURNAL OF PATHOLOGY 2016; 186:212-24. [PMID: 26776074 DOI: 10.1016/j.ajpath.2015.07.025] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/10/2015] [Revised: 07/23/2015] [Accepted: 07/30/2015] [Indexed: 12/20/2022]
Abstract
Mast cells and IgE are so inextricably linked to the pathology of allergic disorders, including fatal anaphylaxis, that it can be difficult to think of them in other contexts. Surely, we do not have mast cells and IgE so that we can eat a peanut and die! It is thought that mast cells and IgE and basophils (circulating granulocytes, whose functions partially overlap with those of mast cells) can contribute to host defense as components of adaptive T helper cell type 2 immune responses to helminths, ticks, and certain other parasites. Accordingly, it was suggested that allergies are misdirected type 2 immune responses in which IgE antibodies are produced against any of a broad variety of apparently harmless antigens. However, components of animal venoms also can sensitize individuals to develop severe IgE-associated allergic reactions, including fatal anaphylaxis, on subsequent venom exposure. Here, I describe evidence that mast cells can enhance innate host resistance to reptile or arthropod venoms during responses to an initial exposure to such venoms and that acquired type 2 immune responses, IgE antibodies, the high-affinity IgE receptor FcεRI, and mast cells can contribute toward acquired resistance in mice to the lethal effects of honeybee or Russell's viper venom. These findings support the hypothesis that mast cells and IgE can help protect the host against noxious substances.
Collapse
Affiliation(s)
- Stephen J Galli
- Department of Pathology, Stanford University School of Medicine, Stanford, California; Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, California; Sean N. Parker Center for Allergy Research, Stanford University School of Medicine, Stanford, California.
| |
Collapse
|
14
|
Galli SJ, Starkl P, Marichal T, Tsai M. Mast cells and IgE in defense against venoms: Possible "good side" of allergy? Allergol Int 2016; 65:3-15. [PMID: 26666482 DOI: 10.1016/j.alit.2015.09.002] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Accepted: 09/03/2015] [Indexed: 01/05/2023] Open
Abstract
Physicians think of mast cells and IgE primarily in the context of allergic disorders, including fatal anaphylaxis. This 'bad side' of mast cells and IgE is so well accepted that it can be difficult to think of them in other contexts, particularly those in which they may have beneficial functions. However, there is evidence that mast cells and IgE, as well as basophils (circulating granulocytes whose functions partially overlap with those of mast cells), can contribute to host defense as components of adaptive type 2 immune responses to helminths, ticks and certain other parasites. Accordingly, allergies often are conceptualized as "misdirected" type 2 immune responses, in which IgE antibodies are produced against any of a diverse group of apparently harmless antigens, as well as against components of animal venoms. Indeed, certain unfortunate patients who have become sensitized to venoms develop severe IgE-associated allergic reactions, including fatal anaphylaxis, upon subsequent venom exposure. In this review, we will describe evidence that mast cells can enhance innate resistance to reptile or arthropod venoms during a first exposure to such venoms. We also will discuss findings indicating that, in mice which survive an initial encounter with venom, acquired type 2 immune responses, IgE antibodies, the high affinity IgE receptor (FcɛRI), and mast cells can contribute to acquired resistance to the lethal effects of both honeybee venom and Russell's viper venom. These findings support the hypothesis that mast cells and IgE can help protect the host against venoms and perhaps other noxious substances.
Collapse
Affiliation(s)
- Stephen J Galli
- Department of Pathology and the Sean N. Parker Center for Allergy Research, Stanford University School of Medicine, Stanford, CA, USA; Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA.
| | - Philipp Starkl
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria; Department of Medicine 1, Laboratory of Infection Biology, Medical University of Vienna, Vienna, Austria
| | - Thomas Marichal
- GIGA-Research and Faculty of Veterinary Medicine, University of Liege, Liege, Belgium
| | - Mindy Tsai
- Department of Pathology and the Sean N. Parker Center for Allergy Research, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
15
|
Ibarra-Coronado EG, Pantaleón-Martínez AM, Velazquéz-Moctezuma J, Prospéro-García O, Méndez-Díaz M, Pérez-Tapia M, Pavón L, Morales-Montor J. The Bidirectional Relationship between Sleep and Immunity against Infections. J Immunol Res 2015; 2015:678164. [PMID: 26417606 PMCID: PMC4568388 DOI: 10.1155/2015/678164] [Citation(s) in RCA: 129] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2014] [Accepted: 12/24/2014] [Indexed: 12/31/2022] Open
Abstract
Sleep is considered an important modulator of the immune response. Thus, a lack of sleep can weaken immunity, increasing organism susceptibility to infection. For instance, shorter sleep durations are associated with a rise in suffering from the common cold. The function of sleep in altering immune responses must be determined to understand how sleep deprivation increases the susceptibility to viral, bacterial, and parasitic infections. There are several explanations for greater susceptibility to infections after reduced sleep, such as impaired mitogenic proliferation of lymphocytes, decreased HLA-DR expression, the upregulation of CD14+, and variations in CD4+ and CD8+ T lymphocytes, which have been observed during partial sleep deprivation. Also, steroid hormones, in addition to regulating sexual behavior, influence sleep. Thus, we hypothesize that sleep and the immune-endocrine system have a bidirectional relationship in governing various physiological processes, including immunity to infections. This review discusses the evidence on the bidirectional effects of the immune response against viral, bacterial, and parasitic infections on sleep patterns and how the lack of sleep affects the immune response against such agents. Because sleep is essential in the maintenance of homeostasis, these situations must be adapted to elicit changes in sleep patterns and other physiological parameters during the immune response to infections to which the organism is continuously exposed.
Collapse
Affiliation(s)
- Elizabeth G. Ibarra-Coronado
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, AP 70228, 04510 México, DF, Mexico
| | - Ana Ma. Pantaleón-Martínez
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, AP 70228, 04510 México, DF, Mexico
| | - Javier Velazquéz-Moctezuma
- Area de Neurociencias, Departmento de Biología de la Reproduccion, CBS, Universidad Autonoma Metropolitana-Iztapalapa, Avenida San Rafael Atlixco No. 186, Col Vicentina, Iztapalapa, 09340 Mexico City, DF, Mexico
| | - Oscar Prospéro-García
- Grupo de Neurociencias, Laboratorio de Canabinoides, Departamento de Fisiología, Facultad de Medicina, Universidad Nacional Autónoma de México, México, DF, Mexico
| | - Mónica Méndez-Díaz
- Grupo de Neurociencias, Laboratorio de Canabinoides, Departamento de Fisiología, Facultad de Medicina, Universidad Nacional Autónoma de México, México, DF, Mexico
| | - Mayra Pérez-Tapia
- Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, IPN Prolongación de Carpio y Plan de Ayala s/n, Col. Sto. Tomás, 11340 México, DF, Mexico
- Unidad de Desarrollo e Investigación en Bioprocesos (UDIBI), Escuela Nacional de Ciencias Biológicas, IPN Prolongación de Carpio y Plan de Ayala s/n, Col. Sto. Tomás, 11340 México, DF, Mexico
| | - Lenin Pavón
- Laboratorio de Psicoinmunología, Dirección de Investigaciones en Neurociencias, Instituto Nacional de Psiquiatría Ramón de la Fuente, 14370 México, DF, Mexico
| | - Jorge Morales-Montor
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, AP 70228, 04510 México, DF, Mexico
| |
Collapse
|
16
|
Tsai M, Starkl P, Marichal T, Galli SJ. Testing the 'toxin hypothesis of allergy': mast cells, IgE, and innate and acquired immune responses to venoms. Curr Opin Immunol 2015. [PMID: 26210895 DOI: 10.1016/j.coi.2015.07.001] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Work in mice indicates that innate functions of mast cells, particularly degradation of venom toxins by mast cell-derived proteases, can enhance resistance to certain arthropod or reptile venoms. Recent reports indicate that acquired Th2 immune responses associated with the production of IgE antibodies, induced by Russell's viper venom or honeybee venom, or by a component of honeybee venom, bee venom phospholipase 2 (bvPLA2), can increase the resistance of mice to challenge with potentially lethal doses of either of the venoms or bvPLA2. These findings support the conclusion that, in contrast to the detrimental effects associated with allergic type 2 (Th2) immune responses, mast cells and IgE-dependent immune responses to venoms can contribute to innate and adaptive resistance to venom-induced pathology and mortality.
Collapse
Affiliation(s)
- Mindy Tsai
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Philipp Starkl
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Thomas Marichal
- GIGA-Research and Faculty of Veterinary Medicine, University of Liege, 4000 Liege, Belgium
| | - Stephen J Galli
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Microbiology & Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA.
| |
Collapse
|
17
|
Abstract
CD4(+) T cells are key cells of the adaptive immune system that use T cell antigen receptors to recognize peptides that are generated in endosomes or phagosomes and displayed on the host cell surface bound to major histocompatibility complex molecules. These T cells participate in immune responses that protect hosts from microbes such as Mycobacterium tuberculosis, Cryptococcus neoformans, Leishmania major, and Salmonella enterica, which have evolved to live in the phagosomes of macrophages and dendritic cells. Here, we review studies indicating that CD4(+) T cells control phagosomal infections asymptomatically in most individuals by secreting cytokines that activate the microbicidal activities of infected phagocytes but in a way that inhibits the pathogen but does not eliminate it. Indeed, we make the case that localized, controlled, persistent infection is necessary to maintain large numbers of CD4(+) effector T cells in a state of activation needed to eradicate systemic and more pathogenic forms of the infection. Finally, we posit that current vaccines for phagosomal infections fail because they do not produce this "periodic reminder" form of CD4(+) T cell-mediated immune control.
Collapse
|
18
|
Marichal T, Starkl P, Reber LL, Kalesnikoff J, Oettgen HC, Tsai M, Metz M, Galli SJ. A beneficial role for immunoglobulin E in host defense against honeybee venom. Immunity 2013; 39:963-75. [PMID: 24210352 PMCID: PMC4164235 DOI: 10.1016/j.immuni.2013.10.005] [Citation(s) in RCA: 143] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2013] [Accepted: 08/21/2013] [Indexed: 10/26/2022]
Abstract
Allergies are widely considered to be misdirected type 2 immune responses, in which immunoglobulin E (IgE) antibodies are produced against any of a broad range of seemingly harmless antigens. However, components of insect venoms also can sensitize individuals to develop severe IgE-associated allergic reactions, including fatal anaphylaxis, upon subsequent venom exposure. We found that mice injected with amounts of honeybee venom similar to that which could be delivered in one or two stings developed a specific type 2 immune response that increased their resistance to subsequent challenge with potentially lethal amounts of the venom. Our data indicate that IgE antibodies and the high affinity IgE receptor, FcεRI, were essential for such acquired resistance to honeybee venom. The evidence that IgE-dependent immune responses against venom can enhance survival in mice supports the hypothesis that IgE, which also contributes to allergic disorders, has an important function in protection of the host against noxious substances.
Collapse
Affiliation(s)
- Thomas Marichal
- Department of Pathology, Stanford University School of Medicine, Stanford, California, 94305; USA
| | - Philipp Starkl
- Department of Pathology, Stanford University School of Medicine, Stanford, California, 94305; USA
| | - Laurent L. Reber
- Department of Pathology, Stanford University School of Medicine, Stanford, California, 94305; USA
| | - Janet Kalesnikoff
- Department of Pathology, Stanford University School of Medicine, Stanford, California, 94305; USA
| | - Hans C. Oettgen
- Department of Microbiology & Immunology, Stanford University School of Medicine, Stanford, California, 94305; USA
| | - Mindy Tsai
- Department of Pathology, Stanford University School of Medicine, Stanford, California, 94305; USA
| | - Martin Metz
- Department of Pathology, Stanford University School of Medicine, Stanford, California, 94305; USA
- Division of Immunology, Boston Children’s Hospital, Boston, Massachusetts, 02115; USA
| | - Stephen J. Galli
- Department of Pathology, Stanford University School of Medicine, Stanford, California, 94305; USA
| |
Collapse
|
19
|
Bronevetsky Y, Villarino AV, Eisley CJ, Barbeau R, Barczak AJ, Heinz GA, Kremmer E, Heissmeyer V, McManus MT, Erle DJ, Rao A, Ansel KM. T cell activation induces proteasomal degradation of Argonaute and rapid remodeling of the microRNA repertoire. ACTA ACUST UNITED AC 2013; 210:417-32. [PMID: 23382546 PMCID: PMC3570096 DOI: 10.1084/jem.20111717] [Citation(s) in RCA: 168] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
CD4+ T cell activation–induced Argonaute degradation and global miRNA downregulation promotes acquisition of helper T cell effector functions. Activation induces extensive changes in the gene expression program of naive CD4+ T cells, promoting their differentiation into helper T cells that coordinate immune responses. MicroRNAs (miRNAs) play a critical role in this process, and miRNA expression also changes dramatically during T cell differentiation. Quantitative analyses revealed that T cell activation induces global posttranscriptional miRNA down-regulation in vitro and in vivo. Argonaute (Ago) proteins, the core effector proteins of the miRNA-induced silencing complex (miRISC), were also posttranscriptionally down-regulated during T cell activation. Ago2 was inducibly ubiquitinated in activated T cells and its down-regulation was inhibited by the proteasome inhibitor MG132. Therefore, activation-induced miRNA down-regulation likely occurs at the level of miRISC turnover. Measurements of miRNA-processing intermediates uncovered an additional layer of activation-induced, miRNA-specific transcriptional regulation. Thus, transcriptional and posttranscriptional mechanisms cooperate to rapidly reprogram the miRNA repertoire in differentiating T cells. Altering Ago2 expression in T cells revealed that Ago proteins are limiting factors that determine miRNA abundance. Naive T cells with reduced Ago2 and miRNA expression differentiated more readily into cytokine-producing helper T cells, suggesting that activation-induced miRNA down-regulation promotes acquisition of helper T cell effector functions by relaxing the repression of genes that direct T cell differentiation.
Collapse
Affiliation(s)
- Yelena Bronevetsky
- Sandler Asthma Basic Research Center, University of California San Francisco, San Francisco, CA 94143, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
|
21
|
Innate immunity modulation by the IL-33/ST2 system in intestinal mucosa. BIOMED RESEARCH INTERNATIONAL 2012; 2013:142492. [PMID: 23484079 PMCID: PMC3591220 DOI: 10.1155/2013/142492] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/14/2012] [Accepted: 10/29/2012] [Indexed: 12/23/2022]
Abstract
Innate immunity prevents pathogens from entering and spreading within the body. This function is especially important in the gastrointestinal tract and skin, as these organs have a large surface contact area with the outside environment. In the intestine, luminal commensal bacteria are necessary for adequate food digestion and play a crucial role in tolerance to benign antigens. Immune system damage can create an intestinal inflammatory response, leading to chronic disease including inflammatory bowel diseases (IBD). Ulcerative colitis (UC) is an IBD of unknown etiology with increasing worldwide prevalence. In the intestinal mucosa of UC patients, there is an imbalance in the IL-33/ST2 axis, an important modulator of the innate immune response. This paper reviews the role of the IL-33/ST2 system in innate immunity of the intestinal mucosa and its importance in inflammatory bowel diseases, especially ulcerative colitis.
Collapse
|
22
|
Chi H, Zhang Z, Inami M, Bøgwald J, Zhan W, Dalmo RA. Molecular characterizations and functional assessments of GATA-3 and its splice variant in Atlantic cod (Gadus morhua L.). DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2012; 36:491-501. [PMID: 21978454 DOI: 10.1016/j.dci.2011.09.004] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/06/2011] [Revised: 09/08/2011] [Accepted: 09/09/2011] [Indexed: 05/31/2023]
Abstract
GATA-3 is a master transcription factor of the Th2 cells. We have identified GATA-3 cDNA and its splice variant in Atlantic cod. Cod GATA-3 (GmGATA-3) has a 1320 b p open reading frame encoding a polypeptide of 440 amino acids with two zinc finger domains that are well conserved within teleosts and higher vertebrates. The GATA-3 cDNA splice variant without zinc finger domains was shown to contain an 828 b p open reading frame encoding a polypeptide of 276 amino acids. Both GATA-3 proteins fused with RFP-tag were identified in or close to the nuclei 48 h after the plasmids were transfected in CHSE-214 cells. The full length GATA-3 with two zinc finger domains has a transcriptional function confirmed by transfection with GATA-3 reporter vector along with expression constructs of GATA-3 plasmids in CHSE-214 cells, whereas the GATA-3 splice variant without zinc finger domain did not enhance the activity of the GATA-3 reporter vector, and no interference was found between these two GATA-3 variants. RT-PCR analysis revealed that the two Atlantic cod GATA-3 variants were strongly expressed in the gills and infection with live Vibrio anguillarum induced the spleen expression of both GmGATA-3L and GmGATA-3S. Unexpectedly, PMA increased the expression of the GATA-3 splice variant in vivo and especially in vitro, with an increase of more than 100,000-fold in head kidney leukocytes at 24 and 48 h. On the other hand, there were no significant increases at the transcript level of full length GATA-3 between Poly I:C and β-glucan treatment groups compared to controls.
Collapse
Affiliation(s)
- Heng Chi
- Norwegian College of Fishery Science, University of Tromsø, N-9037 Tromsø, Norway
| | | | | | | | | | | |
Collapse
|
23
|
Abstract
Initial recognition of bacteria by the innate immune system is thought to occur primarily by germline-encoded pattern recognition receptors (PRRs). These receptors are present in multiple compartments of host cells and are thus capable of surveying both the intracellular and extracellular milieu for bacteria. It has generally been presumed that the cellular location of these receptors dictates what type of bacteria they respond to: extracellular bacteria being recognized by cell surface receptors, such as certain Toll-like receptors, and bacteria that are capable of breaching the plasma membrane and entering the cytoplasm, being sensed by cytoplasmic receptors, including the Nod-like receptors (NLRs). Increasingly, it is becoming apparent that this is a false dichotomy and that extracellular bacteria can be sensed by cytoplasmic PRRs and this is crucial for controlling the levels of these bacteria. In this review, we discuss the role of two NLRs, Nod1 and Nod2, in the recognition of and response to extracellular bacteria.
Collapse
Affiliation(s)
- Thomas B Clarke
- Department of Microbiology, University of Pennsylvania School of Medicine, Philadelphia, PA 19104-6076, USA
| | | |
Collapse
|
24
|
Chegini N. Proinflammatory and profibrotic mediators: principal effectors of leiomyoma development as a fibrotic disorder. Semin Reprod Med 2010; 28:180-203. [PMID: 20414842 PMCID: PMC3057653 DOI: 10.1055/s-0030-1251476] [Citation(s) in RCA: 105] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Leiomyomas are believed to derive from the transformation of myometrial smooth muscle cells/connective tissue fibroblasts. Although the identity of the molecule(s) that initiate such cellular transformation and orchestrate subsequent growth is still unknown, conventional evidence indicates that ovarian steroids are essential for leiomyoma growth. Ovarian steroid action in their target cell/tissue is mediated in part through local expression of various growth factors, cytokines, and chemokines. These autocrine/paracrine molecules with proinflammatory and profibrotic activities serve as major contributing factors in regulating cellular transformation, cell growth and apoptosis, angiogenesis, cellular hypertrophy, and excess tissue turnover, events central to leiomyoma growth. This review addresses the key regulatory functions of proinflammatory and profibrotic mediators and their molecular mechanisms, downstream signaling that regulates cellular events that result in transformation, and commitments of specific cells into forming a cellular environment with a possible role in development and subsequent growth of leiomyomas.
Collapse
Affiliation(s)
- Nasser Chegini
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, University of Florida, Gainesville, Florida 32610, USA.
| |
Collapse
|
25
|
Sierra-Puente RE, Campos-Rodríguez R, Jarillo-Luna RA, Muñoz-Fernández L, Rodríguez MG, Muñoz-Ortega MH, Ventura-Juárez J. Expression of immune modulator cytokines in human fulminant amoebic colitis. Parasite Immunol 2009; 31:384-91. [PMID: 19527454 DOI: 10.1111/j.1365-3024.2009.01118.x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Human fulminant amoebic colitis (FAC) is characterized by ulceration and inflammation of the colon. The specific mixture of pro-inflammatory and anti-inflammatory cytokines may participate in either the host defense or in the pathogenesis of amoebic colitis. Therefore, we studied the expression of IL-8, IL-10, IL-4, TGF-beta and IFN-gamma in human FAC patients and controls through immunohistochemistry analysis. The number of cells expressing IL-8, IL-4 and IL-10 was significantly enhanced in all FAC samples compared to the control samples. However, the expression of TGF- beta in patients was low in the colonic mucosa and high in the lamina propria compared with the control. No expression of IFN-gamma was found in the controls or FAC samples. The production of IL-8 by intestinal epithelial cells may play a role in the pathogenesis of amoebic infection, because this cytokine attracts neutrophils, which lead to an inflammatory reaction that results in tissue damage. The predominant expression of the macrophage down-regulating cytokines, IL-4, IL-10 and TGF-beta, or the Th2-type immune response could inhibit a cell-mediated immune response, which in turn would facilitate parasite invasion in these tissues.
Collapse
Affiliation(s)
- R E Sierra-Puente
- Departamento de Histología, Facultad de Medicina, Universidad Juárez del Estado de Durango, Durango, México
| | | | | | | | | | | | | |
Collapse
|
26
|
Abstract
In this Commentary, McKee et al. highlight the properties of extrinsic vaccine adjuvants that must be considered to achieve the most protective immune response, as occurs naturally with many intrinsic pathogen-derived adjuvants.
Collapse
|
27
|
|
28
|
|
29
|
Hattori H, Rosas LE, Okano M, Durbin JE, Nishizaki K, Satoskar AR. STAT1 is involved in the pathogenesis of murine allergic rhinitis. ACTA ACUST UNITED AC 2007; 21:241-7. [PMID: 17424888 DOI: 10.2500/ajr.2007.21.2970] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
BACKGROUND Signal transducer and activator of transcription (STAT) 1 signaling pathway mediates biological functions of interferon (IFN) gamma, which is a key cytokine-regulating T helper 1 (Thl) differentiation. Although constitutive activation of STAT1 has been reported in the airway epithelium of patients with chronic asthma, its in vivo role in the pathogenesis of allergic rhinitis is not clear. We determined the role of STAT1 in the pathogenesis of allergic rhinitis in vivo using STAT1 gene-deficient (STAT1-/-) mice and a murine model of Schistosoma mansoni egg antigen (SEA)-induced allergic rhinitis. METHODS STATI -/- BALB/c and wild-type (WT) mice were sensitized by intranasal administration of SEA, and their immunologic responses were examined. RESULTS STATI-1- mice showed impaired nasal eosinophilia and markedly reduced histamine-induced nasal hyperresponsiveness after SEA sensitization. Moreover, levels of Th2-associated SEA-specific IgG1 and IgE antibodies were lower in STAT1-/- mice. Anti-CD3stimulated nasal lymphocytes from STAT1-/-mice also produced less amounts of Th2-associated cytokines IL-4, IL-5, IL-10, and IL-13 compared with WT mice, but both produced comparable levels of IFN-gamma. CONCLUSION These results show that STAT1 is involved in the pathogenesis of SEA-induced allergic rhinitis in BALB/c mice. Furthermore, they reveal a surprising role of STAT1 in induction of nasal eosinophilia, and Th2-type cytokine production from nasal lymphocytes during allergic rhinitis.
Collapse
Affiliation(s)
- Hisashi Hattori
- Department of Microbiology, The Ohio State University, Columbus, Ohio, USA
| | | | | | | | | | | |
Collapse
|
30
|
Fritz JH, Le Bourhis L, Sellge G, Magalhaes JG, Fsihi H, Kufer TA, Collins C, Viala J, Ferrero RL, Girardin SE, Philpott DJ. Nod1-mediated innate immune recognition of peptidoglycan contributes to the onset of adaptive immunity. Immunity 2007; 26:445-59. [PMID: 17433730 DOI: 10.1016/j.immuni.2007.03.009] [Citation(s) in RCA: 236] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2006] [Revised: 02/22/2007] [Accepted: 03/05/2007] [Indexed: 01/07/2023]
Abstract
Recent evidence has suggested that signals other than those from Toll-like receptors (TLRs) could contribute to the elicitation of antigen-specific immunity. Therefore, we examined the role of the Nod-like receptor (NLR) family member, Nod1, in the generation of adaptive immune responses. Our findings show that innate immune sensing of peptidoglycan by Nod1 is key for priming antigen-specific T cell immunity and subsequent antibody responses in vivo. Nod1 stimulation alone was sufficient to drive antigen-specific immunity with a predominant Th2 polarization profile. In conjunction with TLR stimulation, however, Nod1 triggering was required to instruct the onset of Th1 and Th2 as well as Th17 immune pathways. Cells outside of the hematopoietic lineage provided the early signals necessary to orchestrate the development of Nod1-dependent immune responses. These findings highlight Nod1 as a key innate immune trigger in the local tissue microenvironment that drives the development of adaptive immunity.
Collapse
Affiliation(s)
- Jörg H Fritz
- Department of Immunology, Medical Sciences Building, University of Toronto, 1 King's College Circle, M5S 1A8 Toronto, Ontario, Canada
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Djuretic IM, Levanon D, Negreanu V, Groner Y, Rao A, Ansel KM. Transcription factors T-bet and Runx3 cooperate to activate Ifng and silence Il4 in T helper type 1 cells. Nat Immunol 2006; 8:145-53. [PMID: 17195845 DOI: 10.1038/ni1424] [Citation(s) in RCA: 422] [Impact Index Per Article: 22.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2006] [Accepted: 11/14/2006] [Indexed: 11/08/2022]
Abstract
Cell differentiation involves activation and silencing of lineage-specific genes. Here we show that the transcription factor Runx3 is induced in T helper type 1 (T(H)1) cells in a T-bet-dependent manner, and that both transcription factors T-bet and Runx3 are required for maximal production of interferon-gamma (IFN-gamma) and silencing of the gene encoding interleukin 4 (Il4) in T(H)1 cells. T-bet does not repress Il4 in Runx3-deficient T(H)2 cells, but coexpression of Runx3 and T-bet induces potent repression in those cells. Both T-bet and Runx3 bind to the Ifng promoter and the Il4 silencer, and deletion of the silencer decreases the sensitivity of Il4 to repression by either factor. Our data indicate that cytokine gene expression in T(H)1 cells may be controlled by a feed-forward regulatory circuit in which T-bet induces Runx3 and then 'partners' with Runx3 to direct lineage-specific gene activation and silencing.
Collapse
Affiliation(s)
- Ivana M Djuretic
- Harvard Medical School and the CBR Institute for Biomedical Research, Boston, Massachusetts 02115, USA
| | | | | | | | | | | |
Collapse
|
32
|
Abstract
Helper T cells coordinate immune responses through the production of cytokines. Th2 cells express the closely linked Il4, Il13, and Il5 cytokine genes, whereas these same genes are silenced in the Th1 lineage. The Th1/Th2 lineage choice has become a textbook example for the regulation of cell differentiation, and recent discoveries have further refined and expanded our understanding of how Th2 differentiation is initiated and reinforced by signals from antigen-presenting cells and cytokine-driven feedback loops. Epigenetic changes that stabilize the active or silent state of the Il4 locus in differentiating helper T cells have been a major focus of recent research. Overall, the field is progressing toward an integrated model of the signaling and transcription factor networks, cis-regulatory elements, epigenetic modifications, and RNA interference mechanisms that converge to determine the lineage fate and gene expression patterns of differentiating helper T cells.
Collapse
Affiliation(s)
- K Mark Ansel
- Harvard Medical School, CBR Institute for Biomedical Research, Boston, Massachusetts 02115, USA.
| | | | | | | |
Collapse
|
33
|
Owyang AM, Zaph C, Wilson EH, Guild KJ, McClanahan T, Miller HRP, Cua DJ, Goldschmidt M, Hunter CA, Kastelein RA, Artis D. Interleukin 25 regulates type 2 cytokine-dependent immunity and limits chronic inflammation in the gastrointestinal tract. ACTA ACUST UNITED AC 2006; 203:843-9. [PMID: 16606667 PMCID: PMC1800834 DOI: 10.1084/jem.20051496] [Citation(s) in RCA: 303] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
The cytokine interleukin (IL) 25 has been implicated in the initiation of type 2 immunity by driving the expression of type 2 cytokines such as IL-5 and IL-13, although its role in the regulation of immunity and infection-induced inflammation is unknown. Here, we identify a dual function for IL-25: first, in promoting type 2 cytokine-dependent immunity to gastrointestinal helminth infection and, second, in limiting proinflammatory cytokine production and chronic intestinal inflammation. Treatment of genetically susceptible mice with exogenous IL-25 promoted type 2 cytokine responses and immunity to Trichuris. IL-25 was constitutively expressed by CD4+ and CD8+ T cells in the gut of mouse strains that are resistant to Trichuris, and IL-25–deficient mice on a genetically resistant background failed to develop a type 2 immune response or eradicate infection. Furthermore, chronically infected IL-25−/− mice developed severe infection-induced intestinal inflammation associated with heightened expression of interferon-γ and IL-17, identifying a role for IL-25 in limiting pathologic inflammation at mucosal sites. Therefore, IL-25 is not only a critical mediator of type 2 immunity, but is also required for the regulation of inflammation in the gastrointestinal tract.
Collapse
Affiliation(s)
- Alexander M Owyang
- Department of Pathobiology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Abstract
This article discusses the potential role of bacterial superantigens (SAgs) in chronic rhinosinusitis with nasal polyposis (CRS/NP). First, it briefly describes SAgs, focusing on how they interact with the immune system by binding to T-cell receptors (TCR) and major histocompatibility complex (MHC) class II molecules. Second, it discusses the role of SAgs in other chronic inflammatory diseases.Finally, it presents evidence for the role of SAgs in the pathogenesis and maintenance of CRS/NP focusing on current research and future considerations.
Collapse
Affiliation(s)
- Kristin A Seiberling
- Department of Otolaryngology-Head and Neck Surgery, Northwestern University Feinberg School of Medicine, 303 E. Chicago Avenue, Searle Building 12-561, Chicago, IL 60611, USA
| | | | | |
Collapse
|
35
|
Seiberling KA, Conley DB, Tripathi A, Grammer LC, Shuh L, Haines GK, Schleimer R, Kern RC. Superantigens and chronic rhinosinusitis: detection of staphylococcal exotoxins in nasal polyps. Laryngoscope 2005; 115:1580-5. [PMID: 16148698 DOI: 10.1097/01.mlg.0000168111.11802.9c] [Citation(s) in RCA: 87] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
OBJECTIVE/HYPOTHESIS The role of infectious agents in the etiology of chronic rhinosinusitis with nasal polyposis (CRSwNP) remains unclear. Recent studies have provided indirect evidence of exposure to staphylococcal exotoxins in the blood and polyp tissue of patients with CRSwNP. These exotoxins have the capacity to act as superantigens, bypassing normal antigen processing and directly stimulating a massive inflammatory response. The objective of the study was to analyze mucus and polyp tissue samples from patients with CRSwNP for the presence of staphylococcal exotoxins. STUDY DESIGN Prospective study. METHODS Tissue and mucus samples were obtained from 42 patients undergoing endoscopic sinus surgery for chronic rhinosinusitis and 11 normal control patients. Twenty-nine of 42 patients had chronic rhinosinusitis with bilateral nasal polyposis, 2 had antrochoanal polyps, and 11 had chronic rhinosinusitis without nasal polyps. Eleven patients without chronic rhinosinusitis or polyps served as normal control patients. Specimens were analyzed for the presence of five staphylococcal exotoxins (SEA, SEB, SEC, SED, and toxic shock syndrome toxin type 1 [TSST-1]) using enzyme-linked immunosorbent assay (ELISA). Histological analysis of specimens and mean eosinophil counts were correlated with the presence of toxin. RESULTS At least one toxin was detected in 14 of 29 patients with bilateral nasal polyposis. Nine of the 14 patients also had positive findings for additional toxins. The dominant histological pattern in the CRSwNP patient group was polypoid mucosa with edema, which was found in both ELISA-positive and ELISA-negative patients. Mean eosinophil counts tended to be higher in ELISA-positive patients with polyps compared with patients without toxin detection. No toxin was detected in the 11 specimens taken from normal control patients. Only 1 of the 13 patients with CRS without polyps had positive ELISA results for toxin. CONCLUSION The current study demonstrates the presence of superantigen toxins in 14 of 29 patients with CRSwNP, with SEB and TSST-1 being the most common. Further studies are necessary to correlate the presence of toxin with the pathological changes present in polyp tissue.
Collapse
Affiliation(s)
- Kristin A Seiberling
- Division of Allergy and Immunology, Department of Medicine, Northwestern University Feinberg School of Medicine, 676 North St. Clair, Chicago, IL 60611, USA
| | | | | | | | | | | | | | | |
Collapse
|