1
|
Nair K, Bhat AR. Applications of Gene Therapy in Dentistry: A Review Article. JOURNAL OF HEALTH AND ALLIED SCIENCES NU 2023. [DOI: 10.1055/s-0042-1759711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
AbstractGene therapy promises to possess a good prospect in bridging the gap between dental applications and medicine. The dynamic therapeutic modalities of gene therapy have been advancing rapidly. Conventional approaches are being revamped to be more comprehensive and pre-emptive, which could do away with the need for surgery and medicine altogether. The complementary base sequences known as genes convey the instructions required to manufacture proteins. The oral cavity is one of the most accessible locations for the therapeutic intervention of gene therapy for several oral tissues. In 1990, the first significant trial of gene therapy was overseen to alleviate adenosine deaminase deficiency. The notion of genetic engineering has become increasingly appealing as a reflection of its benefits over conventional treatment modalities. An example of how this technology may alter dentistry is the implementation of gene therapy for dental and oral ailments. The objective of this article is to examine the effects of gene therapy on the field of dentistry, periodontology and implantology. Furthermore, the therapeutic factors of disease therapy, minimal invasion, and appropriate outcome have indeed been taken into consideration.
Collapse
Affiliation(s)
- Karthika Nair
- Department of Periodontology, A B Shetty Memorial Institute of Dental Sciences, NITTE Deemed to be University, Mangaluru, Karnataka, India
| | - Amitha Ramesh Bhat
- Department of Periodontology, A B Shetty Memorial Institute of Dental Sciences, NITTE Deemed to be University, Mangaluru, Karnataka, India
| |
Collapse
|
2
|
Wang W, Fang K, Wang X, Li M, Wu Y, Chen F, Shahzad KA, Gu N, Shen C. Antigen-Specific Killer Polylactic-Co-Glycolic Acid (PLGA) Microspheres Can Prolong Alloskin Graft Survival in a Murine Model. Immunol Invest 2015; 44:385-99. [DOI: 10.3109/08820139.2015.1014098] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
3
|
Donate A, Voigt A, Nguyen CQ. The value of animal models to study immunopathology of primary human Sjögren's syndrome symptoms. Expert Rev Clin Immunol 2014; 10:469-81. [PMID: 24506531 PMCID: PMC5769146 DOI: 10.1586/1744666x.2014.883920] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Sjögren's syndrome (SjS) is a complex chronic autoimmune disease of multifactorial etiology that results in eventual loss of secretory function in the exocrine glands. The challenges towards finding a therapeutic prevention or treatment for SjS are due primarily to a lack of understanding in the pathophysiological and clinical progression of the disease. In order to circumnavigate this problem, there is a need for appropriate animal models that resemble the major phenotypes of human SjS and deliver a clear underlying biological or molecular mechanism capable of defining various aspects for the disease. Here, we present an overview of SjS mouse models that are providing insight into the autoimmune process of SjS and advance our focus on potential diagnostic and therapeutic targets.
Collapse
Affiliation(s)
- Amy Donate
- Department of Infectious Diseases and Pathology, University of Florida College of Veterinary Medicine, 2015 SW 16 Ave, Gainesville, Florida 32611, USA
| | - Alexandria Voigt
- Department of Infectious Diseases and Pathology, University of Florida College of Veterinary Medicine, 2015 SW 16 Ave, Gainesville, Florida 32611, USA
| | - Cuong Q. Nguyen
- Department of Infectious Diseases and Pathology, University of Florida College of Veterinary Medicine, 2015 SW 16 Ave, Gainesville, Florida 32611, USA
- Center for Orphan Autoimmune Disorders, University of Florida College of Dentistry, 1600 SW Archer Rd, Gainesville, Florida 32610, USA
- Department of Oral Biology, University of Florida College of Dentistry, 1600 SW Archer Rd, Gainesville, Florida 32610, USA
| |
Collapse
|
4
|
Liu KC, Huynh K, Grubbs J, Davis RM. Autoimmunity in the pathogenesis and treatment of keratoconjunctivitis sicca. Curr Allergy Asthma Rep 2014; 14:403. [PMID: 24395332 DOI: 10.1007/s11882-013-0403-7] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Dry eye is a chronic corneal disease that impacts the quality of life of many older adults. Keratoconjunctivitis sicca (KCS), a form of aqueous-deficient dry eye, is frequently associated with Sjögren's syndrome and mechanisms of autoimmunity. For KCS and other forms of dry eye, current treatments are limited, with many medications providing only symptomatic relief rather than targeting the pathophysiology of disease. Here, we review proposed mechanisms in the pathogenesis of autoimmune-based KCS: genetic susceptibility and disruptions in antigen recognition, immune response, and immune regulation. By understanding the mechanisms of immune dysfunction through basic science and translational research, potential drug targets can be identified. Finally, we discuss current dry eye therapies as well as promising new treatment options and drug therapy targets.
Collapse
Affiliation(s)
- Katy C Liu
- Department of Ophthalmology, University of North Carolina at Chapel Hill School of Medicine, CB# 7040, 5151 Bioinformatics Building, Chapel Hill, NC, 27599, USA,
| | | | | | | |
Collapse
|
5
|
Kurien BT, Dsouza A, Igoe A, Lee YJ, Maier-Moore JS, Gordon T, Jackson M, Scofield RH. Immunization with 60 kD Ro peptide produces different stages of preclinical autoimmunity in a Sjögren's syndrome model among multiple strains of inbred mice. Clin Exp Immunol 2013; 173:67-75. [PMID: 23607771 PMCID: PMC3694536 DOI: 10.1111/cei.12094] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/13/2013] [Indexed: 11/27/2022] Open
Abstract
Sjögren's syndrome is a chronic illness manifested characteristically by immune injury to the salivary and lacrimal glands, resulting in dry mouth/eyes. Anti-Ro [Sjögren's syndrome antigen A (SSA)] and anti-La [Sjögren's syndrome antigen B (SSB)] autoantibodies are found frequently in Sjögren's subjects as well as in individuals who will go on to develop the disease. Immunization of BALB/c mice with Ro60 peptides results in epitope spreading with anti-Ro and anti-La along with lymphocyte infiltration of salivary glands similar to human Sjögren's. In addition, these animals have poor salivary function/low saliva volume. In this study, we examined whether Ro-peptide immunization produces a Sjögren's-like illness in other strains of mice. BALB/c, DBA-2, PL/J, SJL/J and C57BL/6 mice were immunized with Ro60 peptide-274. Sera from these mice were studied by immunoblot and enzyme-linked immunosorbent assay for autoantibodies. Timed salivary flow was determined after pharmacological stimulation, and salivary glands were examined pathologically. We found that SJL/J mice had no immune response to the peptide from Ro60, while C57BL/6 mice produced antibodies that bound the peptide but had no epitope spreading. PL/J mice had epitope spreading to other structures of Ro60 as well as to La, but like C57BL/6 and SJL/J had no salivary gland lymphocytic infiltration and no decrement of salivary function. DBA-2 and BALB/c mice had infiltration but only BALB/c had decreased salivary function. The immunological processes leading to a Sjögren's-like illness after Ro-peptide immunization were interrupted in a stepwise fashion in these differing mice strains. These data suggest that this is a model of preclinical disease with genetic control for epitope spreading, lymphocytic infiltration and glandular dysfunction.
Collapse
MESH Headings
- Amino Acid Sequence
- Animals
- Antibodies, Antinuclear/biosynthesis
- Antibodies, Antinuclear/immunology
- Autoantigens/immunology
- Autoimmunity/genetics
- Autoimmunity/immunology
- Carbachol/pharmacology
- Disease Models, Animal
- Epitopes/immunology
- Freund's Adjuvant
- H-2 Antigens/genetics
- H-2 Antigens/immunology
- Haplotypes
- Immunization
- Lymphocyte Subsets/immunology
- Lymphocyte Subsets/pathology
- Male
- Mice
- Mice, Inbred Strains/genetics
- Mice, Inbred Strains/immunology
- Molecular Sequence Data
- Muscle, Smooth/drug effects
- Muscle, Smooth/immunology
- Peptide Fragments/immunology
- Prodromal Symptoms
- RNA, Small Cytoplasmic/immunology
- Receptor, Muscarinic M3/drug effects
- Receptor, Muscarinic M3/immunology
- Ribonucleoproteins/immunology
- Salivary Glands/pathology
- Salivation
- Sjogren's Syndrome/etiology
- Sjogren's Syndrome/immunology
- Specific Pathogen-Free Organisms
- Urinary Bladder
- Xerostomia/etiology
- Xerostomia/immunology
- SS-B Antigen
Collapse
Affiliation(s)
- B T Kurien
- Arthritis & Clinical Immunology Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | | | | | | | | | | | | | | |
Collapse
|
6
|
Bombardieri M, Barone F, Lucchesi D, Nayar S, van den Berg WB, Proctor G, Buckley CD, Pitzalis C. Inducible tertiary lymphoid structures, autoimmunity, and exocrine dysfunction in a novel model of salivary gland inflammation in C57BL/6 mice. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2012; 189:3767-76. [PMID: 22942425 PMCID: PMC3448973 DOI: 10.4049/jimmunol.1201216] [Citation(s) in RCA: 89] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Salivary glands in patients with Sjögren's syndrome (SS) develop ectopic lymphoid structures (ELS) characterized by B/T cell compartmentalization, the formation of high endothelial venules, follicular dendritic cell networks, functional B cell activation with expression of activation-induced cytidine deaminase, as well as local differentiation of autoreactive plasma cells. The mechanisms that trigger ELS formation, autoimmunity, and exocrine dysfunction in SS are largely unknown. In this article, we present a novel model of inducible ectopic lymphoid tissue formation, breach of humoral self-tolerance, and salivary hypofunction after delivery of a replication-deficient adenovirus-5 in submandibular glands of C57BL/6 mice through retrograde excretory duct cannulation. In this model, inflammation rapidly and consistently evolves from diffuse infiltration toward the development of SS-like periductal lymphoid aggregates within 2 wk from AdV delivery. These infiltrates progressively acquire ELS features and support functional GL7(+)/activation-induced cytidine deaminase(+) germinal centers. Formation of ELS is preceded by ectopic expression of lymphoid chemokines CXCL13, CCL19, and lymphotoxin-β, and is associated with development of anti-nuclear Abs in up to 75% of mice. Finally, reduction in salivary flow was observed over 3 wk post-AdV infection, consistent with exocrine gland dysfunction as a consequence of the inflammatory response. This novel model has the potential to unravel the cellular and molecular mechanisms that regulate ELS formation and their role in exocrine dysfunction and autoimmunity in SS.
Collapse
Affiliation(s)
- Michele Bombardieri
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
| | - Francesca Barone
- Rheumatology Research Group, University of Birmingham, Birmingham, United Kingdom
| | - Davide Lucchesi
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
| | - Saba Nayar
- Rheumatology Research Group, University of Birmingham, Birmingham, United Kingdom
| | - Wim B van den Berg
- Department of Rheumatology, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Gordon Proctor
- Oral Medicine and Pathology, King’s College, London, United Kingdom
| | | | - Costantino Pitzalis
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
| |
Collapse
|
7
|
Lee BH, Gauna AE, Pauley KM, Park YJ, Cha S. Animal models in autoimmune diseases: lessons learned from mouse models for Sjögren's syndrome. Clin Rev Allergy Immunol 2012; 42:35-44. [PMID: 22105703 DOI: 10.1007/s12016-011-8288-5] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/15/2022]
Abstract
The mouse model is the one of the most frequently used and well-established animal models, and is currently used in many research areas. To date, various mouse models have been utilized to elucidate underlying causes of multifactorial autoimmune conditions, including pathological immune components and specific signaling pathways. This review summarizes the more recent mouse models for Sjögren's syndrome, a systemic autoimmune disease characterized by lymphocytic infiltration in the exocrine glands, such as the salivary and lacrimal glands, and loss of secretory function, resulting in dry mouth and dry eyes in patients. Although every Sjögren's syndrome mouse model resembles the major symptoms or phenotypes of Sjögren's syndrome conditions in humans, the characteristics of each model are variable. Moreover, to date, there is no single mouse model that can completely replicate the human conditions. However, unique features of each mouse model provide insights into the roles of potential etiological and immunological factors in the development and progression of Sjögren's syndrome. Here, we will overview the Sjögren's syndrome mouse models. Lessons from these mouse models will aid us to understand underlying immune dysregulation in autoimmune diseases in general, and will guide us to direct future research towards appropriate diagnostic and therapeutic strategies.
Collapse
Affiliation(s)
- Byung Ha Lee
- Department of Oral and Maxillofacial Diagnostic Sciences, University of Florida College of Dentistry, JHMHSC, Gainesville, FL 32610, USA
| | | | | | | | | |
Collapse
|
8
|
Delaleu N, Nguyen CQ, Peck AB, Jonsson R. Sjögren's syndrome: studying the disease in mice. Arthritis Res Ther 2011; 13:217. [PMID: 21672284 PMCID: PMC3218871 DOI: 10.1186/ar3313] [Citation(s) in RCA: 78] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Sjögren's syndrome (SS), a systemic autoimmune disease, is characterized by inflammation of exocrine tissues accompanied by a significant loss of their secretory function. Clinical symptoms develop late and there are no diagnostic tests enabling early diagnosis of SS. Thus, particularly to study these covert stages, researchers turn to studying animal models where mice provide great freedom for genetic manipulation and testing the effect of experimental intervention. The present review summarizes current literature pertaining to both spontaneous and extrinsic-factor induced SS-like diseases in mouse models, discussing advantages and disadvantages related to the use of murine models in SS research.
Collapse
Affiliation(s)
- Nicolas Delaleu
- Broegelmann Research Laboratory, The Gade Institute, University of Bergen, New Laboratory Building, 5th floor, #5305, 5021 Bergen, Norway.
| | | | | | | |
Collapse
|
9
|
Shen C, He Y, Cheng K, Zhang D, Miao S, Zhang A, Meng F, Miao F, Zhang J. Killer artificial antigen-presenting cells deplete alloantigen-specific T cells in a murine model of alloskin transplantation. Immunol Lett 2011; 138:144-55. [PMID: 21513739 DOI: 10.1016/j.imlet.2011.04.002] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2010] [Revised: 03/28/2011] [Accepted: 04/05/2011] [Indexed: 11/19/2022]
Abstract
FasL-expressing killer antigen-presenting cells (KAPCs) have the ability to delete antigen-specific T cells and, therefore, could potentially be used for the treatment of allograft rejection and autoimmunity; however, their cellular nature markedly limits their clinical use. Novel bead-based killer artificial antigen-presenting cells (KaAPCs), which are generated by coupling major histocompatibility complex (MHC) class I antigens together with the apoptosis-inducing anti-Fas monoclonal antibody (mAb) onto magnetic beads, have recently attracted more attention. KaAPCs have a number of advantages over KAPCs and are able to deplete specific T cells in cocultures. However, it remains unknown whether bead-based KaAPCs can also induce apoptosis of alloreactive or autoreactive T cells and, consequently, generate hyporesponsiveness in vivo. In this study, H-2K(b)/peptide monomers and anti-Fas mAb have been covalently coupled to latex beads and administered intravenously into BALB/c mice (H-2K(d)) that had previously been grafted with skin squares from C57BL/6 mice (H-2K(b)). Alloskin graft survival was prolonged for 6 days. A 60% decrease of H-2K(b) antigen-alloreactive T cells was demonstrated by several measures 2 days after each injection of KaAPCs, but intact immune function, including antitumor activity, was maintained. These data provide the first in vivo evidence that bead-based KaAPCs can selectively deplete antigen-specific T cells without the loss of overall immune responsiveness and, therefore, highlight the therapeutic potential of this novel strategy for the treatment of allograft rejection and autoimmune disorders.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal/chemistry
- Antibodies, Monoclonal/immunology
- Antibodies, Monoclonal/pharmacology
- Antibodies, Monoclonal, Murine-Derived
- Antigen-Presenting Cells/chemistry
- Antigen-Presenting Cells/cytology
- Antigen-Presenting Cells/immunology
- Apoptosis/drug effects
- Artificial Cells/chemistry
- Artificial Cells/cytology
- Artificial Cells/immunology
- CD4-Positive T-Lymphocytes/immunology
- Cell Proliferation/drug effects
- Fas Ligand Protein/antagonists & inhibitors
- Fas Ligand Protein/immunology
- Fas Ligand Protein/metabolism
- Graft Rejection/immunology
- Graft Rejection/prevention & control
- Graft Survival/immunology
- Histocompatibility Antigens Class I/immunology
- Immune Tolerance
- Immunoconjugates/chemistry
- Immunoconjugates/immunology
- Immunoconjugates/pharmacology
- Injections, Intravenous
- Lymphocyte Depletion
- Magnetics/methods
- Male
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Microspheres
- Models, Animal
- Skin Transplantation/immunology
- Transplantation, Homologous
Collapse
Affiliation(s)
- Chuanlai Shen
- Key Laboratory of Developmental Genes and Human Disease, Ministry of Education, Southeast University School of Medicine, Nanjing, Jiangsu, China
| | | | | | | | | | | | | | | | | |
Collapse
|
10
|
The role of FasL and Fas in health and disease. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2010; 647:64-93. [PMID: 19760067 DOI: 10.1007/978-0-387-89520-8_5] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The FS7-associated cell surface antigen (Fas, also named CD95, APO-1 or TNFRSF6) attracted considerable interest in the field of apoptosis research since its discovery in 1989. The groups of Shin Yonehara and Peter Krammer were the first reporting extensive apoptotic cell death induction upon treating cells with Fas-specific monoclonal antibodies.1,2 Cloning of Fas3 and its ligand,4,5 FasL (also known as CD178, CD95L or TNFSF6), laid the cornerstone in establishing this receptor-ligand system as a central regulator of apoptosis in mammals. Therapeutic exploitation of FasL-Fas-mediated cytotoxicity was soon an ambitous goal and during the last decade numerous strategies have been developed for its realization. In this chapter, we will briefly introduce essential general aspects of the FasL-Fas system before reviewing its physiological and pathophysiological relevance. Finally, FasL-Fas-related therapeutic tools and concepts will be addressed.
Collapse
|
11
|
Wang Y, Shnyra A, Africa C, Warholic C, McArthur C. Activation of the extrinsic apoptotic pathway by TNF-alpha in human salivary gland (HSG) cells in vitro, suggests a role for the TNF receptor (TNF-R) and intercellular adhesion molecule-1 (ICAM-1) in Sjögren's syndrome-associated autoimmune sialadenitis. Arch Oral Biol 2009; 54:986-96. [PMID: 19716548 DOI: 10.1016/j.archoralbio.2009.07.011] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2009] [Revised: 07/27/2009] [Accepted: 07/31/2009] [Indexed: 10/20/2022]
Abstract
UNLABELLED Studies suggest that apoptosis plays a major role in destruction of salivary glands in Sjögren's syndrome. We hypothesise that apoptosis results in the exposure of cryptic T cell epitopes and an autoimmune response which is pathway-specific. OBJECTIVE To activate the extrinsic or intrinsic apoptotic pathway to examine morphology, adhesion molecules, markers of antigen processing, and autoantigens on apoptotic bodies in vitro. METHODS Tumor necrosis factor-alpha (TNF-alpha) or staurosporine, a protein kinase inhibitor, was used to trigger the extrinsic or intrinsic apoptotic pathway in a Human Salivary Gland cell line (HSG), in vitro. Activated genes were profiled by cDNA array. Apoptotic bodies were visualised using light and SEM. Proteins were evaluated by immunofluorescence and confirmed by functional binding assays with Jurkat lymphocytes. RESULTS TNF-alpha-triggered extrinsic apoptosis resulted in "sticky" aggregated, apoptotic bodies which displayed cleaved alpha-fodrin autoantigen. In contrast, intrinsic apoptosis induced by staurosporine, resulted in dispersed cell blebs which were alpha-fodrin-negative. cDNA arrays revealed that TNF-alpha, but not staurosporine, upregulated transcriptional expression of Intercellular Adhesion Molecule-1 (ICAM-1) and Macrophage Inflammatory Protein-3 (CCL20). CONCLUSION The apoptotic pathway controls morphological, structural and functional properties of apoptotic bodies. Collectively, TNF-alpha-dependent activation of the extrinsic apoptotic pathway leads to upregulation of ICAM-1 and CCL20 in HSG in vitro. This suggests that pathogenesis in Sjögren's syndrome may involve a TNF-controlled cross-talk between apoptotic ductal and CCL20-attracted dendritic cells via the CD137/CD137L signalling pathway.
Collapse
Affiliation(s)
- Yan Wang
- Department of Oral Biology, University of Missouri-Kansas City School of Dentistry, 650 E 25th Street, Kansas City, MO 64108, United States
| | | | | | | | | |
Collapse
|
12
|
Kasman LM, London LL, London SD, Pilgrim MJ. A mouse model linking viral hepatitis and salivary gland dysfunction. Oral Dis 2009; 15:587-95. [PMID: 19656314 DOI: 10.1111/j.1601-0825.2009.01600.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
OBJECTIVE Viral hepatitis is known to cause xerostomia in humans, but this has not been reported in an animal model. We report a severe, acute, highly reproducible saliva deficiency occurring in BALB/c mice as a result of experimental viral hepatitis. MATERIALS AND METHODS BALB/c mice, splenectomized or carrying genetic mutations to detect immunological contributions to the saliva deficiency syndrome, were infected intraperitoneally with a non-lethal dose of murine cytomegalovirus. Pilocarpine-stimulated saliva volumes were determined between 0 and 15 days after infection. Salivary gland, liver, spleen, and sera were analyzed for the presence of virus, cytokines, inflammatory infiltrates, and tissue damage. RESULTS Saliva deficiency was detectable 2 days after cytomegalovirus infection, peaked at 88% below normal by day 7, and resolved partially in all mice by 15 days postinfection as sialoadenitis increased. Neither salivary gland viral titers, sialoadenitis, splenectomy, nor systemic inflammatory markers correlated with hyposalivation severity. Elevated liver enzymes did correlate with hyposalivation, and mice genetically resistant to murine cytomegalovirus-induced hepatitis were significantly protected. CONCLUSIONS Murine cytomegalovirus-induced salivary gland dysfunction is biphasic, with an acute hepatitis-associated phase and a later sialoadenitis-associated phase. Acute murine cytomegalovirus infection of BALB/c mice may provide a model for investigation of hepatitis-associated xerostomia.
Collapse
Affiliation(s)
- L M Kasman
- Department of Microbiology and Immunology, Medical University of South Carolina, BSB-201, PO Box 250504, 173 Ashley Ave, Charleston, SC 29403, USA.
| | | | | | | |
Collapse
|
13
|
Abstract
Sjögren's syndrome is an autoimmune, chronic inflammatory disease characterized by focal mononuclear cell infiltration of exocrine tissues, accompanied by loss of secretory function. The pathogenesis of autoimmune diseases is complex and, therefore, difficult to study in vitro. As of today, the role of initiating factors remains obscure, clinical symptoms develop late, and there are no tests for early diagnosis of SS. Hence, the disease is difficult to detect and treat. Animal models may provide insights into the identification of target antigens, narrowing the relevant pathological immune mechanisms, and to study the evolution of tissue pathology. This review summarizes current knowledge on murine strains, both spontaneous and induced models, used to study Sjögren's syndrome. Special attention is paid to the characteristics of different strains regarding their properties to mimic specific aspects or stages of the disease.
Collapse
Affiliation(s)
- Malin V Jonsson
- Broegelmann Research Laboratory, The Gade Institute, University of Bergen, Armauer Hansen Building, 5021, Bergen, Norway
| | | | | |
Collapse
|
14
|
Abstract
Several cell-based immunotherapy strategies have been developed to specifically modulate T cell-mediated immune responses. These methods frequently rely on the utilization of tolerogenic cell-based antigen-presenting cells (APCs). However, APCs are highly sensitive to cytotoxic T-cell responses, thus limiting their therapeutic capacity. Here, we describe a novel bead-based approach to modulate T-cell responses in an antigen-specific fashion. We have generated killer artificial APCs (kappaaAPCs) by coupling an apoptosis-inducing alpha-Fas (CD95) IgM mAb together with HLA-A2 Ig molecules onto beads. These kappaaAPCs deplete targeted antigen-specific T cells in a Fas/Fas ligand (FasL)-dependent fashion. T-cell depletion in cocultures is rapidly initiated (30 minutes), dependent on the amount of kappaaAPCs and independent of activation-induced cell death (AICD). kappaaAPCs represent a novel technology that can control T cell-mediated immune responses, and therefore has potential for use in treatment of autoimmune diseases and allograft rejection.
Collapse
|
15
|
The Role of Epithelial Cells in the Pathogenesis of Sjögren’s Syndrome. Clin Rev Allergy Immunol 2007; 32:225-30. [DOI: 10.1007/s12016-007-8007-4] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
16
|
Lodde BM, Baum BJ, Tak PP, Illei G. Experience with experimental biological treatment and local gene therapy in Sjogren's syndrome: implications for exocrine pathogenesis and treatment. Ann Rheum Dis 2006; 65:1406-13. [PMID: 16880196 PMCID: PMC1798364 DOI: 10.1136/ard.2006.052761] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/22/2006] [Indexed: 12/13/2022]
Abstract
Sjögren's syndrome is an autoimmune exocrinopathy, mainly affecting the lacrimal and salivary glands, and resulting in ocular and oral dryness (keratoconjunctivitis sicca and xerostomia). The aetiology and pathogenesis are largely unknown, and only palliative treatment is currently available. Data obtained from experimental animal and human studies using biological agents or gene therapeutics can offer insight into the disease process of Sjögren's syndrome. This article reviews the current literature on these approaches and assesses the lessons learnt about the pathogenesis of Sjögren's syndrome.
Collapse
Affiliation(s)
- B M Lodde
- Gene Therapy and Therapeutics Branch/NIDCR, National Institutes of Health, 10 Center Drive, Building 10, Room 1N114, Bethesda, MD 20892-1190, USA
| | | | | | | |
Collapse
|
17
|
Abstract
Sjögren's syndrome (SS) is a complex autoimmune disorder, characterised by mononuclear cell infiltration of exocrine glands, principally the lacrimal and salivary glands. Both cellular, in the form of autoreactive immune cells, and humoral factors, such as autoantibodies, contribute to the expression of the disease. SS can also occur as a systemic disease affecting several organs, and approximately 5% of the patients develop malignant lymphoproliferation. Today SS is considered uncurable. The treatment available is only palliative, and the treatment goals are to manage symptoms and prevent or limit tissue damage. This may involve both local and systemic measures. However, the existing systemic treatment of chronic inflammatory autoimmune diseases has several limitations and unwanted side effects. In recent years the possibility to treat diseases with gene therapy has gained interest and has become a subject of investigation. Given the multitude of factors contributing to the pathogenesis of SS, gene therapy is a major challenge, but may elicit great benefits if successful. Keeping this in mind, the possibility for gene therapeutics in SS in general and potential targets for gene therapy are discussed.
Collapse
Affiliation(s)
- Anne Isine Bolstad
- Department of Oral Sciences-Periodontics, Faculty of Dentistry, University of Bergen, Aarstadveien 17, N-5009 Bergen, Norway.
| | | |
Collapse
|
18
|
Scofield RH, Asfa S, Obeso D, Jonsson R, Kurien BT. Immunization with short peptides from the 60-kDa Ro antigen recapitulates the serological and pathological findings as well as the salivary gland dysfunction of Sjogren's syndrome. THE JOURNAL OF IMMUNOLOGY 2006; 175:8409-14. [PMID: 16339583 DOI: 10.4049/jimmunol.175.12.8409] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Sjögren's syndrome is a poorly understood autoimmune inflammatory illness that affects the salivary and lacrimal glands as well as other organ systems. We undertook the present study to determine whether mice immunized with short peptides from the 60-kDa Ro (or SSA) Ag, which is a common target of the autoimmunity of Sjögren's syndrome, develop an illness similar to Sjögren's syndrome. BALB/c mice were immunized with one of two short peptides from 60-kDa Ro that are know to induce epitope spreading. The animals were analyzed for the presence of anti-Ro and anti-La (or SSB) in the sera by immunoblot and ELISA. Salivary glands were collected and examined by histology after H&E staining. Salivary lymphocytes were purified and studied for cell surface makers by fluorescence-activated cell sorting. Timed stimulated salivary flow was measured. As reported previously, BALB/c mice immunized with 60-kDa Ro peptides developed an immune response directed against the entire Ro/La ribonucleoprotein particle that was similar to that found in humans with lupus or Sjögren's syndrome. Functional studies showed a statistical decrease in salivary flow in immunized mice compared with controls. Furthermore, there were lymphocytic infiltrates in the salivary glands of immunized animals that were not present in controls. The infiltrates consisted of both CD4- and CD8+ T lymphocytes as well as B lymphocytes. BALB/c mice immunized with 60-kDa Ro peptides develop anti-Ro, salivary gland lymphocyte infiltrates, and salivary dysfunction that is highly reminiscent of human Sjögren's syndrome.
Collapse
Affiliation(s)
- R Hal Scofield
- Arthritis and Immunology Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA.
| | | | | | | | | |
Collapse
|
19
|
Samarkos M, Moutsopoulos HM. Recent advances in the management of ocular complications of Sjögren's syndrome. Curr Allergy Asthma Rep 2005; 5:327-32. [PMID: 15967078 DOI: 10.1007/s11882-005-0077-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Sjögren's syndrome (SS) is an autoimmune disorder, the principal ocular manifestation of which is decreased tear production leading to chronic irritation and damage to the corneal and conjunctival epithelium. The most important advance in the treatment of ocular manifestations of SS is the introduction of topical anti-inflammatory agents such as cyclosporine A, which increases tear production and decreases symptoms without any significant side effect. Stimulators of tear secretion, both topical, such as diquafosol, and systemic, such as pilocarpine and cevimeline, are also effective, although they have been associated with frequent side effects. Topical use of autologous serum is another new and effective form of treatment, but problems in the preparations prevent their widespread use. Additionally, nonpharmacologic treatments, such as insertion of punctal plugs, are beneficial in the dry eye of SS, whereas several other modalities, such as anti-CD4 monoclonal antibody eye drops and gene transfer, are still in experimental phases.
Collapse
Affiliation(s)
- Michael Samarkos
- Department of Pathophysiology, University of Athens School of Medicine, 25 Mikras Asias street, 115 27, Athens, Greece.
| | | |
Collapse
|
20
|
Bohana-Kashtan O, Civin CI. Fas Ligand as a Tool for Immunosuppression and Generation of Immune Tolerance. Stem Cells 2004; 22:908-24. [PMID: 15536183 DOI: 10.1634/stemcells.22-6-908] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The role of Fas ligand (FasL) in physiologically limiting immune responses and maintaining immune-privileged sites has led to a body of research aiming to confer protection to allogeneic grafts by expressing FasL on the allogeneic tissue or by administrating FasL-transduced donor dendritic cells. In addition, several studies have used FasL to abrogate autoimmune responses. This review presents the results of these studies and discusses the problems associated with FasL usage.
Collapse
Affiliation(s)
- Osnat Bohana-Kashtan
- Department of Oncology, Immunology and Hematopoiesis Division, Sidney Kimmel Comprehensive Cancer at Johns Hopkins, 1650 Orleans Street, Baltimore, MD 21231, USA
| | | |
Collapse
|
21
|
Kok MR, Yamano S, Lodde BM, Wang J, Couwenhoven RI, Yakar S, Voutetakis A, Leroith D, Schmidt M, Afione S, Pillemer SR, Tsutsui MT, Tak PP, Chiorini JA, Baum BJ. Local adeno-associated virus-mediated interleukin 10 gene transfer has disease-modifying effects in a murine model of Sjögren's syndrome. Hum Gene Ther 2004; 14:1605-18. [PMID: 14633403 DOI: 10.1089/104303403322542257] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Female nonobese diabetic (NOD) mice develop spontaneous autoimmune sialadenitis and loss of salivary flow, and are a widely used model of Sjögren's syndrome. We examined the feasibility of local salivary gland immunomodulatory gene delivery to alter these sequelae in NOD mice. We constructed recombinant adeno-associated virus (rAAV) vectors encoding either human interleukin 10 (rAAVhIL-10) or beta-galactosidase (rAAVLacZ, control vector). Mice received rAAVhIL-10 or rAAVLacZ by retrograde submandibular ductal instillation either at age 8 weeks (early, before onset of sialadenitis), or at 16 weeks (late, after onset of sialadenitis). As a systemic treatment control, separate mice received intramuscular delivery of rAAVhIL-10 at each time point. Both submandibular and intramuscular delivery of vector led to low circulating levels of hIL-10. After submandibular administration of rAAVhIL-10, salivary flow rates at 20 weeks for both the early and late treatment groups were significantly higher than for both rAAVLacZ-administered and untreated mice. Systemic delivery of rAAVhIL-10 led to improved salivary flow in the late treatment group. Inflammatory infiltrates in submandibular glands, however, were significantly reduced only in mice receiving rAAVhIL-10 locally in the salivary gland compared with mice receiving this vector intramuscularly, or rAAVLacZ or no treatment. In addition, after submandibular rAAVhIL-10 delivery, NOD mice exhibited significantly lower blood glucose, and higher serum insulin, levels than all other groups, indicating some systemic benefit of this treatment. These studies show that expression of hIL-10 by rAAV vectors can have disease-modifying effects in the salivary glands of NOD mice, and suggest that local immunomodulatory gene transfer may be useful for managing the salivary gland pathology in Sjögren's syndrome.
Collapse
Affiliation(s)
- Marc R Kok
- Gene Therapy and Therapeutics Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Denman AM, Rager-Zisman B. Viruses and autoimmune diseases—adapting Koch's postulates. Autoimmun Rev 2004; 3:355-61. [PMID: 15288001 DOI: 10.1016/j.autrev.2004.03.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2004] [Accepted: 03/03/2004] [Indexed: 12/21/2022]
Abstract
Viral hypotheses for the aetiopathogenesis of autoimmune diseases are unproven. Indeed, a primary role for virus-induced autoimmune disease has yet to be fully established even in experimental systems. However new insights into virus-host interactions and improved technology justify fresh evaluation of these issues. We suggest modified Koch's postulates to test the viral hypothesis.
Collapse
Affiliation(s)
- A M Denman
- Northwick Park Hospital, Harrow HA1 3UJ, UK
| | | |
Collapse
|
23
|
Li L, Hsu HC, Stockard CR, Yang P, Zhou J, Wu Q, Grizzle WE, Mountz JD. IL-12 Inhibits Thymic Involution by Enhancing IL-7- and IL-2-Induced Thymocyte Proliferation. THE JOURNAL OF IMMUNOLOGY 2004; 172:2909-16. [PMID: 14978093 DOI: 10.4049/jimmunol.172.5.2909] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
IL-12 has been reported to affect thymic T cell selection, but the role of IL-12 in thymic involution has not been studied. We found that in vivo, IL-12b knockout (IL-12b(-/-)) mice exhibited accelerated thymic involution compared with wild-type (WT) B6 mice. This is characterized by an increase in thymocytes with the early development stage phenotype of CD25(-)CD44(+)CD4(-)CD8(-) in aged IL-12b(-/-) mice. Histologically, there were accelerated degeneration of thymic extracellular matrix and blood vessels, a significantly decreased thymic cortex/medulla ratio, and increased apoptotic cells in aged IL-12b(-/-) mice compared with WT mice. There was, however, no apparent defect in thymic structure and thymocyte development in young IL-12(-/-) mice. These results suggest the importance of IL-12 in maintaining thymic integrity and function during the aging process. Surprisingly, in WT B6 mice, there was no age-related decrease in the levels of IL-12 produced from thymic dendritic cells. Stimulation of thymocytes with IL-12 alone also did not enhance the thymocyte proliferative response in vitro. IL-12, however, provided a strong synergistic effect to augment the IL-7 or IL-2 induced thymocyte proliferative response, especially in aged WT and IL-12b(-/-) mice. Our data strongly support the role of IL-12 as an enhancement cytokine, which acts through its interactions with other cytokines to maintain thymic T cell function and development during aging.
Collapse
Affiliation(s)
- Lina Li
- Department of Pathology, Veterans Administration Medical Center, Birmingham, AL 35233, USA
| | | | | | | | | | | | | | | |
Collapse
|
24
|
Hoves S, Krause SW, Herfarth H, Halbritter D, Zhang HG, Mountz JD, Schölmerich J, Fleck M. Elimination of activated but not resting primary human CD4 and CD8 T cells by Fas ligand (FasL/CD95L)-expressing Killer-dendritic cells. Immunobiology 2004; 208:463-75. [PMID: 15124861 DOI: 10.1078/0171-2985-00293] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Dendritic cells (DC) genetically engineered to express high levels of Fas ligand (FasL/CD95L) have been demonstrated to delete T cells in an antigen specific manner in several different animal models in vivo. However, the immunomodulatory capacity of primary human FasL-expressing Killer-DC has not been determined. Therefore, human Killer-DC were generated from mature monocyte-derived DC using the inducible CRE/LoxP adenoviral vector system, and the immunoregulatory capacity of these cells was analyzed in cocultures with primary human T cells in vitro. Combined transductions of DC by AdloxPFasL and AxCANCre resulted in FasL expression in > 70% of DC without affecting the mature phenotype. Proliferation of activated primary human T cells was inhibited up to 80% in cocultures with FasL-expressing DC but not EGFP-transduced DC, which was due to induction of apoptosis in activated but not resting CD4+ and CD8+ T cells. Apoptosis induced by Killer-DC could be blocked by an anti-FasL-antibody in a dose dependent fashion. The present results demonstrate that FasL-expressing Killer-DC eliminate activated but not resting primary human CD4+ and CD8+ T cells by induction of Fas-mediated apoptosis supporting the concept to apply Killer-DC as a novel strategy for the treatment of T cell-dependent autoimmune disease and allograft rejection in humans.
Collapse
Affiliation(s)
- Sabine Hoves
- The University of Regensburg, Department of Internal Medicine I, 93042 Regensburg, Germany
| | | | | | | | | | | | | | | |
Collapse
|
25
|
Hoves S, Krause SW, Halbritter D, Zhang HG, Mountz JD, Schölmerich J, Fleck M. Mature but not immature Fas ligand (CD95L)-transduced human monocyte-derived dendritic cells are protected from Fas-mediated apoptosis and can be used as killer APC. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2003; 170:5406-13. [PMID: 12759415 DOI: 10.4049/jimmunol.170.11.5406] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Several in vitro and animal studies have been performed to modulate the interaction of APCs and T cells by Fas (CD95/Apo-1) signaling to delete activated T cells in an Ag-specific manner. However, due to the difficulties in vector generation and low transduction frequencies, similar studies with primary human APC are still lacking. To evaluate whether Fas ligand (FasL/CD95L) expressing killer APC could be generated from primary human APC, monocyte-derived dendritic cells (DC) were transduced using the inducible Cre/Loxp adenovirus vector system. Combined transduction of DC by AdLoxpFasL and AxCANCre, but not single transduction with these vectors, resulted in dose- and time-dependent expression of FasL in >70% of mature DC (mDC), whereas <20% of immature DC (iDC) expressed FasL. In addition, transduction by AdLoxpFasL and AxCANCre induced apoptosis in >80% of iDC, whereas FasL-expressing mDC were protected from FasL/Fas (CD95/Apo-1)-mediated apoptosis despite coexpression of Fas. FasL-expressing mDC eliminated Fas(+) Jurkat T cells as well as activated primary T cells by apoptosis, whereas nonactivated primary T cells were not deleted. Induction of apoptosis in Fas(+) target cells required expression of FasL in DC and cell-to-cell contact between effector and target cell, and was not dependent on soluble FasL. Induction of apoptosis in Fas(+) target cells required expression of FasL in DC, cell-to-cell contact between effector and target cell, and was not dependent on soluble FasL. The present results demonstrate that FasL-expressing killer APC can be generated from human monocyte-derived mDC using adenoviral gene transfer. Our results support the strategy to use killer APCs as immunomodulatory cells for the treatment of autoimmune disease and allograft rejection.
Collapse
Affiliation(s)
- Sabine Hoves
- Department of Internal Medicine I, Regensburg, Germany
| | | | | | | | | | | | | |
Collapse
|
26
|
Abstract
Gene therapy has been applied in a variety of experimental models of autoimmunity with some success. In this article, we outline recent developments in gene therapy vectors, discuss advantages and disadvantages of each, and highlight their recent applications in autoimmune models. We also consider progress in vector targeting and components for regulating transgene expression, which will both improve gene therapy safety and empower gene therapy to fullfil its potential as a therapeutic modality. In conclusion, we consider candidate vectors that satisfy requirements for application in the principal therapeutic strategies in which gene therapy will be applied to autoimmune conditions.
Collapse
Affiliation(s)
- D J Gould
- 1Bone & Joint Research Unit, Barts & The London, Queen Mary's Medical School, University of London, London, UK
| | | |
Collapse
|
27
|
Zhan HG, Mountz JD, Fleck M, Zhou T, Hsu HC. Specific deletion of autoreactive T cells by adenovirus-transfected, Fas ligand-producing antigen-presenting cells. Immunol Res 2003; 26:235-46. [PMID: 12403361 DOI: 10.1385/ir:26:1-3:235] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Immune privilege is a unique strategy developed in several internal organs that can prevent the development of immune attack against these vital organs. One critical mechanism of immune privilege is utilization of Fas/FasL-mediated apoptosis to delete the invading T cells at the immune privilege sites. In this article, we describe the development and application of a unique cell-gene therapy to correct defective FasL-mediated apoptosis and autoimmune disease in autoimmune mice. This cell-gene therapy strategy using antigen-presenting cells (APCs) to express FasL is not only a therapeutic tool, but also has allowed us to understand the complexity of T cell regulation and the concept of eliminating T cells in the spleen, lymph node, or elsewhere in vivo to regulate the homeostasis of the peripheral T cell response. In this regard, the FasL-expressing APCs can be considered as circulating and regulatable immune privilege sites. Our studies provide substantial evidence that FasL-expressing APCs can be introduced exogenously without liver toxicity to eliminate infiltrating T cells and prevent the development of immune attack in lung, liver, kidney, joint, and salivary gland. Therefore, given the hazardous potential of persistent T cell invasion at the local inflammatory site, it is tempting to speculate that such an endogenous control mechanism occurs normally in vivo to limit a chronic T cell inflammatory response.
Collapse
Affiliation(s)
- Huang-Ge Zhan
- Department of Medicine, University of Alabama at Birmingham, 35294, USA
| | | | | | | | | |
Collapse
|
28
|
Abstract
Autoimmune disease in the elderly is hypothesized to be caused by an imbalance in T-cell expansion and deletion after an encounter with self-antigens. A decrease in thymic output leads to a decreased pool of naive T cells in the periphery and to increased oligoclonal expansion of T cells. This expansion may be caused by stimulation with autoantigens that drive high-affinity interactions with self-antigens. Accumulation of presenescent, apoptosis-resistant, and proinflammatory T cells results in the growth of these autoreactive T cells. A decreased T-cell activation response that occurs with age leads to several defects that diminish the immune response.
Collapse
Affiliation(s)
- Hui-Chen Hsu
- Division of Clinical Immunology and Rheumatology, Department of Medicine, University of Alabama at Birmingham, 701 South 19th Street, Lyons Harrison Research Building 473, Birmingham, AL 35294, USA
| | | |
Collapse
|
29
|
Tarner IH, Fathman CG. The potential for gene therapy in the treatment of autoimmune disease. Clin Immunol 2002; 104:204-16. [PMID: 12217329 DOI: 10.1006/clim.2002.5235] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Ingo H Tarner
- Department of Rheumatology, Stanford University School of Medicine, California 94305-5166, USA
| | | |
Collapse
|
30
|
Affiliation(s)
- J M Vitolo
- Gene Therapy and Therapeutics Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892, USA
| | | |
Collapse
|
31
|
Zhang HG, Yang P, Xie J, Liu Z, Liu D, Xiu L, Zhou T, Wang Y, Hsu HC, Mountz JD. Depletion of collagen II-reactive T cells and blocking of B cell activation prevents collagen II-induced arthritis in DBA/1j mice. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2002; 168:4164-72. [PMID: 11937577 DOI: 10.4049/jimmunol.168.8.4164] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Collagen II (CII)-induced arthritis in DBA/1j mice is mediated by both CII-reactive T cells and anti-CII Ab-producing B cells. To determine the relative role of these processes in the development of arthritis, we specifically eliminated CII-reactive T cells by treating the mice with CII-pulsed syngeneic macrophages that had been transfected with a binary adenovirus system. These macrophages express murine Fas ligand in a doxycycline-inducible manner with autocrine suicide inhibited by concomitant expression of p35. The mice were treated i.v. with four doses of CII-APC-AdFasLp35Tet or a single dose of AdCMVsTACI (5 x 10(9) PFU), or both simultaneously, beginning 2 wk after priming with CII in CFA. Treatment with CII-APC-AdFasLp35Tet alone or in combination with a single dose of AdCMVsTACI prevented the development of CII-induced arthritis and T cell infiltration in the joint. The elimination of T cells was specific in that a normal T cell response was observed on stimulation with OVA after treatment with CII-APC-AdFasLp35Tet. Treatment with AdCMVsTACI alone prevented production of detectable levels of circulating anti-CII autoantibodies and reduced the severity of arthritis but did not prevent its development. These results indicate that the CII-reactive T cells play a crucial role in the development of CII-induced arthritis and that the anti-CII Abs act to enhance the development of CII-induced arthritis.
Collapse
MESH Headings
- Adenoviridae/genetics
- Administration, Oral
- Animals
- Antigen-Presenting Cells/transplantation
- Apoptosis/genetics
- Apoptosis/immunology
- Arthritis, Experimental/immunology
- Arthritis, Experimental/pathology
- Arthritis, Experimental/prevention & control
- Autoantibodies/biosynthesis
- Autocrine Communication/genetics
- Autocrine Communication/immunology
- B-Lymphocytes/immunology
- Cartilage, Articular/metabolism
- Cartilage, Articular/pathology
- Cell Migration Inhibition
- Collagen Type II/administration & dosage
- Collagen Type II/immunology
- Cytomegalovirus/genetics
- Down-Regulation/genetics
- Down-Regulation/immunology
- Drug Therapy, Combination
- Fas Ligand Protein
- Female
- Genetic Vectors/administration & dosage
- Genetic Vectors/therapeutic use
- Inhibitor of Apoptosis Proteins
- Lymphocyte Activation/genetics
- Lymphocyte Depletion/methods
- Macrophages, Peritoneal/transplantation
- Membrane Glycoproteins/biosynthesis
- Membrane Glycoproteins/genetics
- Membrane Proteins
- Mice
- Mice, Inbred DBA
- Receptors, Tumor Necrosis Factor/biosynthesis
- Receptors, Tumor Necrosis Factor/genetics
- Receptors, Tumor Necrosis Factor/physiology
- Solubility
- T-Lymphocyte Subsets/immunology
- T-Lymphocyte Subsets/pathology
- Tetracycline Resistance/genetics
- Trans-Activators/genetics
- Transmembrane Activator and CAML Interactor Protein
- Viral Proteins/genetics
Collapse
|
32
|
Baum BJ, Kok M, Tran SD, Yamano S. The impact of gene therapy on dentistry: a revisiting after six years. J Am Dent Assoc 2002; 133:35-44. [PMID: 11811741 DOI: 10.14219/jada.archive.2002.0019] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
BACKGROUND Gene therapy is an emerging field of biomedicine that has commanded considerable scientific and popular attention. The procedure involves the transfer of genes to patients for clinical benefit. Transferred genes can b e used for either reparative or pharmacological purposes. OVERVIEW In 1995, the first author and a colleague described the potential impact of gene therapy on dentistry, on the basis of initial studies of gene transfer applications to salivary glands, keratinocytes and cancer cells. Their conclusion was that gene therapy would have a significant impact on the nature of dental practice within 20 years. In this article, the authors consider research progress since 1995 and reexamine the earlier conclusion. PRACTICE IMPLICATIONS In the past six years, remarkable progress has been made in the field of gene therapy, including seven areas relevant to dental practice: bone repair, salivary glands, autoimmune disease, pain, DNA vaccinations, keratinocytes and cancer. While considerable problems remain, thus impeding the routine clinical use of gene transfer, gene therapy will have a pervasive and significant impact on areas of dental practice that are based in biological science. By 2015, this will translate into practitioners' having a wide range of novel biological treatment options for their patients.
Collapse
Affiliation(s)
- Bruce J Baum
- Gene Therapy and Therapeutics Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892-1190, USA.
| | | | | | | |
Collapse
|
33
|
Abstract
Recent work on gene therapies for autoimmune disease has continued to provide insight into the pathogenesis of autoimmunity. Reliable, effective and targeted gene therapy applications have been achieved by using transduced dendritic cells and antigen-specific T cells as delivery vehicles. Bioluminescence imaging has been implemented to visualize cell trafficking and homing in vivo. As a first step into human gene therapy, a phase I clinical trial for assessing the feasibility and safety of gene transfer has been completed in a group of rheumatoid arthritis patients.
Collapse
Affiliation(s)
- I H Tarner
- Stanford University School of Medicine, Department of Medicine, Division of Immunology and Rheumatology, CCSR Building, Stanford, CA 94305-5166, USA.
| | | |
Collapse
|