1
|
Le PM, Mattapallil MJ, Caspi RR, Stepp MA, Menko AS. Immunoregulatory Properties of Immune Cells that Associate with the Lens Capsule Surface during Acute and Resolution Phases of Experimental Autoimmune Uveitis. THE AMERICAN JOURNAL OF PATHOLOGY 2024; 194:2194-2211. [PMID: 39159867 PMCID: PMC11627221 DOI: 10.1016/j.ajpath.2024.07.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Revised: 06/07/2024] [Accepted: 07/31/2024] [Indexed: 08/21/2024]
Abstract
Inflammation in the eye is tightly regulated to prevent vision impairment and irreversible blindness. Emerging evidence shows that immune cells are specifically recruited to the lens capsule in response to autoimmune uveitis, yet the potential that they have a role in regulating this inflammatory disease remained unexplored. Here, an immunolocalization approach combined with high-resolution confocal microscopy was used to investigate whether the immune cells that become stably associated with the lens capsule in the eyes of C57BL/6J mice with experimental autoimmune uveitis (EAU) have an immunoregulatory phenotype. These studies revealed that during the acute phase of uveitis, at day 18 after disease induction, the immune cells specifically recruited to the lens capsule, such as regulatory T cells [forkhead box P3 (FoxP3)+CD4+] and M2 macrophages (CD68+ arginase 1+IL-10+), included those with putative anti-inflammatory, proresolution roles. The frequency of these lens capsule-associated immunomodulatory phenotypes increased at day 35 after induction, during the resolution phase of EAU inflammation. At this later stage of resolution, most of the macrophages expressed CD206, a mannose receptor responsible for removing inflammatory molecules, in addition to arginase 1 and IL-10. These results suggest a previously unknown role for the lens as a site for recruitment of immune cells whose role is to suppress inflammation, promote resolution, and maintain remission of EAU.
Collapse
Affiliation(s)
- Phuong M Le
- Department of Pathology and Genomic Medicine, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Mary J Mattapallil
- Laboratory of Immunology, National Eye Institute, National Institutes of Health, Bethesda, Maryland
| | - Rachel R Caspi
- Laboratory of Immunology, National Eye Institute, National Institutes of Health, Bethesda, Maryland
| | - Mary Ann Stepp
- Department of Anatomy and Cell Biology, George Washington University School of Medicine and Health Sciences, Washington, District of Columbia; Department of Ophthalmology, George Washington University School of Medicine and Health Sciences, Washington, District of Columbia
| | - A Sue Menko
- Department of Pathology and Genomic Medicine, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania; Department of Ophthalmology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania.
| |
Collapse
|
2
|
Yang SN, Shi Y, Berggren PO. The anterior chamber of the eye technology and its anatomical, optical, and immunological bases. Physiol Rev 2024; 104:881-929. [PMID: 38206586 PMCID: PMC11381035 DOI: 10.1152/physrev.00024.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 11/30/2023] [Accepted: 01/05/2024] [Indexed: 01/12/2024] Open
Abstract
The anterior chamber of the eye (ACE) is distinct in its anatomy, optics, and immunology. This guarantees that the eye perceives visual information in the context of physiology even when encountering adverse incidents like inflammation. In addition, this endows the ACE with the special nursery bed iris enriched in vasculatures and nerves. The ACE constitutes a confined space enclosing an oxygen/nutrient-rich, immune-privileged, and less stressful milieu as well as an optically transparent medium. Therefore, aside from visual perception, the ACE unexpectedly serves as an excellent transplantation site for different body parts and a unique platform for noninvasive, longitudinal, and intravital microimaging of different grafts. On the basis of these merits, the ACE technology has evolved from the prototypical through the conventional to the advanced version. Studies using this technology as a versatile biomedical research platform have led to a diverse range of basic knowledge and in-depth understanding of a variety of cells, tissues, and organs as well as artificial biomaterials, pharmaceuticals, and abiotic substances. Remarkably, the technology turns in vivo dynamic imaging of the morphological characteristics, organotypic features, developmental fates, and specific functions of intracameral grafts into reality under physiological and pathological conditions. Here we review the anatomical, optical, and immunological bases as well as technical details of the ACE technology. Moreover, we discuss major achievements obtained and potential prospective avenues for this technology.
Collapse
Affiliation(s)
- Shao-Nian Yang
- The Rolf Luft Research Center for Diabetes and Endocrinology, Karolinska Institutet, Stockholm, Sweden
| | - Yue Shi
- The Rolf Luft Research Center for Diabetes and Endocrinology, Karolinska Institutet, Stockholm, Sweden
| | - Per-Olof Berggren
- The Rolf Luft Research Center for Diabetes and Endocrinology, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
3
|
Teabagy S, Wood E, Bilsbury E, Doherty S, Janardhana P, Lee DJ. Ocular immunosuppressive microenvironment and novel drug delivery for control of uveitis. Adv Drug Deliv Rev 2023; 198:114869. [PMID: 37172782 PMCID: PMC10330522 DOI: 10.1016/j.addr.2023.114869] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 04/17/2023] [Accepted: 05/07/2023] [Indexed: 05/15/2023]
Abstract
Ocular immune privilege is a phenomenon described by Peter Medawar in relation to the indefinite survival of the placement of foreign tissue grafts into the eye. Several mechanisms have been described that contribute to ocular immune privilege, such as a blood-ocular barrier and lack of lymphatics in the eye, the production of immune-suppressing molecules inside the ocular microenvironment, and the induction of systemic regulatory immunity against antigens found in the eye. Because ocular immune privilege is not absolute, failure of it can result in uveitis. Uveitis is a group of inflammatory disorders that can lead to vision loss if not treated properly. The current uveitis treatments involve the use of immunosuppressive and anti-inflammatory medications. Researching mechanisms of ocular immune privilege and the development of novel treatments for uveitis is ongoing. This review discusses mechanisms of ocular immune privilege, followed by an overview of uveitis treatments and ongoing clinical trials.
Collapse
Affiliation(s)
- Sean Teabagy
- Department of Ophthalmology and Visual Sciences, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Emma Wood
- Department of Ophthalmology and Visual Sciences, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Evan Bilsbury
- Department of Ophthalmology and Visual Sciences, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Sean Doherty
- Department of Ophthalmology and Visual Sciences, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Priya Janardhana
- Department of Ophthalmology and Visual Sciences, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Darren J Lee
- Department of Ophthalmology and Visual Sciences, University of Massachusetts Chan Medical School, Worcester, MA, USA.
| |
Collapse
|
4
|
Lasagni Vitar RM, Bonelli F, Rama P, Ferrari G. Immunity and pain in the eye: focus on the ocular surface. Clin Exp Immunol 2021; 207:149-163. [PMID: 35020868 PMCID: PMC8982975 DOI: 10.1093/cei/uxab032] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 10/21/2021] [Accepted: 12/10/2021] [Indexed: 02/03/2023] Open
Abstract
Most ocular diseases are associated with pain. While pain has been generally considered a mere (deleterious) additional symptom, it is now emerging that it is a key modulator of innate/adaptive immunity. Because the cornea receives the highest nerve density of the entire body, it is an ideal site to demonstrate interactions between pain and the immune response. Indeed, most neuropeptides involved in pain generation are also potent regulators of innate and adaptive leukocyte physiology. On the other hand, most inflammatory cells can modulate the generation of ocular pain through release of specific mediators (cytokines, chemokines, growth factors, and lipid mediators). This review will discuss the reciprocal role(s) of ocular surface (and specifically: corneal) pain on the immune response of the eye. Finally, we will discuss the clinical implications of such reciprocal interactions in the context of highly prevalent corneal diseases.
Collapse
Affiliation(s)
- Romina Mayra Lasagni Vitar
- Cornea and Ocular Surface Disease Unit, Eye Repair Lab, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Filippo Bonelli
- Cornea and Ocular Surface Disease Unit, Eye Repair Lab, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Paolo Rama
- Cornea and Ocular Surface Disease Unit, Eye Repair Lab, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Giulio Ferrari
- Cornea and Ocular Surface Disease Unit, Eye Repair Lab, IRCCS San Raffaele Scientific Institute, Milan, Italy,Correspondence: Giulio Ferrari, Cornea and Ocular Surface Unit, Eye Repair Lab, IRCCS San Raffaele Scientific Institute, Milan, Italy. E-mail:
| |
Collapse
|
5
|
Corneal Allografts: Factors for and against Acceptance. J Immunol Res 2021; 2021:5372090. [PMID: 34642632 PMCID: PMC8502534 DOI: 10.1155/2021/5372090] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 08/26/2021] [Accepted: 09/21/2021] [Indexed: 12/21/2022] Open
Abstract
Cornea is one of the most commonly transplanted tissues worldwide. However, it is usually omitted in the field of transplantology. Transplantation of the cornea is performed to treat many ocular diseases. It restores eyesight significantly improving the quality of life. Advancements in banking of explanted corneas and progressive surgical techniques increased availability and outcomes of transplantation. Despite the vast growth in the field of transplantation laboratory testing, standards for corneal transplantation still do not include HLA typing or alloantibody detection. This standard practice is based on immune privilege dogma that accounts for high success rates of corneal transplantation. However, the increasing need for retransplantation in high-risk patients with markedly higher risk of rejection causes ophthalmology transplantation centers to reevaluate their standard algorithms. In this review we discuss immune privilege mechanisms influencing the allograft acceptance and factors disrupting the natural immunosuppressive environment of the eye. Current developments in testing and immunosuppressive treatments (including cell therapies), when applied in corneal transplantation, may give very good results, decrease the possibility of rejection, and reduce the need for retransplantation, which is fairly frequent nowadays.
Collapse
|
6
|
Szeligo BM, Ivey AD, Boone BA. Poor Response to Checkpoint Immunotherapy in Uveal Melanoma Highlights the Persistent Need for Innovative Regional Therapy Approaches to Manage Liver Metastases. Cancers (Basel) 2021; 13:3426. [PMID: 34298647 PMCID: PMC8307800 DOI: 10.3390/cancers13143426] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 06/24/2021] [Accepted: 07/02/2021] [Indexed: 12/12/2022] Open
Abstract
Uveal melanoma is a cancer that develops from melanocytes in the posterior uveal tract. Metastatic uveal melanoma is an extremely rare disease that has a poor long-term prognosis, limited treatment options and a strong predilection for liver metastasis. Median overall survival has been reported to be 6 months and 1 year mortality of 80%. Traditional chemotherapy used in cutaneous melanoma is ineffective in uveal cases. Surgical resection and ablation is the preferred therapy for liver metastasis but is often not feasible due to extent of disease. In this review, we will explore treatment options for liver metastases from uveal melanoma, with a focus on isolated hepatic perfusion (IHP). IHP offers an aggressive regional therapy approach that can be used in bulky unresectable disease and allows high-dose chemotherapy with melphalan to be delivered directly to the liver without systemic effects. Long-term median overall survival has been reported to be as high as 27 months. We will also highlight the poor responses associated with checkpoint inhibitors, including an overview of the biological rationale driving this lack of immunotherapy effect for this disease. The persistent failure of traditional treatments and immunotherapy suggest an ongoing need for regional surgical approaches such as IHP in this disease.
Collapse
Affiliation(s)
- Brett M. Szeligo
- Division of Surgical Oncology, Department of Surgery, West Virginia University, Morgantown, WV 26508, USA;
| | - Abby D. Ivey
- Cancer Cell Biology, West Virginia University, Morgantown, WV 26508, USA;
| | - Brian A. Boone
- Division of Surgical Oncology, Department of Surgery, West Virginia University, Morgantown, WV 26508, USA;
- Department of Microbiology, Immunology and Cell Biology, West Virginia University, Morgantown, WV 26508, USA
| |
Collapse
|
7
|
Niederkorn JY. "Corneal Nerves, CD11c + Dendritic Cells and Their Impact on Ocular Immune Privilege". Front Immunol 2021; 12:701935. [PMID: 34220866 PMCID: PMC8253307 DOI: 10.3389/fimmu.2021.701935] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Accepted: 05/19/2021] [Indexed: 11/19/2022] Open
Abstract
The eye and the brain have limited capacities for regeneration and as such, immune-mediated inflammation can produce devastating consequences in the form of neurodegenerative diseases of the central nervous system or blindness as a result of ocular inflammatory diseases such as uveitis. Accordingly, both the eye and the brain are designed to limit immune responses and inflammation - a condition known as "immune privilege". Immune privilege is sustained by physiological, anatomical, and regulatory processes that conspire to restrict both adaptive and innate immune responses.
Collapse
Affiliation(s)
- Jerry Y. Niederkorn
- Department of Ophthalmology, University of Texas Southwestern Medical Center, Dallas, TX, United States
| |
Collapse
|
8
|
Abstract
Clear vision is dependent on features that protect the anatomical integrity of the eye (cornea and sclera) and those that contribute to internal ocular homeostasis by conferring hemangiogenic (avascular tissues and antiangiogenic factors), lymphangiogenic (lack of draining lymphatics), and immunologic (tight junctions that form blood-ocular barriers, immunosuppressive cells, and modulators) privileges. The later examples are necessary components that enable the eye to maintain an immunosuppressive environment that responds to foreign invaders in a deviated manner, minimizing destructive inflammation that would impair vision. These conditions allowed for the observations made by Medawar, in 1948, of delayed rejection of allogenic tissue grafts in the anterior chamber of mouse eye and permit the sequestration of foreign invaders (eg, Toxoplasma gondii) within the retina of healthy individuals. Yet successful development of intraocular drugs (biologics and delivery devices) has been stymied by adverse ocular pathology, much of which is driven by immune pathways. The eye can be intolerant of foreign protein irrespective of delivery route, and endogenous ocular cells have remarkable plasticity when recruited to preserve visual function. This article provides a review of current understanding of ocular immunology and the potential role of immune mechanisms in pathology observed with intraocular drug delivery.
Collapse
Affiliation(s)
| | | | - Sharmila Masli
- 12259Boston University School of Medicine, Boston, MA, USA
| |
Collapse
|
9
|
Cayabyab F, Nih LR, Yoshihara E. Advances in Pancreatic Islet Transplantation Sites for the Treatment of Diabetes. Front Endocrinol (Lausanne) 2021; 12:732431. [PMID: 34589059 PMCID: PMC8473744 DOI: 10.3389/fendo.2021.732431] [Citation(s) in RCA: 73] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Accepted: 08/13/2021] [Indexed: 01/08/2023] Open
Abstract
Diabetes is a complex disease that affects over 400 million people worldwide. The life-long insulin injections and continuous blood glucose monitoring required in type 1 diabetes (T1D) represent a tremendous clinical and economic burdens that urges the need for a medical solution. Pancreatic islet transplantation holds great promise in the treatment of T1D; however, the difficulty in regulating post-transplantation immune reactions to avoid both allogenic and autoimmune graft rejection represent a bottleneck in the field of islet transplantation. Cell replacement strategies have been performed in hepatic, intramuscular, omentum, and subcutaneous sites, and have been performed in both animal models and human patients. However more optimal transplantation sites and methods of improving islet graft survival are needed to successfully translate these studies to a clinical relevant therapy. In this review, we summarize the current progress in the field as well as methods and sites of islet transplantation, including stem cell-derived functional human islets. We also discuss the contribution of immune cells, vessel formation, extracellular matrix, and nutritional supply on islet graft survival. Developing new transplantation sites with emerging technologies to improve islet graft survival and simplify immune regulation will greatly benefit the future success of islet cell therapy in the treatment of diabetes.
Collapse
Affiliation(s)
- Fritz Cayabyab
- Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, United States
| | - Lina R. Nih
- Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, United States
- David Geffen School of Medicine at University of California, Los Angeles, CA, United States
| | - Eiji Yoshihara
- Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, United States
- David Geffen School of Medicine at University of California, Los Angeles, CA, United States
- *Correspondence: Eiji Yoshihara,
| |
Collapse
|
10
|
Capacity of Retinal Ganglion Cells Derived from Human Induced Pluripotent Stem Cells to Suppress T-Cells. Int J Mol Sci 2020; 21:ijms21217831. [PMID: 33105725 PMCID: PMC7660053 DOI: 10.3390/ijms21217831] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Revised: 10/08/2020] [Accepted: 10/20/2020] [Indexed: 02/06/2023] Open
Abstract
Retinal ganglion cells (RGCs) are impaired in patients such as those with glaucoma and optic neuritis, resulting in permanent vision loss. To restore visual function, development of RGC transplantation therapy is now underway. Induced pluripotent stem cells (iPSCs) are an important source of RGCs for human allogeneic transplantation. We therefore analyzed the immunological characteristics of iPSC-derived RGCs (iPSC-RGCs) to evaluate the possibility of rejection after RGC transplantation. We first assessed the expression of human leukocyte antigen (HLA) molecules on iPSC-RGCs using immunostaining, and then evaluated the effects of iPSC-RGCs to activate lymphocytes using the mixed lymphocyte reaction (MLR) and iPSC-RGC co-cultures. We observed low expression of HLA class I and no expression of HLA class II molecules on iPSC-RGCs. We also found that iPSC-RGCs strongly suppressed various inflammatory immune cells including activated T-cells in the MLR assay and that transforming growth factor-β2 produced by iPSC-RGCs played a critical role in suppression of inflammatory cells in vitro. Our data suggest that iPSC-RGCs have low immunogenicity, and immunosuppressive capacity on lymphocytes. Our study will contribute to predicting immune attacks after RGC transplantation.
Collapse
|
11
|
Neelam S, Niederkorn JY. Corneal Nerve Ablation Abolishes Ocular Immune Privilege by Downregulating CD103 on T Regulatory Cells. Invest Ophthalmol Vis Sci 2020; 61:25. [PMID: 32305043 PMCID: PMC7401639 DOI: 10.1167/iovs.61.4.25] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Accepted: 02/28/2020] [Indexed: 11/24/2022] Open
Abstract
Purpose Severing corneal nerves during orthotopic corneal transplantation elicits the elaboration of the neuropeptide substance P (SP), which induces the generation of CD11c+ contrasuppressor (CS) cells. CS cells disable T regulatory cells (Tregs) that are induced when antigens enter the anterior chamber (AC), either by direct injection or by orthotopic corneal transplantation. This study examined the crucial cell surface molecules on Tregs that are adversely affected by CS cells that are generated by severing corneal nerves. Methods CS cells were induced by producing shallow 2.0-mm circular incisions in the corneal epithelium in BALB/c mice. CD8+ Tregs were generated by injecting ovalbumin into the AC. The effects of CS cells and SP on the expression and function of two cell surface molecules (CD103 and the receptor of interferon-γ) that are crucial for the induction and function of CD8+ Tregs were analyzed. Results SP converted CD11c+, but not CD11c- , dendritic cells (DCs) to CS cells. Severing corneal nerves resulted in a 66% reduction in the expression of CD103 on CD8+ AC-associated immune deviation (ACAID) Tregs, and a 50% reduction in the interferon-γ receptor (IFN-γR). These effects could be mimicked in vitro by coculturing CS cells with CD8+ ACAID Tregs. Conclusions The elaboration of SP in response to corneal nerve ablation converts CD11c+ DCs to CS cells. CS cells disable CD8+ ACAID Tregs by downregulating two crucial cell surface molecules, CD103 and IFN-γR, by an SP-dependent pathway. Blocking this pathway may provide a means of restoring ocular immune privilege in corneas subjected to corneal nerve injury.
Collapse
Affiliation(s)
- Sudha Neelam
- Department of Ophthalmology, University of Texas Southwestern Medical Center, Dallas, Texas, United States
| | - Jerry Y. Niederkorn
- Department of Ophthalmology, University of Texas Southwestern Medical Center, Dallas, Texas, United States
| |
Collapse
|
12
|
Abstract
Autoimmune uveitis is a sight-threatening, rare disease, potentially leading to blindness. Uveitis is a synonym for intraocular inflammation, presenting as various clinical phenotypes with different underlying immune responses in patients, whereas different animal models usually represent one certain clinical and immunological type of uveitis due to genetic uniformity and the method of disease induction. T cells recognizing intraocular antigens initiate the disease, recruiting inflammatory cells (granulocytes, monocytes/macrophages) to the eyes, which cause the damage of the tissue. The treatment of uveitis so far aims at downregulation of inflammation to protect the ocular tissues from damage, and at immunosuppression to stop fueling T cell reactivity. Uveitis is usually prevented by specific mechanisms of the ocular immune privilege and the blood-eye-barriers, but once the disease is induced, mechanisms of the immune privilege as well as a variety of novel regulatory features including new Treg cell populations and suppressive cytokines are induced to downregulate the ocular inflammation and T cell responses and to avoid relapses and chronicity. Here we describe mechanisms of regulation observed in experimental animal models as well as detected in studies with peripheral lymphocytes from patients.
Collapse
|
13
|
Tsagkaraki IM, Kourouniotis CD, Gomatou GL, Syrigos NK, Kotteas EA. Orbital metastases of invasive lobular breast carcinoma. Breast Dis 2019; 38:85-91. [PMID: 31640079 DOI: 10.3233/bd-190398] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Breast cancer is the main site of origin of orbital metastatic disease. Although invasive lobular breast carcinoma accounts for 10-15% of all breast cancer cases, it has been noticed that it metastasizes to the orbit more often compared to breast cancer of no special type (NST). The pathogenesis of this metastasis is not entirely understood; however, it seems that the unique tissue-specific characteristics of orbital microenvironment might contribute to metastatic disease in this particular site. Given the increasing survival of breast cancer patients and the prolonged metastatic potential of invasive lobular breast carcinoma, it is possible that the incidence of this rare metastasis might increase in the future. The purpose of this review is to present clinical manifestations, immunohistochemical characteristics and therapeutic options for orbital metastases from invasive lobular carcinoma.
Collapse
Affiliation(s)
- Ismini Michail Tsagkaraki
- Third Department of Medicine, Oncology Unit, Sotiria General Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | | | - Georgia Leonidas Gomatou
- Third Department of Medicine, Oncology Unit, Sotiria General Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Nikolaos Konstantinos Syrigos
- Third Department of Medicine, Oncology Unit, Sotiria General Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Elias Alexandros Kotteas
- Third Department of Medicine, Oncology Unit, Sotiria General Hospital, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
14
|
Taylor AW, Ng TF. Negative regulators that mediate ocular immune privilege. J Leukoc Biol 2018; 103:1179-1187. [PMID: 29431864 PMCID: PMC6240388 DOI: 10.1002/jlb.3mir0817-337r] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Revised: 12/15/2017] [Accepted: 01/10/2018] [Indexed: 08/13/2023] Open
Abstract
The ocular microenvironment has adapted several negative regulators of inflammation to maintain immune privilege and health of the visual axis. Several constitutively produced negative regulators within the eye TGF-β2, α-melanocyte stimulating hormone (α-MSH), Fas ligand (FasL), and PD-L1 standout because of their capacity to influence multiple pathways of inflammation, and that they are part of promoting immune tolerance. These regulators demonstrate the capacity of immune privilege to prevent the activation of inflammation, and to suppress activation of effector immune cells even under conditions of ocular inflammation induced by endotoxin and autoimmune disease. In addition, these negative regulators promote and expand immune cells that mediate regulatory and tolerogenic immunity. This in turn makes the immune cells themselves negative regulators of inflammation. This provides for a greater understanding of immune privilege in that it includes both molecular and cellular negative regulators of inflammation. This would mean that potentially new approaches to the treatment of autoimmune disease can be developed through the use of molecules and cells as negative regulators of inflammation.
Collapse
Affiliation(s)
- Andrew W Taylor
- Boston University School of Medicine, Boston, Massachusetts, USA
| | - Tat Fong Ng
- Boston University School of Medicine, Boston, Massachusetts, USA
| |
Collapse
|
15
|
Kim DW, Anderson J, Patel SP. Immunotherapy for uveal melanoma. Melanoma Manag 2016; 3:125-135. [PMID: 30190881 DOI: 10.2217/mmt-2015-0006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2015] [Accepted: 04/13/2016] [Indexed: 11/21/2022] Open
Abstract
Uveal melanoma (UM) is a rare cancer with a high mortality rate. In comparison to cutaneous melanoma, UM has unique immunological features. Arising in the immune suppressive environment of the eye, it maintains immune resistance once metastatic. This is considered a major obstacle for successful immunotherapy in UM. However, a growing body of evidence suggests strategies that may abrogate resistance and enhance antitumor immunity in UM. Recently, three new immune agents have been approved for melanoma. While these drugs demonstrate durable clinical responses with long-term remissions in metastatic cutaneous melanoma, only limited data exist in metastatic UM. In this review, immunological aspects of UM and data from clinical studies of immunotherapeutic agents and regimens for UM will be discussed.
Collapse
Affiliation(s)
- Dae Won Kim
- Moffitt Cancer Center, Tampa, FL 33612, USA.,Moffitt Cancer Center, Tampa, FL 33612, USA
| | - Jaime Anderson
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd Unit 0430, Houston, TX 77030, USA.,Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd Unit 0430, Houston, TX 77030, USA
| | - Sapna P Patel
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd Unit 0430, Houston, TX 77030, USA.,Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd Unit 0430, Houston, TX 77030, USA
| |
Collapse
|
16
|
Taylor AW. Ocular Immune Privilege and Transplantation. Front Immunol 2016; 7:37. [PMID: 26904026 PMCID: PMC4744940 DOI: 10.3389/fimmu.2016.00037] [Citation(s) in RCA: 114] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Accepted: 01/25/2016] [Indexed: 11/30/2022] Open
Abstract
Allografts are afforded a level of protection from rejection within immune-privileged tissues. Immune-privileged tissues involve mechanisms that suppress inflammation and promote immune tolerance. There are anatomical features, soluble factors, membrane-associated proteins, and alternative antigen-presenting cells (APC) that contribute to allograft survival in the immune-privileged tissue. This review presents the current understanding of how the mechanism of ocular immune privilege promotes tolerogenic activity by APC, and T cells in response to the placement of foreign antigen within the ocular microenvironment. Discussed will be the unique anatomical, cellular, and molecular mechanisms that lessen the chance for graft destroying immune responses within the eye. As more is understood about the molecular mechanisms of ocular immune privilege greater is the potential for using these molecular mechanisms in therapies to prevent allograft rejection.
Collapse
Affiliation(s)
- Andrew W Taylor
- Department of Ophthalmology, Boston University School of Medicine , Boston, MA , USA
| |
Collapse
|
17
|
Bolaños-Jiménez R, Navas A, López-Lizárraga EP, de Ribot FM, Peña A, Graue-Hernández EO, Garfias Y. Ocular Surface as Barrier of Innate Immunity. Open Ophthalmol J 2015; 9:49-55. [PMID: 26161163 PMCID: PMC4484240 DOI: 10.2174/1874364101509010049] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2014] [Revised: 04/06/2015] [Accepted: 04/13/2015] [Indexed: 01/08/2023] Open
Abstract
Sight is one of the most important senses that human beings possess. The ocular system is a complex structure equipped with mechanisms that prevent or limit damage caused by physical, chemical, infectious and environmental factors. These mechanisms include a series of anatomical, cellular and humoral factors that have been a matter of study. The cornea is not only the most powerful and important lens of the optical system, but also, it has been involved in many other physiological and pathological processes apart from its refractive nature; the morphological and histological properties of the cornea have been thoroughly studied for the last fifty years; drawing attention in its molecular characteristics of immune response. This paper will review the anatomical and physiological aspects of the cornea, conjunctiva and lacrimal apparatus, as well as the innate immunity at the ocular surface.
Collapse
Affiliation(s)
- Rodrigo Bolaños-Jiménez
- Research Unit, Institute of Ophthalmology, Conde de Valenciana Foundation, Chimalpopoca 14, Obrera, CP 06800, Mexico City, México ; Ophthalmology Department, Regional Hospital, Adolfo López Mateos, ISSSTE, México City, México
| | - Alejandro Navas
- Research Unit, Institute of Ophthalmology, Conde de Valenciana Foundation, Chimalpopoca 14, Obrera, CP 06800, Mexico City, México ; Department of Cataract and refractive surgery, Institute of Ophthalmology, Conde de Valenciana Foundation, Mexico City, México
| | - Erika Paulina López-Lizárraga
- Department of Cataract and refractive surgery, Institute of Ophthalmology, Conde de Valenciana Foundation, Mexico City, México
| | - Francesc March de Ribot
- Department of Retina, Institute of Ophthalmology, Conde de Valenciana Foundation, Mexico City, México
| | - Alexandra Peña
- Ophthalmology Department, Regional Hospital, Adolfo López Mateos, ISSSTE, México City, México
| | - Enrique O Graue-Hernández
- Research Unit, Institute of Ophthalmology, Conde de Valenciana Foundation, Chimalpopoca 14, Obrera, CP 06800, Mexico City, México ; Department of Cataract and refractive surgery, Institute of Ophthalmology, Conde de Valenciana Foundation, Mexico City, México
| | - Yonathan Garfias
- Research Unit, Institute of Ophthalmology, Conde de Valenciana Foundation, Chimalpopoca 14, Obrera, CP 06800, Mexico City, México ; Department of Biochemistry, Faculty of Medicine, Universidad Nacional Autónoma de México, 04510. Mexico City, Mexico
| |
Collapse
|
18
|
Morgan JT, Wood JA, Walker NJ, Raghunathan VK, Borjesson DL, Murphy CJ, Russell P. Human trabecular meshwork cells exhibit several characteristics of, but are distinct from, adipose-derived mesenchymal stem cells. J Ocul Pharmacol Ther 2014; 30:254-66. [PMID: 24456002 DOI: 10.1089/jop.2013.0175] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
PURPOSE To support the growing promise of regenerative medicine in glaucoma, we characterized the similarities and differences between human trabecular meshwork (HTM) cells and human mesenchymal stem cells (hMSCs). METHODS HTM cells and hMSCs were phenotypically characterized by flow cytometry. Using quantitative polymerase chain reaction, the expression of myoc, angptl7, sox2, pou5f1, and notch1 was determined in both cell types with and without dexamethasone (Dex). Immunosuppressive behavior of HTM cells and hMSCs was determined using T cells activated with phytohemagglutinin. T-cell proliferation was determined using BrdU incorporation and flow cytometry. Multipotency of HTM cells and hMSCs was determined using adipogenic and osteogenic differentiation media as well as aqueous humor (AH). Alpha-smooth muscle actin (αSMA) expression was determined in HTM cells, hMSCs, and HTM tissue. RESULTS Phenotypically, HTM and hMSCs expressed CD73, CD90, CD105, and CD146 but not CD31, CD34, and CD45 and similar sox2, pou5f1, and notch1 expression. Both cell types suppressed T-cell proliferation. However, HTM cells, but not hMSCs, upregulated myoc and angptl7 in response to Dex. Additionally, HTM cells did not differentiate into adipocytes or osteocytes. Culture of hMSCs in 20%, but not 100%, AH potently induced alkaline phosphatase activity. HTM cells in culture possessed uniformly strong expression of αSMA, which contrasted with the limited expression in hMSCs and spatially discrete expression in HTM tissue. CONCLUSIONS HTM cells possess a number of important similarities with hMSCs but lack multipotency, one of the defining characteristics of stem cells. Further work is needed to explore the molecular mechanisms and functional implications underlying the phenotypic similarities.
Collapse
Affiliation(s)
- Joshua T Morgan
- 1 Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California , Davis, California
| | | | | | | | | | | | | |
Collapse
|
19
|
Somatostatin Expression in Human Hair Follicles and Its Potential Role in Immune Privilege. J Invest Dermatol 2013; 133:1722-30. [DOI: 10.1038/jid.2013.53] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
20
|
Shaharuddin B, Ahmad S, Meeson A, Ali S. Concise review: immunological properties of ocular surface and importance of limbal stem cells for transplantation. Stem Cells Transl Med 2013; 2:614-24. [PMID: 23817133 DOI: 10.5966/sctm.2012-0143] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Cornea transplantation has been considered to be different from other solid organ transplantation because of the assumed immune-privileged state of the anterior chamber of the eye. Three major lines of thought regarding the molecular mechanisms of immune privilege in the eye are as follows: (a) anatomical, cellular, and molecular barriers in the eye; (b) anterior chamber-associated immune deviation; and (c) immunosuppressive microenvironment in the eye. However, cornea transplants suffer allograft rejection when breached by vascularization. In recent developments, cellular corneal transplantation from cultivated limbal epithelial cells has shown impressive advances as a future therapy. The limbal stem cell niche contains stem cells that promote proliferation and migration and have immunosuppressive mechanisms to protect them from immunological reactions. Limbal stem cells are also noted to display an enhanced expression of genes for the antiapoptotic proteins, a property that is imperative for the survival of transplanted tissues. Further investigation of the molecular mechanisms regulating the immune regulation of limbal stem cells is relevant in the clinical setting to promote the survival of whole corneal and limbal stem cell transplantation.
Collapse
Affiliation(s)
- Bakiah Shaharuddin
- Institute of Genetic Medicine, Newcastle University, Newcastle-Upon-Tyne, United Kingdom
| | | | | | | |
Collapse
|
21
|
Huntjens B, Charman WN, Workman H, Hosking SL, O’Donnell C. Short-term stability in refractive status despite large fluctuations in glucose levels in diabetes mellitus type 1 and 2. PLoS One 2012; 7:e52947. [PMID: 23285232 PMCID: PMC3532445 DOI: 10.1371/journal.pone.0052947] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2012] [Accepted: 11/26/2012] [Indexed: 11/26/2022] Open
Abstract
PURPOSE This work investigates how short-term changes in blood glucose concentration affect the refractive components of the diabetic eye in patients with long-term Type 1 and Type 2 diabetes. METHODS Blood glucose concentration, refractive error components (mean spherical equivalent MSE, J0, J45), central corneal thickness (CCT), anterior chamber depth (ACD), crystalline lens thickness (LT), axial length (AL) and ocular aberrations were monitored at two-hourly intervals over a 12-hour period in: 20 T1DM patients (mean age ± SD) 38±14 years, baseline HbA1c 8.6±1.9%; 21 T2DM patients (mean age ± SD) 56±11 years, HbA1c 7.5±1.8%; and in 20 control subjects (mean age ± SD) 49±23 years, HbA1c 5.5±0.5%. The refractive and biometric results were compared with the corresponding changes in blood glucose concentration. RESULTS Blood glucose concentration at different times was found to vary significantly within (p<0.0005) and between groups (p<0.0005). However, the refractive error components and ocular aberrations were not found to alter significantly over the day in either the diabetic patients or the control subjects (p>0.05). Minor changes of marginal statistical or optical significance were observed in some biometric parameters. Similarly there were some marginally significant differences between the baseline biometric parameters of well-controlled and poorly-controlled diabetic subjects. CONCLUSION This work suggests that normal, short-term fluctuations (of up to about 6 mM/l on a timescale of a few hours) in the blood glucose levels of diabetics are not usually associated with acute changes in refractive error or ocular wavefront aberrations. It is therefore possible that factors other than refractive error fluctuations are sometimes responsible for the transient visual problems often reported by diabetic patients.
Collapse
Affiliation(s)
- Byki Huntjens
- Division of Optometry and Visual Science, City University London, London, United Kingdom
| | - W. Neil Charman
- Faculty of Life Sciences, The University of Manchester, London, United Kingdom
| | - Helena Workman
- School of Life and Health Sciences, Aston University, Birmingham, United Kingdom
| | - Sarah L. Hosking
- Division of Optometry and Visual Science, City University London, London, United Kingdom
- Department of Ophthalmology, University of Melbourne, Melbourne, Australia
| | - Clare O’Donnell
- Faculty of Life Sciences, The University of Manchester, London, United Kingdom
- School of Life and Health Sciences, Aston University, Birmingham, United Kingdom
- Optegra Eye Sciences, Manchester, United Kingdom
| |
Collapse
|
22
|
McKenna KC, Previte DM. Influence of CD8+ T regulatory cells on intraocular tumor development. Front Immunol 2012; 3:303. [PMID: 23060881 PMCID: PMC3460369 DOI: 10.3389/fimmu.2012.00303] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2012] [Accepted: 09/10/2012] [Indexed: 11/13/2022] Open
Abstract
The interior of the eye, or uvea, is a site of immune privilege where certain immune responses are attenuated or completely excluded to protect non-regenerating tissues essential for vision. One consequence of this immunoregulation is compromised immune mediated elimination of intraocular tumors. For example, certain murine tumor cell lines which are rejected by host immune responses when transplanted in the skin grow progressively when placed in the anterior chamber (a.c.) of the eye. Progressive ocular tumor growth occurs despite induction of tumor-specific CD8+ T cell responses capable of eliminating a subsequent tumor challenge in the skin or opposite eye. Why these CD8+ T effectors fail to eliminate established ocular tumors is not known. It is well appreciated that growth of tumors in the a.c. induces the generation of immunosuppressive CD8+ T regulatory (Treg) cells. However, the contribution of CD8+ Treg in ocular tumor progression remains unclear. Several studies indicate that these CD8+ Treg target responding CD4+ T cells to inhibit their induction of macrophage-dependent delayed type hypersensitivity (DTH) responses to tumor antigens (Ags). However, induction of tumor-specific CD4+ T cell responses does not assure intraocular tumor elimination. This review is focused on how CD8+ Treg could influence the tumoricidal activity of ocular tumor-specific CD8+ T effector cells.
Collapse
Affiliation(s)
- Kyle C McKenna
- Departments of Ophthalmology and Immunology/Medicine, University of Pittsburgh, University of Pittsburgh Cancer Institute Pittsburgh, PA, USA
| | | |
Collapse
|
23
|
Fonollosa A, Coronado E, Catalan R, Gutierrez M, Macia C, Zapata MA, Martinez-Alday N, Simo R, Garcia-Arumi J. Vitreous levels of somatostatin in patients with chronic uveitic macular oedema. Eye (Lond) 2012; 26:1378-83. [PMID: 22878444 DOI: 10.1038/eye.2012.161] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
PURPOSE Intravitreal somatostatin (SST) levels are decreased in patients with diabetic macular oedema. This deficit may be involved in the pathogenesis of this condition. The aim of the present study was to determine SST concentration in the vitreous fluid of patients with chronic uveitic macular oedema (CUMO) and quiescent intraocular inflammation. METHODS Plasma and vitreous fluid samples were obtained during vitrectomy from 11 eyes of patients with CUMO and from 42 eyes of control subjects (idiopathic epiretinal membrane, macular hole). SST concentration was measured by radioimmunoassay. STATISTICS χ(2)-square test, Mann-Whitney U-test, Wilcoxon test, Spearman's rank correlation coefficient, and multivariant linear regression models. RESULTS Plasma SST concentrations were similar in uveitic patients and controls (28.25 pg/ml (21.3-31) vs 28.7 pg/ml (22-29.5); P=0.869). A higher vitreous concentration of proteins was found in uveitic patients (1.59±0.38 mg/ml vs 0.73±0.32 mg/ml, P<0.0001). Vitreous SST was markedly lower in uveitic patients, both in absolute terms and after adjusting for total intravitreous protein concentration (39.37 pg/ml (6.16-172) vs 486.73 pg/ml (4.7-1833), P<0.0001; 33.1 pg/mg (3.9-215.74) vs 629.75 pg/mg (6.91-2024), P<0.0001). No correlations were found between plasma and vitreous concentration of SST in either group (ρ=0.191, P=0.57 and ρ=0.49, P=0.66). There were no correlations between vitreous SST concentration and visual acuity or macular thickness in uveitic patients (ρ=0.302, P=0.31 and ρ=0.45, P=0.13). CONCLUSIONS Intravitreous SST is decreased in patients with CUMO and quiescent intraocular inflammation. The deficit of SST may have a role in the pathogenesis of this condition.
Collapse
Affiliation(s)
- A Fonollosa
- Department of Ophthalmology, Hospital de Cruces, Universidad del País Vasco, Barakaldo, Spain.
| | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Niederkorn JY. Ocular immune privilege and ocular melanoma: parallel universes or immunological plagiarism? Front Immunol 2012; 3:148. [PMID: 22707951 PMCID: PMC3374415 DOI: 10.3389/fimmu.2012.00148] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2012] [Accepted: 05/21/2012] [Indexed: 12/31/2022] Open
Abstract
Evidence of immune privilege in the eye was recorded almost 140 years ago, yet interest in immune privilege languished for almost a century. However, the past 35 years have witnessed a plethora of research and a rekindled interest in the mechanisms responsible for immune privilege in the anterior chamber of the eye. This research has demonstrated that multiple anatomical, structural, physiological, and immunoregulatory processes contribute to immune privilege and remind us of the enormous complexity of this phenomenon. It is widely accepted that immune privilege is an adaptation for reducing the risk of immune-mediated inflammation in organs such as the eye and brain whose tissues have a limited capacity to regenerate. Recent findings suggest that immune privilege also occurs in sites where stem cells reside and raise the possibility that immune privilege is also designed to prevent the unwitting elimination of stem cells by immune-mediated inflammation at these sites. Uveal melanoma arises within the eye and as such, benefits from ocular immune privilege. A significant body of research reveals an intriguing parallel between the mechanisms that contribute to immune privilege in the eye and those strategies used by uveal melanoma cells to evade immune elimination once they have disseminated from the eye and establish metastatic foci in the liver. Uveal melanoma metastases seem to have “plagiarized” the blueprints used for ocular immune privilege to create “ad hoc immune privileged sites” in the liver.
Collapse
Affiliation(s)
- Jerry Y Niederkorn
- Department of Ophthalmology, University of Texas Southwestern Medical Center Dallas, TX, USA
| |
Collapse
|
25
|
Zhou R, Horai R, Mattapallil MJ, Caspi RR. A new look at immune privilege of the eye: dual role for the vision-related molecule retinoic acid. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2011; 187:4170-7. [PMID: 21918194 PMCID: PMC3186879 DOI: 10.4049/jimmunol.1101634] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The eye is an immunologically privileged and profoundly immunosuppressive environment. Early studies reported inhibition of T cell proliferation, IFN-γ production, and generation of regulatory T cells (Tregs) by aqueous humor (AH) and identified TGF-β as a critical factor. However, T cell subsets including Foxp3(+) Treg and Th17 were unknown at that time, as was the role of retinoic acid (RA) in Treg induction. Consequently, the effect of the ocular microenvironment on T cell lineage commitment and function, and the role of RA in this process, had not been explored. We now use gene-manipulated mice and highly purified T cell populations to demonstrate that AH suppresses lineage commitment and acquisition of Th1 and Th17 effector function of naive T cells, manifested as reduction of lineage-specific transcription factors and cytokines. Instead, AH promoted its massive conversion to Foxp3(+) Tregs that expressed CD25, GITR, CTLA-4, and CD103 and were functionally suppressive. TGF-β and RA were both needed and synergized for Treg conversion by AH, with TGF-β-enhancing T cell expression of RA receptor α. Newly converted Foxp3(+) Tregs were unstable, but were stabilized upon continued exposure to AH or by the DNA demethylating agent 5-aza-2'-deoxycytidine. In contrast, T cells already committed to effector function were resistant to the suppressive and Treg-inducing effects of AH. We conclude that RA in the eye plays a dual role: in vision and in immune privilege. Nevertheless, primed effector T cells are relatively insensitive to AH, helping to explain their ability to induce uveitis despite an inhibitory ocular microenvironment.
Collapse
Affiliation(s)
- Ru Zhou
- Laboratory of Immunology, National Eye Institute, National Institutes of Health, Bethesda, MD 20892-1857, USA
| | | | | | | |
Collapse
|
26
|
Hori J, Vega JL, Masli S. Review of ocular immune privilege in the year 2010: modifying the immune privilege of the eye. Ocul Immunol Inflamm 2011; 18:325-33. [PMID: 20849282 DOI: 10.3109/09273948.2010.512696] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The original evidence for the existence of immunologically privileged sites in the body was based on the prolonged survival of genetically disparate transplanted tissue in the anterior chamber of the eye. The failure of the immune system to elicit an immune response in this and other such sites constitutes the hallmark of the immune privilege status. The remarkably successful field of corneal transplantation in clinical practice is undoubtedly associated with corneal immune privilege. Several investigations have addressed the regulatory mechanisms governing this phenomenon, which involves a complex interplay between multiple molecular and cellular pathways. Furthermore, the use of various transgenic mouse models has facilitated the identification of critical pathways, which upon disruption can modify the immune privileged status of the eye. Understanding these pathways not only reveals the mechanisms underlying various ocular inflammatory disease conditions, but also has clinical implications for the transplantation field and for the treatment of autoimmunity.
Collapse
Affiliation(s)
- Junko Hori
- Department of Ophthalmology, Nippon Medical School, Tokyo, Japan
| | | | | |
Collapse
|
27
|
Abstract
The eye is one of the immune privilege sites of the body that is consequently protected from the detrimental and potentially blinding influences of immunologic inflammation. Within the eye, the anterior chamber has been recognized for its immune privilege property for many years now; however, a similar property detectable in the subretinal space has only recently been appreciated. These ocular sites are not only equipped with specialized mechanisms that barricade local inflammatory responses, but also induce systemic regulatory immune response. Numerous studies have characterized molecular and cellular mechanisms involved in conferring both these sites with an immune privilege status. Pigmented epithelial cells lining the anterior chamber in the iris and ciliary body area as well as those in the retina are endowed with immunomodulatory properties that contribute to ocular immune privilege. These cells, via expression of either soluble factors or membrane molecules, inhibit inflammatory T cell activation and promote the generation of regulatory T cells. In the anterior chamber resident antigen-presenting cells, influenced by the various immunosuppressive factors present in the aqueous humor, capture ocular antigens and present them in the spleen to T cells in association with NKT cells and marginal zone B cells. Immunomodulatory microenvironment created by these cells helps generate regulatory T cells, capable of interrupting the induction as well as expression of inflammatory responses. Furthermore, neural regulation of both intraocular and systemic regulatory mechanisms also contributes to ocular immune privilege.
Collapse
Affiliation(s)
- Sharmila Masli
- Department of Ophthalmology, Harvard Medical School, Schepens Eye Research Institute, Boston, MA, USA.
| | | |
Collapse
|
28
|
Abstract
The phrase "immune privilege" was coined by Peter Medawar to describe the absence of an immune response to allografts placed into the anterior chamber of the eye or brain. We now understand that immune privilege is more than a passive microenvironment with a distinctive anatomical structure that holds back immunity. The ocular microenvironment actively engages the immune system with immunosuppressive biochemical mechanisms. The unique characteristics of ocular immune privilege appear designed to protect the eye from damage while preserving foveal vision, thus providing the host with a definite survival advantage. However, the protection is not always sufficient and the eye becomes susceptible to uveitis. Uveitis is an intraocular inflammatory disorder that encompasses a wide range of underlying etiologies. It may be idiopathic or associated with systemic disease or infection. Understanding the biochemistry of immune privilege has the potential to identify its weaknesses that allow for immunity to break through.
Collapse
Affiliation(s)
- Andrew W Taylor
- Department of Ophthalmology, Boston University School of Medicine, Boston, Massachusetts, USA
| | | |
Collapse
|
29
|
Denniston AK, Kottoor SH, Khan I, Oswal K, Williams GP, Abbott J, Wallace GR, Salmon M, Rauz S, Murray PI, Curnow SJ. Endogenous Cortisol and TGF-β in Human Aqueous Humor Contribute to Ocular Immune Privilege by Regulating Dendritic Cell Function. THE JOURNAL OF IMMUNOLOGY 2010; 186:305-11. [DOI: 10.4049/jimmunol.1001450] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
30
|
Abstract
Immune privilege is the condition in which selected immune responses are suppressed or excluded in certain organs, such as the eye. Immune privilege in the eye was described over 130 years ago, but its significance was not appreciated until the early 1950s. Investigations beginning in the 1970s ushered in a new era and revealed that ocular immune privilege is due to anatomical, physiological, and immunoregulatory processes that prevent the induction and expression of immune-mediated inflammation. It is widely believed that immune privilege is an adaptation for reducing immune-mediated injury to ocular cells that have limited or no capacity for regeneration.
Collapse
Affiliation(s)
- Jerry Y Niederkorn
- Department of Ophthalmology, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA.
| | | |
Collapse
|
31
|
Abstract
Mechanisms that maintain ocular immune privilege may contribute to ocular tumor progression by inhibiting tumoricidal immune responses. Consistent with that notion are observations from transplantable tumor models in mice demonstrating that the tumoricidal activity of CD8(+) cytolytic T lymphocytes (CTL) may be inhibited directly by interfering with CTL effector function in the eye or indirectly by abrogating the effector function of CD8+ T cell-activated intratumoral macrophages that are critical for ocular tumor rejection. In addition, epigenetic gene regulation by factors within the ocular tumor environment favors the generation of tumor variants that are resistant to CD8(+) CTL. Intratumoral macrophages may be essential for eliminating these variants because, unlike CTL, their tumoricidal activity is nonspecific. Hence, the inhibition of macrophage effector function within the eye, presumably to preserve immune privilege by minimizing ocular immunopathology, may hasten the outgrowth of tumor escape variants which contributes to ocular tumor progression.
Collapse
Affiliation(s)
- Kyle C McKenna
- Department of Ophthalmology, University of Pittsburgh, Pittsburgh, PA 15213, USA.
| | | |
Collapse
|
32
|
Abstract
It has been over 60 years since the phrase immune privilege was used by Sir Peter Medawar to describe the lack of an immune response against allografts placed into the ocular microenvironment. Since then, we have come to understand that the mechanisms of ocular immune privilege include unique anatomical features of a blood barrier and a lack of direct lymphatic drainage. Also, we know that the ocular microenvironment is rich with immunosuppressive molecules that influence the activity of immune cells. Moreover, the placement of foreign antigen into the ocular microenvironment can induce a systemic form of tolerance to the foreign antigen called anterior chamber-associated immune deviation (ACAID). Many soluble immunomodulators are found in aqueous humour, and are a mixture of growth factors, cytokines, neuropeptides, and soluble receptors. This is a continuously growing list. The mechanisms of ocular immune privilege induce apoptosis, promote the production of anti-inflammatory cytokines, and mediate the activation of antigen-specific regulatory immunity. These mechanisms of immune privilege also attempt to impose themselves upon immunity within the uveitic eye. The adaptation of several anatomical and biochemical mechanisms to establish an immune privileged microenvironment within the eye makes the eye immunologically unique. It is a tissue site where we may learn how immunity is regulated in inflammation and at rest. Success in translating the lessons of ocular immune privilege to other tissues has the potential to drastically change the therapy and clinical outcomes of autoimmune diseases and allograft survival.
Collapse
Affiliation(s)
- A W Taylor
- Department of Ophthalmology, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
33
|
Abstract
The notion that the immune system might control the growth of tumors was suggested over 100 years ago by the eminent microbiologist Paul Ehrlich. This concept was refined and expanded by Burnet and Thomas 50 years later with their articulation of the "immune surveillance" hypothesis. In its simplest form, the immune surveillance hypothesis suggests that neoplasms arise spontaneously and express novel antigens that are recognized by the immune system, which either eliminates the tumors or restrains their growth. Within the eye, immune responses are controlled and sometimes profoundly inhibited - a condition known as immune privilege. Immune privilege in the eye is the result of a complex array of anatomical, physiological, and immunoregulatory mechanisms that prevent the induction and expression of many immune responses. Tumors arising in the eye would seem to have an advantage in evading immune surveillance due to ocular immune privilege. Uveal melanoma, the most common and malignant intraocular tumor in adults, not only benefits from the immune privilege of the eye but also has adopted many of the mechanisms that contribute to ocular immune privilege as a strategy for protecting uveal melanoma cells once they leave the sanctuary of the eye and are disseminated systemically in the form of metastases. Although the immune system possesses a battery of effector mechanisms designed to rid the body of neoplasms, tumors are capable of rapidly evolving and countering even the most sophisticated immunological effector mechanisms. To date, tumors seem to be winning this arms race, but an increased understanding of these mechanisms should provide insights for designing immunotherapy that was envisioned over half a century ago, but has failed to materialize to date.
Collapse
Affiliation(s)
- Jerry Y Niederkorn
- Department of Ophthalmology, University of Texas Southwestern Medical Center, Dallas, TX 75390-9057, USA.
| |
Collapse
|
34
|
Joachim SC, Bruns K, Lackner KJ, Pfeiffer N, Grus FH. Antibodies to α B-Crystallin, Vimentin, and Heat Shock Protein 70 in Aqueous Humor of Patients with Normal Tension Glaucoma and IgG Antibody Patterns Against Retinal Antigen in Aqueous Humor. Curr Eye Res 2009; 32:501-9. [PMID: 17612966 DOI: 10.1080/02713680701375183] [Citation(s) in RCA: 97] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
PURPOSE To show the existence of IgG antibodies against retinal antigens in aqueous humor of normal tension glaucoma patients. METHODS Forty-two patients were included in this study. Aqueous humor was collected from control subjects (CO; n = 21) and patients with normal tension glaucoma (NTG; n = 21). Western blot methods against bovine retinal antigens were used to detect the IgG antibody patterns. The complex antibody repertoires were analyzed by multivariate statistical techniques. Mass spectrometry was used to identify the most important antigens. RESULTS Very complex IgG antibody patterns against retinal antigens were found in all analyzed aqueous humor samples. Our multivariate approach could quantify differences in immunoreactivities, and including all peaks, the analysis of discriminance revealed a statistical significant difference between the patterns of the NTG and the CO group (p < 0.001). The antigen band at 21 kDa was identified as alpha B-crystallin, the 57-kDa antigen band as vimentin, and one at 70 kDa as heat shock protein 70. CONCLUSIONS We could demonstrate that complex IgG antibody patterns against retina exist in aqueous humor. The significant differences in the antibody pattern of the glaucoma group compared with the nonglaucoma group in aqueous humor confirm the results of previous studies using sera of glaucoma patients. These differences in antibody patterns might be further evidence for an autoimmune involvement in the pathogenesis of some glaucoma patients.
Collapse
|
35
|
Leung KW, Barnstable CJ, Tombran-Tink J. Bacterial endotoxin activates retinal pigment epithelial cells and induces their degeneration through IL-6 and IL-8 autocrine signaling. Mol Immunol 2009; 46:1374-86. [DOI: 10.1016/j.molimm.2008.12.001] [Citation(s) in RCA: 85] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2008] [Revised: 11/27/2008] [Accepted: 12/02/2008] [Indexed: 01/21/2023]
|
36
|
ter Veld F, Rose B, Mussmann R, Martin S, Herder C, Kempf K. Effects of somatostatin and octreotide on cytokine and chemokine production by lipopolysaccharide-activated peripheral blood mononuclear cells. J Endocrinol Invest 2009; 32:123-9. [PMID: 19411809 DOI: 10.1007/bf03345700] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
BACKGROUND Somatostatin plays an important role in the communication between the nervous, endocrine, and immune systems. Although somatostatin or its analogues have been shown to modulate a number of immune functions, their immunomodulatory effects are not uniform and are strongly dependent on the underlying cell system. AIM The aim of our study was to analyze the immunomodulatory effects of somatostatin and its analogue octreotide on peripheral blood mononuclear cells (PBMC) in vitro. MATERIALS/SUBJECTS:We used lipopolysaccharide-activated cells from normal glucose tolerant (NGT) subjects and from Type 2 diabetes mellitus (T2DM) patients as T2DM is associated with chronic, low-grade inflammation, and measured immune mediator release with multiplex bead-based assays. RESULTS Our data showed no statistically significant effects on the secretion of the cytokines interleukin (IL)-1beta, IL-6, IL-10, IL-12, interferon-gamma and tumor necrosis factor-alpha as well as the chemokines IL-8 and monocyte chemoattractant protein (MCP)-1, either on PBMC from T2DM patients or on those from NGT controls. However, a trend towards a dose-dependent biphasic effect was observed for IL- 6, IL-10 and MCP-1 with reduced immune mediator levels at low and increased/unaltered levels at higher somatostatin or octreotide concentrations. These observations could not be explained by interference with cell viability or proliferation. CONCLUSIONS We could not confirm immunomodulatory properties of somatostatin and octreotide on PBMC. Further analyses are necessary to explain the interaction between neuropeptides and the immune system.
Collapse
Affiliation(s)
- F ter Veld
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center at Heinrich Heine University Düsseldorf, Auf'm Hennekamp 65, 40225 Düsseldorf, Germany
| | | | | | | | | | | |
Collapse
|
37
|
Abstract
Elevated intraocular pressure does not explain glaucoma in all patients, but there is information that autoimmune mechanisms may be involved in this disorder. This review attempts to reveal the findings about specific changes in autoantibody profiles in glaucoma patients and their possible role in glaucoma. Considering that these changes in natural autoimmunity can be found consistently among different study populations, it might be a promising new tool for glaucoma detection.
Collapse
|
38
|
Missotten T, van Laar JAM, van der Loos TL, van Daele PLA, Kuijpers RWAM, Baarsma GS, van Hagen PM. Octreotide long-acting repeatable for the treatment of chronic macular edema in uveitis. Am J Ophthalmol 2007; 144:838-843. [PMID: 17916316 DOI: 10.1016/j.ajo.2007.07.039] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2006] [Revised: 07/29/2007] [Accepted: 07/30/2007] [Indexed: 11/19/2022]
Abstract
PURPOSE To report on the efficacy of the somatostatin analog octreotide long-acting repeatable (LAR), in the treatment of uveitic chronic macular edema (CME). DESIGN Case series, retrospective analysis. METHODS In 20 patients, 20 episodes of recurrent CME during otherwise quiescent uveitis were treated with intramuscular octreotide LAR injections. Patients were included if CME control with acetazolamide or systemic and periocular steroids had failed during previous CME episodes or if contraindications existed for persistent use of these therapies. Mean outcome points were CME and visual acuity changes. Correlation of prognostic factors with these outcomes was analyzed. RESULTS The included CME episodes occurred 7.6 +/- 1.4 years after onset of uveitis. Octreotide LAR treatment started 7.0 +/- 7.3 months after diagnosis of CME. CME decreased in 70% of episodes, after 2.7 +/- 1.3 months of treatment. After arrest of successful treatment, CME recurred instantly (27.2%) or within six months (36.4%). In 36.4% of successfully treated episodes, CME was absent for more than one year. A probable prognostic factor for success was the duration of CME before treatment. CONCLUSIONS Octreotide LAR had an edema-reducing effect in 70% of treated CME episodes. Successful response was related to duration of CME before start of treatment. The early recurrence of CME (63.6%) after arrest of octreotide LAR advocates a long-term treatment in recent episodes of macular edema in otherwise quiescent uveitis.
Collapse
Affiliation(s)
- Tom Missotten
- Department of Uveitis, Rotterdam Eye Hospital, Rotterdam, The Netherlands.
| | | | | | | | | | | | | |
Collapse
|
39
|
Decreased active TGF-beta2 levels in the aqueous humour during immune reactions following penetrating keratoplasty. Eye (Lond) 2007; 22:569-75. [PMID: 17975559 DOI: 10.1038/sj.eye.6703028] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
PURPOSE The secretion of transforming growth factor (TGF)-beta (2) into the aqueous humour is important for maintaining the immunological privilege of the anterior ocular segment that promotes corneal allograft survival. Levels of total TGF-beta (2), however, were similar in eyes with and without immune reactions following penetrating keratoplasty (PK). In this study, we tested the hypothesis whether the activated form of TGF-beta (2) is decreased in eyes with immune reactions following PK. METHODS Anterior chamber puncture was performed in 38 cataract patients without PK (group I), in 10 patients without immune reaction following PK (group II), and in 10 patients following PK with newly diagnosed endothelial immune reaction (group III). About 0.05-0.1 ml of aqueous humour were harvested from each patient. Analysis of active TGF-beta (2) was started via ELISA within 3 h following puncture. RESULTS Average active TGF-beta (2) concentration was 38.9 pg/ml+/-24.3 in group I, 47.4 pg/ml+/-23.1 in group II, and 12.7+/-26.0 pg/ml in group III. The differences between groups I and III (P=0.005) and groups II and III (P=0.008) were statistically significant. CONCLUSION The lowest levels of active TGF-beta (2) were measured in eyes with newly diagnosed endothelial immune reactions. This suggests that TGF-beta (2) has a protective effect on corneal grafts following PK. Further (prospective) studies are being carried out to discover whether active TGF-beta (2)--determined prior to PK--might serve as a predictive parameter for the occurrence of immune reactions.
Collapse
|
40
|
Grus FH, Joachim SC, Pfeiffer N. Proteomics in ocular fluids. Proteomics Clin Appl 2007; 1:876-88. [DOI: 10.1002/prca.200700105] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2007] [Indexed: 12/14/2022]
|
41
|
Peptidergic nerves in the eye, their source and potential pathophysiological relevance. ACTA ACUST UNITED AC 2006; 53:39-62. [PMID: 16872680 DOI: 10.1016/j.brainresrev.2006.06.002] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2005] [Revised: 06/09/2006] [Accepted: 06/13/2006] [Indexed: 12/17/2022]
Abstract
Over the last five decades, several neuropeptides have been discovered which subsequently have been found to be highly conserved during evolution, to be widely distributed both in the central and peripheral nervous system and which act as neurotransmitters and/or neuromodulators. In the eye, the first peptide to be explored was substance P which was reported to be present in the retina but also in peripherally innervated tissues of the eye. Substance P is certainly the best characterized peptide which has been found in sensory neurons innervating the eye. Functionally, it has been shown to act trophically on corneal wound healing and to participate in the irritative response in lower mammals, a model for neurogenic inflammation, where it mediates the noncholinergic nonadrenergic contraction of the sphincter muscle. Over the last three decades, the interest has extended to investigate the presence and distribution of other neuropeptides including calcitonin gene-related peptide, vasoactive intestinal polypeptide, neuropeptide Y, pituitary adenylate cyclase-activating polypeptides, cholecystokinin, somatostatin, neuronal nitric oxide, galanin, neurokinin A or secretoneurin and important functional results have been obtained for these peptides. This review focuses on summarizing the current knowledge about neuropeptides in the eye excluding the retina and retinal pigment epithelium and to elucidate their potential functional significance.
Collapse
|
42
|
Ghosh S, Choritz L, Geibel J, Coca-Prados M. Somatostatin modulates PI3K-Akt, eNOS and NHE activity in the ciliary epithelium. Mol Cell Endocrinol 2006; 253:63-75. [PMID: 16764985 DOI: 10.1016/j.mce.2006.05.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2006] [Revised: 04/10/2006] [Accepted: 05/02/2006] [Indexed: 12/20/2022]
Abstract
Somatostatin (SST) is a biologically active peptide produced in neuroendocrine cells. In the present study, we provide evidence of pro-SST and SST receptor (SSTR1 and 2A) mRNA expression in ocular ciliary epithelium (CE). SST or SST-like immunoreactivity was detected by radioimmunoassay in tissue extract from ciliary processes and in aqueous humor. The distinct immunolabeling of CE with SST and proprotein convertases PC1 and PC2 antibodies suggested a tissue and cell-specific processing of pro-SST. SST (10(-8) to 10(-4)M) added exogenously to the CE, elicited the following effects: (i) a dose-dependent attenuation of Na+/H+-exchanger (NHE) activity; (ii) up to a two-fold increase phosphorylation of p-Akt-Ser473 and of p-eNOS-Ser617, and (iii) lack of response on intracellular cyclic GMP production. LY294002, a PI3K-inhibitor, blocked SST-induced p-Akt-Ser473 and partially p-eNOS-Ser617, however, it did not reverse SST-induced NHE attenuation. Collectively, these results suggested involvement of SST in multiple intracellular signaling pathways in the CE.
Collapse
Affiliation(s)
- Sikha Ghosh
- Department of Ophthalmology and Visual Science, Yale University School of Medicine, New Haven, CT 06510, USA
| | | | | | | |
Collapse
|
43
|
Abstract
Immune-mediated inflammation and allograft rejection are greatly reduced in certain organs, a phenomenon called 'immune privilege'. Immune privilege is well developed in three regions of the body: the eye, the brain and the pregnant uterus. Immune-mediated inflammation has devastating consequences in the eye and brain, which have limited capacity for regeneration. Likewise, loss of immune privilege at the maternal-fetal interface culminates in abortion in rodents. However, all three regions share many adaptations that restrict the induction and expression of immune-mediated inflammation. A growing body of evidence from rodent studies suggests that a breakdown in immune privilege contributes to multiple sclerosis, uveitis, corneal allograft rejection and possibly even immune abortion.
Collapse
Affiliation(s)
- Jerry Y Niederkorn
- Department of Ophthalmology, University of Texas Southwestern Medical Center, Dallas, Texas 75390-9057, USA.
| |
Collapse
|
44
|
Kezuka T, Takeuchi M, Keino H, Usui Y, Takeuchi A, Yamakawa N, Usui M. Peritoneal Exudate Cells Treated with Calcitonin Gene-Related Peptide Suppress Murine Experimental Autoimmune Uveoretinitis via IL-10. THE JOURNAL OF IMMUNOLOGY 2004; 173:1454-62. [PMID: 15240742 DOI: 10.4049/jimmunol.173.2.1454] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Immunization with retinal Ag induces experimental autoimmune uveoretinitis (EAU) in mice. We investigated the suppression of murine EAU by peritoneal exudate cells (PEC) cultured with calcitonin gene-related peptide (CGRP). PEC derived from mice were treated with CGRP and residues 1-20 of human interphotoreceptor retinoid-binding protein (hIRBP 1-20). The hIRBP 1-20-immunized mice were injected i.v. with PEC treated with CGRP and hIRBP 1-20. After immunization, Ag-specific delayed hypersensitivity (DH) was measured and EAU was assessed histopathologically. Both EAU- and Ag-specific DH were suppressed by injection of PEC treated with CGRP (100 ng/ml) and hIRBP 1-20. However, hIRBP 1-20-mediated EAU was not suppressed by injection of PEC treated with CGRP and BSA. Both EAU- and Ag-specific DH were not suppressed by injection of PEC treated with CGRP and hIRBP 1-20 into splenectomized mice. In mice adoptively transferred spleen cells from hIRBP 1-20-immunized mice, EAU was also suppressed by injection of CGRP-treated PEC. EAU was markedly inhibited in hIRBP 1-20-immunized mice adoptively transferred T cells obtained from mice injected with hIRBP 1-20-pulsed, CGRP-treated PEC. Furthermore, EAU- and Ag-specific DH were not suppressed by injection of PEC treated with CGRP and hIRBP 1-20 when the recipient mice were given anti-IL-10 Ab i.p., or when the PEC were derived from IL-10 knockout mice. The present results indicate that PEC treated with CGRP suppress murine EAU in an Ag-specific manner, even in the efferent phase, and IL-10 secreted from PEC might play an important role in the CGRP-mediated suppression of murine EAU.
Collapse
Affiliation(s)
- Takeshi Kezuka
- Department of Ophthalmology, Tokyo Medical University, Tokyo, Japan.
| | | | | | | | | | | | | |
Collapse
|
45
|
Abstract
Regulation of immunity within the immune-privileged ocular microenvironment is a dynamic interaction of anatomical features, factors, and cells that work toward suppressing the induction inflammation. Immunosuppressive neuropeptides found in aqueous humor are central to this immunoregulation. These neuropeptides are alpha-melanocyte-stimulating hormone, vasoactive intestinal peptide, calcitonin gene-related peptide, and somatostatin. Along with transforming growth factor-beta2, the neuropeptides target specific cells and pathways in innate and adaptive immunity. These aqueous humor factors prevent pathogen-induced inflammation and activation of Th1 cells, while promoting induction of regulatory T cells. Therefore, the ocular microenvironment, through the constitutive production of immunosuppressive factors found in aqueous humor, maintains immune privilege by manipulating regional innate and adaptive immunity away from inflammatory responses.
Collapse
Affiliation(s)
- Andrew Taylor
- The Schepens Eye Research Institute and the Department of Ophthalmology, Harvard Medical School, Boston, MA 02114, USA
| |
Collapse
|
46
|
Streilein JW. Ocular immune privilege: therapeutic opportunities from an experiment of nature. Nat Rev Immunol 2004; 3:879-89. [PMID: 14668804 DOI: 10.1038/nri1224] [Citation(s) in RCA: 539] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Affiliation(s)
- J Wayne Streilein
- Schepens Eye Research Institute, Harvard Medical School, Boston, Massachusetts 02114, USA.
| |
Collapse
|