Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
1.
medRxiv ; 2024 Apr 27.
Article in English | MEDLINE | ID: mdl-38712112

ABSTRACT

Background: Variability in treatment response may be attributable to organ-level heterogeneity in tumor lesions. Radiomic analysis of medical images can elucidate non-invasive biomarkers of clinical outcome. Organ-specific radiomic comparison across immunotherapies and targeted therapies has not been previously reported. Methods: We queried UPMC Hillman Cancer Center registry for patients with metastatic melanoma (MEL) treated with immune checkpoint inhibitors (ICI) (anti-PD1/CTLA4 [ipilimumab+nivolumab; I+N] or anti-PD1 monotherapy) or BRAF targeted therapy. Best overall response was measured using RECIST v1.1. Lesions were segmented into discrete volume-of-interest with 400 radiomics features extracted. Overall and organ-specific machine-learning models were constructed to predict disease control (DC) versus progressive disease (PD) using XGBoost. Results: 291 MEL patients were identified, including 242 ICI (91 I+N, 151 PD1) and 49 BRAF. 667 metastases were analyzed, including 541 ICI (236 I+N, 305 PD1) and 126 BRAF. Across cohorts, baseline demographics included 39-47% female, 24-29% M1C, 24-46% M1D, and 61-80% with elevated LDH. Among patients experiencing DC, the organs with the greatest reduction were liver (-88%±12%, I+N; mean±S.E.M.) and lung (-72%±8%, I+N). For patients with multiple same-organ target lesions, the highest inter-lesion heterogeneity was observed in brain among patients who received ICI while no intra-organ heterogeneity was observed in BRAF. 267 patients were kept for radiomic modeling, including 221 ICI (86 I+N, 135 PD1) and 46 BRAF. Models consisting of optimized radiomic signatures classified DC/PD across I+N (AUC=0.85) and PD1 (0.71) and within individual organ sites (AUC=0.72∼0.94). Integration of clinical variables improved the models' performance. Comparison of models between treatments and across organ sites suggested mostly non-overlapping DC or PD features. Skewness, kurtosis, and informational measure of correlation (IMC) were among the radiomic features shared between overall response models. Kurtosis and IMC were also utilized by multiple organ-site models. Conclusions: Differential organ-specific response was observed across BRAF and ICI with within organ heterogeneity observed for ICI but not for BRAF. Radiomic features of organ-specific response demonstrated little overlap. Integrating clinical factors with radiomics improves the prediction of disease course outcome and prediction of tumor heterogeneity.

2.
Am Soc Clin Oncol Educ Book ; 44(3): e433694, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38781565

ABSTRACT

The management of brain metastases, a potentially devastating complication of advanced cancers, has become increasingly complex with advancements in local and systemic therapies. Improved outcomes and extended survival for patients with metastatic solid tumors have led to a surge in the prevalence and possibly incidence of brain metastases, affecting up to 40% of individuals with solid tumors. Enhanced imaging technologies contribute to more accurate and early detection, shaping the understanding of the intricate landscape of this condition. Traditionally, surgery and radiation stood as the mainstays of treatment because of the limited efficacy of systemic therapies within the brain. However, emerging clinical data, particularly in melanoma, lung, and breast cancers, reveal promising results with novel systemic treatments such as immunotherapy and targeted therapies. Despite the historical exclusion of patients with active brain metastases from clinical trials, a shift is occurring toward a more inclusive approach. This chapter delves into the multifaceted challenges associated with managing brain metastases, with a focus on the evolving landscape of systemic approaches as well as the intricacies of shared decision making, providing a comprehensive overview of the current state and future directions in navigating the complexities of brain metastases management.


Subject(s)
Brain Neoplasms , Disease Management , Humans , Brain Neoplasms/secondary , Brain Neoplasms/therapy , Combined Modality Therapy
3.
Cancer J ; 30(2): 102-107, 2024.
Article in English | MEDLINE | ID: mdl-38527263

ABSTRACT

ABSTRACT: Development of brain metastasis is one of the most serious complications of advanced melanoma, carrying a significant burden of morbidity and mortality. Although advances in local treatment modalities such as stereotactic radiosurgery and breakthrough systemic therapies including immunotherapy and targeted therapies have improved the outcomes of patients with metastatic melanoma, management of patients with melanoma brain metastases (MBMs) remains challenging. Notably, patients with MBMs have historically been excluded from clinical trials, limiting insights into their specific treatment responses. Encouragingly, a growing body of evidence shows the potential of systemic therapies to yield durable intracranial responses in these patients, highlighting the need for inclusion of patients with MBMs in future clinical trials. This is pivotal for expediting the advancement of novel therapies tailored to this distinct patient population. In this review, we will highlight the evolving landscape of MBM management, focusing on local and systemic treatment strategies.


Subject(s)
Brain Neoplasms , Melanoma , Radiosurgery , Humans , Melanoma/drug therapy , Brain Neoplasms/therapy , Combined Modality Therapy , Immunotherapy
4.
Transplantation ; 107(6): 1302-1310, 2023 06 01.
Article in English | MEDLINE | ID: mdl-36584368

ABSTRACT

BACKGROUND: Corneal transplantation outcomes are generally less favorable in young children compared with adults. The purpose of this study was to determine the immunological mechanisms underlying this difference. METHODS: A murine model of allogeneic corneal transplantation was used in the study, and graft survival was determined by evaluating opacity scores for 8 wk. Syngeneic transplantation in the very young host served as a surgical control. The frequencies of total and activated natural killer (NK) cells in cornea posttransplantation were kinetically evaluated using flow cytometry. The regulatory T cell (Treg) frequency and function in naive animals were assessed by flow cytometry and in vitro suppression assays, respectively. Finally, graft survival and immune responses were determined in NK cell-depleted, or adult naive Treg-transferred, young hosts. RESULTS: Corneal allograft survival in the very young recipients was significantly lower than in adult hosts. The frequencies of total NK cells and their interferon gamma-expressing subset in the cornea were significantly higher in the very young mice posttransplantation. In ungrafted mice, frequencies of Treg in draining lymph nodes as well as their capabilities to suppress NK-cell secretion of interferon gamma were lower in the very young compared with adults. In NK cell-depleted or adult Treg--transferred very young recipients, the allograft survival was significantly improved along with the suppressed NK-cell response. CONCLUSIONS: Our data demonstrate that amplified activity of NK cells, together with lower suppressive function of Treg, contributes to early rejection of corneal allografts in very young graft recipients.


Subject(s)
Corneal Transplantation , T-Lymphocytes, Regulatory , Mice , Animals , Interferon-gamma , Cornea , Killer Cells, Natural , Graft Rejection , Mice, Inbred C57BL , Mice, Inbred BALB C
5.
Curr Oncol Rep ; 23(10): 116, 2021 08 03.
Article in English | MEDLINE | ID: mdl-34342752

ABSTRACT

PURPOSE OF REVIEW: For patients with metastatic melanoma, immune checkpoint inhibition has drastically changed outcomes. Here, we review the current and next generations of immune-based anti-cancer therapeutics for patients with metastatic melanoma. RECENT FINDINGS: The need for new anti-cancer therapeutics in patients with metastatic melanoma who have progression of disease despite immune checkpoint blockade is evident. Several novel agents are expected to have FDA approval within the next few years, as they have yielded impressive responses. Despite these optimistic agents, the field of immuno-oncology continues to expand and produce agents with novel mechanisms of action. The next generation of immunotherapy is based upon years of thoroughly researched immuno-oncology. Many of these agents are currently being evaluated in early phase clinical trials, and much of the preliminary data looks promising.


Subject(s)
Immunotherapy/trends , Melanoma/therapy , Cell- and Tissue-Based Therapy , Cytokines/immunology , Cytokines/therapeutic use , Humans , Immune Checkpoint Inhibitors/therapeutic use , Immune Checkpoint Proteins/immunology , Immunity, Innate , Melanoma/immunology , Melanoma/pathology , T-Lymphocytes/immunology , T-Lymphocytes/transplantation , Toll-Like Receptors/agonists , Toll-Like Receptors/immunology
6.
Cancers (Basel) ; 13(11)2021 May 30.
Article in English | MEDLINE | ID: mdl-34070756

ABSTRACT

The interrogation of intrinsic and adaptive resistance to cancer immunotherapy has identified lack of antigen presentation and type I interferon signaling as biomarkers of non-T-cell-inflamed tumors and clinical progression. A myriad of pre-clinical studies have implicated the cGAS/stimulator of interferon genes (STING) pathway, a cytosolic DNA-sensing pathway that drives activation of type I interferons and other inflammatory cytokines, in the host immune response against tumors. The STING pathway is also increasingly understood to have other anti-tumor functions such as modulation of the vasculature and augmentation of adaptive immunity via the support of tertiary lymphoid structure development. Many natural and synthetic STING agonists have entered clinical development with the first generation of intra-tumor delivered cyclic dinucleotides demonstrating safety but only modest systemic activity. The development of more potent and selective STING agonists as well as novel delivery systems that would allow for sustained inflammation in the tumor microenvironment could potentially augment response rates to current immunotherapy approaches and overcome acquired resistance. In this review, we will focus on the latest developments in STING-targeted therapies and provide an update on the clinical development and application of STING agonists administered alone, or in combination with immune checkpoint blockade or other approaches.

7.
Mucosal Immunol ; 14(1): 177-186, 2021 01.
Article in English | MEDLINE | ID: mdl-32327706

ABSTRACT

Long-lived memory T-helper 17 (Th17) cells actively mediate the chronic inflammation in autoimmune disorders, including dry eye disease (DED). The mechanisms responsible for the maintenance and reactivation of these cells in autoimmunity have been subject of investigation. However, the process through which memory Th17 are generated from their effector precursors remains to be elucidated. Herein, using our murine model of DED, we detect a linear transition from effector-to-memory Th17 cells during the abatement phase of acute inflammation, which is accompanied by persistently high levels of IL-23 and diminished levels of IL-2. In addition, in vitro culture of effector Th17 cells derived from the DED animals with IL-23, but not IL-2, leads to significant generation of memory Th17 cells, along with upregulated expression levels of IL-7R and IL-15R by these cells. Furthermore, supplementation of IL-2 abolishes and blockade of IL-2 enhances IL-23-induced generation of memory Th17 cells in vitro. Finally, in vivo blockade of IL-23 signaling during the contraction phase of primary response inhibits the generation of memory Th17 cells from their effector precursors. Together, our data demonstrate a new dichotomy between IL-23 and IL-2 in driving effector Th17 cells into the memory pool in autoimmune-mediated ocular surface inflammation.


Subject(s)
Autoimmunity , Dry Eye Syndromes/etiology , Dry Eye Syndromes/metabolism , Immunologic Memory , Interleukin-23/metabolism , Interleukin-2/metabolism , Th17 Cells/immunology , Th17 Cells/metabolism , Animals , Apoptosis/genetics , Apoptosis/immunology , Autoimmune Diseases/etiology , Autoimmune Diseases/metabolism , Autoimmune Diseases/pathology , Biomarkers , Disease Models, Animal , Disease Susceptibility , Dry Eye Syndromes/pathology , Humans , Interleukin-2/genetics , Interleukin-23/genetics , Mice , Signal Transduction
8.
Exp Eye Res ; 198: 108152, 2020 09.
Article in English | MEDLINE | ID: mdl-32721424

ABSTRACT

Over the past century, corneal transplantation has become the most commonly performed allogeneic solid tissue transplantation. Although more than 80% of the corneal transplantations have favorable outcomes, immune-mediated rejection continues to be the major cause of failure in well over 50% of graft recipients that have inflamed and vascularized host beds. Over the past two decades, the progress in our understanding of the immunological pathways that mediate graft rejection has aided in the development of novel therapeutic strategies. In order to successfully test the efficacy of these interventions, it is essential to model the immunological processes occurring as a consequence of corneal transplantation. Herein, we have comprehensively reviewed the established animal models used for replicating the immunopathological processes causing graft rejection in high-risk corneal transplantation settings. We have also discussed the practical and technical differences, as well as biological and immunological variations in different animal models.


Subject(s)
Corneal Diseases/surgery , Corneal Transplantation/methods , Graft Rejection/prevention & control , Animals , Disease Models, Animal
9.
Am J Pathol ; 190(9): 1859-1866, 2020 09.
Article in English | MEDLINE | ID: mdl-32473919

ABSTRACT

Substance P (SP) is a tachykinin neuropeptide, implicated in the pathogenesis of various inflammatory conditions and a critical mediator in pain transmission. Recently, the role of SP was described in the pathogenesis of dry eye disease (DED) through its role in the maturation of antigen-presenting cells at the ocular surface after exposure to desiccating stress. However, the effect of SP on regulatory T cells (Tregs), which are functionally impaired in DED, remains unclear. This study examined the phenotypic and functional changes in Tregs in response to SP in DED. The in vitro cultures of normal Tregs in the presence of SP led to a significant reduction in both Treg frequencies and their suppressive function, which was prevented by the addition of an SP receptor (neurokinin-1 receptor) antagonist. Furthermore, in vivo treatment with the neurokinin-1 receptor antagonist in DED mice effectively restored Treg function, suppressed pathogenic T helper 17 response, and significantly ameliorated the disease. Our results show that a significant increase in SP levels promotes Treg dysfunction in DED, and blockade of SP effectively restores Treg function and suppresses DED severity.


Subject(s)
Dry Eye Syndromes/immunology , Dry Eye Syndromes/metabolism , Receptors, Neurokinin-1/metabolism , T-Lymphocytes, Regulatory/immunology , Animals , Female , Mice , Mice, Inbred C57BL , Neurokinin-1 Receptor Antagonists/pharmacology , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/metabolism
10.
Am J Transplant ; 20(2): 389-398, 2020 02.
Article in English | MEDLINE | ID: mdl-31587452

ABSTRACT

The functional competence of corneal endothelial cells (CEnCs) is critical for survival of corneal allografts, but these cells are often targets of the immune response mediated by graft-attacking effector T cells. Although regulatory T cells (Tregs) have been studied for their role in regulating the host's alloimmune response towards the graft, the cytoprotective function of these cells on CEnCs has not been investigated. The aim of this study was to determine whether Tregs suppress effector T cell-mediated and inflammatory cytokine-induced CEnC death, and to elucidate the mechanism by which this cytoprotection occurs. Using 2 well-established models of corneal transplantation (low-risk and high-risk models), we show that Tregs derived from low-risk graft recipients have a superior capacity in protecting CEnCs against effector T cell-mediated and interferon-γ and tumor necrosis factor-α-induced cell death compared to Tregs derived from high-risk hosts. We further demonstrate that the cytoprotective function of Tregs derived from low-risk hosts occurs independently of direct cell-cell contact and is mediated by the immunoregulatory cytokine IL-10. Our study is the first to report that Tregs provide cytoprotection for CEnCs through secretion of IL-10, indicating potentially novel therapeutic targets for enhancing CEnC survival following corneal transplantation.


Subject(s)
Cornea/immunology , Corneal Transplantation , Endothelial Cells/immunology , Graft Survival/immunology , Interleukin-10/metabolism , T-Lymphocytes, Regulatory/immunology , Animals , Biomarkers/metabolism , Cell Survival/immunology , Cornea/cytology , Immunohistochemistry , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Real-Time Polymerase Chain Reaction , Transplantation, Homologous
11.
Am J Pathol ; 190(1): 125-133, 2020 01.
Article in English | MEDLINE | ID: mdl-31669306

ABSTRACT

Neuroinflammation plays an important role in the pathogenesis of ocular surface disease, including dry eye disease (DED), but little is known about the contribution of substance P (SP) to DED. In this study, we investigated the expression of SP at the ocular surface and evaluated its effect on maturation of antigen-presenting cells (APCs), the key cell component involved in the induction of type 17 helper T-cell (Th17) response in DED. The effect of topical blockade of SP signaling was further investigated using neurokinin-1 receptor (NK1R) inhibitors on APC maturation, Th17 cell activation, and disease severity in a mouse model of DED. The results demonstrate that SP is constitutively expressed at the ocular surface, and trigeminal ganglion neurons are the major source of SP in DED. SP derived from trigeminal ganglion enhanced the expression of major histocompatibility complex class II maturation marker by bone marrow-derived dendritic cells, an effect that is abrogated by blockade of SP signaling using NK1R antagonist spantide. Finally, using a well-established murine model of DED, topical treatment of DED mice with NK1R antagonists CP-99,994 and L-733,060 suppressed APC acquisition of major histocompatibility complex class II, reduced Th17 cell activity, and ameliorated DED severity. These findings are of translational value, as they suggest that antagonizing NK1R-mediated SP signaling may be an effective strategy in suppressing Th17-mediated ocular surface disease.


Subject(s)
Antigen-Presenting Cells/immunology , Disease Models, Animal , Dry Eye Syndromes/prevention & control , Neurokinin-1 Receptor Antagonists/pharmacology , Receptors, Neurokinin-1/chemistry , Th17 Cells/immunology , Animals , Antigen-Presenting Cells/drug effects , Cornea/drug effects , Cornea/immunology , Dry Eye Syndromes/immunology , Dry Eye Syndromes/metabolism , Dry Eye Syndromes/pathology , Female , Lymph Nodes/drug effects , Lymph Nodes/immunology , Mice , Mice, Inbred C57BL , Th17 Cells/drug effects
12.
Ocul Surf ; 17(2): 198-207, 2019 04.
Article in English | MEDLINE | ID: mdl-30695735

ABSTRACT

The therapeutic potential of mesenchymal stem cells (MSCs) has been heralded by their multipotentiality and immunomodulatory capacity. MSCs migrate toward sites of tissue damage, where specific pro-inflammatory factors 'license' their immunosuppressive functions. Recent studies in animal models of ocular surface disease have demonstrated the potential of MSC-derived therapies to limit inflammation and promote tissue repair. Herein, we review the immunoregulatory mechanisms of MSCs, as well as strategies to harness their regenerative function at the cornea. We examine reports of the therapeutic application of MSCs in the setting of ocular surface inflammation; including corneal injury, transplantation, ocular surface autoimmunity and allergy.


Subject(s)
Autoimmunity , Corneal Injuries/surgery , Mesenchymal Stem Cell Transplantation/methods , Mesenchymal Stem Cells/immunology , Animals , Corneal Injuries/immunology , Humans
13.
Transfus Med Rev ; 33(1): 43-50, 2019 01.
Article in English | MEDLINE | ID: mdl-30528986

ABSTRACT

Although peripheral blood stem cells (PBSC) have worldwide become the predominant source of progenitor cells for hematopoietic stem cell transplantation (HSCT), debate about their role compared with bone marrow (BM) has recently intensified, in large part based on the results of a multicenter Clinical Trials Network study which showed lower incidence of chronic graft-versus-host disease (cGVHD) and improved quality of life in recipients of myeloablative HLA-matched unrelated BM compared with PBSC transplants. However, in certain patient populations, PBSC may lead to improved clinical outcomes due to faster hematologic recovery, a lower risk of graft failure, and possibly a lower probability of relapse. This review will provide a comprehensive summary of studies comparing PBSC with BM as the graft source in terms of acute and chronic GVHD incidence, time to engraftment, and disease-free and overall survival probabilities after HLA-matched related and unrelated donor transplantation and haploidentical donor transplantation. Recommendations based on these studies regarding the use of PBSC versus BM for HSCT are offered.


Subject(s)
Bone Marrow Cells/cytology , Hematopoietic Stem Cell Transplantation/adverse effects , Hematopoietic Stem Cell Transplantation/methods , Hematopoietic Stem Cells/cytology , Blood Donors , Bone Marrow Transplantation/methods , Disease-Free Survival , Graft vs Host Disease/etiology , HLA Antigens/chemistry , Humans , Multicenter Studies as Topic , Quality of Life , Randomized Controlled Trials as Topic , Recurrence , Stem Cells/cytology , T-Lymphocytes/cytology , Transplantation, Homologous/adverse effects , Unrelated Donors
14.
Transplantation ; 103(1): 182-190, 2019 01.
Article in English | MEDLINE | ID: mdl-30247445

ABSTRACT

BACKGROUND: Regulatory T (Treg) cell-based immunotherapies have been studied as potential cell-based modalities for promoting transplant survival. However, the efficacy of local delivery of Treg cells in corneal transplantation has not been fully elucidated. Herein, we investigated the kinetics of migration of subconjunctivally injected Treg cells and their role in promoting corneal allograft survival. METHODS: GFPCD4CD25Foxp3 Treg cells were isolated from draining lymph nodes (DLNs) of GFP transgenic mice and were subconjunctivally injected to corneal allograft recipients. Next, Treg cells, conventional T cells (Tconv) or a combination of both was locally injected to graft recipients, and graft survival was determined by evaluating opacity scores for 10 weeks. Transplanted mice without treatment served as controls. The frequencies of major histocompatibility complex-IICD11b antigen-presenting cells, IFNγCD4 Th1 cells, and CD45 cells in the DLNs and cornea were evaluated at week 2 posttransplantation using flow cytometry. Expressions of IFNγ, IL-10 and TGF-ß in the grafts were assessed using reverse transcription polymerase chain reaction and enzyme-linked immunosorbent assay. RESULTS: GFP Treg cells were detected in the ipsilateral cornea and DLNs of recipients 6 hours after injection. Subconjunctival injection of Treg cells significantly decreased the frequencies of mature antigen-presenting cells in the graft and DLNs, suppressed Th1 frequencies in DLNs, and inhibited CD45 cell infiltration to the graft. Finally, locally delivered Treg cells significantly reduced the expression of IFN-γ, enhanced the levels of IL-10 and TGF-ß in the graft, and promoted long-term allograft survival. CONCLUSIONS: Our study elucidates the kinetics of migration of locally delivered Treg cells and shows their role in suppressing host immune response against the allograft.


Subject(s)
Adoptive Transfer/methods , Cornea/surgery , Corneal Transplantation , Graft Survival , Green Fluorescent Proteins/metabolism , T-Lymphocytes, Regulatory/transplantation , Allografts , Animals , Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/metabolism , Chemotaxis, Leukocyte , Cornea/immunology , Cornea/pathology , Cytokines/immunology , Cytokines/metabolism , Green Fluorescent Proteins/genetics , Kinetics , Lymph Nodes/immunology , Lymph Nodes/metabolism , Lymphocyte Activation , Male , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Th1 Cells/immunology , Th1 Cells/metabolism
15.
Am J Transplant ; 19(3): 662-673, 2019 03.
Article in English | MEDLINE | ID: mdl-30129280

ABSTRACT

Beyond their historical role as the effector cells in allergic disorders, mast cells have been implicated in regulating both innate and adaptive immune responses. Possessing considerable functional plasticity, mast cells are abundant at mucosal surfaces, where the host and external environments interface. The purpose of this study was to evaluate the contribution of mast cells to allograft rejection at the ocular surface. Using a well-characterized murine model of corneal transplantation, we report that mast cells promote allosensitization. Our data show mast cell frequencies and activation are increased following transplantation. We demonstrate that mast cell inhibition (a) limits the infiltration of inflammatory cells and APC maturation at the graft site; (b) reduces allosensitization and the generation of Th1 cells in draining lymphoid tissues; (c) decreases graft infiltration of alloimmune-inflammatory cells; and (d) prolongs allograft survival. Our data demonstrate a novel function of mast cells in promoting allosensitization at the ocular surface.


Subject(s)
Antigen-Presenting Cells/immunology , Corneal Transplantation/methods , Eye/immunology , Graft Rejection/prevention & control , Mast Cells/immunology , Mucous Membrane/immunology , Th1 Cells/immunology , Animals , Graft Rejection/immunology , Graft Survival/immunology , Immunization , Lymphoid Tissue/immunology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Transplantation, Homologous
16.
Am J Pathol ; 188(9): 2016-2024, 2018 09.
Article in English | MEDLINE | ID: mdl-30097165

ABSTRACT

Corneal transplantation is the most prevalent form of tissue transplantation. The success of corneal transplantation mainly relies on the integrity of corneal endothelial cells (CEnCs), which maintain graft transparency. CEnC density decreases significantly after corneal transplantation even in the absence of graft rejection. To date, different strategies have been used to enhance CEnC survival. The neuropeptide vasoactive intestinal peptide (VIP) improves CEnC integrity during donor cornea tissue storage and protects CEnCs against oxidative stress-induced apoptosis. However, little is known about the effect of exogenous administration of VIP on corneal transplant outcomes. We found that VIP significantly accelerates endothelial wound closure and suppresses interferon-γ- and tumor necrosis factor-α-induced CEnC apoptosis in vitro in a dose-dependent manner. In addition, we found that intracameral administration of VIP to mice undergoing syngeneic corneal transplantation with endothelial injury increases CEnC density and decreases graft opacity scores. Finally, using a mouse model of allogeneic corneal transplantation, we found for the first time that treatment with VIP significantly suppresses posttransplantation CEnC loss and improves corneal allograft survival.


Subject(s)
Corneal Transplantation , Endothelium, Corneal/drug effects , Gastrointestinal Agents/pharmacology , Graft Rejection/prevention & control , Graft Survival/drug effects , Vasoactive Intestinal Peptide/pharmacology , Wound Healing/drug effects , Allografts , Animals , Cells, Cultured , Endothelium, Corneal/injuries , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL
17.
Ocul Surf ; 16(4): 470-477, 2018 10.
Article in English | MEDLINE | ID: mdl-30055331

ABSTRACT

PURPOSE: In this study, we examine the expression of corneal epithelium-derived thrombospondin-1 (TSP-1) and its immunomodulatory functions in a validated murine model of dry eye disease (DED). METHODS: DED was induced in female C57BL/6 using a controlled environment chamber (CEC) for 14 days. mRNA and protein expression of TSP-1 by corneal epithelial cells was quantified using real-time PCR and flow cytometry. Corneal epithelial cells from either naïve or DED mice were cultured with bone marrow derived dendritic cells (BMDCs) in the presence of IFNγ for 48 h, and BMDC expression of MHC-II and CD86 was determined using flow cytometry. Next, either recombinant TSP-1 or anti-TSP-1 antibody was added to the co-culture, and BMDC expression of above activation markers was evaluated. Finally, either DED mice were topically treated with either recombinant TSP-1 or human serum albumin (HSA), and maturation of corneal DCs, expression of inflammatory cytokines, and DED severity were investigated. RESULTS: mRNA expression of TSP-1 by the corneal epithelium was upregulated in DED. Corneal epithelial cells derived from mice with DED demonstrated an enhanced capacity in suppressing BMDC expression of MHC-II and CD86 relative to wild type mice, and this effect was abrogated by TSP-1 blockade and potentiated by recombinant TSP-1. Finally, topical application of recombinant TSP-1 significantly suppressed corneal DC maturation and mRNA expression of pro-inflammatory cytokines, and ameliorated disease severity in mice with DED. CONCLUSIONS: Our study elucidates the function of epithelium-derived TSP-1 in inhibiting DC maturation and shows its translational potential to limit corneal epitheliopathy in DED.


Subject(s)
Dry Eye Syndromes/immunology , Epithelium, Corneal/immunology , Thrombospondin 1/physiology , Animals , Cornea/metabolism , Dendritic Cells/immunology , Dry Eye Syndromes/metabolism , Epithelial Cells/metabolism , Epithelium, Corneal/metabolism , Female , Humans , Mice , Mice, Inbred C57BL , Thrombospondin 1/metabolism
18.
Invest Ophthalmol Vis Sci ; 59(3): 1191-1198, 2018 03 01.
Article in English | MEDLINE | ID: mdl-29625439

ABSTRACT

Purpose: Neutrophil-secreted effector molecules are one of the primary causes of tissue damage during corneal inflammation. In the present study, we have investigated the effect of stromal cells in regulating neutrophil expression of tissue-damaging enzymes, myeloperoxidase (MPO), and N-elastase (ELANE). Methods: Bone marrow-purified nonhematopoietic mesenchymal stromal cells and formyl-methionyl-leucyl-phenylalanine-activated neutrophils were cocultured in the presence or absence of Transwell inserts for 1 hour. Neutrophil effector molecules, MPO and ELANE, were quantified using ELISA. In mice, corneal injury was created by mechanical removal of the corneal epithelium and anterior stroma approximating one third of total corneal thickness, and mesenchymal stromal cells were then intravenously injected 1 hour post injury. Corneas were harvested to evaluate MPO expression and infiltration of CD11b+Ly6G+ neutrophils. Results: Activated neutrophils cocultured with mesenchymal stromal cells showed a significant 2-fold decrease in secretion of MPO and ELANE compared to neutrophils activated alone (P < 0.05). This suppressive effect was cell-cell contact dependent, as stromal cells cocultured with neutrophils in the presence of Transwell failed to suppress the secretion of neutrophil effector molecules. Following corneal injury, stromal cell-treated mice showed a significant 40% decrease in MPO expression by neutrophils and lower neutrophil frequencies compared to untreated injured controls (P < 0.05). Reduced MPO expression by neutrophils was also accompanied by normalization of corneal tissue structure following stromal cell treatment. Conclusions: Mesenchymal stromal cells inhibit neutrophil effector functions via direct cell-cell contact interaction during inflammation. The current findings could have implications for the treatment of inflammatory ocular disorders caused by excessive neutrophil activation.


Subject(s)
Corneal Injuries/enzymology , Leukocyte Elastase/metabolism , Mesenchymal Stem Cells/metabolism , Neutrophils/enzymology , Peroxidase/metabolism , Animals , CD11b Antigen/metabolism , Cell Communication , Coculture Techniques , Disease Models, Animal , Inflammation/metabolism , Mice , Serine Proteases/metabolism
19.
Cell Mol Life Sci ; 75(9): 1509-1520, 2018 05.
Article in English | MEDLINE | ID: mdl-29307015

ABSTRACT

The cornea is the most commonly transplanted tissue in the body. Corneal grafts in low-risk recipients enjoy high success rates, yet over 50% of high-risk grafts (with inflamed and vascularized host beds) are rejected. As our understanding of the cellular and molecular pathways that mediate rejection has deepened, a number of novel therapeutic strategies have been unveiled. This manuscript reviews therapeutic approaches to promote corneal transplant survival through targeting (1) corneal lymphangiogenesis and hemangiogenesis, (2) antigen presenting cells, (3) effector and regulatory T cells, and (4) mesenchymal stem cells.


Subject(s)
Corneal Transplantation/methods , Graft Survival , Adaptive Immunity , Animals , Antigen-Presenting Cells/immunology , Cornea/blood supply , Cornea/immunology , Cornea/physiology , Humans , Immune Tolerance , Lymphangiogenesis , Mesenchymal Stem Cells/immunology , Neovascularization, Physiologic , T-Lymphocytes/immunology
20.
Trends Immunol ; 39(4): 288-301, 2018 04.
Article in English | MEDLINE | ID: mdl-29248310

ABSTRACT

The ocular surface is a unique mucosal immune compartment in which anatomical, physiological, and immunological features act in concert to foster a particularly tolerant microenvironment. These mechanisms are vital to the functional competence of the eye, a fact underscored by the devastating toll of excessive inflammation at the cornea - blindness. Recent data have elucidated the contributions of specific anatomical components, immune cells, and soluble immunoregulatory factors in promoting homeostasis at the ocular surface. We highlight research trends at this distinctive mucosal barrier and identify crucial gaps in our current knowledge.


Subject(s)
Eye/immunology , Immune Privilege , Immunity, Mucosal , Inflammation/immunology , Mucous Membrane/immunology , Animals , Homeostasis , Humans , Immune Tolerance
SELECTION OF CITATIONS
SEARCH DETAIL