Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
1.
J Leukoc Biol ; 2024 May 24.
Article in English | MEDLINE | ID: mdl-38785333

ABSTRACT

T cell immunoglobulin and ITIM domain (TIGIT) is an immune checkpoint molecule that suppresses T cell activation and promotes an immunosuppressive environment to suppress autoimmune diseases. However, the impact of a TIGIT agonist as a treatment for ocular autoimmune disease has not been investigated. We examined TIGIT expression on Th17 and Treg cells, the role of TIGIT on experimental autoimmune uveitis (EAU) and Th17 cells, and the impact of Treg generation following TIGIT stimulation. TIGIT stimulation at the onset of clinical symptoms reduced the severity of uveitis and suppressed infiltration of Th17 cells into the eye. Further, Tregs from mice treated with the TIGIT agonist were capable of suppressing EAU in recipient mice. This report demonstrates that stimulation of TIGIT at onset of disease suppresses symptoms and allows for induction of regulatory immunity that provides resistance to uveitis.

2.
Neurogastroenterol Motil ; 36(1): e14691, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37849439

ABSTRACT

BACKGROUND: Patients with obesity often report esophageal symptoms, with abnormal reflux and esophageal motility suggested as potential mechanisms. However, prior studies showed varying results, often limited by study design/size and esophageal function/symptom measures utilized. We aimed to examine the relationship between obesity and objective esophageal function testing and patient-reported outcomes, utilizing prospective symptom, manometric and reflux monitoring data with impedance. METHODS: Adults referred for high-resolution impedance-manometry (HRiM) and multichannel intraluminal impedance-pH monitoring (MII-pH) to evaluate esophageal symptoms were enrolled. Validated symptom and health-related quality of life (HR-QOL) instruments were prospectively collected: GERDQ, reflux symptoms index (RSI), dominant symptom intensity (DSI, multiplied 5-point Likert scales for symptom frequency/severity), global symptom severity (GSS, 100-point visual analog scale), and Short Form-12 (SF-12) for HR-QOL. Esophageal function testing measures were compared across body mass index (BMI) categories and correlated with patient-reported outcomes. KEY RESULTS: Seven hundred and fifty four patients were included (Normal:281/Overweight:253/Class I obesity:137/Class II/III obesity:83). Reflux burden measures on MII-pH (acid exposure time, total reflux episodes, bolus exposure time), conclusive pathologic reflux (Lyon), and hiatal hernia were increased in higher obesity classes compared to normal BMI. Class II/III obesity was associated with more normal/hypercontractile swallows, less ineffective swallows, and better bolus transit on HRiM. BMI correlated positively with GERDQ/RSI/DSI/GSS, and negatively with physical component score (SF-12). Esophageal symptom severity and HR-QOL correlated strongly with MII-pH findings, but not HRiM measures. CONCLUSIONS/INFERENCES: Obesity is associated with increased esophageal symptom burden and worse physical HR-QOL, which correlate with higher acid/bolus reflux burden but not altered esophageal motility/transit/contractile reserve.


Subject(s)
Gastroesophageal Reflux , Quality of Life , Adult , Humans , Prospective Studies , Esophageal pH Monitoring , Gastroesophageal Reflux/complications , Gastroesophageal Reflux/diagnosis , Manometry/methods , Obesity/complications , Electric Impedance
3.
Mucosal Immunol ; 16(4): 422-431, 2023 08.
Article in English | MEDLINE | ID: mdl-37164238

ABSTRACT

The proper function of regulatory T cells (Tregs) to suppress inflammation requires homing to the correct tissue site. Resolution of autoimmune uveitis generates distinct programmed death receptor 1 (PD-1+) and T-cell immunoreceptor with immunoglobulin and immunoreceptor tyrosine-based inhibitory motif domains (TIGIT+) Tregs in an adenosine 2A receptor (A2Ar)-dependent manner found in the spleen. Where and how these Tregs migrate from the spleen to prevent uveitis is not known. In this work, we show that A2Ar-dependent Tregs migrated to the eye and secondary lymphoid tissue and expressed chemokine receptor (CCR)6 and CCR7. Suppression of autoimmune uveitis required CCR6 and CCR7 expression for TIGIT+ Tregs but not PD-1+ Tregs. Moreover, stimulation of A2Ar on T cells from patients showed a decreased capacity to induce TIGIT+ Tregs that expressed CCR6 or CCR7, and PD-1+ Treg that expressed CCR6. This work provides a mechanistic understanding of the homing requirements of each of these Treg populations. Importantly, this work is clinically relevant because patients with chronic autoimmune uveitis are unable to induce the Treg populations identified in mice that home to the target tissue.


Subject(s)
Autoimmune Diseases , T-Lymphocytes, Regulatory , Uveitis , Animals , Mice , Inflammation/metabolism , Receptors, CCR7/metabolism , Receptors, Immunologic/genetics , Receptors, Immunologic/metabolism , Uveitis/metabolism , Autoimmune Diseases/metabolism , Receptor, Adenosine A2A/metabolism
4.
Adv Drug Deliv Rev ; 198: 114869, 2023 07.
Article in English | MEDLINE | ID: mdl-37172782

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.


Subject(s)
Eye , Uveitis , Humans , Uveitis/drug therapy , Antigens
5.
J Clin Gastroenterol ; 56(3): 228-233, 2022 03 01.
Article in English | MEDLINE | ID: mdl-33988353

ABSTRACT

GOAL: The goal of this study was to evaluate whether a history of eating disorders (EDs) or psychiatric disorders (PDs) are risk factors for rumination syndrome (RS). BACKGROUND: RS is a disorder of gut-brain interaction characterized by an effortless postprandial retrograde flow of ingested contents. Disorder of gut-brain interactions have been associated with psychiatric and behavioral comorbidities. No prior comparative study has assessed the relationship between RS and ED or PD. METHODS: This was a case-control study of adults with RS at a tertiary center in January 2013 to January 2018. Two age-matched/gender-matched controls per RS case were identified. The Fisher exact test (categorical)/Student t test (continuous) and forward stepwise logistic regression were performed for univariate and multivariable analyses, respectively. RESULTS: Seventy-two patients (24 cases/48 controls) were included. Baseline demographics and characteristics were similar between cases and controls. Among RS patients, 9 (37.5%) had a history of ED, including 3 (12.5%) anorexia nervosa and 4 (16.7%) bulimia nervosa; and 20 (83.3%) had a PD, including 9 (37.5%) anxiety and 7 (29.2%) depression. Prevalence of ED (37.5% vs. 4.2%, P=0.0002) and PD (83.3% vs. 50.0%, P=0.0062) were higher among RS patients than controls. Specifically, the risks of anorexia nervosa (16.7% vs. 0%, P=0.005) and bulimia nervosa (21.1% vs. 0%, P=0.001) were both increased in RS patients. On multivariable analysis, ED (adjusted odds ratio=16.4, P=0.0033) and PD (adjusted odds ratio=4.47, P=0.029) remained independent predictors for RS. CONCLUSIONS: A history of ED and PD were independent risk factors for RS. Abnormal eating behaviors and psychiatric comorbidities may contribute to the pathogenesis of RS. Evaluation of RS should include a detailed history for ED and PD.


Subject(s)
Bulimia Nervosa , Feeding and Eating Disorders , Rumination Syndrome , Adult , Bulimia Nervosa/epidemiology , Bulimia Nervosa/psychology , Case-Control Studies , Feeding and Eating Disorders/epidemiology , Humans , Risk Factors
6.
Front Immunol ; 12: 742154, 2021.
Article in English | MEDLINE | ID: mdl-34867964

ABSTRACT

Regulatory immunity that provides resistance to relapse emerges during resolution of experimental autoimmune uveitis (EAU). This post-EAU regulatory immunity requires a melanocortin 5 receptor (MC5r)-dependent suppressor antigen presenting cell (APC), as shown using a MC5r single knock-out mouse. The MC5r-dependent APC activates an adenosine 2A receptor (A2Ar)-dependent regulatory Treg cell, as shown using an A2Ar single knock-out mouse. Unexpectedly, when MC5r-/- post-EAU APC were used to activate A2Ar-/- post-EAU T cells the combination of cells significantly suppressed EAU, when transferred to EAU mice. In contrast, transfer of the reciprocal activation scheme did not suppress EAU. In order to explain this finding, MC5r-/-A2Ar-/- double knock-out (DKO) mice were bred. Naïve DKO mice had no differences in the APC populations, or inflammatory T cell subsets, but did have significantly more Treg cells. When we examined the number of CD4 and CD8 T cell subsets, we found significantly fewer CD8 T cells in the DKO mice compared to WT and both single knock-out mice. DKO mice also had significantly reduced EAU severity and accelerated resolution. In order to determine if the CD8 T cell deficiency contributed to the resistance to EAU in the DKO mice, we transferred naïve CD8 T cells from WT mice, that were immunized for EAU. Susceptibility to EAU was restored in DKO mice that received a CD8 T cell transfer. While the mechanism that contributed to the CD8 T cell deficiency in the DKO mice remains to be determined, these observations indicate an importance of CD8 T cells in the initiation of EAU. The involvement of CD4 and CD8 T cells suggests that both class I and class II antigen presentation can trigger an autoimmune response, suggesting a much wider range of antigens may trigger autoimmune disease.


Subject(s)
Autoimmune Diseases/immunology , CD8-Positive T-Lymphocytes/immunology , Receptor, Adenosine A2A/immunology , Receptors, Melanocortin/immunology , Uveitis/immunology , Animals , Autoimmunity/immunology , CD4-Positive T-Lymphocytes/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptor, Adenosine A2A/deficiency , Receptors, Melanocortin/deficiency
8.
J Clin Gastroenterol ; 55(6): 499-504, 2021 07 01.
Article in English | MEDLINE | ID: mdl-32649446

ABSTRACT

GOAL: The goal of this study was to compare the clinical presentations of esophagogastric junction outflow obstruction (EGJOO) with coexisting abnormal esophageal body motility (EBM) to isolated EGJOO. BACKGROUND: The clinical significance and management of EGJOO remain debated, as patients may have varied to no symptoms. The effect of coexisting abnormal EBM in EGJOO is unclear. We hypothesized that a concomitant EBM disorder is associated with clinical symptoms of EGJOO. STUDY: This was a retrospective cohort study of consecutive adults diagnosed with EGJOO on high-resolution impedance-manometry (HRIM) at 2 academic centers in March 2018 to September 2018. Patients with prior treatment for achalasia, foregut surgery, or evidence of obstruction were excluded. Subjects were divided into EGJOO with abnormal EBM per Chicago classification v3.0 and isolated EGJOO. Statistical analyses were performed using Fisher-exact or Student t test (univariate) and logistic or linear regression (multivariate). RESULTS: Eighty-two patients (72% women, age 61.1±10.7 y) were included. Thirty-one (37.8%) had abnormal EBM, including 16 (19.5%) ineffective esophageal motility and 15 (18.2%) hypercontractile esophagus. Esophageal symptoms (heartburn, regurgitation, chest pain, dysphagia) were more prevalent among those with abnormal EBM (90.3% vs. 64.7%, P=0.01). On logistic regression adjusting for age, gender, body mass index, and opioid use, abnormal EBM remained predictive of esophageal symptoms (adjusted odds ratio [aOR] 7.51, P=0.007). On separate models constructed, HE was associated with chest pain (aOR 7.45, P=0.01) and regurgitation (aOR 4.06, P=0.046), while ineffective esophageal motility was predictive of heartburn (aOR 5.84, P=0.009) and decreased complete bolus transit (ß-coefficient -0.177, P=0.04). CONCLUSION: Coexisting abnormal EBM is associated with esophageal symptoms and bolus transit in patients with EGJOO.


Subject(s)
Esophageal Motility Disorders , Adult , Aged , Chicago , Esophageal Motility Disorders/diagnosis , Esophagogastric Junction , Female , Humans , Male , Manometry , Middle Aged , Retrospective Studies
9.
Front Immunol ; 11: 975, 2020.
Article in English | MEDLINE | ID: mdl-32508841

ABSTRACT

Experimental autoimmune uveoretinitis (EAU) is a mouse model of human autoimmune uveitis. EAU spontaneously resolves and is marked by ocular autoantigen-specific regulatory immunity in the spleen. Kallikrein binding protein (KBP) or kallistatin is a serine proteinase inhibitor that inhibits angiogenesis and inflammation, but its role in autoimmune uveitis has not been explored. We report that T cells activation is inhibited and EAU is attenuated in human KBP (HKBP) mice with no significant difference in the Treg population that we previously identified both before and after recovery from EAU. Moreover, following EAU immunization HKBP mice have potent ocular autoantigen specific regulatory immunity that is functionally suppressive.


Subject(s)
Autoimmune Diseases/prevention & control , Autoimmunity , Lymphocyte Activation , Serpins/metabolism , Spleen/metabolism , T-Lymphocytes/metabolism , Uvea/metabolism , Uveitis/prevention & control , Animals , Autoimmune Diseases/genetics , Autoimmune Diseases/immunology , Autoimmune Diseases/metabolism , Disease Models, Animal , Mice, Inbred C57BL , Mice, Transgenic , Serpins/genetics , Spleen/immunology , T-Lymphocytes/immunology , Uvea/immunology , Uvea/pathology , Uveitis/genetics , Uveitis/immunology , Uveitis/metabolism
10.
J Autoimmun ; 111: 102441, 2020 07.
Article in English | MEDLINE | ID: mdl-32201225

ABSTRACT

Regulatory T cells (Tregs) are necessary to prevent autoimmune disease. As such, stable FoxP3 expression is required for the proper function of Tregs in the control of autoimmune disease. Different Treg subsets that utilize different mechanisms of suppression have been identified. The T-cell immunoglobulin immunoreceptor tyrosine-based inhibitory motif (TIGIT) is a relatively new Treg cell marker that has a suppressive function. We have previously identified the adenosine 2A receptor (A2Ar) as a requirement for the emergence of Tregs following resolution of autoimmune disease. Using a FoxP3-GFP-Cre reporter mouse, we identify FoxP3 and 'exFoxP3' cells, show FoxP3 and not exFoxP3 cells are suppressive. We further show FoxP3 cells express TIGIT, and are induced through A2Ar in healthy volunteers, but not patients with autoimmune disease. Furthermore, we show Tregs emerge in the target tissue at the onset of autoimmune disease in an A2Ar-dependent manner. In summary, we identify a novel subset of TIGIT+ Tregs that are induced through stimulation of the A2Ar.


Subject(s)
Autoimmune Diseases/immunology , Receptor, Adenosine A2A/metabolism , Receptors, Immunologic/metabolism , Retinitis/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocytes, Regulatory/immunology , Uveitis/immunology , Animals , Cells, Cultured , Disease Models, Animal , Forkhead Transcription Factors/genetics , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptor, Adenosine A2A/genetics
11.
Int Immunopharmacol ; 83: 106378, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32172208

ABSTRACT

The role of regulatory T cells (Treg cell) in controlling autoimmune disease is an area of intense study. As such, the characterization and understanding the function of Treg markers has the potential to provide a considerable impact in developing treatments and understanding the pathogenesis of autoimmune diseases. One such inhibitory Treg cell marker that has been recently discovered is T cell immunoglobulin and ITIM domain (TIGIT). In this review, we discuss what is known about the expression and function of TIGIT on Treg cells, and we discuss the relationship between TIGIT expressing Treg cells and different autoimmune diseases such as atopic dermatitis, autoimmune thyroiditis, type 1 diabetes, autoimmune uveitis, aplastic anemia, multiple sclerosis, systemic lupus erythematosus, arthritis, and colitis.


Subject(s)
Autoimmune Diseases/immunology , Receptors, Immunologic/metabolism , T-Lymphocytes, Regulatory/immunology , Animals , Humans
12.
Sci Rep ; 9(1): 16941, 2019 11 15.
Article in English | MEDLINE | ID: mdl-31729418

ABSTRACT

Experimental autoimmune uveoretinitis (EAU) is a mouse model of human autoimmune uveitis marked by ocular autoantigen-specific regulatory immunity in the spleen. The melanocortin 5 receptor (MC5r) and adenosine 2 A receptor (A2Ar) are required for induction of post-EAU regulatory T cells (Tregs) which provide resistance to EAU. We show that blocking the PD-1/PD-L1 pathway prevented suppression of EAU by post-EAU Tregs. A2Ar induction of PD-1+FoxP3+ Tregs in uveitis patients was similar compared to healthy controls, but was significantly reduced with melanocortin stimulation. Further, lower body mass index correlated with responsiveness to stimulation of this pathway. These observations indicate an importance of the PD-1/PD-L1 pathway to provide resistance to relapsing uveitis and shows a reduced capacity of uveitis patients to induce Tregs when stimulated through melanocortin receptors, but that it is possible to bypass this part of the pathway through direct stimulation of A2Ar.


Subject(s)
Autoimmune Diseases/metabolism , Autoimmune Diseases/prevention & control , Programmed Cell Death 1 Receptor/metabolism , Receptors, Melanocortin/metabolism , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Adult , Animals , Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/metabolism , Autoantigens/immunology , Autoimmune Diseases/immunology , Autoimmunity , Biomarkers , Cytokines/metabolism , Disease Models, Animal , Disease Susceptibility , Female , Humans , Immunomodulation , Inflammation Mediators/metabolism , Male , Mice , Middle Aged , Uveitis/etiology , Uveitis/metabolism , Uveitis/pathology
13.
J Leukoc Biol ; 106(4): 879-887, 2019 10.
Article in English | MEDLINE | ID: mdl-31287586

ABSTRACT

Autoimmune uveitis is a leading cause of blindness with a complex etiology. Obesity is considered a chronic disease with a connection with autoimmune diseases through systemic inflammation. However, an obesity and autoimmune disease connection is not consistently true in rodent models of autoimmune disease. A mouse model of human autoimmune uveitis, experimental autoimmune uveitis (EAU) has been used to better understand the immunobiology of uveitis. In this study, we assessed EAU in a high-fat diet (HFD) obesity model and found that the EAU severity is significantly higher in wild-type mice, but not in HFD melanocortin 5 receptor deficient mice. We find a decrease in CD11b+ F4/80+ Ly-6Clo Ly-6G+ Mϕs, previously shown to be suppressive, and an enhancement of a Th1 response at the onset of EAU in obese mice. We further demonstrate that at recovery of EAU, obese mice lack regulatory immunity that provides protection from EAU. This report demonstrates that obesity exacerbates autoimmune uveitis and inhibits the promotion of post-EAU regulatory immunity through the melanocortin 5 receptor. The implication of this work is that obesity may contribute to the prevalence of autoimmune uveitis.


Subject(s)
Autoimmune Diseases/pathology , Disease Progression , Obesity/pathology , Receptors, Melanocortin/metabolism , Uveitis/pathology , Animals , Autoimmune Diseases/immunology , CD11 Antigens/metabolism , Diet, High-Fat , Immunity , Interferon-gamma/biosynthesis , Interleukin-17/biosynthesis , Mice, Inbred C57BL , Spleen/pathology , Th1 Cells/immunology , Uveitis/immunology
14.
Eur J Immunol ; 49(11): 2074-2082, 2019 11.
Article in English | MEDLINE | ID: mdl-31177529

ABSTRACT

Tissue-specific immune regulation is an important component of the immune response relevant to many areas of immunology. The focus of this study is on tissue-specific mechanisms that contribute to autoimmune uveitis. Precise gene regulation is necessary for the proper expression of an inflammatory or regulatory response. This precision gene regulation can be accomplished by microRNA at the level of the mRNA transcript. miR-155, in particular, has a complicated role in the immune response with positive and negative inflammatory effects. In this work, we identify a decrease in miR-155 in suppressor macrophages and further examine how tissue-specific production of miR-155 impacts experimental autoimmune uveitis. Importantly, we show that eliminating miR-155 expression by the target tissue before initiation reduces disease severity, but elimination of miR-155 after the onset of inflammation does not alter the course of disease. Additionally, expression of miR-155 by the target tissue before initiation is necessary for the induction of regulatory immunity that protects from further autoimmune disease, but not after the onset of inflammation. In summary, we find a MC5r-dependent decrease in miR-155 in postexperimental autoimmune uveitis APC, miR-155 production by the target tissue is necessary for the initiation of autoimmune uveitis, and may have a role in establishing protective regulatory immunity.


Subject(s)
Autoimmune Diseases/genetics , Macrophages/immunology , MicroRNAs/genetics , Receptors, Melanocortin/genetics , Retinal Pigment Epithelium/immunology , Uveitis/genetics , Animals , Autoimmune Diseases/chemically induced , Autoimmune Diseases/immunology , Autoimmune Diseases/pathology , Freund's Adjuvant/administration & dosage , Gene Expression Regulation/immunology , Humans , Macrophages/drug effects , Macrophages/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , MicroRNAs/immunology , Organ Specificity , Receptors, Melanocortin/immunology , Retinal Pigment Epithelium/drug effects , Retinal Pigment Epithelium/pathology , Signal Transduction , Spleen/drug effects , Spleen/immunology , Spleen/pathology , Uveitis/chemically induced , Uveitis/immunology , Uveitis/pathology
15.
Sci Rep ; 6: 37790, 2016 11 25.
Article in English | MEDLINE | ID: mdl-27886238

ABSTRACT

Autoantigen-specific regulatory immunity emerges in the spleen of mice recovering from experimental autoimmune uveitis (EAU), a murine model for human autoimmune uveoretinitis. This regulatory immunity provides induced tolerance to ocular autoantigen, and requires melanocortin 5 receptor (MC5r) expression on antigen presenting cells with adenosine 2 A receptor (A2Ar) expression on T cells. During EAU it is not well understood what roles MC5r and A2Ar have on promoting regulatory immunity. Cytokine profile analysis during EAU revealed MC5r and A2Ar each mediate distinct T cell responses, and are responsible for a functional regulatory immune response in the spleen. A2Ar stimulation at EAU onset did not augment this regulatory response, nor bypass the MC5r requirement to induce regulatory immunity. The importance of this pathway in human autoimmune uveitis was assayed. PBMC from uveitis patients were assayed for MC5r expression on monocytes and A2Ar on T cells, and comparison between uveitis patients and healthy controls had no significant difference. The importance for MC5r and A2Ar expression in EAU to promote the induction of protective regulatory immunity, and the expression of MC5r and A2Ar on human immune cells, suggests that it may be possible to utilize the melanocortin-adenosinergic pathways to induce protective immunity in uveitic patients.


Subject(s)
Autoimmune Diseases/immunology , Cell Polarity/genetics , Receptors, Adenosine A2/genetics , Receptors, Melanocortin/genetics , T-Lymphocytes, Regulatory/immunology , Uveitis/immunology , Animals , Autoimmune Diseases/blood , Case-Control Studies , Cytokines/metabolism , Humans , Mice , Mice, Inbred C57BL , Uveitis/blood
16.
Biomed Res Int ; 2016: 3801570, 2016.
Article in English | MEDLINE | ID: mdl-27119078

ABSTRACT

Diabetes mellitus (DM) is a metabolic disease defined by elevated blood glucose (BG). DM is a global epidemic and the prevalence is anticipated to continue to increase. The ocular complications of DM negatively impact the quality of life and carry an extremely high economic burden. While systemic control of BG can slow the ocular complications they cannot stop them, especially if clinical symptoms are already present. With the advances in biodegradable polymers, implantable ocular devices can slowly release medication to stop, and in some cases reverse, diabetic complications in the eye. In this review we discuss the ocular complications associated with DM, the treatments available with a focus on localized treatments, and what promising treatments are on the horizon.


Subject(s)
Diabetes Complications/therapy , Diabetic Retinopathy/pathology , Glaucoma/pathology , Macular Edema/pathology , Blood Glucose , Diabetes Complications/pathology , Diabetic Retinopathy/therapy , Eye/pathology , Glaucoma/etiology , Glaucoma/therapy , Humans , Insulin/metabolism , Macular Edema/etiology , Macular Edema/therapy , Quality of Life , Risk Factors
17.
J Funct Biomater ; 6(3): 650-66, 2015 Jul 31.
Article in English | MEDLINE | ID: mdl-26264035

ABSTRACT

Uveitis is the third leading cause of blindness in developed countries. Currently, the most widely used treatment of non-infectious uveitis is corticosteroids. Posterior uveitis and macular edema can be treated with intraocular injection of corticosteroids, however, this is problematic in chronic cases because of the need for repeat injections. Another option is systemic immunosuppressive therapies that have their own undesirable side effects. These systemic therapies result in a widespread suppression of the entire immune system, leaving the patient susceptible to infection. Therefore, an effective localized treatment option is preferred. With the recent advances in bioengineering, biodegradable polymers that allow for a slow sustained-release of a medication. These advances have culminated in drug delivery implants that are food and drug administration (FDA) approved for the treatment of non-infectious uveitis. In this review, we discuss the types of ocular implants available and some of the polymers used, implants used for the treatment of non-infectious uveitis, and bioengineered alternatives that are on the horizon.

18.
J Leukoc Biol ; 97(6): 1101-9, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25877928

ABSTRACT

The recovery of EAU, a mouse model of endogenous human autoimmune uveitis, is marked with the emergence of autoantigen-specific regulatory immunity in the spleen that protects the mice from recurrence of EAU. This regulatory immunity is mediated by a melanocortin-driven suppressor APC that presents autoantigen and uses adenosine to activate an antigen-specific CD4(+) Tregs through the A2Ar. These cells are highly effective in suppressing uveitis, and they appear to be inducible Tregs. In this study, we determined whether they are inducible or natural Tregs and identified the dependent mechanism for the function of these post-EAU Tregs. The post-EAU spleen CD25(+)CD4(+) T cells were sorted for NRP-1 expression and transferred to recipient mice immunized for EAU. The sorted NRP-1(-), but not the NRP-1(+), Tregs suppressed EAU. These NRP-1(-) Tregs coexpress PD-1 and PD-L1. Treatment of naive APCs with α-MSH promoted a regulatory APC that induced CD25(+) CD4(+) Tregs in a CD73-dependent manner. These Tregs were PD-L1(+) PD-1(+) NRP-1(-) FOXP3(+) HELIOS(-) and suppressed EAU when transferred to recipient mice. In contrast, PD-1(-) T cells did not suppress EAU, indicating that PD-1 is necessary for the suppressive activity of iTregs. Moreover, these Tregs did not suppress effector T cells when the PD/-1/PD-L1 pathway was blocked. These results demonstrate that post-EAU Tregs are inducible Tregs, which use a PD-1/PD-L1 mechanism to suppress disease.


Subject(s)
Autoimmune Diseases/immunology , B7-H1 Antigen/immunology , Dendritic Cells/immunology , Programmed Cell Death 1 Receptor/immunology , T-Lymphocytes, Regulatory/immunology , Uveitis/immunology , Adenosine/immunology , Animals , Antigens, CD/genetics , Antigens, CD/immunology , Autoimmune Diseases/genetics , Autoimmune Diseases/pathology , B7-H1 Antigen/genetics , Convalescence , DNA-Binding Proteins/genetics , DNA-Binding Proteins/immunology , Dendritic Cells/pathology , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/immunology , Gene Expression Regulation , Humans , Melanocortins/immunology , Mice , Neuropilin-1/genetics , Neuropilin-1/immunology , Programmed Cell Death 1 Receptor/genetics , Signal Transduction , Spleen/immunology , Spleen/pathology , T-Lymphocytes, Regulatory/pathology , T-Lymphocytes, Regulatory/transplantation , Transcription Factors/genetics , Transcription Factors/immunology , Uveitis/genetics , Uveitis/pathology , alpha-MSH/pharmacology
19.
J Immunol ; 191(8): 4103-11, 2013 Oct 15.
Article in English | MEDLINE | ID: mdl-24043903

ABSTRACT

The ocular microenvironment uses a poorly defined mela5 receptor (MC5r)-dependent pathway to recover immune tolerance following intraocular inflammation. This dependency is seen in experimental autoimmune uveoretinitis (EAU), a mouse model of endogenous human autoimmune uveitis, with the emergence of autoantigen-specific regulatory immunity in the spleen that protects the mice from recurrence of EAU. In this study, we found that the MC5r-dependent regulatory immunity increased CD11b(+)F4/80(+)Ly-6C(low)Ly-6G(+)CD39(+)CD73(+) APCs in the spleen of post-EAU mice. These MC5r-dependent APCs require adenosine 2A receptor expression on T cells to activate EAU-suppressing CD25(+)CD4(+)Foxp3(+) regulatory T cells. Therefore, in the recovery from autoimmune disease, the ocular microenvironment induces tolerance through a melanocortin-mediated expansion of Ly-6G(+) regulatory APCs in the spleen that use the adenosinergic pathway to promote activation of autoantigen-specific regulatory T cells.


Subject(s)
Receptor, Adenosine A2A/metabolism , Receptors, Melanocortin/metabolism , Self Tolerance/immunology , T-Lymphocytes, Regulatory/immunology , Uveitis/immunology , 5'-Nucleotidase/metabolism , Animals , Antigens, CD/metabolism , Antigens, Ly/metabolism , Apyrase/metabolism , Autoantigens/immunology , Autoimmune Diseases , CD11b Antigen/metabolism , CD4 Antigens/metabolism , Disease Models, Animal , Eye/immunology , Eye Proteins/immunology , Forkhead Transcription Factors/metabolism , Inflammation/immunology , Interleukin-2 Receptor alpha Subunit/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Melanocortin/genetics , Retinol-Binding Proteins/immunology , Spleen/immunology , T-Lymphocytes, Regulatory/metabolism
20.
Mol Genet Genomics ; 288(9): 401-12, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23793387

ABSTRACT

The poly(A)-binding protein PAB1 from the yeast Saccharomyces cerevisiae plays an important role in controlling mRNA deadenylation rates. Deletion of either its RRM1 or proline-rich domain (P domain) severely restricts deadenylation and slows mRNA degradation. Because these large deletions could be having unknown effects on the structure of PAB1, different strategies were used to determine the importance of the RRM1 and P domains to deadenylation. Since the P domain is quite variable in size and sequence among eukaryotes, P domains from two human PABPCs and from Xenopus were substituted for that of PAB1. The resultant PAB1 hybrid proteins, however, displayed limited or no difference in mRNA deadenylation as compared with PAB1. In contrast to the P domain, the RRM1 domain is highly conserved across species, and a systematic mutagenesis of the RRM1 domain was undertaken to identify its functional regions. Several mutations along the RNA-binding surface of RRM1 inhibited deadenylation, whereas one set of mutations on its exterior non-RNA binding surface shifted deadenylation from a slow distributive process to a rapid processive deadenylation. These results suggest that the RRM1 domain is the more critical region of PAB1 for controlling deadenylation and consists of at least two distinguishable functional regions.


Subject(s)
Poly(A)-Binding Proteins/metabolism , RNA, Fungal/metabolism , RNA, Messenger/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Animals , Binding Sites , Humans , Poly(A)-Binding Proteins/genetics , Protein Structure, Tertiary , RNA, Fungal/genetics , RNA, Messenger/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics , Xenopus Proteins/genetics , Xenopus Proteins/metabolism , Xenopus laevis
SELECTION OF CITATIONS
SEARCH DETAIL
...