Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
1.
Kidney Int ; 104(3): 463-469, 2023 09.
Article in English | MEDLINE | ID: mdl-37391039

ABSTRACT

Kidney damage due to ischemia or rejection results in the accumulation of unfolded and misfolded proteins in the endoplasmic reticulum (ER) lumen, a condition known as "ER stress." Inositol-requiring enzyme 1α (IRE1α), the first ER stress sensor found, is a type I transmembrane protein with kinase and endoribonuclease activity. On activation, IRE1α nonconventionally splices an intron from unspliced X-box-binding protein 1 (XBP1) mRNA to produce XBP1s mRNA that encodes the transcription factor, XBP1s, for the expression of genes encoding proteins that mediate the unfolded protein response. The unfolded protein response promotes the functional fidelity of ER and is required for secretory cells to sustain protein folding and secretory capability. Prolonged ER stress can lead to apoptosis, which may result in detrimental repercussions to organ health and has been implicated in the pathogenesis and progression of kidney diseases. The IRE1α-XBP1 signaling acts as a major arm of unfolded protein response and is involved in regulating autophagy, cell differentiation, and cell death. IRE1α also interacts with activator protein-1 and nuclear factor-κB pathways to regulate inflammatory responses. Studies using transgenic mouse models highlight that the roles of IRE1α differ depending on cell type and disease setting. This review covers these cell-specific roles of IRE1α signaling and the potential for therapeutic targeting of this pathway in the context of ischemia and rejection affecting the kidneys.


Subject(s)
Endoribonucleases , Protein Serine-Threonine Kinases , Animals , Endoplasmic Reticulum Stress/genetics , Endoribonucleases/genetics , Endoribonucleases/metabolism , Graft Rejection , Inositol/metabolism , Kidney/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , RNA, Messenger/metabolism , Unfolded Protein Response , Humans
2.
Am J Transplant ; 22(11): 2502-2508, 2022 11.
Article in English | MEDLINE | ID: mdl-35612993

ABSTRACT

Connexins are a class of membrane proteins widely distributed throughout the body and have various functions based on their location and levels of expression. More specifically, connexin proteins expressed in endothelial cells (ECs) have unique roles in maintaining EC barrier integrity and function-a highly regulated process that is critical for pro-inflammatory and pro-coagulant reactions. In this minireview, we discuss the regulatory influence connexin proteins have in maintaining EC barrier integrity and their role in ischemia-reperfusion injury as it relates to organ transplantation. It is evident that certain isoforms of the connexin protein family are uniquely positioned to have far-reaching effects on preserving organ function; however, there is still much to be learned of their roles in transplant immunology and the application of this knowledge to the development of targeted therapeutics.


Subject(s)
Organ Transplantation , Reperfusion Injury , Humans , Endothelial Cells/metabolism , Connexins/therapeutic use , Reperfusion Injury/metabolism , Organ Preservation
3.
Am J Transplant ; 22(5): 1293-1298, 2022 05.
Article in English | MEDLINE | ID: mdl-35224837

ABSTRACT

Patients undergoing organ transplantation transition from one life-altering issue (organ dysfunction) to a lifelong commitment-immunosuppression. Regimens of immunosuppressive agents (ISAs) come with significant side effects and comorbidities. Recently, the use of nanoparticles (NPs) as a solution to the problems associated with the long-term and systemic use of ISAs in transplantation has emerged. This minireview describes the role of NPs in organ transplantation and discusses obstacles to clinical implementation and pathways to clinical translation.


Subject(s)
Immunosuppressive Agents , Organ Transplantation , Graft Rejection/drug therapy , Graft Rejection/prevention & control , Humans , Immune Tolerance , Immunosuppression Therapy , Immunosuppressive Agents/therapeutic use
4.
Am J Respir Cell Mol Biol ; 62(6): 793-804, 2020 06.
Article in English | MEDLINE | ID: mdl-32078336

ABSTRACT

Patients with lymphangioleiomyomatosis (LAM) develop pulmonary cysts associated with neoplastic, smooth muscle-like cells that feature neuroendocrine cell markers. The disease preferentially affects premenopausal women. Existing therapeutics do not cure LAM. As gp100 is a diagnostic marker expressed by LAM lesions, we proposed to target this immunogenic glycoprotein using TCR transgenic T cells. To reproduce the genetic mutations underlying LAM, we cultured Tsc2-/- kidney tumor cells from aged Tsc2 heterozygous mice and generated a stable gp100-expressing cell line by lentiviral transduction. T cells were isolated from major histocompatibility complex-matched TCR transgenic pmel-1 mice to measure cytotoxicity in vitro, and 80% cytotoxicity was observed within 48 hours. Antigen-specific cytotoxicity was likewise observed using pmel-1 TCR-transduced mouse T cells, suggesting that transgenic T cells may likewise be useful to treat LAM in vivo. On intravenous injection, slow-growing gp100+ LAM-like cells formed lung nodules that were readily detectable in severe combined immunodeficient/beige mice. Adoptive transfer of gp100-reactive but not wild-type T cells into mice significantly shrunk established lung tumors, even in the absence of anti-PD-1 therapy. These results demonstrate the treatment potential of adoptively transferred T cells to eliminate pulmonary lesions in LAM.


Subject(s)
Immunotherapy, Adoptive , Lymphangioleiomyomatosis/therapy , T-Lymphocyte Subsets/transplantation , Animals , Cell Line , Cell Line, Tumor , Coculture Techniques , Gene Knockout Techniques , Immunocompetence , Kidney Neoplasms , Lymphangioleiomyomatosis/immunology , Male , Melanoma/immunology , Melanoma/therapy , Mice , Mice, Mutant Strains , Mice, SCID , Mice, Transgenic , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Programmed Cell Death 1 Receptor/immunology , Receptors, Antigen, T-Cell/immunology , Recombinant Proteins/immunology , T-Cell Antigen Receptor Specificity , T-Lymphocyte Subsets/immunology , Tuberous Sclerosis Complex 2 Protein/deficiency , Tuberous Sclerosis Complex 2 Protein/genetics , Vesicular Transport Proteins/deficiency , gp100 Melanoma Antigen/genetics , gp100 Melanoma Antigen/immunology
5.
Mol Vis ; 23: 26-38, 2017.
Article in English | MEDLINE | ID: mdl-28275313

ABSTRACT

PURPOSE: Infection of the human cornea by herpes simplex virus type-1 (HSV-1) can cause significant vision loss. The purpose of this study was to develop an ex vivo model to visualize viral growth and spread in the cornea. The model was also used to analyze cytokine production and study the antiviral effects of zinc oxide tetrapods. METHODS: A ß-galactosidase-expressing recombinant virus, HSV-1(KOS)tk12, was used to demonstrate the ability of the virus to enter and develop blue plaques on human corneal epithelial (HCE) cells and corneal tissues. Freshly obtained porcine corneas were cultured and then scratched before infection with HSV-1(KOS)tk12. The blue plaques on the corneas were imaged using a stereomicroscope. Western blot analysis for HSV-1 proteins was performed to verify HSV-1 infection of the cornea. Using the ex vivo model, zinc oxide tetrapods were tested for their anti-HSV-1 potential, and a cytokine profile was developed to assess the effects of the treatment. RESULTS: Cultured corneas and the use of ß-galactosidase-expressing HSV-1(KOS)tk12 virus can provide an attractive ex vivo model to visualize and study HSV-1 entry and spread of the infection in tissues. We found that unlike cultured HCE cells, which demonstrated nearly 100% infectivity, HSV-1 infection of the cultured cornea was more restrictive and took longer to develop. We also found that the zinc oxide tetrapod-shaped nano- and microstructures inhibited HSV infection of the cultured cells, as well as the cultured corneas. The cytokine profile of the infected samples was consistent with previous studies of HSV-1 corneal infection. CONCLUSIONS: The ability to visualize HSV-1 growth and spread in corneal tissues can provide new details about HSV-1 infection of the cornea and the efficacy of new cornea-specific antiviral drug candidates. The ex vivo model also demonstrates antiviral effects of zinc oxide tetrapods and adequately portrays the drug delivery issues that cornea-specific treatments face.


Subject(s)
Cornea/pathology , Cornea/virology , Herpes Simplex/virology , Zinc Oxide/pharmacology , Animals , Cells, Cultured , Cornea/drug effects , Cytokines/metabolism , Epithelial Cells/drug effects , Epithelial Cells/pathology , Epithelial Cells/virology , Epithelium, Corneal/pathology , Epithelium, Corneal/virology , Herpes Simplex/immunology , Herpes Simplex/pathology , Herpesvirus 1, Human/drug effects , Humans , Immunity, Innate/drug effects , Sus scrofa , Thymidine Kinase/metabolism , Virus Internalization/drug effects
6.
J Virol ; 89(3): 1932-8, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25428865

ABSTRACT

Uncontrolled herpes simplex virus 1 (HSV-1) infection can advance to serious conditions, including corneal blindness or fatal encephalitis. Here, we describe a highly potent anti-HSV-1 peptide (DG2) that inhibits HSV-1 entry into host cells and blocks all aspects of infection. Importantly, DG2 is highly resistant to proteases and shows minimal toxicity, paving the way for prophylactic or therapeutic application of the peptide in vivo.


Subject(s)
Antiviral Agents/pharmacology , Herpesvirus 1, Human/drug effects , Herpesvirus 1, Human/physiology , Oligopeptides/pharmacology , Virus Internalization/drug effects , Antiviral Agents/metabolism , Antiviral Agents/toxicity , Cells, Cultured , Humans , Oligopeptides/metabolism , Oligopeptides/toxicity , Peptide Hydrolases/metabolism , Proteolysis
7.
Shock ; 61(3): 346-359, 2024 Mar 01.
Article in English | MEDLINE | ID: mdl-38517237

ABSTRACT

ABSTRACT: Severe traumatic brain injury (TBI) often initiates a systemic inflammatory response syndrome, which can potentially culminate into multiorgan dysfunction. A central player in this cascade is endotheliopathy, caused by perturbations in homeostatic mechanisms governed by endothelial cells due to injury-induced coagulopathy, heightened sympathoadrenal response, complement activation, and proinflammatory cytokine release. Unique to TBI is the potential disruption of the blood-brain barrier, which may expose neuronal antigens to the peripheral immune system and permit neuroinflammatory mediators to enter systemic circulation, propagating endotheliopathy systemically. This review aims to provide comprehensive insights into the "neuroendothelial axis" underlying endothelial dysfunction after TBI, identify potential diagnostic and prognostic biomarkers, and explore therapeutic strategies targeting these interactions, with the ultimate goal of improving patient outcomes after severe TBI.


Subject(s)
Brain Injuries, Traumatic , Endothelial Cells , Humans , Endothelial Cells/metabolism , Brain Injuries, Traumatic/therapy , Cytokines/metabolism , Blood-Brain Barrier/metabolism , Complement Activation
8.
Front Transplant ; 2: 1082227, 2023.
Article in English | MEDLINE | ID: mdl-38993857

ABSTRACT

Mitochondrial dynamics are central to the pathophysiology of cellular damage and inflammatory responses. In the context of solid organ transplantation, mitochondria are implicated in immune activation in donor organs that occurs after brain death, as they are critical to the regulation of cellular stress response, cell death, and display energetic adaptations through the adjustment of respiratory capacity depending on the cellular milieu. Mitochondrial damage activates mitochondrial systems of fission, fusion, biogenesis, and mitochondrial autophagy, or mitophagy. The mechanistic pathways as well as therapies targeting mitochondrial physiology have been studied as plausible ways to mitigate the negative effects of brain death on donor organs, though there is no summative evaluation of the multiple efforts across the field. This mini-review aims to discuss the interplay of donor brain death, mitochondrial dynamics, and impact on allograft function as it pertains to heart, lung, liver, and kidney transplants.

9.
Comput Struct Biotechnol J ; 21: 1030-1040, 2023.
Article in English | MEDLINE | ID: mdl-36733705

ABSTRACT

The structural diversity of metazoic heparan sulfate (HS) composed of unique sulfated domains is remarkably preserved among various vertebrates and invertebrate species. Interestingly the sulfated moieties of HS have been known as the key determinants generating extraordinary ligand binding sites in the HS chain to regulate multiple biological functions and homeostasis. One such ligand for 3-O sulfation in the HS chain is a glycoprotein D (gD) from an ancient herpesvirus, herpes simplex virus (HSV). This interaction between gD and 3-O sulfated HS leads to virus-cell fusion to promote HSV entry. It is quite astonishing that HSV-1, which infects two-thirds of the world population, is also capable of causing severe diseases in primates and non-primates including primitive zebrafish. Supporting evidence that HSV may cross the species barrier comes from the fact that an enzymatic modification in HS encoded by 3-O sulfotransferase-3 (3-OST-3) from a vertebrate zoonotic species enhances HSV-1 infectivity. The latter phenomenon suggests the possible role of sulfated-HS as an entry receptor during reverse zoonosis, especially during an event when humans encounter domesticated animals in proximity. In this mini-review, we explore the possibility that structural diversity in HS may have played a substantial role in species-specific adaptability for herpesviruses in general including their potential role in promoting cross-species transmission.

10.
JID Innov ; 3(5): 100190, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37554516

ABSTRACT

Checkpoint inhibitors treat a variety of tumor types with significant benefits. Unfortunately, these therapies come with diverse adverse events. Skin rash is observed early into treatment and might serve as an indicator of downstream responses to therapy. We studied the cellular composition of cutaneous eruptions and whether their contribution varies with the treatment applied. Skin samples from 18 patients with cancer and 11 controls were evaluated by mono- and multiplex imaging, quantification, and statistical analysis. T cells were the prime contributors to skin rash, with T cells and macrophages interacting and proliferating on site. Among T cell subsets examined, type 1 and 17 T cells were relatively increased among inflammatory skin infiltrates. A combination of increased cytotoxic T cell content and decreased macrophage abundance was associated with dual checkpoint inhibition over PD1 inhibition alone. Importantly, responders significantly separated from nonresponders by greater CD68+ macrophage and either CD11c+ antigen-presenting cell or CD4+ T cell abundance in skin rash. The microenvironment promoted epidermal proliferation and thickening as well. The combination of checkpoint inhibitors used affects the development and composition of skin infiltrates, whereas the combined abundance of two cell types in cutaneous eruptions aligns with responses to checkpoint inhibitor therapy.

11.
Cancers (Basel) ; 13(4)2021 Feb 16.
Article in English | MEDLINE | ID: mdl-33669271

ABSTRACT

Cytokines are key molecules within the tumor microenvironment (TME) that can be used as biomarkers to predict the magnitude of anti-tumor immune responses. During immune monitoring, it has been customary to predict outcomes based on the abundance of a single cytokine, in particular IFN-γ or TGF-ß, as a readout of ongoing anti-cancer immunity. However, individual cytokines within the TME can exhibit dual opposing roles. For example, both IFN-γ and TGF-ß have been associated with pro- and anti-tumor functions. Moreover, cytokines originating from different cellular sources influence the crosstalk between CD4+ and CD8+ T cells, while the array of cytokines expressed by T cells is also instrumental in defining the mechanisms of action and efficacy of treatments. Thus, it becomes increasingly clear that a reliable readout of ongoing immunity within the TME will have to include more than the measurement of a single cytokine. This review focuses on defining a panel of cytokines that could help to reliably predict and analyze the outcomes of T cell-based anti-tumor therapies.

12.
Cell Stress Chaperones ; 26(5): 845-857, 2021 09.
Article in English | MEDLINE | ID: mdl-34542825

ABSTRACT

Developing immunosuppressive therapies for autoimmune diseases comes with a caveat that immunosuppression may promote the risk of developing other conditions or diseases. We have previously shown that biolistic delivery of an expression construct encoding inducible HSP70 (HSP70i) with one amino acid modification in the dendritic cell (DC) activating moiety 435-445 (HSP70iQ435A) to mouse skin resulted in significant immunosuppressive activity of autoimmune vitiligo, associated with fewer tissue infiltrating T cells. To prepare HSP70iQ435A as a potential therapeutic for autoimmune vitiligo, in this study we evaluated whether and how biolistic delivery of HSP70iQ435A in mice affects anti-tumor responses. We found that HSP70iQ435A in fact supports anti-tumor responses in melanoma-challenged C57BL/6 mice. Biolistic delivery of the HSP70iQ435A-encoding construct to mice elicited significant anti-HSP70 titers, and anti-HSP70 IgG and IgM antibodies recognize surface-expressed and cytoplasmic HSP70i in human and mouse melanoma cells. A peptide scan revealed that the anti-HSP70 antibodies recognize a specific C-terminal motif within the HSP70i protein. The antibodies elicited surface CD107A expression among mouse NK cells, representative of antibody-mediated cellular cytotoxicity (ADCC), supporting the concept, that HSP70iQ435A-encoding DNA elicits a humoral response to the stress protein expressed selectively on the surface of melanoma cells. Thus, besides limiting autoimmunity and inflammation, HSP70iQ435A elicits humoral responses that limit tumor growth and may be used in conjunction with immune checkpoint inhibitors to not only control tumor but to also limit adverse events following tumor immunotherapy.


Subject(s)
Autoimmunity , HSP70 Heat-Shock Proteins/genetics , Melanoma/genetics , Melanoma/immunology , Mutation/genetics , Skin Neoplasms/genetics , Skin Neoplasms/immunology , Animals , Antibodies/metabolism , Autoimmunity/genetics , Cell Degranulation , Cell Line, Tumor , DNA, Neoplasm/metabolism , HSP70 Heat-Shock Proteins/metabolism , Humans , Killer Cells, Natural/metabolism , Killer Cells, Natural/physiology , Male , Melanoma/pathology , Mice, Inbred C57BL , Models, Biological , Skin Neoplasms/pathology
13.
JCI Insight ; 6(7)2021 04 08.
Article in English | MEDLINE | ID: mdl-33621216

ABSTRACT

The drive to withstand environmental stresses and defend against invasion is a universal trait extant in all forms of life. While numerous canonical signaling cascades have been characterized in detail, it remains unclear how these pathways interface to generate coordinated responses to diverse stimuli. To dissect these connections, we followed heparanase (HPSE), a protein best known for its endoglycosidic activity at the extracellular matrix but recently recognized to drive various forms of late-stage disease through unknown mechanisms. Using herpes simplex virus-1 (HSV-1) infection as a model cellular perturbation, we demonstrate that HPSE acts beyond its established enzymatic role to restrict multiple forms of cell-intrinsic defense and facilitate host cell reprogramming by the invading pathogen. We reveal that cells devoid of HPSE are innately resistant to infection and counteract viral takeover through multiple amplified defense mechanisms. With a unique grasp of the fundamental processes of transcriptional regulation and cell death, HPSE represents a potent cellular intersection with broad therapeutic potential.


Subject(s)
Glucuronidase/metabolism , Herpes Simplex/metabolism , Host-Pathogen Interactions/physiology , Animals , Cell Survival , Female , Glucuronidase/genetics , Herpes Simplex/genetics , Herpes Simplex/pathology , Herpes Simplex/virology , Herpesvirus 1, Human/pathogenicity , Immunity, Innate , Inflammation/genetics , Inflammation/pathology , Inflammation/virology , Interferon Type I/genetics , Interferon Type I/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout , Necroptosis , Transcription Factors/genetics , Transcription Factors/metabolism
14.
JCI Insight ; 6(22)2021 11 22.
Article in English | MEDLINE | ID: mdl-34806651

ABSTRACT

Mutations underlying disease in tuberous sclerosis complex (TSC) give rise to tumors with biallelic mutations in TSC1 or TSC2 and hyperactive mammalian target of rapamycin complex 1 (mTORC1). Benign tumors might exhibit de novo expression of immunogens, targetable by immunotherapy. As tumors may rely on ganglioside D3 (GD3) expression for mTORC1 activation and growth, we compared GD3 expression in tissues from patients with TSC and controls. GD3 was overexpressed in affected tissues from patients with TSC and also in aging Tsc2+/- mice. As GD3 overexpression was not accompanied by marked natural immune responses to the target molecule, we performed preclinical studies with GD3 chimeric antigen receptor (CAR) T cells. Polyfunctional CAR T cells were cytotoxic toward GD3-overexpressing targets. In mice challenged with Tsc2-/- tumor cells, CAR T cells substantially and durably reduced the tumor burden, correlating with increased T cell infiltration. We also treated aged Tsc2+/- heterozygous (>60 weeks) mice that carry spontaneous Tsc2-/- tumors with GD3 CAR or untransduced T cells and evaluated them at endpoint. Following CAR T cell treatment, the majority of mice were tumor free while all control animals carried tumors. The outcomes demonstrate a strong treatment effect and suggest that targeting GD3 can be successful in TSC.


Subject(s)
Immunotherapy, Adoptive/methods , Immunotherapy/methods , Tuberous Sclerosis/genetics , Animals , Female , Humans , Mice
15.
Nat Cancer ; 2(12): 1372-1386, 2021 12.
Article in English | MEDLINE | ID: mdl-35121903

ABSTRACT

Only a subset of recurrent glioblastoma (rGBM) responds to anti-PD-1 immunotherapy. Previously, we reported enrichment of BRAF/PTPN11 mutations in 30% of rGBM that responded to PD-1 blockade. Given that BRAF and PTPN11 promote MAPK/ERK signaling, we investigated whether activation of this pathway is associated with response to PD-1 inhibitors in rGBM, including patients that do not harbor BRAF/PTPN11 mutations. Here we show that immunohistochemistry for ERK1/2 phosphorylation (p-ERK), a marker of MAPK/ERK pathway activation, is predictive of overall survival following adjuvant PD-1 blockade in two independent rGBM patient cohorts. Single-cell RNA-sequencing and multiplex immunofluorescence analyses revealed that p-ERK was mainly localized in tumor cells and that high-p-ERK GBMs contained tumor-infiltrating myeloid cells and microglia with elevated expression of MHC class II and associated genes. These findings indicate that ERK1/2 activation in rGBM is predictive of response to PD-1 blockade and is associated with a distinct myeloid cell phenotype.


Subject(s)
Glioblastoma , Glioblastoma/drug therapy , Humans , Immunotherapy , MAP Kinase Signaling System , Neoplasm Recurrence, Local/drug therapy , Phosphorylation
16.
J Vis Exp ; (157)2020 03 30.
Article in English | MEDLINE | ID: mdl-32281983

ABSTRACT

Multispectral fluorescence imaging on formalin-fixed paraffin-embedded (FFPE) tissues enables the detection of multiple markers in a single tissue sample that can provide information about antigen coexpression and spatial distribution of the markers. However, a lack of suitable antibodies for formalin-fixed tissues may restrict the nature of markers that can be detected. In addition, the staining method is time-consuming. Here we describe a rapid method to perform multispectral fluorescence imaging on frozen tissues. The method includes the fluorophore combinations used, detailed steps for the staining of mouse and human frozen tissues, and the scanning, acquisition, and analysis procedures. For staining analysis, a commercially available semiautomated multispectral fluorescence imaging system is used. Through this method, up to six different markers were stained and detected in a single frozen tissue section. The machine learning analysis software can phenotype cells that can be used for quantitative analysis. The method described here for frozen tissues is useful for the detection of markers that cannot be detected in FFPE tissues or for which antibodies are not available for FFPE tissues.


Subject(s)
Fluorescence , Frozen Sections/methods , Tissue Fixation/methods , Animals , Humans , Mice
17.
PLoS One ; 15(1): e0227909, 2020.
Article in English | MEDLINE | ID: mdl-31986193

ABSTRACT

Vitiligo is a T-cell mediated skin disorder characterized by progressive loss of skin color. In individuals genetically predisposed to the disease, various triggers contribute to the initiation of vitiligo. Precipitating factors can stress the skin, leading to T-cell activation and recruitment. Though hereditary factors are implicated in the pathogenesis of vitiligo, it is unknown whether precipitating, stressful events play a role in vitiligo. To understand this, we utilized a validated perceived stress scale (PSS) to measure this parameter in vitiligo patients compared to persons without vitiligo. Additionally, we probed a clinical database, using a knowledge linking software called ROCKET, to gauge stress-related conditions in the vitiligo patient population. From a pool of patients in an existing database, a hundred individuals with vitiligo and twenty-five age- and sex-matched comparison group of individuals without vitiligo completed an online survey to quantify their levels of perceived stress. In parallel, patients described specifics of their disease condition, including the affected body sites, the extent, duration and activity of their vitiligo. Perceived stress was significantly higher among vitiligo individuals compared to those without vitiligo. ROCKET analyses suggested signs of metabolic-related disease (i.e., 'stress') preceding vitiligo development. No correlation was found between perceived stress and the stage or the extent of disease, suggesting that elevated stress may not be a consequence of pigment loss alone. The data provide further support for stress as a precipitating factor in vitiligo development.


Subject(s)
Stress, Physiological , Stress, Psychological/physiopathology , Vitiligo/physiopathology , Vitiligo/psychology , Adolescent , Adult , Aged , Child , Child, Preschool , Female , Heat-Shock Response/genetics , Humans , Infant , Infant, Newborn , Male , Melanocytes/metabolism , Melanocytes/pathology , Middle Aged , Patients/psychology , Stress, Psychological/complications , Stress, Psychological/genetics , Stress, Psychological/metabolism , Surveys and Questionnaires , T-Lymphocytes/metabolism , T-Lymphocytes/pathology , Vitiligo/complications , Vitiligo/metabolism , Young Adult
18.
Front Immunol ; 11: 581433, 2020.
Article in English | MEDLINE | ID: mdl-33335528

ABSTRACT

Vitiligo is an autoimmune skin disease characterized by melanocyte destruction. Regulatory T cells (Tregs) are greatly reduced in vitiligo skin, and replenishing peripheral skin Tregs can provide protection against depigmentation. Ganglioside D3 (GD3) is overexpressed by perilesional epidermal cells, including melanocytes, which prompted us to generate GD3-reactive chimeric antigen receptor (CAR) Tregs to treat vitiligo. Mice received either untransduced Tregs or GD3-specific Tregs to test the hypothesis that antigen specificity contributes to reduced autoimmune reactivity in vitro and in vivo. CAR Tregs displayed increased IL-10 secretion in response to antigen, provided superior control of cytotoxicity towards melanocytes, and supported a significant delay in depigmentation compared to untransduced Tregs and vehicle control recipients in a TCR transgenic mouse model of spontaneous vitiligo. The latter findings were associated with a greater abundance of Tregs and melanocytes in treated mice versus both control groups. Our data support the concept that antigen-specific Tregs can be prepared, used, and stored for long-term control of progressive depigmentation.


Subject(s)
Antigens/immunology , T-Lymphocytes, Regulatory/immunology , Vitiligo/immunology , Animals , Autoimmune Diseases/immunology , Autoimmunity/immunology , Epidermal Cells/immunology , Humans , Interleukin-10/immunology , Melanocytes/immunology , Mice , Mice, Transgenic , Receptors, Chimeric Antigen/immunology , Skin/immunology
19.
Sci Adv ; 5(8): eaax0780, 2019 08.
Article in English | MEDLINE | ID: mdl-31453334

ABSTRACT

Current drug-delivery systems are designed primarily for parenteral applications and are either lipid or polymer drug conjugates. In our quest to inhibit herpes simplex virus infection via the compounds found in commonly used cosmetic products, we found that activated carbon particles inhibit infection and, in addition, substantially improve topical delivery and, hence, the efficacy of a common antiviral drug, acyclovir (ACV). Our in vitro studies demonstrate that highly porous carbon structures trapped virions, blocked infection and substantially improved efficacy when ACV was loaded onto them. Also, using murine models of corneal and genital herpes infections, we show that the topical use of drug-encapsulated carbon (DECON) reduced dosing frequency, shortened treatment duration, and exhibited higher therapeutic efficacy than currently approved topical or systemic antivirals alone. DECON is a nontoxic, cost-effective and nonimmunogenic alternative to current topical drug-delivery systems that is uniquely triggered for drug release by virus trapping.


Subject(s)
Acyclovir/administration & dosage , Acyclovir/therapeutic use , Antiviral Agents/therapeutic use , Corneal Diseases/drug therapy , Drug Carriers/therapeutic use , Herpes Genitalis/drug therapy , Herpesvirus 1, Human/drug effects , Animals , Antiviral Agents/administration & dosage , CHO Cells , Carbon/chemistry , Cell Line, Tumor , Charcoal/chemistry , Chlorocebus aethiops , Corneal Diseases/virology , Cricetulus , Disease Models, Animal , Female , HeLa Cells , Herpes Genitalis/virology , Humans , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Vero Cells
20.
Front Pharmacol ; 9: 1315, 2018.
Article in English | MEDLINE | ID: mdl-30555321

ABSTRACT

An extraordinary binding site generated in heparan sulfate (HS) structures, during its biosynthesis, provides a unique opportunity to interact with multiple protein ligands including viral proteins, and therefore adds tremendous value to this master molecule. An example of such a moiety is the sulfation at the C3 position of glucosamine residues in HS chain via 3-O sulfotransferase (3-OST) enzymes, which generates a unique virus-cell fusion receptor during herpes simplex virus (HSV) entry and spread. Emerging evidence now suggests that the unique patterns in HS sulfation assist multiple viruses in invading host cells at various steps of their life cycles. In addition, sulfated-HS structures are known to assist in invading host defense mechanisms and initiating multiple inflammatory processes; a critical event in the disease development. All these processes are detrimental for the host and therefore raise the question of how HS-sulfation is regulated. Epigenetic modulations have been shown to be implicated in these reactions during HSV infection as well as in HS modifying enzyme sulfotransferases, and therefore pose a critical component in answering it. Interestingly, heparanase (HPSE) activity is shown to be upregulated during virus infection and multiple other diseases assisting in virus replication to promote cell and tissue damage. These phenomena suggest that sulfotransferases and HPSE serve as key players in extracellular matrix remodeling and possibly generating unique signatures in a given disease. Therefore, identifying the epigenetic regulation of OST genes, and HPSE resulting in altered yet specific sulfation patterns in HS chain during virus infection, will be a significant a step toward developing potential diagnostic markers and designing novel therapies.

SELECTION OF CITATIONS
SEARCH DETAIL