Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Front Immunol ; 15: 1398120, 2024.
Article in English | MEDLINE | ID: mdl-38903528

ABSTRACT

Psoriasis is an inflammatory disease with systemic manifestations that most commonly presents as itchy, erythematous, scaly plaques on extensor surfaces. Activation of the IL-23/IL-17 pro-inflammatory signaling pathway is a hallmark of psoriasis and its inhibition is key to clinical management. Granzyme K (GzmK) is an immune cell-secreted serine protease elevated in inflammatory and proliferative skin conditions. In the present study, human psoriasis lesions exhibited elevated GzmK levels compared to non-lesional psoriasis and healthy control skin. In an established murine model of imiquimod (IMQ)-induced psoriasis, genetic loss of GzmK significantly reduced disease severity, as determined by delayed plaque formation, decreased erythema and desquamation, reduced epidermal thickness, and inflammatory infiltrate. Molecular characterization in vitro revealed that GzmK contributed to macrophage secretion of IL-23 as well as PAR-1-dependent keratinocyte proliferation. These findings demonstrate that GzmK enhances IL-23-driven inflammation as well as keratinocyte proliferation to exacerbate psoriasis severity.


Subject(s)
Cell Proliferation , Granzymes , Inflammation , Interleukin-23 , Keratinocytes , Psoriasis , Psoriasis/immunology , Psoriasis/pathology , Animals , Keratinocytes/metabolism , Keratinocytes/immunology , Keratinocytes/pathology , Humans , Mice , Granzymes/metabolism , Granzymes/genetics , Interleukin-23/metabolism , Inflammation/immunology , Inflammation/pathology , Imiquimod , Disease Models, Animal , Mice, Knockout , Female , Male , Mice, Inbred C57BL
2.
Lab Invest ; 103(6): 100123, 2023 06.
Article in English | MEDLINE | ID: mdl-36849037

ABSTRACT

Age-related macular degeneration (AMD) is a leading cause of irreversible central vision loss in the elderly. The pathology of neovascular age-related macular degeneration (nAMD), also known as wet AMD, is associated with an abnormal blood vessel growth in the eye and involves an imbalance of proangiogenic and antiangiogenic factors. Thrombospondin (TSP)-1 and TSP-2 are endogenous matricellular proteins that inhibit angiogenesis. TSP-1 is significantly diminished in eyes with AMD, although the mechanisms involved in its reduction are unknown. Granzyme B (GzmB) is a serine protease with an increased extracellular activity in the outer retina and choroid of human eyes with nAMD-related choroidal neovascularization (CNV). This study investigated whether TSP-1 and TSP-2 are GzmB substrates using in silico and cell-free cleavage assays and explored the relationship between GzmB and TSP-1 in human eyes with nAMD-related CNV and the effect of GzmB on TSP-1 in retinal pigment epithelial culture and an explant choroid sprouting assay (CSA). In this study, TSP-1 and TSP-2 were identified as GzmB substrates. Cell-free cleavage assays substantiated the GzmB proteolysis of TSP-1 and TSP-2 by showing dose-dependent and time-dependent cleavage products. TSP-1 and TSP-2 proteolysis were hindered by the inhibition of GzmB. In the retinal pigment epithelium and choroid of human eyes with CNV, we observed a significant inverse correlation between TSP-1 and GzmB, as indicated by lower TSP-1 and higher GzmB immunoreactivity. In CSA, the vascular sprouting area increased significantly with GzmB treatment and reduced significantly with TSP-1 treatment. Western blot showed significantly reduced expression of TSP-1 in GzmB-treated retinal pigment epithelial cell culture and CSA supernatant compared with that in controls. Together, our findings suggest that the proteolysis of antiangiogenic factors such as TSP-1 by extracellular GzmB might represent a mechanism through which GzmB may contribute to nAMD-related CNV. Future studies are needed to investigate whether pharmacologic inhibition of extracellular GzmB can mitigate nAMD-related CNV by preserving intact TSP-1.


Subject(s)
Choroidal Neovascularization , Macular Degeneration , Humans , Aged , Thrombospondin 1/metabolism , Granzymes/metabolism , Proteolysis , Macular Degeneration/complications , Macular Degeneration/metabolism , Macular Degeneration/pathology , Choroidal Neovascularization/drug therapy , Choroidal Neovascularization/etiology , Choroidal Neovascularization/metabolism
3.
Br J Dermatol ; 189(3): 279-291, 2023 08 24.
Article in English | MEDLINE | ID: mdl-36652225

ABSTRACT

BACKGROUND: Granzyme K (GzmK) is a serine protease with minimal presence in healthy tissues while abundant in inflamed tissues. Initially thought to play an exclusive role in immune-mediated cell death, extracellular GzmK can also promote inflammation. OBJECTIVES: To evaluate the role of GzmK in the pathogenesis of atopic dermatitis (AD), the most common inflammatory skin disease. METHODS: A panel of human AD and control samples was analysed to determine if GzmK is elevated. Next, to determine a pathological role for GzmK in AD-like skin inflammation, oxazolone-induced dermatitis was induced in GzmK-/- and wild-type (WT) mice. RESULTS: In human lesional AD samples, there was an increase in the number of GzmK+ cells compared with healthy controls. GzmK-/- mice exhibited reduced overall disease severity characterized by reductions in scaling, erosions and erythema. Surprisingly, the presence of GzmK did not notably increase the overall pro-inflammatory response or epidermal barrier permeability in WT mice; rather, GzmK impaired angiogenesis, increased microvascular damage and microhaemorrhage. Mechanistically, GzmK contributed to vessel damage through cleavage of syndecan-1, a key structural component of the glycocalyx, which coats the luminal surface of vascular endothelia. CONCLUSIONS: GzmK may provide a potential therapeutic target for skin conditions associated with persistent inflammation, vasculitis and pathological angiogenesis.


Subject(s)
Dermatitis, Atopic , Granzymes , Animals , Humans , Mice , Dermatitis, Atopic/pathology , Epidermis/metabolism , Granzymes/metabolism , Inflammation , Skin/pathology
4.
Sci Rep ; 12(1): 12622, 2022 07 23.
Article in English | MEDLINE | ID: mdl-35871073

ABSTRACT

Pressure injuries, also known as pressure ulcers, are regions of localized damage to the skin and/or underlying tissue. Repeated rounds of ischemia-reperfusion (I/R) have a major causative role for tissue damage in pressure injury. Ischemia prevents oxygen/nutrient supply, and restoration of blood flow induces a burst of reactive oxygen species that damages blood vessels, surrounding tissues and can halt blood flow return. Minimizing the consequences of repeated I/R is expected to provide a protective effect against pressure injury. Sulfaphenazole (SP), an off patent sulfonamide antibiotic, is a potent CYP 2C6 and CYP 2C9 inhibitor, functioning to decrease post-ischemic vascular dysfunction and increase blood flow. The therapeutic effect of SP on pressure injury was therefore investigated in apolipoprotein E knockout mice, a model of aging susceptible to ischemic injury, which were subjected to repeated rounds of I/R-induced skin injury. SP reduced overall severity, improved wound closure and increased wound tensile strength compared to vehicle-treated controls. Saliently, SP restored tissue perfusion in and around the wound rapidly to pre-injury levels, decreased tissue hypoxia, and reduced both inflammation and fibrosis. SP also demonstrated bactericidal activity through enhanced M1 macrophage activity. The efficacy of SP in reducing thermal injury severity was also demonstrated. SP is therefore a potential therapeutic option for pressure injury and other ischemic skin injuries.


Subject(s)
Pressure Ulcer , Reperfusion Injury , Sulfaphenazole , Animals , Mice , Ischemia , Perfusion , Reactive Oxygen Species , Reperfusion Injury/drug therapy , Sulfaphenazole/pharmacology
5.
Physiology (Bethesda) ; 37(6): 323-348, 2022 11 01.
Article in English | MEDLINE | ID: mdl-35820180

ABSTRACT

Granzymes are serine proteases previously believed to play exclusive and somewhat redundant roles in lymphocyte-mediated target cell death. However, recent studies have challenged this paradigm. Distinct substrate profiles and functions have since emerged for each granzyme while their dysregulated proteolytic activities have been linked to diverse pathologies.


Subject(s)
Granzymes , Humans , Granzymes/metabolism , Wound Healing , Serine Proteases , Inflammation
6.
NPJ Aging Mech Dis ; 7(1): 6, 2021 Mar 05.
Article in English | MEDLINE | ID: mdl-33674592

ABSTRACT

Pressure injuries (PIs), also known as bedsores or pressure ulcers, are a major cause of death and morbidity in the elderly. The serine protease, Granzyme B (GzmB), contributes to skin aging and impaired wound healing. Aging is a major risk factor for PIs; thus, the role of GzmB in PI pathogenesis was investigated. GzmB levels in human PI tissue and wound fluids were markedly elevated. A causative role for GzmB was assessed in GzmB knockout (GzmB-/-) and wild-type (WT) mice using a murine model of PI. An apolipoprotein E knockout (ApoE-/-) model of aging and vascular dysfunction was also utilized to assess GzmB in a relevant age-related model better resembling tissue perfusion in the elderly. PI severity displayed no difference between young GzmB-/- and WT mice. However, in aged mice, PI severity was reduced in mice lacking GzmB. Mechanistically, GzmB increased vascular wall inflammation and impaired extracellular matrix remodeling. Together, GzmB is an important contributor to age-dependent impaired PI healing.

7.
Nat Commun ; 12(1): 302, 2021 01 12.
Article in English | MEDLINE | ID: mdl-33436591

ABSTRACT

Pemphigoid diseases refer to a group of severe autoimmune skin blistering diseases characterized by subepidermal blistering and loss of dermal-epidermal adhesion induced by autoantibody and immune cell infiltrate at the dermal-epidermal junction and upper dermis. Here, we explore the role of the immune cell-secreted serine protease, granzyme B, in pemphigoid disease pathogenesis using three independent murine models. In all models, granzyme B knockout or topical pharmacological inhibition significantly reduces total blistering area compared to controls. In vivo and in vitro studies show that granzyme B contributes to blistering by degrading key anchoring proteins in the dermal-epidermal junction that are necessary for dermal-epidermal adhesion. Further, granzyme B mediates IL-8/macrophage inflammatory protein-2 secretion, lesional neutrophil infiltration, and lesional neutrophil elastase activity. Clinically, granzyme B is elevated and abundant in human pemphigoid disease blister fluids and lesional skin. Collectively, granzyme B is a potential therapeutic target in pemphigoid diseases.


Subject(s)
Autoimmune Diseases/enzymology , Autoimmune Diseases/pathology , Granzymes/antagonists & inhibitors , Granzymes/metabolism , Animals , Autoantigens/metabolism , Blister , Chemokine CXCL2/metabolism , Chemotactic Factors/pharmacology , Disease Models, Animal , Epidermolysis Bullosa/enzymology , Epidermolysis Bullosa/pathology , Humans , Inflammation/pathology , Integrin alpha6/metabolism , Interleukin-8/metabolism , Neutrophil Infiltration/drug effects , Non-Fibrillar Collagens/metabolism , Pemphigoid, Bullous/enzymology , Pemphigoid, Bullous/pathology , Severity of Illness Index , Collagen Type XVII
8.
J Invest Dermatol ; 141(1): 36-47, 2021 01.
Article in English | MEDLINE | ID: mdl-32504614

ABSTRACT

Atopic dermatitis (AD) is the most common inflammatory skin condition. Skin barrier dysfunction is of major importance in AD because it facilitates allergen sensitization and systemic allergic responses. Long regarded as a pro-apoptotic protease, emerging studies indicate granzyme B (GzmB) to have extracellular roles involving the proteolytic cleavage of extracellular matrix, cell adhesion proteins, and basement membrane proteins. Minimally expressed in normal skin, GzmB is elevated in AD and is positively correlated with disease severity and pruritus. We hypothesized that GzmB contributes to AD through extracellular protein cleavage. A causative role for GzmB was assessed in an oxazolone-induced murine model of dermatitis, comparing GzmB-/- mice with wild-type mice, showing significant reductions in inflammation, epidermal thickness, and lesion formation in GzmB-/- mice. Topical administration of a small-molecule GzmB inhibitor reduced disease severity compared with vehicle-treated controls. Mechanistically, GzmB impaired epithelial barrier function through E-cadherin and FLG cleavage. GzmB proteolytic activity contributes to impaired epidermal barrier function and represents a valid therapeutic target for AD.


Subject(s)
Cadherins/metabolism , Dermatitis, Atopic/metabolism , Granzymes/metabolism , Oxazolone/adverse effects , S100 Proteins/metabolism , Animals , Dermatitis, Atopic/chemically induced , Dermatitis, Atopic/pathology , Epidermis/metabolism , Extracellular Matrix/metabolism , Filaggrin Proteins , Humans
10.
Front Immunol ; 11: 574, 2020.
Article in English | MEDLINE | ID: mdl-32318066

ABSTRACT

Granzymes are a family of serine proteases first shown to be intracellular initiators of immune-mediated cell death in target pathogenic cells. In addition to its intracellular role, Granzyme B (GzmB) has important extracellular functions in immune regulation and extracellular matrix (ECM) degradation. Verified substrates of extracellular GzmB activity include tight junctional and ECM proteins. Interestingly, little is known about the activity of GzmB in the outer human retina, a tissue in which the degradation of the tight junctional contacts of retinal pigment epithelial (RPE) cells and within the external limiting membrane, as well as remodeling of the ECM in Bruch's membrane, cause the breakdown of the blood-retinal barrier and slowing of metabolite transport between neuroretina and choroidal blood supply. Such pathological changes in outer retina signal early events in the development of age-related macular degeneration (AMD), a multifactorial, chronic inflammatory eye disease. This study is the first to focus on the distribution of GzmB in the outer retina of the healthy and diseased post-mortem human eye. Our results revealed that GzmB is present in RPE and choroidal mast cells. More immunoreactive cells are present in older (>65 years) compared to younger (<55 years) donor eyes, and choroidal immunoreactive cells are more numerous in eyes with choroidal neovascularization (CNV), while RPE immunoreactive cells are more numerous in eyes with soft drusen, an early AMD event. In vitro studies demonstrated that RPE-derived tight junctional and ECM proteins are cleaved by exogenous GzmB stimulation. These results suggest that the increased presence of GzmB immunoreactive cells in outer retina of older (healthy) eyes as well as in diseased eyes with CNV (from AMD) and eyes with soft drusen exacerbate ECM remodeling in the Bruch's membrane and degradation of the blood-retinal barrier. Currently there are no treatments that prevent remodeling of the Bruch's membrane and/or the loss of function of the outer blood-retinal barrier, known to promote early AMD changes, such as drusen deposition, RPE dysfunction and pro-inflammation. Specific inhibitors of GzmB, already in preclinical studies for non-ocular diseases, may provide new strategies to stop these early events associated with the development of AMD.


Subject(s)
Choroid/enzymology , Choroidal Neovascularization/enzymology , Extracellular Matrix/enzymology , Granzymes/metabolism , Retinal Pigment Epithelium/enzymology , Adult , Aged , Extracellular Matrix Proteins/metabolism , Female , Humans , Male , Mast Cells/enzymology , Middle Aged , Retina/enzymology , Tight Junctions/metabolism
11.
Expert Opin Ther Targets ; 23(9): 745-754, 2019 09.
Article in English | MEDLINE | ID: mdl-31461387

ABSTRACT

Introduction: Granzyme B is a serine protease traditionally understood as having a role in immune-mediated cytotoxicity. Over the past decade, this dogma has been challenged, with a new appreciation that granzyme B can exert alternative extracellular roles detrimental to wound closure and remodeling. Granzyme B is elevated in response to tissue injury, chronic inflammation and/or autoimmune skin diseases, resulting in impaired wound healing. Areas covered: This review provides a historical background of granzyme B and a description of how it is regulated. Details are provided on the role of granzyme B in apoptosis as well as newly identified extracellular roles, focusing on those affecting wound healing, including on inflammation, dermal-epidermal junction separation, re-epithelialization, scarring and fibrosis, and autoimmunity. Finally, the use of pharmacological granzyme B inhibitors as potential therapeutic options for wound treatment is discussed. Expert opinion: Endogenous extracellular granzyme B inhibitors have not been identified in human bio-fluids, thus in chronic wound environments granzyme B appears to remain uncontrolled and unregulated. In response, targeted granzyme B inhibitors have been developed for therapeutic applications in wounds. Animal studies trialing inhibitors of granzyme B show improved healing outcomes, and may therefore provide a novel therapeutic approach for wound treatment.


Subject(s)
Granzymes/antagonists & inhibitors , Wound Healing/drug effects , Wounds and Injuries/drug therapy , Animals , Autoimmune Diseases/drug therapy , Autoimmune Diseases/pathology , Granzymes/metabolism , Humans , Inflammation/drug therapy , Inflammation/pathology , Skin Diseases/drug therapy , Skin Diseases/pathology , Wounds and Injuries/pathology
12.
Sci Rep ; 9(1): 10027, 2019 07 11.
Article in English | MEDLINE | ID: mdl-31296909

ABSTRACT

Wildfire smoke induces acute pulmonary distress and is of particular concern to risk groups such as the sick and elderly. Wood smoke (WS) contains many of the same toxic compounds as those found in cigarette smoke (CS) including polycyclic aromatic hydrocarbons, carbon monoxide, and free radicals. CS is a well-established risk factor for respiratory diseases such as asthma and COPD. Limited studies investigating the biological effects of WS on the airway epithelium have been performed. Using a cell culture-based model, we assessed the effects of a WS-infused solution on alveolar epithelial barrier function, cell migration, and survival. The average geometric mean of particles in the WS was 178 nm. GC/MS analysis of the WS solution identified phenolic and cellulosic compounds. WS exposure resulted in a significant reduction in barrier function, which peaked after 24 hours of continuous exposure. The junctional protein E-cadherin showed a prominent reduction in response to increasing concentrations of WS. Furthermore, WS significantly repressed cell migration following injury to the cell monolayer. There was no difference in cell viability following WS exposure. Mechanistically, WS exposure induced activation of the p44/42, but not p38, MAPK signaling pathway, and inhibition of p44/42 phosphorylation prevented the disruption of barrier function and loss of E-cadherin staining. Thus, WS may contribute to the breakdown of alveolar structure and function through a p44/42 MAPK-dependent pathway and may lead to the development and/or exacerbation of respiratory pathologies with chronic exposure.


Subject(s)
Alveolar Epithelial Cells/pathology , Epithelium/physiopathology , MAP Kinase Signaling System/drug effects , Smoke/adverse effects , Tight Junctions/pathology , A549 Cells , Alveolar Epithelial Cells/drug effects , Cadherins/metabolism , Cell Line , Cell Movement/drug effects , Cell Survival/drug effects , Humans , Lung Diseases/chemically induced , Tight Junctions/drug effects , Wildfires
13.
Front Immunol ; 10: 1454, 2019.
Article in English | MEDLINE | ID: mdl-31297118

ABSTRACT

Pemphigoid diseases are a subgroup of autoimmune skin diseases characterized by widespread tense blisters. Standard of care typically involves immunosuppressive treatments, which may be insufficient and are often associated with significant adverse events. As such, a deeper understanding of the pathomechanism(s) of pemphigoid diseases is necessary in order to identify improved therapeutic approaches. A major initiator of pemphigoid diseases is the accumulation of autoantibodies against proteins at the dermal-epidermal junction (DEJ), followed by protease activation at the lesion. The contribution of proteases to pemphigoid disease pathogenesis has been investigated using a combination of in vitro and in vivo models. These studies suggest proteolytic degradation of anchoring proteins proximal to the DEJ is crucial for dermal-epidermal separation and blister formation. In addition, proteases can also augment inflammation, expose autoantigenic cryptic epitopes, and/or provoke autoantigen spreading, which are all important in pemphigoid disease pathology. The present review summarizes and critically evaluates the current understanding with respect to the role of proteases in pemphigoid diseases.


Subject(s)
Autoantibodies/immunology , Autoantigens/immunology , Dermis/immunology , Epidermis/immunology , Pemphigoid, Bullous/immunology , Peptide Hydrolases/immunology , Dermis/pathology , Epidermis/pathology , Humans , Pemphigoid, Bullous/pathology
14.
Matrix Biol ; 75-76: 126-140, 2019 01.
Article in English | MEDLINE | ID: mdl-29247692

ABSTRACT

Granzyme B (GzmB) is a serine protease emerging as an important mediator of skin injury, inflammation and repair. Found at low levels in healthy skin, GzmB is dramatically elevated in chronic disease and inflammatory skin disorders, including diabetic ulcers, hypertrophic scarring, autoimmune skin disorders, cutaneous leishmaniasis and aging skin. Traditionally known for its pro-apoptotic function, the role of GzmB in disease has been redefined due to the discovery of additional activities involving the cleavage of extracellular matrix proteins, epithelial barrier disruption, fibrosis, vascular permeability, anoikis, inflammation and autoimmunity. In addition to the accumulation of GzmB+ cells in diseased tissue, and critical to the mechanistic redefinition, is the realization that GzmB often accumulates in the extracellular milieu, retains its activity in plasma, and is expressed by both immune and non-immune cells that may or may not express perforin, the pore-forming protein required for GzmB internalization into target cells. As GzmB is not normally found in the extracellular milieu, and does not appear to be regulated, GzmB-mediated proteolysis can impact processes such as tissue remodelling, barrier function, autoantigen generation and angiogenesis. The present review will summarize and critically examine the current knowledge regarding GzmB in inflammatory skin disease, providing an overview of both apoptotic and extracellular mechanisms, but with a focus on the extracellular roles of GzmB in skin health and disease.


Subject(s)
Granzymes/genetics , Inflammation/genetics , Skin Aging/genetics , Skin Diseases/genetics , Animals , Apoptosis/genetics , Chronic Disease , Fibrosis/genetics , Fibrosis/pathology , Humans , Inflammation/pathology , Skin/growth & development , Skin/pathology , Skin Aging/pathology , Skin Diseases/pathology
15.
JACC Cardiovasc Imaging ; 12(1): 135-145, 2019 01.
Article in English | MEDLINE | ID: mdl-30448122

ABSTRACT

OBJECTIVES: This study investigated processes causing leaflet thickening and structural valve degeneration (SVD). BACKGROUND: Although transcatheter aortic valve replacement (TAVR) has changed the treatment of aortic stenosis, concerns remain regarding SVD, potentially related to valve thrombosis and thickening, based on studies using computed tomography (CT). Detailed histological analyses are provided to help attain insights into these processes. METHODS: Explanted transcatheter heart valves (THVs) were evaluated for thrombosis, fibrosis, and calcification for quantification of leaflet thickness. Immunohistochemical and microscopy approaches were used to investigate SVD-associated mechanisms. RESULTS: THVs (n = 23) were obtained from 22 patients (median 81 years of age; 50% male) from 0 to 2,583 days post TAVR. Maximal leaflet thickness increased relative to implant duration (ρ = 0.427; p = 0.027). THVs explanted after >2 years were thicker than those explanted after <2 years (p = 0.007). All THVs had adherent thrombus on both aortic and ventricular sides, which beyond 60 days was seen in combination with fibrosis and beyond 4 years had calcification. Early thrombus formation (<60 days) occurred despite rapid endothelialization with an abnormal hyperplastic phenotype. Fibrosis was observed in 6 patients on both the aortic and the ventricular THV surfaces, remodeled over time, and was associated with matrix metalloproteinase-1 expression. Five THVs showed overt calcification associated with adherent thrombus and fibrosis. CONCLUSIONS: There is a time-dependent degeneration of THVs consisting of thrombus formation, endothelial hyperplasia, fibrosis, tissue remodeling, proteinase expression, and calcification. Future investigation is needed to further understand these mechanisms contributing to leaflet thickening and SVD.


Subject(s)
Aortic Valve/pathology , Aortic Valve/surgery , Heart Valve Prosthesis , Prosthesis Failure , Transcatheter Aortic Valve Replacement/instrumentation , Aged , Aged, 80 and over , Aortic Valve/enzymology , Calcinosis/etiology , Calcinosis/pathology , Device Removal , Endothelial Cells/pathology , Female , Fibrosis , Humans , Male , Matrix Metalloproteinase 1/metabolism , Prosthesis Design , Registries , Retrospective Studies , Thrombosis/etiology , Thrombosis/pathology , Time Factors , Transcatheter Aortic Valve Replacement/adverse effects , Treatment Outcome
16.
J Invest Dermatol ; 139(4): 930-939, 2019 04.
Article in English | MEDLINE | ID: mdl-30395844

ABSTRACT

Granzyme K (GzmK), traditionally described as a pro-apoptotic, granule-secreted serine protease, has been proposed to promote inflammation. Found at low levels in the plasma of healthy individuals, GzmK is markedly elevated in response to sepsis and infection. In this study we investigated the role of GzmK in inflammation and remodeling in response to thermal injury. In human burn tissue, GzmK was elevated compared with normal skin, with expression predominantly found in macrophages. GzmK was expressed and secreted by cultured human classically activated macrophages. To assess the role of GzmK in response to skin wounding, wild-type or GzmK-/- mice were subjected to grade 2 thermal injury. GzmK-/- mice exhibited improved wound closure, matrix organization, and tensile strength compared with wild-type mice. Reduced proinflammatory IL-6, ICAM-1, VCAM-1, and MCP-1 expressions were observed at 3 days after injury. Additionally, GzmK induced IL-6 expression in keratinocytes and skin fibroblasts that was dependent on PAR-1 activation. Re-epithelialization showed the greatest degree of improvement of all healing parameters, suggesting that keratinocytes are sensitive to GzmK-mediated proteolysis. In support, keratinocytes, but not skin fibroblasts, exposed to GzmK showed impaired wound healing in vitro. In summary, GzmK influences wound healing by augmenting inflammation and impeding epithelialization.


Subject(s)
Burns/genetics , Gene Expression Regulation , Granzymes/genetics , Inflammation/genetics , Macrophages/metabolism , Re-Epithelialization/physiology , Animals , Burns/metabolism , Burns/pathology , Cytokines/biosynthesis , Cytokines/genetics , Disease Models, Animal , Fibroblasts/metabolism , Fibroblasts/pathology , Granzymes/biosynthesis , Humans , Immunohistochemistry , Inflammation/metabolism , Inflammation/pathology , Keratinocytes/metabolism , Keratinocytes/pathology , Macrophages/pathology , Mice , RNA/genetics
17.
Cell Rep ; 25(9): 2401-2416.e5, 2018 11 27.
Article in English | MEDLINE | ID: mdl-30485809

ABSTRACT

Cells in multicellular organisms are arranged in complex three-dimensional patterns. This requires both transient and stable adhesions with the extracellular matrix (ECM). Integrin adhesion receptors bind ECM ligands outside the cell and then, by binding the protein talin inside the cell, assemble an adhesion complex connecting to the cytoskeleton. The activity of talin is controlled by several mechanisms, but these have not been well studied in vivo. By generating mice containing the activating point mutation E1770A in talin (Tln1), which disrupts autoinhibition, we show that talin autoinhibition controls cell-ECM adhesion, cell migration, and wound healing in vivo. In particular, blocking autoinhibition gives rise to more mature, stable focal adhesions that exhibit increased integrin activation. Mutant cells also show stronger attachment to ECM and decreased traction force. Overall, these results demonstrate that modulating talin function via autoinhibition is an important mechanism for regulating multiple aspects of integrin-mediated cell-ECM adhesion in vivo.


Subject(s)
Extracellular Matrix/metabolism , Talin/metabolism , Wound Healing , Actins/metabolism , Animals , Biomechanical Phenomena , Cell Adhesion , Cell Movement , Embryo, Mammalian/metabolism , Fibroblasts/metabolism , Focal Adhesions/metabolism , Integrins/metabolism , Mice , Mutation/genetics , Phenotype , Signal Transduction , Talin/genetics
18.
Exp Mol Med ; 50(5): 1-11, 2018 05 30.
Article in English | MEDLINE | ID: mdl-29849046

ABSTRACT

Granzyme B (GzmB) is a serine protease that has long been thought to function exclusively in lymphocyte-mediated apoptosis. In recent years, this paradigm has been revisited due to the recognition that GzmB accumulates in the extracellular milieu in many autoimmune and chronic inflammatory disorders, and contributes to impaired tissue remodeling due to the cleavage of extracellular matrix proteins. Knockout studies suggest that GzmB-mediated cleavage of decorin (DCN) contributes to impaired collagen fibrillogenesis and remodeling. As DCN is anti-fibrotic and contributes to reduced hypertrophic scarring, GzmB-induced DCN cleavage could play a role in wound healing following burn injury. In the present study, a novel, gel-formulated, first-in-class small-molecule inhibitor of GzmB, VTI-1002, was assessed in a murine model of impaired, diabetic burn wound healing. VTI-1002 exhibited high specificity, potency, and target selectivity. Gel-formulated VTI-1002 was able to penetrate the stratum corneum and was retained in the skin with minimal systemic absorption. Daily topical administration of VTI-1002 gel for 30 days following thermal injury showed significantly accelerated wound closure, increased DCN protein levels, and collagen organization that was translated into significantly increased wound tensile strength compared to controls. Overall, VTI-1002 gel was well-tolerated in vivo and no adverse events were observed. Topical application of VTI-1002 represents a novel therapeutic approach for the treatment of cutaneous burn wounds.


Subject(s)
Burns/pathology , Granzymes/antagonists & inhibitors , Small Molecule Libraries/administration & dosage , Small Molecule Libraries/pharmacology , Wound Healing/drug effects , Administration, Topical , Animals , Cicatrix/pathology , Diabetes Mellitus, Experimental/pathology , Disease Models, Animal , Granzymes/metabolism , Male , Mice, Inbred C57BL
19.
Adv Healthc Mater ; 6(2)2017 Jan.
Article in English | MEDLINE | ID: mdl-27869355

ABSTRACT

Flightless I (Flii) is elevated in human chronic wounds and is a negative regulator of wound repair. Decreasing its activity improves healing responses. Flii neutralizing antibodies (FnAbs) decrease Flii activity in vivo and hold significant promise as healing agents. However, to avoid the need for repeated application in a clinical setting and to protect the therapeutic antibody from the hostile environment of the wound, suitable delivery vehicles are required. In this study, the use of porous silicon nanoparticles (pSi NPs) is demonstrated for the controlled release of FnAb to diabetic wounds. We achieve FnAb loading regimens exceeding 250 µg antibody per mg of vehicle. FnAb-loaded pSi NPs increase keratinocyte proliferation and enhance migration in scratch wound assays. Release studies confirm the functionality of the FnAb in terms of Flii binding. Using a streptozotocin-induced model of diabetic wound healing, a significant improvement in healing is observed for mice treated with FnAb-loaded pSi NPs compared to controls, including FnAb alone. FnAb-loaded pSi NPs treated with proteases show intact and functional antibody for up to 7 d post-treatment, suggesting protection of the antibodies from proteolytic degradation in wound fluid. pSi NPs may therefore enable new therapeutic approaches for the treatment of diabetic ulcers.


Subject(s)
Antibodies, Neutralizing/pharmacology , Cytoskeletal Proteins/antagonists & inhibitors , Diabetes Complications/drug therapy , Diabetes Mellitus, Experimental/drug therapy , Nanoparticles , Wound Healing/drug effects , Wounds and Injuries/drug therapy , Animals , Carrier Proteins , Delayed-Action Preparations/pharmacology , Humans , Mice, Inbred BALB C , Microfilament Proteins , Silicon , Trans-Activators
20.
Biochem Biophys Res Commun ; 479(4): 721-727, 2016 Oct 28.
Article in English | MEDLINE | ID: mdl-27693584

ABSTRACT

Pompe disease is caused by a deficiency in the lysosomal enzyme α-glucosidase, and this leads to glycogen accumulation in the autolysosomes of patient cells. Glycogen storage material is exocytosed at a basal rate in cultured Pompe cells, with one study showing up to 80% is released under specific culture conditions. Critically, exocytosis induction may reduce glycogen storage in Pompe patients, providing the basis for a therapeutic strategy whereby stored glycogen is redirected to an extracellular location and subsequently degraded by circulating amylases. The focus of the current study was to identify compounds capable of inducing rapid glycogen exocytosis in cultured Pompe cells. Here, calcimycin, lysophosphatidylcholine and α-l-iduronidase each significantly increased glycogen exocytosis compared to vehicle-treated controls. The most effective compound, calcimycin, induced exocytosis through a Ca2+-dependent mechanism, although was unable to release a pool of vesicular glycogen larger than the calcimycin-induced exocytic pore. There was reduced glycogen release from Pompe compared to unaffected cells, primarily due to increased granule size in Pompe cells. Drug induced exocytosis therefore shows promise as a therapeutic approach for Pompe patients but strategies are required to enhance the release of large molecular weight glycogen granules.


Subject(s)
Calcimycin/pharmacology , Exocytosis/drug effects , Glycogen Storage Disease Type II/metabolism , Glycogen/metabolism , Iduronidase/pharmacology , Lysophosphatidylcholines/pharmacology , Calcimycin/therapeutic use , Cells, Cultured , Drug Evaluation, Preclinical , Fibroblasts/drug effects , Fibroblasts/metabolism , Glycogen Storage Disease Type II/drug therapy , Humans , Iduronidase/therapeutic use , Lysophosphatidylcholines/therapeutic use , Lysosomes/drug effects , Phagosomes/drug effects , Pharmaceutical Vehicles/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...