Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 15 de 15
1.
Front Immunol ; 14: 1099529, 2023.
Article En | MEDLINE | ID: mdl-37228593

Over the past thirty years, the complexity of the αß-T cell compartment has been enriched by the identification of innate-like T cells (ITCs), which are composed mainly of invariant natural killer T (iNKT) cells and mucosal-associated invariant T (MAIT) cells. Based on animal studies using ischemia-reperfusion (IR) models, a key role has been attributed to iNKT cells in close connection with the alarmin/cytokine interleukin (IL)-33, as early sensors of cell-stress in the initiation of acute sterile inflammation. Here we have investigated whether the new concept of a biological axis of circulating iNKT cells and IL-33 applies to humans, and may be extended to other ITC subsets, namely MAIT and γδ-T cells, in the acute sterile inflammation sequence occurring during liver transplant (LT). From a prospective biological collection of recipients, we reported that LT was accompanied by an early and preferential activation of iNKT cells, as attested by almost 40% of cells having acquired the expression of CD69 at the end of LT (i.e. 1-3 hours after portal reperfusion), as opposed to only 3-4% of conventional T cells. Early activation of iNKT cells was positively correlated with the systemic release of the alarmin IL-33 at graft reperfusion. Moreover, in a mouse model of hepatic IR, iNKT cells were activated in the periphery (spleen), and recruited in the liver in WT mice, as early as the first hour after reperfusion, whereas this phenomenon was virtually missing in IL-33-deficient mice. Although to a lesser degree than iNKT cells, MAIT and γδ-T cells also seemed targeted during LT, as attested by 30% and 10% of them acquiring CD69 expression, respectively. Like iNKT cells, and in clear contrast to γδ-T cells, activation of MAIT cells during LT was closely associated with both release of IL-33 immediately after graft reperfusion and severity of liver dysfunction occurring during the first three post-operative days. All in all, this study identifies iNKT and MAIT cells in connection with IL-33 as new key cellular factors and mechanisms of acute sterile inflammation in humans. Further investigations are required to confirm the implication of MAIT and iNKT cell subsets, and to precisely assess their functions, in the clinical course of sterile inflammation accompanying LT.


Liver Diseases , Natural Killer T-Cells , Animals , Humans , Mice , Alarmins/metabolism , Inflammation/metabolism , Interleukin-33/metabolism , Ischemia/metabolism , Liver Diseases/metabolism , Natural Killer T-Cells/metabolism , Prospective Studies , Reperfusion
2.
Front Immunol ; 12: 744927, 2021.
Article En | MEDLINE | ID: mdl-34621275

Ischemia and reperfusion injury is an early inflammatory process during liver transplantation that impacts on graft function and clinical outcomes. Interleukin (IL)-33 is a danger-associated molecular pattern involved in kidney ischemia/reperfusion injury and several liver diseases. The aims were to assess whether IL-33 was released as an alarmin responsible for ischemia/reperfusion injury in a mouse model of warm hepatic ischemia, and whether this hypothesis could also apply in the setting of human liver transplantation. First, a model of warm hepatic ischemia/reperfusion was used in wild-type and IL-33-deficient mice. Severity of ischemia/reperfusion injury was assessed with ALT and histological analysis. Then, serum IL-33 was measured in a pilot cohort of 40 liver transplant patients. Hemodynamic postreperfusion syndrome, graft dysfunction (assessed by model for early allograft scoring >6), renal failure, and tissue lesions on time-zero biopsies were assessed. In the mouse model, IL-33 was constitutively expressed in the nucleus of endothelial cells, immediately released in response to hepatic pedicle clamping without neosynthesis, and participated in the recruitment of neutrophils and tissue injury on site. The kinetics of IL-33 in liver transplant patients strikingly matched the ones in the animal model, as attested by serum levels reaching a peak immediately after reperfusion, which correlated to clinical outcomes including postreperfusion syndrome, posttransplant renal failure, graft dysfunction, and histological lesions of ischemia/reperfusion injury. IL-33 was an independent factor of graft dysfunction with a cutoff of IL-33 at 73 pg/ml after reperfusion (73% sensitivity, area under the curve of 0.76). Taken together, these findings establish the immediate implication of IL-33 acting as an alarmin in liver I/R injury and provide evidence of its close association with cardinal features of early liver injury-associated disorders in LT patients.


Alarmins/immunology , Interleukin-33/immunology , Liver Transplantation , Liver/pathology , Reperfusion Injury/pathology , Alarmins/metabolism , Animals , Cohort Studies , Humans , Interleukin-33/metabolism , Liver/immunology , Liver/metabolism , Mice , Pilot Projects , Reperfusion Injury/immunology , Reperfusion Injury/metabolism
3.
Nephrol Ther ; 16(6): 388-399, 2020 Nov.
Article Fr | MEDLINE | ID: mdl-32571740

Ischemia-reperfusion injury is an inescapable phenomenon in kidney transplantation. It combines lesional processes of biochemical origin associated with oxydative stress and of immunological origin in connection with the recruitment and activation of innate immunity cells. Histological lesions associate acute tubular necrosis and interstitial œdema, which can progress to interstitial fibrosis. The extent of these lesions depends on donor characteristics (age, expanded criteria donor, etc.) and cold ischemia time. In the short term, ischemia-reperfusion results in delayed recovery of graft function. Cold ischemia time also impacts long-term graft survival. Preclinical models, such as murine and porcine models, have furthered understanding of the pathophysiological mechanisms of ischemia-reperfusion injury. Due to its renal anatomical proximity to humans, the porcine model is relevant to assessment of the molecules administered to a donor or recipient, and also of additives to preservation solutions. Different donor resuscitation and graft perfusion strategies can be studied. In humans, prevention of ischemia-reperfusion injury is a research subject as concerns donor conditioning, additive molecules in preservation solutions, graft reperfusion modalities and choice of the molecules administered to the recipient. Pending significant advances in research, the goal is to achieve the shortest possible cold ischemia time.


Kidney Transplantation , Reperfusion Injury , Animals , Humans , Kidney , Kidney Transplantation/adverse effects , Mice , Organ Preservation , Perfusion , Reperfusion Injury/etiology , Swine
4.
Front Immunol ; 10: 2014, 2019.
Article En | MEDLINE | ID: mdl-31507607

The pyrogenic property being the first activity described, members of the interleukin-1 superfamily (IL-1α, IL-1ß, IL-18, and the newest members: IL-33, IL-36, IL-37, and IL-38) are now known to be involved in several inflammatory diseases such as obesity, atherosclerosis, cancer, viral and parasite infections, and auto-inflammatory syndromes as well as liver diseases. Inflammation processes are keystones of chronic liver diseases, of which the etiology may be viral or toxic, as in alcoholic or non-alcoholic liver diseases. Inflammation is also at stake in acute liver failure involving massive necrosis, and in ischemia-reperfusion injury in the setting of liver transplantation. The role of the IL-1 superfamily of cytokines and receptors in liver diseases can be either protective or pro-inflammatory, depending on timing and the environment. Our review provides an overview of current understanding of the IL-1 family members in liver inflammation, highlighting recent key investigations, and therapeutic perspectives. We have tried to apply the concept of trained immunity to liver diseases, based on the role of the members of the IL-1 superfamily, first of all IL-1ß but also IL-18 and IL-33, in modulating innate lymphoid immunity carried by natural killer cells, innate lymphoid cells or innate T-αß lymphocytes.


Disease Susceptibility , Hepatitis/etiology , Hepatitis/metabolism , Interleukin-1/genetics , Interleukin-1/metabolism , Multigene Family , Animals , Cytokines/genetics , Cytokines/metabolism , Disease Susceptibility/immunology , Genetic Predisposition to Disease , Hepatitis/pathology , Hepatitis/therapy , Humans , Liver Diseases/etiology , Liver Diseases/metabolism , Liver Diseases/pathology , Liver Diseases/therapy , Receptors, Interleukin-1/genetics , Receptors, Interleukin-1/metabolism
5.
Front Immunol ; 9: 2308, 2018.
Article En | MEDLINE | ID: mdl-30374349

Although the contribution of iNKT cells to induction of sterile inflammation is now well-established, the nature of the endogenous compounds released early after cellular stress or damage that drive their activation and recruitment remains poorly understood. More precisely, iNKT cells have not been described as being reactive to endogenous non-protein damage-associated molecular-pattern molecules (DAMPs). A second subset of DAMPs, called alarmins, are tissue-derived nuclear proteins, constitutively expressed at high levels in epithelial barrier tissues and endothelial barriers. These potent immunostimulants, immediately released after tissue damage, include the alarmin IL-33. This factor has aroused interest due to its singular action as an alarmin during infectious, allergic responses and acute tissue injury, and as a cytokine, contributing to the latter resolutive/repair phase of sterile inflammation. IL-33 targets iNKT cells, inducing their recruitment in an inflammatory state, and amplifying their regulatory and effector functions. In the present review, we introduce the new concept of a biological axis of iNKT cells and IL-33, involved in alerting and controlling the immune cells in experimental models of sterile inflammation. This review will focus on acute organ injury models, especially ischemia-reperfusion injury, in the kidneys, liver and lungs, where iNKT cells and IL-33 have been presumed to mediate and/or control the injury mechanisms, and their potential relevance in human pathophysiology.


Inflammation/etiology , Inflammation/metabolism , Natural Killer T-Cells/immunology , Natural Killer T-Cells/metabolism , Alarmins/metabolism , Animals , Biomarkers , Cytokines/metabolism , Humans , Inflammation/pathology , Interleukin-33/metabolism
6.
J Am Soc Nephrol ; 29(4): 1272-1288, 2018 04.
Article En | MEDLINE | ID: mdl-29436517

Inflammation is a prominent feature of ischemia-reperfusion injury (IRI), which is characterized by leukocyte infiltration and renal tubular injury. However, signals that initiate these events remain poorly understood. We examined the role of the nuclear alarmin IL-33 in tissue injury and innate immune response triggered by experimental kidney ischemia-reperfusion. In wild-type mice, we found that IL-33 was constitutively expressed throughout the kidney in peritubular and periglomerular spaces, mainly by microvascular endothelial cells, from which it was released immediately during IRI. Compared with wild-type mice, mice lacking IL-33 (IL-33Gt/Gt) exhibited reductions in early tubular cell injury and subsequent renal infiltration of IFN-γ/IL-17A-producing neutrophils, with preservation of renal functions. This protection associated with decreased renal recruitment of myeloid dendritic cells, natural killer (NK) cells, and invariant natural killer T (iNKT) cells, the latter of which were reported as deleterious in IRI. Increases in the level of circulating IL-12, a key IL-33 cofactor, and the expression of ST2, an IL-33-specific receptor, on the surface of iNKT cells preceded the IL-33- and iNKT cell-dependent phase of neutrophil infiltration. Furthermore, IL-33 directly targeted iNKT cells in vitro, inducing IFN-γ and IL-17A production. We propose that endogenous IL-33 is released as an alarmin and contributes to kidney IRI by promoting iNKT cell recruitment and cytokine production, resulting in neutrophil infiltration and activation at the injury site. Our findings show a novel molecular mediator contributing to innate immune cell recruitment induced by renal ischemia-reperfusion and may provide therapeutic insights into AKI associated with renal transplantation.


Alarmins/physiology , Interleukin-33/physiology , Kidney/blood supply , Reperfusion Injury/metabolism , Alarmins/deficiency , Alarmins/genetics , Animals , Cytokines/biosynthesis , Cytokines/genetics , Dendritic Cells/immunology , Gene Expression Regulation/immunology , Immunity, Innate , Interferon-gamma/biosynthesis , Interferon-gamma/genetics , Interleukin-1 Receptor-Like 1 Protein/deficiency , Interleukin-1 Receptor-Like 1 Protein/physiology , Interleukin-12/blood , Interleukin-17/biosynthesis , Interleukin-17/genetics , Interleukin-33/biosynthesis , Interleukin-33/deficiency , Interleukin-33/genetics , Kidney/immunology , Kidney/metabolism , Kidney/pathology , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Neutrophil Activation , Neutrophils/immunology , Reperfusion Injury/immunology
7.
Front Immunol ; 7: 688, 2016.
Article En | MEDLINE | ID: mdl-28138330

We recently identified a new human subset of NK-like [KIR/NKG2A(+)] CD8(+) T cells with a marked/memory phenotype, high Eomesodermin expression, potent antigen-independent cytotoxic activity, and the capacity to generate IFN-γ rapidly after exposure to pro-inflammatory cytokines. These features support the hypothesis that this new member of the innate T cell family in humans, hereafter referred to as innate CD8(+) T cells, has a role in cancer immune surveillance analogous to invariant natural killer T (iNKT) cells. Here, we report the first quantitative and functional analysis of innate CD8(+) T cells in a physiopathological context in humans, namely chronic myeloid leukemia (CML), a well-characterized myeloproliferative disorder. We have chosen CML based on our previous report that IL-4 production by iNKT cells was deficient in CML patients at diagnosis and considering the recent evidence in mice that IL-4 promotes the generation/differentiation of innate CD8(+) T cells. We found that the pool of innate CD8(+) T cells was severely reduced in the blood of CML patients at diagnosis. Moreover, like iNKT and NK cells, innate CD8(+) T cells were functionally impaired, as attested by their loss of antigen-independent cytotoxic activity and IFN-γ production in response to innate-like stimulation with IL-12 + IL-18. Remarkably, as previously reported for IL-4 production by iNKT cells, both quantitative and functional deficiencies of innate CD8(+) T cells were at least partially corrected in patients having achieved complete cytogenetic remission following tyrosine kinase inhibitor therapy. Finally, direct correlation between the functional potential of innate CD8(+) T and iNKT cells was found when considering all healthy donors and CML patients in diagnosis and remission, in accordance with the iNKT cell-dependent generation of innate CD8(+) T cells reported in mice. All in all, our data demonstrate that CML is associated with deficiencies of innate CD8(+) T cells that are restored upon remission, thereby suggesting their possible contribution to disease control. More generally, our study strongly supports the existence of an innate iNKT/innate CD8(+) T-cell axis in humans and reveals its potential contribution to the restoration of tumor immune surveillance.

8.
Eur J Immunol ; 45(7): 1926-33, 2015 Jul.
Article En | MEDLINE | ID: mdl-25903796

Polyclonal CD8(+) T cells, with a marked innate/memory phenotype, high eomesodermin (Eomes) expression, and the capacity to generate IFN-γ rapidly without prior exposure to antigen, have been described in mice. However, even though a pool of human CD8(+) T cells expressing killer Ig-like receptors (KIRs) was recently documented, the existence of a human equivalent of murine innate/memory CD8(+) T cells remains to be established. Here, we provide evidence for a population of KIR/NKG2A(+) CD8(+) T cells in healthy human adults sharing the same features, namely increased Eomes expression, prompt IFN-γ production in response to innate-like stimulation by IL-12+IL-18, and a potent antigen-independent cytotoxic activity along with a preferential terminally differentiated effector memory phenotype. None of the above functional characteristics applied to the KIR/NKG2A(-) fraction of the Eomes(+) CD8(+) T-cell population, thereby underlining the ability of KIR/NKG2A to distinguish between "innate/memory-like" and "conventional/memory" pools of CD8(+) T cells. Remarkably, KIR/NKG2A(+) Eomes(+) CD8(+) T cells with innate-like functions and a memory/terminally differentiated effector memory phenotype were also identified in human cord blood, suggesting that their development did not depend on cognate antigens. Taken together, our results support the conclusion that CD8(+) T cells co-expressing Eomes and KIR/NKG2A may represent a new, functionally distinct "innate/memory-like" subset in humans.


CD8-Positive T-Lymphocytes/immunology , Immunity, Innate/immunology , Immunologic Memory/immunology , T-Box Domain Proteins/immunology , T-Lymphocyte Subsets/immunology , Adult , Fetal Blood/cytology , Fetal Blood/immunology , Flow Cytometry , Humans , NK Cell Lectin-Like Receptor Subfamily C/immunology
9.
PLoS One ; 9(2): e88742, 2014.
Article En | MEDLINE | ID: mdl-24586382

The endogenous molecules high mobility group box 1 (HMGB1) and interleukin-33 (IL-33) have been identified as alarmins, capable of mediating danger signals during tissue damage. Here, we address their possible role as innate-immune mediators in ischemia-reperfusion injury (IRI) following human kidney transplantation. We analysed serum and urinary HMGB1 and IL-33 levels, all determined by enzyme-linked immunosorbent assay, in a cohort of 26 deceased renal transplant recipients. Urinary HMGB1 and IL-33 levels were significantly increased as soon as 30 min after reperfusion, as compared to those before treatment. Moreover, both serum and urinary IL-33 (but not HMGB1) increase was positively correlated with cold ischemia time, from 30 min to 3 days post-transplantation. In vitro, human umbilical vein endothelial cells subjected to hypoxia conditions released both HMGB-1 and IL-33, while only the latter was further increased upon subsequent re-oxygenation. Finally, we postulate that leukocytes from renal recipient patients are targeted by both HMGB1 and IL-33, as suggested by increased transcription of their respective receptors (TLR2/4 and ST2L) shortly after transplantation. Consistent with this view, we found that iNKT cells, an innate-like T cell subset involved in IRI and targeted by IL-33 but not by HMGB1 was activated 1 hour post-transplantation. Altogether, these results are in keeping with a potential role of IL-33 as an innate-immune mediator during kidney IRI in humans.


HMGB1 Protein/metabolism , Immunity, Innate/immunology , Interleukins/metabolism , Kidney Transplantation/adverse effects , Reperfusion Injury/immunology , Adult , Aged , Cell Hypoxia/physiology , Cohort Studies , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , HMGB1 Protein/blood , HMGB1 Protein/urine , Human Umbilical Vein Endothelial Cells , Humans , Interleukin-33 , Interleukins/blood , Interleukins/urine , Middle Aged , Prospective Studies , Real-Time Polymerase Chain Reaction , Reperfusion Injury/etiology , Statistics, Nonparametric
10.
J Exp Med ; 211(3): 499-513, 2014 Mar 10.
Article En | MEDLINE | ID: mdl-24534191

Netherton syndrome (NS) is a severe genetic skin disease in which absence of a key protease inhibitor causes congenital exfoliative erythroderma, eczematous-like lesions, and atopic manifestations. Several proteases are overactive in NS, including kallikrein-related peptidase (KLK) 5, KLK7, and elastase-2 (ELA2), which are suggested to be part of a proteolytic cascade initiated by KLK5. To address the role of KLK5 in NS, we have generated a new transgenic murine model expressing human KLK5 in the granular layer of the epidermis (Tg-KLK5). Transgene expression resulted in increased proteolytic activity attributable to KLK5 and its downstream targets KLK7, KLK14, and ELA2. Tg-KLK5 mice developed an exfoliative erythroderma with scaling, growth delay, and hair abnormalities. The skin barrier was defective and the stratum corneum was detached through desmosomal cleavage. Importantly, Tg-KLK5 mice displayed cutaneous and systemic hallmarks of severe inflammation and allergy with pruritus. The skin showed enhanced expression of inflammatory cytokines and chemokines, infiltration of immune cells, and markers of Th2/Th17/Th22 T cell responses. Moreover, serum IgE and Tslp levels were elevated. Our study identifies KLK5 as an important contributor to the NS proteolytic cascade and provides a new and viable model for the evaluation of future targeted therapies for NS or related diseases such as atopic dermatitis.


Disease Models, Animal , Kallikreins/metabolism , Netherton Syndrome/genetics , Netherton Syndrome/pathology , Animals , Blotting, Western , Chemokines/metabolism , Cytokines/metabolism , Flow Cytometry , Fluorescent Antibody Technique , Humans , Image Processing, Computer-Assisted , Immunohistochemistry , Kallikreins/genetics , Mice , Mice, Transgenic , Microscopy, Confocal , Microscopy, Electron, Transmission , Real-Time Polymerase Chain Reaction , Skin/metabolism , Skin/pathology , Statistics, Nonparametric , Water Loss, Insensible/physiology
11.
Eur J Immunol ; 42(7): 1870-5, 2012 Jul.
Article En | MEDLINE | ID: mdl-22585600

Chronic myeloid leukemia (CML) is a clonal hematopoietic stem-cell malignancy characterized by the presence of the chimeric BCR-ABL oncoprotein with deregulated tyrosine-kinase (TK) activity. Although conventional T cells are acknowledged as important players in the control of CML, a possible modification of invariant NKT (iNKT) cells, known for their antitumoral activity, has not been established as yet. Here, we showed that the expression of perforin, CD95L, and promyelocytic leukemia zinc finger, a transcription factor required for maintenance of iNKT cell functions, was reduced or suppressed in CML patients at diagnosis, as compared with healthy individuals. The proliferation rate of blood iNKT cells in response to their cognate ligand was likewise diminished. These functional deficiencies were corrected in patients having achieved complete cytogenetic remission following TK inhibitor or IFN-α therapy. iNKT cells from CML patients in the chronic phase did not display increased TK activity, which argued against a direct autonomous action of BCR-ABL. Instead, we found that their anergic status originated from both intrinsic and APC-dependent dysfunctions. Our data demonstrate that chronic phase CML is associated with functional deficiencies of iNKT cells that are restored upon remission. These results suggest a possible contribution to disease control by TK inhibitor therapies.


Interferon-alpha/pharmacology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/immunology , Natural Killer T-Cells/immunology , Protein-Tyrosine Kinases/immunology , Benzamides , Fas Ligand Protein/blood , Flow Cytometry , Humans , Imatinib Mesylate , Kruppel-Like Transcription Factors/blood , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/enzymology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Natural Killer T-Cells/enzymology , Perforin/blood , Piperazines/pharmacology , Promyelocytic Leukemia Zinc Finger Protein , Protein Kinase Inhibitors/pharmacology , Protein-Tyrosine Kinases/antagonists & inhibitors , Pyrimidines/pharmacology
12.
J Invest Dermatol ; 130(12): 2736-42, 2010 Dec.
Article En | MEDLINE | ID: mdl-20703245

Netherton syndrome (NS) is a severe genodermatosis characterized by abnormal scaling and constant atopic manifestations. NS is caused by mutations in SPINK5 (Serine Protease INhibitor Kazal-type 5), which encodes LEKTI (LymphoEpithelial Kazal Type-related Inhibitor). Lack of LEKTI causes stratum corneum detachment secondary to epidermal proteases hyperactivity. Whereas a skin barrier defect is generally regarded as a major cause for atopy, we previously identified a cell-autonomous signaling cascade that triggers pro-Th2 cytokine thymic stromal lymphopoietin (TSLP) production in LEKTI-deficient epidermis. This signaling is initiated by unrestricted kallikrein 5 (KLK5) activity, which directly activates proteinase-activated receptor 2 (PAR2)-mediated expression of TSLP and favors a cutaneous proallergic microenvironment independently of the environment and of the adaptive immune system. To further confirm these results in vivo, we generated Spink5/Par2 double knockout (DKO) mice. At embryonic day 19.5, these mice display a dramatic decrease in TSLP expression, although stratum corneum detachment persists, confirming the role of the KLK5-PAR2 cascade in TSLP-mediated early proallergic signaling. However, deletion of Par2 in adult DKO-grafted skin does not rescue the inflammatory phenotype probably resulting from stratum corneum detachment. We conclude that several mechanisms trigger and maintain the inflammatory phenotype in NS. These include skin barrier impairment, mechanical stress secondary to stratum corneum detachment, as well as protease-induced proinflammatory and proallergic pathways, including PAR2-mediated overexpression of TSLP.


Dermatitis , Netherton Syndrome , Proteinase Inhibitory Proteins, Secretory/genetics , Receptor, PAR-2/genetics , Receptors, Cytokine/metabolism , Age Factors , Animals , Animals, Newborn , Dermatitis/immunology , Dermatitis/metabolism , Dermatitis/pathology , Disease Models, Animal , Epidermis/immunology , Epidermis/metabolism , Epidermis/pathology , Female , Gene Expression Regulation, Developmental , Mice , Mice, Knockout , Mice, Nude , Netherton Syndrome/immunology , Netherton Syndrome/metabolism , Netherton Syndrome/pathology , Pregnancy , Proteinase Inhibitory Proteins, Secretory/metabolism , Receptor, PAR-2/metabolism , Serine Peptidase Inhibitor Kazal-Type 5 , Signal Transduction/physiology
13.
J Clin Invest ; 120(3): 871-82, 2010 Mar.
Article En | MEDLINE | ID: mdl-20179351

The human epidermis serves 2 crucial barrier functions: it protects against water loss and prevents penetration of infectious agents and allergens. The physiology of the epidermis is maintained by a balance of protease and antiprotease activities, as illustrated by the rare genetic skin disease Netherton syndrome (NS), in which impaired inhibition of serine proteases causes severe skin erythema and scaling. Here, utilizing mass spectrometry, we have identified elastase 2 (ELA2), which we believe to be a new epidermal protease that is specifically expressed in the most differentiated layer of living human and mouse epidermis. ELA2 localized to keratohyalin granules, where it was found to directly participate in (pro-)filaggrin processing. Consistent with the observation that ELA2 was hyperactive in skin from NS patients, transgenic mice overexpressing ELA2 in the granular layer of the epidermis displayed abnormal (pro-)filaggrin processing and impaired lipid lamellae structure, which are both observed in NS patients. These anomalies led to dehydration, implicating ELA2 in the skin barrier defect seen in NS patients. Thus, our work identifies ELA2 as a major new epidermal protease involved in essential pathways for skin barrier function. These results highlight the importance of the control of epidermal protease activity in skin homeostasis and designate ELA2 as a major protease driving the pathogenesis of NS.


Epidermis/enzymology , Gene Expression Regulation, Enzymologic , Intermediate Filament Proteins/metabolism , Lipid Metabolism , Netherton Syndrome/enzymology , Serine Endopeptidases/biosynthesis , Animals , Cytoplasmic Granules/enzymology , Cytoplasmic Granules/genetics , Epidermis/pathology , Filaggrin Proteins , Humans , Intermediate Filament Proteins/genetics , Keratins/genetics , Keratins/metabolism , Mice , Mice, Transgenic , Netherton Syndrome/genetics , Netherton Syndrome/pathology , Serine Endopeptidases/genetics
14.
J Exp Med ; 206(5): 1135-47, 2009 May 11.
Article En | MEDLINE | ID: mdl-19414552

Netherton syndrome (NS) is a severe genetic skin disease with constant atopic manifestations that is caused by mutations in the serine protease inhibitor Kazal-type 5 (SPINK5) gene, which encodes the protease inhibitor lymphoepithelial Kazal-type-related inhibitor (LEKTI). Lack of LEKTI causes stratum corneum detachment secondary to epidermal proteases hyperactivity. This skin barrier defect favors allergen absorption and is generally regarded as the underlying cause for atopy in NS. We show for the first time that the pro-Th2 cytokine thymic stromal lymphopoietin (TSLP), the thymus and activation-regulated chemokine, and the macrophage-derived chemokine are overexpressed in LEKTI-deficient epidermis. This is part of an original biological cascade in which unregulated kallikrein (KLK) 5 directly activates proteinase-activated receptor 2 and induces nuclear factor kappaB-mediated overexpression of TSLP, intercellular adhesion molecule 1, tumor necrosis factor alpha, and IL8. This proinflammatory and proallergic pathway is independent of the primary epithelial failure and is activated under basal conditions in NS keratinocytes. This cell-autonomous process is already established in the epidermis of Spink5(-/-) embryos, and the resulting proinflammatory microenvironment leads to eosinophilic and mast cell infiltration in a skin graft model in nude mice. Collectively, these data establish that uncontrolled KLK5 activity in NS epidermis can trigger atopic dermatitis (AD)-like lesions, independently of the environment and the adaptive immune system. They illustrate the crucial role of protease signaling in skin inflammation and point to new therapeutic targets for NS as well as candidate genes for AD and atopy.


Dermatitis, Atopic/genetics , Kallikreins/deficiency , Serpins/genetics , Skin Transplantation , Animals , Dermatitis, Atopic/pathology , Dermatitis, Atopic/surgery , Dermis/pathology , Epidermis/pathology , Inflammation/genetics , Kallikreins/genetics , Keratin-14/genetics , Mice , Mice, Knockout , Mice, Nude , Serine Peptidase Inhibitor Kazal-Type 5 , Serpins/deficiency , Skin/enzymology , Skin/pathology
15.
Neuropsychopharmacology ; 32(5): 1109-21, 2007 May.
Article En | MEDLINE | ID: mdl-17063155

Striatal dopamine D1 receptors (D1R) are coupled to adenylyl cyclase through Galphaolf. Although this pathway is involved in important brain functions, the consequences of quantitative alterations of its components are not known. We explored the biochemical and behavioral responses to cocaine and D-amphetamine (D-amph) in mice with heterozygous mutations of genes encoding D1R and Galphaolf (Drd1a+/- and Gnal+/-), which express decreased levels of the corresponding proteins in the striatum. Dopamine-stimulated cAMP production in vitro and phosphorylation of AMPA receptor GluR1 subunit in response to D-amph in vivo were decreased in Gnal+/-, but not Drd1a+/- mice. Acute locomotor responses to D1 agonist SKF81259, D-amph and cocaine were altered in Gnal+/- mice, and not in Drd1a+/- mice. This haploinsufficiency showed that Galphaolf but not D1R protein levels are limiting for D1R-mediated biochemical and behavioral responses. Gnal+/- mice developed pronounced locomotor sensitization and conditioned locomotor responses after repeated injections of D-amph (2 mg/kg) or cocaine (20 mg/kg). They also developed normal D-amph-conditioned place preference. The D1R/cAMP pathway remained blunted in repeatedly treated Gnal+/- mice. In contrast, D-amph-induced ERK activation was normal in the striatum of these mice, possibly accounting for the normal development of long-lasting behavioral responses to psychostimulants. Our results clearly dissociate biochemical mechanisms involved in acute and delayed behavioral effects of psychostimulants. They identify striatal levels of Galphaolf as a key factor for acute responses to psychostimulants and suggest that quantitative alterations of its expression may alter specific responses to drugs of abuse, or possibly other behavioral responses linked to dopamine function.


Central Nervous System Stimulants/pharmacology , Corpus Striatum/drug effects , Dopamine/metabolism , GTP-Binding Protein alpha Subunits/genetics , Receptors, Dopamine D1/genetics , Adenylyl Cyclases/metabolism , Animals , Cocaine/pharmacology , Conditioning, Psychological/drug effects , Conditioning, Psychological/physiology , Corpus Striatum/metabolism , Corpus Striatum/physiopathology , Cyclic AMP/metabolism , Dextroamphetamine/pharmacology , Dopamine Agonists/pharmacology , Dopamine Uptake Inhibitors/pharmacology , Extracellular Signal-Regulated MAP Kinases/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , GTP-Binding Protein alpha Subunits/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Motor Activity/drug effects , Motor Activity/physiology , Phosphorylation/drug effects , RNA, Messenger/drug effects , RNA, Messenger/metabolism , Receptors, AMPA/drug effects , Receptors, AMPA/metabolism , Receptors, Dopamine D1/metabolism , Signal Transduction/drug effects , Signal Transduction/physiology
...