Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 18 de 18
Filtrer
Plus de filtres











Base de données
Gamme d'année
1.
Am J Physiol Lung Cell Mol Physiol ; 321(2): L416-L428, 2021 08 01.
Article de Anglais | MEDLINE | ID: mdl-34189964

RÉSUMÉ

Pulmonary hypertension (PH) is a devastating disease characterized by progressive elevation of pulmonary vascular resistance, right ventricular failure, and ultimately death. We have shown previously that insulin receptor substrate 2 (IRS2), a molecule highly critical to insulin resistance and metabolism, has an anti-inflammatory role in Th2-skewed lung inflammation and pulmonary vascular remodeling. Here, we investigated the hypothesis that IRS2 has an immunomodulatory role in human and experimental PH. Expression analysis showed that IRS2 was significantly decreased in the pulmonary vasculature of patients with pulmonary arterial hypertension and in rat models of PH. In mice, genetic ablation of IRS2 enhanced the hypoxia-induced signaling pathway of Akt and Forkhead box O1 (FOXO1) in the lung tissue and increased pulmonary vascular muscularization, proliferation, and perivascular macrophage recruitment. Furthermore, mice with homozygous IRS2 gene deletion showed a significant gene dosage-dependent increase in pulmonary vascular remodeling and right ventricular hypertrophy in response to hypoxia. Functional studies with bone marrow-derived macrophages isolated from homozygous IRS2 gene-deleted mice showed that hypoxia exposure led to enhancement of the Akt and ERK signaling pathway followed by increases in the pro-PH macrophage activation markers, vascular endothelial growth factor-A and arginase 1. Our data suggest that IRS2 contributes to anti-inflammatory effects by regulating macrophage activation and recruitment, which may limit the vascular inflammation, remodeling, and right ventricular hypertrophy that are seen in PH pathology. Restoring the IRS2 pathway may be an effective therapeutic approach for the treatment of PH and right heart failure.


Sujet(s)
Hypertension pulmonaire/métabolisme , Hypoxie/métabolisme , Substrats du récepteur à l'insuline/métabolisme , Remodelage vasculaire , Animaux , Modèles animaux de maladie humaine , Femelle , Protéine O1 à motif en tête de fourche/génétique , Protéine O1 à motif en tête de fourche/métabolisme , Humains , Hypertension pulmonaire/génétique , Hypertension pulmonaire/anatomopathologie , Hypoxie/génétique , Hypoxie/anatomopathologie , Inflammation/génétique , Inflammation/métabolisme , Inflammation/anatomopathologie , Substrats du récepteur à l'insuline/génétique , Mâle , Souris , Souris knockout , Protéines de tissu nerveux/génétique , Protéines de tissu nerveux/métabolisme , Protéines proto-oncogènes c-akt/génétique , Protéines proto-oncogènes c-akt/métabolisme , Rats , Rat nude
2.
Front Cardiovasc Med ; 8: 574708, 2021.
Article de Anglais | MEDLINE | ID: mdl-33981729

RÉSUMÉ

Heart (right) failure is the most frequent cause of death in patients with pulmonary arterial hypertension. Although historically, increased right ventricular afterload has been considered the main contributor to right heart failure in such patients, recent evidence has suggested a potential role of load-independent factors. Here, we tested the hypothesis that resistin-like molecule α (RELMα), which has been implicated in the pathogenesis of vascular remodeling in pulmonary artery hypertension, also contributes to cardiac metabolic remodeling, leading to heart failure. Recombinant RELMα (rRELMα) was generated via a Tet-On expression system in the T-REx 293 cell line. Cultured neonatal rat cardiomyocytes were treated with purified rRELMα for 24 h at a dose of 50 nM. Treated cardiomyocytes exhibited decreased mRNA and protein expression of peroxisome proliferator-activated receptor gamma coactivator 1α (PGC-1α) and transcription factors PPARα and ERRα, which regulate mitochondrial fatty acid metabolism, whereas genes that encode for glycolysis-related proteins were significantly upregulated. Cardiomyocytes treated with rRELMα also exhibited a decreased basal respiration, maximal respiration, spare respiratory capacity, ATP-linked OCR, and increased glycolysis, as assessed with a microplate-based cellular respirometry apparatus. Transmission electron microscopy revealed abnormal mitochondrial ultrastructure in cardiomyocytes treated with rRELMα. Our data indicate that RELMα affects cardiac energy metabolism and mitochondrial structure, biogenesis, and function by downregulating the expression of the PGC-1α/PPARα/ERRα axis.

3.
PLoS One ; 15(7): e0235546, 2020.
Article de Anglais | MEDLINE | ID: mdl-32609743

RÉSUMÉ

Resistin and resistin-like molecules are pleiotropic cytokines that are involved in inflammatory diseases. Our previous work suggested that resistin has the potential to be used as a biomarker and therapeutic target for human pulmonary arterial hypertension. However, data are limited on the distribution of resistin in healthy human organs. In this study, we used our newly developed anti-human resistin (hResistin) antibody to immunohistochemically detect the expression, localization, and intracellular/extracellular compartmentalization of hResistin in a full human tissue panel from healthy individuals. The potential cross reactivity of this monoclonal anti-hResistin IgG1 with normal human tissues also was verified. Results showed that hResistin is broadly distributed and principally localized in the cytoplasmic granules of macrophages scattered in the interstitium of most human tissues. Bone marrow hematopoietic precursor cells also exhibited hResistin signals in their cytoplasmic granules. Additionally, hResistin labeling was observed in the cytoplasm of nervous system cells. Notably, the cytokine activity of hResistin was illustrated by positively stained extracellular material in most human tissues. These data indicate that our generated antibody binds to the secreted hResistin and support its potential use for immunotherapy to reduce circulating hResistin levels in human disease. Our findings comprehensively document the basal expression patterns of hResistin protein in normal human tissues, suggest a critical role of this cytokine in normal and pathophysiologic inflammatory processes, and offer key insights for using our antibody in future pharmacokinetic studies and immunotherapeutic strategies.


Sujet(s)
Anticorps monoclonaux/immunologie , Régulation de l'expression des gènes , Résistine/immunologie , Résistine/métabolisme , Espace extracellulaire/métabolisme , Cellules HEK293 , Humains , Immunohistochimie , Espace intracellulaire/métabolisme , Spécificité d'organe , Transport des protéines
4.
J Immunol ; 205(5): 1393-1405, 2020 09 01.
Article de Anglais | MEDLINE | ID: mdl-32727891

RÉSUMÉ

Intestinal ischemia/reperfusion (I/R) injury is a life-threatening complication that leads to inflammation and remote organ damage. The NLRP3 inflammasome regulates the caspase-1-dependent release of IL-1ß, an early mediator of inflammation after I/R injury. In this study, we investigated the role of the NLRP3 inflammasome in mice with intestinal I/R injury. Deficiency of NLRP3, ASC, caspase-1/11, or IL-1ß prolonged survival after intestinal I/R injury, but neither NLRP3 nor caspase-1/11 deficiency affected intestinal inflammation. Intestinal I/R injury caused acute lung injury (ALI) characterized by inflammation, reactive oxygen species generation, and vascular permeability, which was markedly improved by NLRP3 deficiency. Bone marrow chimeric experiments showed that NLRP3 in non-bone marrow-derived cells was the main contributor to development of intestinal I/R-induced ALI. The NLRP3 inflammasome in lung vascular endothelial cells is thought to be important to lung vascular permeability. Using mass spectrometry, we identified intestinal I/R-derived lipid mediators that enhanced NLRP3 inflammasome activation in lung vascular endothelial cells. Finally, we confirmed that serum levels of these lipid mediators were elevated in patients with intestinal ischemia. To our knowledge, these findings provide new insights into the mechanism underlying intestinal I/R-induced ALI and suggest that endothelial NLRP3 inflammasome-driven IL-1ß is a novel potential target for treating and preventing this disorder.


Sujet(s)
Lésion pulmonaire aigüe/métabolisme , Cellules endothéliales/métabolisme , Inflammasomes/métabolisme , Poumon/métabolisme , Protéine-3 de la famille des NLR contenant un domaine pyrine/métabolisme , Lésion d'ischémie-reperfusion/métabolisme , Animaux , Caspase-1/métabolisme , Inflammation/métabolisme , Interleukine-1 bêta/métabolisme , Mâle , Souris , Souris de lignée C57BL
5.
J Immunol ; 203(11): 2862-2871, 2019 12 01.
Article de Anglais | MEDLINE | ID: mdl-31611261

RÉSUMÉ

Pulmonary hypertension (PH) is a debilitating disease characterized by remodeling of the lung vasculature. In rodents, resistin-like molecule-α (RELMα, also known as HIMF or FIZZ1) can induce PH, but the signaling mechanisms are still unclear. In this study, we used human lung samples and a hypoxia-induced mouse model of PH. We found that the human homolog of RELMα, human (h) resistin, is upregulated in macrophage-like inflammatory cells from lung tissues of patients with idiopathic PH. Additionally, at PH onset in the mouse model, we observed RELMα-dependent lung accumulation of macrophages that expressed high levels of the key damage-associated molecular pattern (DAMP) molecule high-mobility group box 1 (HMGB1) and its receptor for advanced glycation end products (RAGE). In vitro, RELMα/hresistin-induced macrophage-specific HMGB1/RAGE expression and facilitated HMGB1 nucleus-to-cytoplasm translocation and extracellular secretion. Mechanistically, hresistin promoted HMGB1 posttranslational lysine acetylation by preserving the NAD+-dependent deacetylase sirtuin (Sirt) 1 in human macrophages. Notably, the hresistin-stimulated macrophages promoted apoptosis-resistant proliferation of human pulmonary artery smooth muscle cells in an HMGB1/RAGE-dependent manner. In the mouse model, RELMα also suppressed the Sirt1 signal in pulmonary macrophages in the early posthypoxic period. Notably, recruited macrophages in the lungs of these mice carried the RELMα binding partner Bruton tyrosine kinase (BTK). hResistin also mediated the migration of human macrophages by activating BTK in vitro. Collectively, these data reveal a vascular-immune cellular interaction in the early PH stage and suggest that targeting RELMα/DAMP-driven macrophages may offer a promising strategy to treat PH and other related vascular inflammatory diseases.


Sujet(s)
Hypertension pulmonaire/immunologie , Protéines et peptides de signalisation intercellulaire/immunologie , Macrophages alvéolaires/immunologie , Artère pulmonaire/immunologie , Remodelage vasculaire/immunologie , Adolescent , Animaux , Cellules cultivées , Humains , Hypertension pulmonaire/anatomopathologie , Protéines et peptides de signalisation intercellulaire/déficit , Macrophages alvéolaires/anatomopathologie , Souris , Souris de lignée C57BL , Souris knockout , Artère pulmonaire/anatomopathologie
6.
Arterioscler Thromb Vasc Biol ; 39(12): 2505-2519, 2019 12.
Article de Anglais | MEDLINE | ID: mdl-31597444

RÉSUMÉ

OBJECTIVE: HIMF (hypoxia-induced mitogenic factor; also known as FIZZ1 [found in inflammatory zone-1] or RELM [resistin-like molecule-α]) is an etiological factor of pulmonary hypertension (PH) in rodents, but its underlying mechanism is unclear. We investigated the immunomodulatory properties of HIMF signaling in PH pathogenesis. Approach and Results: Gene-modified mice that lacked HIMF (KO [knockout]) or overexpressed HIMF human homolog resistin (hResistin) were used for in vivo experiments. The pro-PH role of HIMF was verified in HIMF-KO mice exposed to chronic hypoxia or sugen/hypoxia. Mechanistically, HIMF/hResistin activation triggered the HMGB1 (high mobility group box 1) pathway and RAGE (receptor for advanced glycation end products) in pulmonary endothelial cells (ECs) of hypoxic mouse lungs in vivo and in human pulmonary microvascular ECs in vitro. Treatment with conditioned medium from hResistin-stimulated human pulmonary microvascular ECs induced an autophagic response, BMPR2 (bone morphogenetic protein receptor 2) defects, and subsequent apoptosis-resistant proliferation in human pulmonary artery (vascular) smooth muscle cells in an HMGB1-dependent manner. These effects were confirmed in ECs and smooth muscle cells isolated from pulmonary arteries of patients with idiopathic PH. HIMF/HMGB1/RAGE-mediated autophagy and BMPR2 impairment were also observed in pulmonary artery (vascular) smooth muscle cells of hypoxic mice, effects perhaps related to FoxO1 (forkhead box O1) dampening by HIMF. Experiments in EC-specific hResistin-overexpressing transgenic mice confirmed that EC-derived HMGB1 mediated the hResistin-driven pulmonary vascular remodeling and PH. CONCLUSIONS: In HIMF-induced PH, HMGB1-RAGE signaling is pivotal for mediating EC-smooth muscle cell crosstalk. The humanized mouse data further support clinical implications for the HIMF/HMGB1 signaling axis and indicate that hResistin and its downstream pathway may constitute targets for the development of novel anti-PH therapeutics in humans.


Sujet(s)
Cellules endothéliales/métabolisme , Régulation de l'expression des gènes , Protéine HMGB1/génétique , Hypertension pulmonaire/génétique , Protéines et peptides de signalisation intercellulaire/génétique , Muscles lisses vasculaires/métabolisme , Animaux , Autophagie , Lignée cellulaire , Modèles animaux de maladie humaine , Cellules endothéliales/anatomopathologie , Femelle , Protéine HMGB1/biosynthèse , Humains , Hypertension pulmonaire/anatomopathologie , Hypertension pulmonaire/physiopathologie , Protéines et peptides de signalisation intercellulaire/biosynthèse , Mâle , Souris , Souris de lignée C57BL , Souris transgéniques , Muscles lisses vasculaires/anatomopathologie , Artère pulmonaire/métabolisme , Artère pulmonaire/anatomopathologie , Artère pulmonaire/physiopathologie , Rats , Rat Sprague-Dawley , Remodelage vasculaire
7.
Sci Signal ; 9(433): ra63, 2016 06 21.
Article de Anglais | MEDLINE | ID: mdl-27330190

RÉSUMÉ

Insulin receptor substrate 2 (IRS2) is an adaptor protein that becomes tyrosine-phosphorylated in response to the cytokines interleukin-4 (IL-4) and IL-13, which results in activation of the phosphoinositide 3-kinase (PI3K)-Akt pathway. IL-4 and IL-13 contribute to allergic lung inflammation. To examine the role of IRS2 in allergic disease, we evaluated the responses of IRS2-deficient (IRS2(-/-)) mice. Unexpectedly, loss of IRS2 resulted in a substantial increase in the expression of a subset of genes associated with the generation of alternatively activated macrophages (AAMs) in response to IL-4 or IL-13 in vitro. AAMs secrete factors that enhance allergic responses and promote airway remodeling. Moreover, compared to IRS2(+/+) mice, IRS2(+/-) and IRS2(-/-) mice developed enhanced pulmonary inflammation, accumulated eosinophils and AAMs, and exhibited airway and vascular remodeling upon allergen stimulation, responses that partially depended on macrophage-intrinsic IRS2 signaling. Both in unstimulated and IL-4-stimulated macrophages, lack of IRS2 enhanced phosphorylation of Akt and ribosomal S6 protein. Thus, we identified a critical inhibitory loop downstream of IRS2, demonstrating an unanticipated and previously unrecognized role for IRS2 in suppressing allergic lung inflammation and remodeling.


Sujet(s)
Asthme/immunologie , Substrats du récepteur à l'insuline/immunologie , Poumon/immunologie , Activation des macrophages , Macrophages/immunologie , Remodelage des voies aériennes/effets des médicaments et des substances chimiques , Remodelage des voies aériennes/génétique , Remodelage des voies aériennes/immunologie , Animaux , Asthme/induit chimiquement , Asthme/génétique , Asthme/anatomopathologie , Substrats du récepteur à l'insuline/génétique , Poumon/anatomopathologie , Macrophages/anatomopathologie , Souris , Souris knockout , Protéines proto-oncogènes c-akt/génétique , Protéines proto-oncogènes c-akt/immunologie
8.
Arterioscler Thromb Vasc Biol ; 36(1): 134-44, 2016 Jan.
Article de Anglais | MEDLINE | ID: mdl-26586659

RÉSUMÉ

OBJECTIVE: Pulmonary hypertension (PH) is characterized by progressive elevation of pulmonary vascular resistance, right ventricular failure, and ultimately death. We have shown that in rodents, hypoxia-induced mitogenic factor (HIMF; also known as FIZZ1 or resistin-like molecule-ß) causes PH by initiating lung vascular inflammation. We hypothesized that hypoxia-inducible factor-1 (HIF-1) is a critical downstream signal mediator of HIMF during PH development. APPROACH AND RESULTS: In this study, we compared the degree of HIMF-induced pulmonary vascular remodeling and PH development in wild-type (HIF-1α(+/+)) and HIF-1α heterozygous null (HIF-1α(+/-)) mice. HIMF-induced PH was significantly diminished in HIF-1α(+/-) mice and was accompanied by a dysregulated vascular endothelial growth factor-A-vascular endothelial growth factor receptor 2 pathway. HIF-1α was critical for bone marrow-derived cell migration and vascular tube formation in response to HIMF. Furthermore, HIMF and its human homolog, resistin-like molecule-ß, significantly increased interleukin (IL)-6 in macrophages and lung resident cells through a mechanism dependent on HIF-1α and, at least to some extent, on nuclear factor κB. CONCLUSIONS: Our results suggest that HIF-1α is a critical downstream transcription factor for HIMF-induced pulmonary vascular remodeling and PH development. Importantly, both HIMF and human resistin-like molecule-ß significantly increased IL-6 in lung resident cells and increased perivascular accumulation of IL-6-expressing macrophages in the lungs of mice. These data suggest that HIMF can induce HIF-1, vascular endothelial growth factor-A, and interleukin-6, which are critical mediators of both hypoxic inflammation and PH pathophysiology.


Sujet(s)
Hypertension pulmonaire/métabolisme , Sous-unité alpha du facteur-1 induit par l'hypoxie/métabolisme , Protéines et peptides de signalisation intercellulaire , Macrophages/métabolisme , Artère pulmonaire/métabolisme , Remodelage vasculaire , Animaux , Apoptose , Transplantation de moelle osseuse , Mouvement cellulaire , Cellules cultivées , Modèles animaux de maladie humaine , Cellules épithéliales/métabolisme , Cellules épithéliales/anatomopathologie , Femelle , Fibroblastes/métabolisme , Génotype , Hémodynamique , Humains , Hypertension pulmonaire/induit chimiquement , Hypertension pulmonaire/génétique , Hypertension pulmonaire/physiopathologie , Hypertension pulmonaire/prévention et contrôle , Sous-unité alpha du facteur-1 induit par l'hypoxie/déficit , Sous-unité alpha du facteur-1 induit par l'hypoxie/génétique , Techniques in vitro , Médiateurs de l'inflammation/métabolisme , Protéines et peptides de signalisation intercellulaire/métabolisme , Interleukine-6/métabolisme , Macrophages/anatomopathologie , Mâle , Souris de lignée C57BL , Souris knockout , Techniques de culture d'organes , Phénotype , Artère pulmonaire/anatomopathologie , Artère pulmonaire/physiopathologie , Transduction du signal , Facteur de croissance endothéliale vasculaire de type A/métabolisme , Récepteur-2 au facteur croissance endothéliale vasculaire/métabolisme
9.
Am J Respir Cell Mol Biol ; 53(3): 303-13, 2015 Sep.
Article de Anglais | MEDLINE | ID: mdl-25569618

RÉSUMÉ

Resistin-like molecule α (RELMα) has mitogenic, angiogenic, vasoconstrictive, and chemokine-like properties and is highly relevant in lung pathology. Here, we used RELMα knockout (Retnla(-/-)) mice to investigate the role of RELMα in pulmonary vascular remodeling after intermittent ovalbumin (OVA) challenge. We compared saline- and OVA-exposed wild-type (WT) mice and found that OVA induced significant increases in right ventricular systolic pressure, cardiac hypertrophy, pulmonary vascular remodeling of intra-alveolar arteries, goblet cell hyperplasia in airway epithelium, and intensive lung inflammation, especially perivascular inflammation. Genetic ablation of Retnla prevented the OVA-induced increase in pulmonary pressure and cardiac hypertrophy seen in WT mice. Histological analysis showed that Retnla(-/-) mice exhibited less vessel muscularization, less perivascular inflammation, reduced medial thickness of intra-alveolar vessels, and fewer goblet cells in upper airway epithelium (250-600 µm) than did WT animals after OVA challenge. Gene expression profiles showed that genes associated with vascular remodeling, including those related to muscle protein, contractile fibers, and actin cytoskeleton, were expressed at a lower level in OVA-challenged Retnla(-/-) mice than in similarly treated WT mice. In addition, bronchoalveolar lavage from OVA-challenged Retnla(-/-) mice had lower levels of cytokines, such as IL-1ß, -1 receptor antagonist, and -16, chemokine (C-X-C motif) ligand 1, -2, -9, -10, and -13, monocyte chemoattractant protein-1, macrophage colony-stimulating factor, TIMP metallopeptidase inhibitor-1, and triggering receptor expressed on myeloid cells-1, than did that from WT mice when analyzed by cytokine array dot blots. Retnla knockout inhibited the OVA-induced T helper 17 response but not the T helper 2 response. Altogether, our results suggest that RELMα is involved in immune response-induced pulmonary vascular remodeling and the associated increase in inflammation typically observed after OVA challenge.


Sujet(s)
Hypertension pulmonaire/métabolisme , Protéines et peptides de signalisation intercellulaire/métabolisme , Remodelage vasculaire/immunologie , Allergènes/immunologie , Animaux , Cytokines/métabolisme , Hypertension pulmonaire/immunologie , Hypertension pulmonaire/physiopathologie , Protéines et peptides de signalisation intercellulaire/génétique , Poumon/immunologie , Poumon/métabolisme , Mâle , Souris de lignée BALB C , Souris knockout , Ovalbumine/immunologie
10.
Am J Physiol Lung Cell Mol Physiol ; 306(12): L1090-103, 2014 Jun 15.
Article de Anglais | MEDLINE | ID: mdl-24793164

RÉSUMÉ

Pulmonary hypertension (PH) is characterized by elevated pulmonary artery pressure that leads to progressive right heart failure and ultimately death. Injury to endothelium and consequent wound repair cascades have been suggested to trigger pulmonary vascular remodeling, such as that observed during PH. The relationship between injury to endothelium and disease pathogenesis in this disorder remains poorly understood. We and others have shown that, in mice, hypoxia-induced mitogenic factor (HIMF, also known as FIZZ1 or RELMα) plays a critical role in the pathogenesis of lung inflammation and the development of PH. In this study, we dissected the mechanism by which HIMF and its human homolog resistin (hRETN) induce pulmonary endothelial cell (EC) apoptosis and subsequent lung inflammation-mediated PH, which exhibits many of the hallmarks of the human disease. Systemic administration of HIMF caused increases in EC apoptosis and interleukin (IL)-4-dependent vascular inflammatory marker expression in mouse lung during the early inflammation phase. In vitro, HIMF, hRETN, and IL-4 activated pulmonary microvascular ECs (PMVECs) by increasing angiopoietin-2 expression and induced PMVEC apoptosis. In addition, the conditioned medium from hRETN-treated ECs had elevated levels of endothelin-1 and caused significant increases in pulmonary vascular smooth muscle cell proliferation. Last, HIMF treatment caused development of PH that was characterized by pulmonary vascular remodeling and right heart failure in wild-type mice but not in IL-4 knockout mice. These data suggest that HIMF contributes to activation of vascular inflammation at least in part by inducing EC apoptosis in the lung. These events lead to subsequent PH.


Sujet(s)
Apoptose/physiologie , Cellules endothéliales/métabolisme , Hypertension pulmonaire/métabolisme , Hypoxie/métabolisme , Protéines et peptides de signalisation intercellulaire/métabolisme , Interleukine-4/métabolisme , Animaux , Prolifération cellulaire , Modèles animaux de maladie humaine , Cellules endothéliales/cytologie , Hypertension pulmonaire/anatomopathologie , Interleukine-4/génétique , Poumon/métabolisme , Poumon/anatomopathologie , Mâle , Souris , Souris de lignée C57BL , Pneumopathie infectieuse/métabolisme , Pneumopathie infectieuse/anatomopathologie
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE