Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 53
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Circulation ; 142(2): 161-174, 2020 07 14.
Article in English | MEDLINE | ID: mdl-32264695

ABSTRACT

BACKGROUND: The cyclic AMP (adenosine monophosphate; cAMP)-hydrolyzing protein PDE4B (phosphodiesterase 4B) is a key negative regulator of cardiac ß-adrenergic receptor stimulation. PDE4B deficiency leads to abnormal Ca2+ handling and PDE4B is decreased in pressure overload hypertrophy, suggesting that increasing PDE4B in the heart is beneficial in heart failure. METHODS: We measured PDE4B expression in human cardiac tissues and developed 2 transgenic mouse lines with cardiomyocyte-specific overexpression of PDE4B and an adeno-associated virus serotype 9 encoding PDE4B. Myocardial structure and function were evaluated by echocardiography, ECG, and in Langendorff-perfused hearts. Also, cAMP and PKA (cAMP dependent protein kinase) activity were monitored by Förster resonance energy transfer, L-type Ca2+ current by whole-cell patch-clamp, and cardiomyocyte shortening and Ca2+ transients with an Ionoptix system. Heart failure was induced by 2 weeks infusion of isoproterenol or transverse aortic constriction. Cardiac remodeling was evaluated by serial echocardiography, morphometric analysis, and histology. RESULTS: PDE4B protein was decreased in human failing hearts. The first PDE4B-transgenic mouse line (TG15) had a ≈15-fold increase in cardiac cAMP-PDE activity and a ≈30% decrease in cAMP content and fractional shortening associated with a mild cardiac hypertrophy that resorbed with age. Basal ex vivo myocardial function was unchanged, but ß-adrenergic receptor stimulation of cardiac inotropy, cAMP, PKA, L-type Ca2+ current, Ca2+ transients, and cell contraction were blunted. Endurance capacity and life expectancy were normal. Moreover, these mice were protected from systolic dysfunction, hypertrophy, lung congestion, and fibrosis induced by chronic isoproterenol treatment. In the second PDE4B-transgenic mouse line (TG50), markedly higher PDE4B overexpression, resulting in a ≈50-fold increase in cardiac cAMP-PDE activity caused a ≈50% decrease in fractional shortening, hypertrophy, dilatation, and premature death. In contrast, mice injected with adeno-associated virus serotype 9 encoding PDE4B (1012 viral particles/mouse) had a ≈50% increase in cardiac cAMP-PDE activity, which did not modify basal cardiac function but efficiently prevented systolic dysfunction, apoptosis, and fibrosis, while attenuating hypertrophy induced by chronic isoproterenol infusion. Similarly, adeno-associated virus serotype 9 encoding PDE4B slowed contractile deterioration, attenuated hypertrophy and lung congestion, and prevented apoptosis and fibrotic remodeling in transverse aortic constriction. CONCLUSIONS: Our results indicate that a moderate increase in PDE4B is cardioprotective and suggest that cardiac gene therapy with PDE4B might constitute a new promising approach to treat heart failure.


Subject(s)
Cyclic Nucleotide Phosphodiesterases, Type 4/genetics , Gene Expression , Heart Failure/etiology , Myocardium/metabolism , Ventricular Remodeling/genetics , Adrenergic beta-Agonists/pharmacology , Animals , Cyclic AMP/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 4/metabolism , Disease Models, Animal , Disease Susceptibility , Genetic Therapy , Genetic Vectors/genetics , Heart Failure/diagnosis , Heart Failure/drug therapy , Heart Failure/metabolism , Heart Function Tests , Humans , Isoproterenol/pharmacology , Mice , Mice, Transgenic , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Phenotype , Receptors, Adrenergic, beta/metabolism , Transduction, Genetic , Ventricular Remodeling/drug effects
2.
J Cell Mol Med ; 20(4): 655-65, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26773707

ABSTRACT

Similar to other adult tissue stem/progenitor cells, bone marrow mesenchymal stem/stromal cells (BM MSCs) exhibit heterogeneity at the phenotypic level and in terms of proliferation and differentiation potential. In this study such a heterogeneity was reflected by the CD200 protein. We thus characterized CD200(pos) cells sorted from whole BM MSC cultures and we investigated the molecular mechanisms regulating CD200 expression. After sorting, measurement of lineage markers showed that the osteoblastic genes RUNX2 and DLX5 were up-regulated in CD200(pos) cells compared to CD200(neg) fraction. At the functional level, CD200(pos) cells were prone to mineralize the extra-cellular matrix in vitro after sole addition of phosphates. In addition, osteogenic cues generated by bone morphogenetic protein 4 (BMP4) or BMP7 strongly induced CD200 expression. These data suggest that CD200 expression is related to commitment/differentiation towards the osteoblastic lineage. Immunohistochemistry of trephine bone marrow biopsies further corroborates the osteoblastic fate of CD200(pos) cells. However, when dexamethasone was used to direct osteogenic differentiation in vitro, CD200 was consistently down-regulated. As dexamethasone has anti-inflammatory properties, we assessed the effects of different immunological stimuli on CD200 expression. The pro-inflammatory cytokines interleukin-1ß and tumour necrosis factor-α increased CD200 membrane expression but down-regulated osteoblastic gene expression suggesting an additional regulatory pathway of CD200 expression. Surprisingly, whatever the context, i.e. pro-inflammatory or pro-osteogenic, CD200 expression was down-regulated when nuclear-factor (NF)-κB was inhibited by chemical or adenoviral agents. In conclusion, CD200 expression by cultured BM MSCs can be induced by both osteogenic and pro-inflammatory cytokines through the same pathway: NF-κB.


Subject(s)
Antigens, CD/genetics , Bone Marrow Cells/drug effects , Mesenchymal Stem Cells/drug effects , NF-kappa B/genetics , Osteoblasts/drug effects , Adult , Antigens, CD/metabolism , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Bone Morphogenetic Protein 4/pharmacology , Bone Morphogenetic Protein 7/pharmacology , Cell Differentiation/drug effects , Cell Lineage/drug effects , Cell Proliferation/drug effects , Core Binding Factor Alpha 1 Subunit/genetics , Core Binding Factor Alpha 1 Subunit/metabolism , Dexamethasone/pharmacology , Extracellular Matrix/metabolism , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Interleukin-1beta/pharmacology , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , NF-kappa B/metabolism , Osteoblasts/cytology , Osteoblasts/metabolism , Phosphates/pharmacology , Primary Cell Culture , Transcription Factors/genetics , Transcription Factors/metabolism , Tumor Necrosis Factor-alpha/pharmacology
3.
Circulation ; 131(4): 390-400; discussion 400, 2015 Jan 27.
Article in English | MEDLINE | ID: mdl-25369805

ABSTRACT

BACKGROUND: Cardiac hypertrophy is an early hallmark during the clinical course of heart failure and is regulated by various signaling pathways. However, the molecular mechanisms that negatively regulate these signal transduction pathways remain poorly understood. METHODS AND RESULTS: Here, we characterized Carabin, a protein expressed in cardiomyocytes that was downregulated in cardiac hypertrophy and human heart failure. Four weeks after transverse aortic constriction, Carabin-deficient (Carabin(-/-)) mice developed exaggerated cardiac hypertrophy and displayed a strong decrease in fractional shortening (14.6±1.6% versus 27.6±1.4% in wild type plus transverse aortic constriction mice; P<0.0001). Conversely, compensation of Carabin loss through a cardiotropic adeno-associated viral vector encoding Carabin prevented transverse aortic constriction-induced cardiac hypertrophy with preserved fractional shortening (39.9±1.2% versus 25.9±2.6% in control plus transverse aortic constriction mice; P<0.0001). Carabin also conferred protection against adrenergic receptor-induced hypertrophy in isolated cardiomyocytes. Mechanistically, Carabin carries out a tripartite suppressive function. Indeed, Carabin, through its calcineurin-interacting site and Ras/Rab GTPase-activating protein domain, functions as an endogenous inhibitor of calcineurin and Ras/extracellular signal-regulated kinase prohypertrophic signaling. Moreover, Carabin reduced Ca(2+)/calmodulin-dependent protein kinase II activation and prevented nuclear export of histone deacetylase 4 after adrenergic stimulation or myocardial pressure overload. Finally, we showed that Carabin Ras-GTPase-activating protein domain and calcineurin-interacting domain were both involved in the antihypertrophic action of Carabin. CONCLUSIONS: Our study identifies Carabin as a negative regulator of key prohypertrophic signaling molecules, calcineurin, Ras, and Ca(2+)/calmodulin-dependent protein kinase II and implicates Carabin in the development of cardiac hypertrophy and failure.


Subject(s)
Calcineurin/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Cardiomegaly/metabolism , Cardiomegaly/prevention & control , GTPase-Activating Proteins/biosynthesis , Genes, ras/physiology , Animals , Calcium-Calmodulin-Dependent Protein Kinase Type 2/antagonists & inhibitors , Cells, Cultured , Female , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocytes, Cardiac/metabolism , Rats , Signal Transduction/physiology
4.
Hum Mol Genet ; 23(5): 1163-74, 2014 Mar 01.
Article in English | MEDLINE | ID: mdl-24122441

ABSTRACT

Carney complex (CNC) is a hereditary disease associating cardiac myxoma, spotty skin pigmentation and endocrine overactivity. CNC is caused by inactivating mutations in the PRKAR1A gene encoding PKA type I alpha regulatory subunit (RIα). Although PKA activity is enhanced in CNC, the mechanisms linking PKA dysregulation to endocrine tumorigenesis are poorly understood. In this study, we used Förster resonance energy transfer (FRET)-based sensors for cAMP and PKA activity to define the role of RIα in the spatiotemporal organization of the cAMP/PKA pathway. RIα knockdown in HEK293 cells increased basal as well as forskolin or prostaglandin E1 (PGE1)-stimulated total cellular PKA activity as reported by western blots of endogenous PKA targets and the FRET-based global PKA activity reporter, AKAR3. Using variants of AKAR3 targeted to subcellular compartments, we identified similar increases in the response to PGE1 in the cytoplasm and at the outer mitochondrial membrane. In contrast, at the plasma membrane, the response to PGE1 was decreased along with an increase in basal FRET ratio. These results were confirmed by western blot analysis of basal and PGE1-induced phosphorylation of membrane-associated vasodilator-stimulated phosphoprotein. Similar differences were observed between the cytoplasm and the plasma membrane in human adrenal cells carrying a RIα inactivating mutation. RIα inactivation also increased cAMP in the cytoplasm, at the outer mitochondrial membrane and at the plasma membrane, as reported by targeted versions of the cAMP indicator Epac1-camps. These results show that RIα inactivation leads to multiple, compartment-specific alterations of the cAMP/PKA pathway revealing new aspects of signaling dysregulation in tumorigenesis.


Subject(s)
Cyclic AMP-Dependent Protein Kinase RIalpha Subunit/genetics , Cyclic AMP-Dependent Protein Kinases/metabolism , Cyclic AMP/metabolism , Alprostadil/pharmacology , Carney Complex/genetics , Carney Complex/metabolism , Cell Membrane/metabolism , Colforsin/pharmacology , Cyclic AMP-Dependent Protein Kinase RIalpha Subunit/metabolism , Enzyme Activation/drug effects , Enzyme Activation/genetics , Gene Silencing , HEK293 Cells , Humans , Intracellular Space/metabolism , Protein Transport , RNA Interference , Signal Transduction
5.
Aesthet Surg J ; 36(5): 609-18, 2016 May.
Article in English | MEDLINE | ID: mdl-26530477

ABSTRACT

BACKGROUND: Liposuction is a very popular technique in plastic surgery that allows for the taking adipose tissue (AT) on large surfaces with little risk of morbidity. Although liposuction was previously shown to preserve large perforator vessels, little is known about the effects of liposuction on the microvasculature network. OBJECTIVES: The aim of this study was to analyze the effect of liposuction on the preservation of microvessels at tissue and cellular levels by flow cytometry and confocal microscopy following abdominoplasty procedure. METHODS: Percentage of endothelial cells in AT from liposuction and en bloc AT was determined by multicolor flow cytometry. Moreover, vessel density and adipocyte content were analyzed in situ in 3 different types of AT (en bloc, from liposuction, and residual AT after liposuction) by confocal microscopy. RESULTS: Flow cytometric analysis showed that en bloc AT contained 30.6% ± 12.9% and AT from liposuction 21.6% ± 9.9% of endothelial cells (CD31(pos)/CD45(neg)/CD235a(neg)/CD11b(neg)) (P = .009). Moreover, analysis of paired AT from the same patients (n = 5) confirmed a lower percentage of endothelial cells in AT from liposuction compared to en bloc AT (17.7% ± 4.5% vs 21.9% ± 3.3%, P = .031). Likewise, confocal microscopy showed that en bloc AT contained 8.2% ± 6.3%, AT from liposuction only 1.6% ± 1.0% (P < .0001), and AT after liposuction 8.9% ± 4.1% (P = .111) of CD31(pos) vessels. Conversely, adipocyte content was 39.5% ± 14.5% in the en bloc AT, 45% ± 18.4% in AT from liposuction (P = .390), and 18.8 ± 14.8% in AT after liposuction (P = .011). CONCLUSIONS: For the first time, we demonstrate that liposuction preserves the microvascular network. Indeed, a low percentage of endothelial cells was found in AT from liposuction and we confirm the persistence of microvessels in the tissue after liposuction.


Subject(s)
Adipocytes/physiology , Adipose Tissue/cytology , Endothelial Cells/physiology , Lipectomy/methods , Microvessels/physiology , Abdominoplasty/methods , Adipose Tissue/pathology , Adult , Female , Flow Cytometry , Humans , Male , Microscopy, Confocal , Microvessels/diagnostic imaging , Microvessels/surgery , Middle Aged
6.
Blood ; 121(9): e57-69, 2013 Feb 28.
Article in English | MEDLINE | ID: mdl-23293084

ABSTRACT

The molecular repertoire of macrophages in health and disease can provide novel biomarkers for diagnosis, prognosis, and treatment. Th2-IL-4­activated macrophages (M2) have been associated with important diseases in mice, yet no specific markers are available for their detection in human tissues. Although mouse models are widely used for macrophage research, translation to the human can be problematic and the human macrophage system remains poorly described. In the present study, we analyzed and compared the transcriptome and proteome of human and murine macrophages under resting conditions (M0) and after IL-4 activation (M2). We provide a resource for tools enabling macrophage detection in human tissues by identifying a set of 87 macrophage-related genes. Furthermore, we extend current understanding of M2 activation in different species and identify Transglutaminase 2 as a conserved M2 marker that is highly expressed by human macrophages and monocytes in the prototypic Th2 pathology asthma.


Subject(s)
Interleukin-4/pharmacology , Macrophage Activation/genetics , Macrophages/drug effects , Macrophages/metabolism , Transcriptome , Animals , Cell Proliferation/drug effects , Cells, Cultured , Cluster Analysis , Gene Expression Profiling , Humans , Macrophage Activation/drug effects , Macrophages/immunology , Macrophages/physiology , Mice , Mice, Inbred C57BL , Models, Biological , Proteome/analysis , Proteome/drug effects , Species Specificity
7.
Blood ; 117(4): 1319-28, 2011 Jan 27.
Article in English | MEDLINE | ID: mdl-21098741

ABSTRACT

Phagocytic and pathogen sensing receptors are responsible for particle uptake and inflammation. It is unclear how these receptors' systems influence each other's function to shape an innate response. The class-A scavenger receptors SR-A (scavenger receptor A) and MARCO (macrophage receptor with collagenous structure) are 2 well-characterized phagocytic receptors that are unable to initiate inflammatory responses by themselves, yet are implicated in the pathogenesis of various inflammatory disorders. However, the mechanism for such an apparent discrepancy is still unclear. We utilized SR-A(-/-), MARCO(-/-), and SR-A(-/-)-MARCO(-/-) mice, along with microbe-derived, environmental, and synthetic polyanions to assess the inflammatory responses following combinatorial ligation of SR-A/MARCO and selected Toll-like receptors (TLRs) and nucleotide-binding oligomerization domain (NOD)-like receptors (NLRs) by their shared ligands. In addition to ligating SR-A and MARCO, these agonists also selectively activated the cell-surface sensor TLR4, endosomal TLR3, and the cytosolic NOD2 and NALP3 (NACHT domain-, leucine-rich repeat-, and pyrin domain-containing protein 3). We show that, following recognition of common ligands, SR-A and MARCO attenuate TLR4-mediated responses while enhancing responses by the intracellular TLR3, NOD2, and NALP3. We conclude that SR-A/MARCO-mediated rapid ligand internalization prevented sensing by surface TLRs while increasing ligand availability in intracellular compartments, thus allowing sensing and robust responses by intracellular sensors.


Subject(s)
Cell Membrane/metabolism , Nod Signaling Adaptor Proteins/physiology , Receptors, Immunologic/physiology , Scavenger Receptors, Class A/physiology , Toll-Like Receptor 4/metabolism , Toll-Like Receptors/physiology , Animals , Antigen Presentation/physiology , Cell Membrane/immunology , Cell Membrane/physiology , Cells, Cultured , Cytokines/metabolism , Ligands , Mice , Mice, Inbred C57BL , Mice, Knockout , Nod Signaling Adaptor Proteins/metabolism , Peritonitis/immunology , Peritonitis/metabolism , Protein Transport/immunology , Receptors, Immunologic/genetics , Receptors, Immunologic/metabolism , Scavenger Receptors, Class A/genetics , Scavenger Receptors, Class A/metabolism , Toll-Like Receptor 4/physiology , Toll-Like Receptors/metabolism
8.
J Clin Med ; 12(13)2023 Jun 22.
Article in English | MEDLINE | ID: mdl-37445249

ABSTRACT

Human-adipose-tissue-derived mesenchymal stromal cells (AD-MSCs) are currently being tested as autologous-cell-based therapies for use in tissue healing and regeneration. Recent studies have also demonstrated that AD-MSC-derived exosomes contribute to tissue repair and peripheral nerve regeneration. Subcutaneous abdominal adipose tissue (AAT) is divided into two layers: the superficial layer (sAAT) and the deep layer (dAAT). However, it is unclear whether there are particular characteristics of each layer in terms of AD-MSC regenerative potential. Using AD-MSCs purified and characterized from three abdominoplasties, we compared their secretomes and exosome functions to identify which layer may be most suitable as a source for cell therapy. Phenotypical analysis of the AD-MSCs containing stromal vascular fraction did not reveal any difference between the two layers. The AD-MSC secretomes showed a very similar pattern of cytokine content and both layers were able to release exosomes with identical characteristics. However, compared to the secretome, the released exosomes showed better biological properties. Interestingly, dAAT exosomes appeared to be more effective on neuromodulation, whereas neither sAAT nor dAAT-derived exosomes had significant effects on endothelial function. It thus appears that AD-MSC-derived exosomes from the two abdominal adipose tissue layers possess different features for cell therapy.

9.
Nat Commun ; 14(1): 80, 2023 01 05.
Article in English | MEDLINE | ID: mdl-36604419

ABSTRACT

Fibro-adipogenic progenitors (FAPs) play a crucial role in skeletal muscle regeneration, as they generate a favorable niche that allows satellite cells to perform efficient muscle regeneration. After muscle injury, FAP content increases rapidly within the injured muscle, the origin of which has been attributed to their proliferation within the muscle itself. However, recent single-cell RNAseq approaches have revealed phenotype and functional heterogeneity in FAPs, raising the question of how this differentiation of regenerative subtypes occurs. Here we report that FAP-like cells residing in subcutaneous adipose tissue (ScAT), the adipose stromal cells (ASCs), are rapidly released from ScAT in response to muscle injury. Additionally, we find that released ASCs infiltrate the damaged muscle, via a platelet-dependent mechanism and thus contribute to the FAP heterogeneity. Moreover, we show that either blocking ASCs infiltration or removing ASCs tissue source impair muscle regeneration. Collectively, our data reveal that ScAT is an unsuspected physiological reservoir of regenerative cells that support skeletal muscle regeneration, underlining a beneficial relationship between muscle and fat.


Subject(s)
Muscle, Skeletal , Muscular Diseases , Humans , Adipose Tissue , Cell Differentiation/genetics , Adipogenesis/genetics
10.
Blood ; 115(2): 353-62, 2010 Jan 14.
Article in English | MEDLINE | ID: mdl-19880493

ABSTRACT

Alternatively activated macrophages play an important role in host defense in the context of a T helper type 2 (Th2) microenvironment such as parasitic infection. However, the role of these macrophages during secondary challenge with Th1 pathogens is poorly defined. In this study, thioglycollate-elicited mouse peritoneal macrophages were treated with interleukin-4 (IL-4) or IL-13 in vitro and challenged with Neisseria meningitidis. After 8 to 12 hours of IL-4 pretreatment, the nonopsonic phagocytic uptake of N meningitidis was markedly reduced, depending on the common IL-4Ralpha chain, but independent of Scavenger receptor A and macrophage receptor with collagenous structure (MARCO), 2 known receptors for N meningitidis. Inhibition of phagocytosis extended to several other microbial particles, zymosan, and other bacteria. Concomitantly, IL-4 potentiated the secretion of proinflammatory cytokines, after additional bacterial stimulation, which depended on the MyD88 signaling pathway. Similar results were obtained after intraperitoneal stimulation of IL-4 and N meningitidis in vivo. Further in vitro studies showed a striking correlation with inhibition of Akt phosphorylation and stimulation of the mitogen-activated protein kinase pathway; inhibition of phagocytosis was associated with inhibition of phagosome formation. These findings are relevant to host defense in mixed infections within a Th2 microenvironment and shed light on immunologic functions associated with alternative priming and full activation of macrophages.


Subject(s)
Interleukin-4/metabolism , Interleukin-4/pharmacology , Macrophage Activation , Macrophages, Peritoneal/metabolism , Neisseria meningitidis, Serogroup B , Phagocytosis , Signal Transduction , Animals , Interleukin-13/genetics , Interleukin-13/immunology , Interleukin-13/metabolism , Interleukin-4/immunology , Macrophages, Peritoneal/immunology , Mice , Mice, Knockout , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/immunology , Myeloid Differentiation Factor 88/metabolism , Phosphorylation/drug effects , Phosphorylation/immunology , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/immunology , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Cell Surface/genetics , Receptors, Cell Surface/immunology , Receptors, Cell Surface/metabolism , Receptors, Immunologic/genetics , Receptors, Immunologic/immunology , Receptors, Immunologic/metabolism , Th2 Cells/immunology , Th2 Cells/metabolism , Time Factors
11.
Front Bioeng Biotechnol ; 10: 895735, 2022.
Article in English | MEDLINE | ID: mdl-36177178

ABSTRACT

Background: Nanofat grafting (NG) is a simple and cost-effective method of lipoaspirates with inter-syringe passages, to produce stromal vascular fraction (SVF) and isolate adipose-derived stem cells (ASCs). This represents a tremendous interest in the future clinical needs of tissue engineering. In this study, we optimized the NG technique to increase the yield of ASC extractions. Methods: We analyzed three groups of SVF obtained by 20, 30, and 40 inter-syringe passages. The control group was an SVF obtained by enzymatic digestion with Celase. We studied their cell composition by flow cytometry, observed their architecture by confocal microscopy, and observed immunomodulatory properties of the ASCs from each of the SVFs by measuring inflammatory markers of macrophages obtained by an ASC monocyte co-culture. Results: We have established the first cell mapping of the stromal vascular fraction of adipose tissue. The results showed that SVF obtained by 20 inter-syringe passages contains more statistically significant total cells, more cells expressing the ASC phenotype, more endothelial cells, and produces more CFU-F than the SVF obtained by 30 and 40 passages and by enzymatic digestion. Confocal microscopy showed the presence of residual adipocytes in SVF obtained by inter-syringe passages but not by enzymatic digestion. The functional study indicates an orientation toward a more anti-inflammatory profile and homogenization of their immunomodulatory properties. Conclusion: This study places mechanically dissociated SVF in the center of approaches to easily extract ASCs and a wide variety and number of other progenitor cells, immediately available in a clinical setting to provide both the amount and quality of cells required for decellularized tissues.

12.
Circ Res ; 105(8): 784-92, 2009 Oct 09.
Article in English | MEDLINE | ID: mdl-19745166

ABSTRACT

RATIONALE: Multiple cyclic nucleotide phosphodiesterases (PDEs) degrade cAMP in cardiomyocytes but the role of PDEs in controlling cAMP signaling during pathological cardiac hypertrophy is poorly defined. OBJECTIVE: Evaluate the beta-adrenergic regulation of cardiac contractility and characterize the changes in cardiomyocyte cAMP signals and cAMP-PDE expression and activity following cardiac hypertrophy. METHODS AND RESULTS: Cardiac hypertrophy was induced in rats by thoracic aortic banding over a time period of 5 weeks and was confirmed by anatomic measurements and echocardiography. Ex vivo myocardial function was evaluated in Langendorff-perfused hearts. Engineered cyclic nucleotide-gated (CNG) channels were expressed in single cardiomyocytes to monitor subsarcolemmal cAMP using whole-cell patch-clamp recordings of the associated CNG current (I(CNG)). PDE variant activity and protein level were determined in purified cardiomyocytes. Aortic stenosis rats exhibited a 67% increase in heart weight compared to sham-operated animals. The inotropic response to maximal beta-adrenergic stimulation was reduced by approximately 54% in isolated hypertrophied hearts, along with a approximately 32% decrease in subsarcolemmal cAMP levels in hypertrophied myocytes. Total cAMP hydrolytic activity as well as PDE3 and PDE4 activities were reduced in hypertrophied myocytes, because of a reduction of PDE3A, PDE4A, and PDE4B, whereas PDE4D was unchanged. Regulation of beta-adrenergic cAMP signals by PDEs was blunted in hypertrophied myocytes, as demonstrated by the diminished effects of IBMX (100 micromol/L) and of both the PDE3 inhibitor cilostamide (1 micromol/L) and the PDE4 inhibitor Ro 201724 (10 micromol/L). CONCLUSIONS: Beta-adrenergic desensitization is accompanied by a reduction in cAMP-PDE and an altered modulation of beta-adrenergic cAMP signals in cardiac hypertrophy.


Subject(s)
Cardiomegaly/enzymology , Cyclic AMP/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 4/metabolism , Gene Expression Regulation, Enzymologic , Myocytes, Cardiac/enzymology , Second Messenger Systems , 1-Methyl-3-isobutylxanthine/pharmacology , 4-(3-Butoxy-4-methoxybenzyl)-2-imidazolidinone/pharmacology , Animals , Aortic Valve Stenosis/enzymology , Aortic Valve Stenosis/pathology , Cardiomegaly/pathology , Cells, Cultured , Dose-Response Relationship, Drug , Male , Myocardial Contraction/drug effects , Myocytes, Cardiac/pathology , Organ Size , Phosphodiesterase 4 Inhibitors , Phosphodiesterase Inhibitors/pharmacology , Quinolones/pharmacology , Rats , Rats, Wistar
13.
Front Immunol ; 12: 626755, 2021.
Article in English | MEDLINE | ID: mdl-33995350

ABSTRACT

Mesenchymal stromal cells (MSCs) are currently widely used in cell based therapy regarding to their remarkable efficacy in controlling the inflammatory status in patients. Despite recent progress and encouraging results, inconstant therapeutic benefits are reported suggesting that significant breakthroughs in the understanding of MSCs immunomodulatory mechanisms of action remains to be investigated and certainly apprehended from original point of view. This review will focus on the recent findings regarding MSCs close relationship with the innate immune compartment, i.e. granulocytes and myeloid cells. The review will also consider the intercellular mechanism of communication involved, such as factor secretion, cell-cell contact, extracellular vesicles, mitochondria transfer and efferocytosis. Immune-like-properties of MSCs supporting part of their therapeutic effect in the clinical setting will be discussed, as well as their potentials (immunomodulatory, anti-bacterial, anti-inflammatory, anti-oxidant defenses and metabolic adaptation…) and effects mediated, such as cell polarization, differentiation, death and survival on various immune and tissue cell targets determinant in triggering tissue regeneration. Their metabolic properties in term of sensing, reacting and producing metabolites influencing tissue inflammation will be highlighted. The review will finally open to discussion how ongoing scientific advances on MSCs could be efficiently translated to clinic in chronic and age-related inflammatory diseases and the current limits and gaps that remain to be overcome to achieving tissue regeneration and rejuvenation.


Subject(s)
Cell Communication , Energy Metabolism , Inflammation/therapy , Macrophages/physiology , Mesenchymal Stem Cells/physiology , Regenerative Medicine , Aging , Exosomes/physiology , Extracellular Vesicles/physiology , Graft vs Host Disease/therapy , Humans , Immunity, Innate , Immunomodulation , Mesenchymal Stem Cell Transplantation , Mitochondria/metabolism
14.
Life Sci ; 283: 119857, 2021 Oct 15.
Article in English | MEDLINE | ID: mdl-34339715

ABSTRACT

AIM: Diabetic cardiomyopathy (DCM) accomodates a spectrum of cardiac abnormalities. This study aims to investigate whether DCM is associated with changes in cyclic adenosine 3'-5' monophosphate (cAMP) signaling, particularly cyclic nucleotide phosphodiesterases (PDEs). MAIN METHODS: Type 1 diabetes (T1D) was induced in rats by streptozotocin (STZ, 65 mg/kg) injection. Myocardial remodeling, structure and function were evaluated by histology and echocardiography, respectively. We delineated the sequential changes affecting cAMP signaling and characterized the expression pattern of the predominant cardiac PDE isoforms (PDE 1-5) and ß-adrenergic (ß-AR) receptors at 4, 8 and 12 weeks following diabetes induction, by real-time quantitative PCR and Western blot. cAMP levels were measured by immunoassays. KEY FINDINGS: T1D-induced DCM was associated with cardiac remodeling, steatosis and fibrosis. Upregulation of ß1-AR receptor transcripts was noted in diabetic hearts at 4 weeks along with an increase in cAMP levels and an upregulation in the ejection fraction and fraction shortening. However, ß2-AR receptors expression remained unchanged regardless of the disease stage. Moreover, we noted an early and specific upregulation of cardiac PDE1A, PDE2A, PDE4B, PDE4D and PDE5A expression at week 4, followed by increases in PDE3A levels in diabetic hearts at week 8. However, DCM was not associated with changes in PDE4A gene expression irrespective of the disease stage. SIGNIFICANCE: We show for the first time differential and time-specific regulations in cardiac PDEs, data that may prove useful in proposing new therapeutic approaches in T1D-induced DCM.


Subject(s)
3',5'-Cyclic-AMP Phosphodiesterases/metabolism , Diabetic Cardiomyopathies/physiopathology , Phosphoric Diester Hydrolases/metabolism , Animals , Cyclic AMP/metabolism , Diabetes Mellitus, Experimental/physiopathology , Diabetic Cardiomyopathies/metabolism , Male , Myocardium/metabolism , Myocytes, Cardiac/metabolism , Phosphoric Diester Hydrolases/physiology , Rats , Rats, Wistar , Receptors, Adrenergic, beta/metabolism , Signal Transduction , Streptozocin/pharmacology
15.
Retrovirology ; 7: 33, 2010 Apr 09.
Article in English | MEDLINE | ID: mdl-20380696

ABSTRACT

Macrophages play a crucial role in innate and adaptative immunity in response to microorganisms and are an important cellular target during HIV-1 infection. Recently, the heterogeneity of the macrophage population has been highlighted. Classically activated or type 1 macrophages (M1) induced in particular by IFN-gamma display a pro-inflammatory profile. The alternatively activated or type 2 macrophages (M2) induced by Th-2 cytokines, such as IL-4 and IL-13 express anti-inflammatory and tissue repair properties. Finally IL-10 has been described as the prototypic cytokine involved in the deactivation of macrophages (dM). Since the capacity of macrophages to support productive HIV-1 infection is known to be modulated by cytokines, this review shows how modulation of macrophage activation by cytokines impacts the capacity to support productive HIV-1 infection. Based on the activation status of macrophages we propose a model starting with M1 classically activated macrophages with accelerated formation of viral reservoirs in a context of Th1 and proinflammatory cytokines. Then IL-4/IL-13 alternatively activated M2 macrophages will enter into the game that will stop the expansion of the HIV-1 reservoir. Finally IL-10 deactivation of macrophages will lead to immune failure observed at the very late stages of the HIV-1 disease.


Subject(s)
HIV-1/immunology , HIV-1/pathogenicity , Macrophage Activation , Macrophages/immunology , Macrophages/virology , Virus Replication , Cytokines/immunology , Humans , Models, Biological
16.
J Immunol ; 181(8): 5350-9, 2008 Oct 15.
Article in English | MEDLINE | ID: mdl-18832691

ABSTRACT

Immune complexes can trigger a SHIP-1-independent proapoptotic signal in mouse class-switched IgG(+) B cells and plasma cells by binding to Fc gammaRIIB, in the absence of concomitant coaggregation with BCR, hence regulating plasma cell survival and participating in the selection of B cells producing high affinity Abs during secondary Ab responses. By contrast, we demonstrate in the present study that the unique aggregation of Fc gammaRIIB on human peripheral IgM(+) B cells does not induce apoptosis but transiently inhibits B cell proliferation and calcium influx triggered by BCR cross-linking. Using human peripheral B cells and IIA1.6 lymphoma B cells expressing wild-type human Fc gammaRIIB (IIA1.6-Fc gammaRIIB), we also show that the unique aggregation of human Fc gammaRIIB induces ITIM phosphorylation. This aggregation provokes the recruitment of phosphorylated SHIP-1 by Fc gammaRIIB and inhibits the constitutive phosphorylation of Akt in human IIA1.6-Fc gammaRIIB cells. This inhibitory signaling pathway is abrogated in IIA1.6 cells expressing ITIM-mutated Fc gammaRIIB (Fc gammaRIIB(Y292G)), suggesting that ITIM phosphorylation is necessary for Fc gammaRIIB-induced B cell blockade. Overall, we demonstrate that the unique aggregation of Fc gammaRIIB on human peripheral IgM(+) B cells is sufficient to transiently down-regulate their activation without inducing apoptosis. Our results suggest that Fc gammaRIIB could negatively regulate IgM(+) B cells before class-switch occurrence and that its unique engagement by immune complexes represents a reversible checkpoint for peripheral IgM(+) B cells.


Subject(s)
Apoptosis/immunology , Immunoglobulin M , Lymphocyte Activation/immunology , Plasma Cells/immunology , Proto-Oncogene Proteins c-bcr/immunology , Receptors, IgG/immunology , Amino Acid Substitution/immunology , Animals , Antigen-Antibody Complex/genetics , Antigen-Antibody Complex/immunology , Antigen-Antibody Complex/metabolism , Apoptosis/genetics , Calcium Signaling/genetics , Calcium Signaling/immunology , Cell Line, Tumor , Cell Proliferation , Cell Survival/genetics , Cell Survival/immunology , Humans , Inositol Polyphosphate 5-Phosphatases , Lymphocyte Activation/genetics , Mice , Mutation, Missense/immunology , Phosphatidylinositol-3,4,5-Trisphosphate 5-Phosphatases , Phosphoric Monoester Hydrolases/genetics , Phosphoric Monoester Hydrolases/immunology , Phosphoric Monoester Hydrolases/metabolism , Phosphorylation , Plasma Cells/metabolism , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/immunology , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-bcr/genetics , Proto-Oncogene Proteins c-bcr/metabolism , Receptors, IgG/genetics
17.
J Leukoc Biol ; 83(3): 718-27, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18070983

ABSTRACT

HIV-1 two-exon transactivator protein (Tat) is a 101-aa protein. We investigated the possible contribution of the extreme C terminus of HIV-1 Tat to maximize nuclear transcription factor NF-kappaB activation, long terminal repeat (LTR) transactivation, and viral replication in T cells. C-terminal deletion and substitution mutants made with the infectious clone HIV-89.6 were assayed for their ability to transactivate NF-kappaB-secreted alkaline phosphatase and HIV-1 LTR-luciferase reporter constructs for low concentrations of Tat. A mutant infectious clone of HIV-89.6 engineered by introducing a stop codon at aa 72 in the Tat open-reading frame (HIVDeltatatexon2) replicated at a significantly lower rate than the wild-type HIV-89.6 in phytohemagglutinin-A/IL-2-stimulated primary peripheral blood lymphocytes. Altogether, our results suggest a critical role for the glutamic acids at positions 92, 94, and 96 or lysines at positions 88, 89, and 90, present in the second encoding Tat exon in activating NF-kappaB, transactivating the HIV-1 LTR and enhancing HIV-1 replication in T cells.


Subject(s)
Genes, tat , HIV-1/physiology , NF-kappa B/physiology , T-Lymphocytes/virology , Transcription, Genetic , Amino Acid Sequence , Exons , HIV-1/genetics , Humans , Jurkat Cells , Kinetics , Molecular Sequence Data , Plasmids , Point Mutation , T-Lymphocytes/physiology , Virus Replication
18.
Cardiovasc Res ; 115(1): 130-144, 2019 01 01.
Article in English | MEDLINE | ID: mdl-29939224

ABSTRACT

Aims: Regulation of vascular tone by 3',5'-cyclic adenosine monophosphate (cAMP) involves many effectors including the large conductance, Ca2+-activated, K+ (BKCa) channels. In arteries, cAMP is mainly hydrolyzed by type 3 and 4 phosphodiesterases (PDE3, PDE4). Here, we examined the specific contribution of BKCa channels to tone regulation by these PDEs in rat coronary arteries, and how this is altered in heart failure (HF). Methods and results: Concomitant application of PDE3 (cilostamide) and PDE4 (Ro-20-1724) inhibitors increased BKCa unitary channel activity in isolated myocytes from rat coronary arteries. Myography was conducted in isolated, U46619-contracted coronary arteries. Cilostamide (Cil) or Ro-20-1724 induced a vasorelaxation that was greatly reduced by iberiotoxin (IBTX), a BKCa channel blocker. Ro-20-1724 and Cil potentiated the relaxation induced by the ß-adrenergic agonist isoprenaline (ISO) or the adenylyl cyclase activator L-858051 (L85). IBTX abolished the effect of PDE inhibitors on ISO but did not on L85. In coronary arteries from rats with HF induced by aortic stenosis, contractility and response to acetylcholine were dramatically reduced compared with arteries from sham rats, but relaxation to PDE inhibitors was retained. Interestingly, however, IBTX had no effect on Ro-20-1724- and Cil-induced vasorelaxations in HF. Expression of the BKCa channel α-subunit, of a 98 kDa PDE3A and of a 80 kDa PDE4D were lower in HF compared with sham coronary arteries, while that of a 70 kDa PDE4B was increased. Proximity ligation assays demonstrated that PDE3 and PDE4 were localized in the vicinity of the channel. Conclusion: BKCa channels mediate the relaxation of coronary artery induced by PDE3 and PDE4 inhibition. This is achieved by co-localization of both PDEs with BKCa channels, enabling tight control of cAMP available for channel opening. Contribution of the channel is prominent at rest and on ß-adrenergic stimulation. This coupling is lost in HF.


Subject(s)
Coronary Vessels/enzymology , Cyclic Nucleotide Phosphodiesterases, Type 3/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 4/metabolism , Heart Failure/enzymology , Large-Conductance Calcium-Activated Potassium Channels/metabolism , Vasodilation , Animals , Coronary Vessels/drug effects , Coronary Vessels/physiopathology , Disease Models, Animal , Heart Failure/physiopathology , Ion Channel Gating , Male , Phosphodiesterase 3 Inhibitors/pharmacology , Phosphodiesterase 4 Inhibitors/pharmacology , Rats, Wistar , Signal Transduction , Vasodilation/drug effects , Vasodilator Agents/pharmacology
19.
Immunology ; 124(4): 562-74, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18284467

ABSTRACT

Tumour necrosis factor receptor associated factor 4 (TRAF4) is a member of the TRAF family of proteins which are cytoplasmic adaptor molecules strongly implicated in multiple immune functions. A previous investigation of TRAF4 biological functions by gene targeting in mice has shown a role for TRAF4 in embryonic development and neurulation in vivo. However, unlike other TRAF family members, the role of TRAF4 in the immune system is still unknown. To address this question, we performed an extensive characterization of the immune development and immune functions of TRAF4-deficient mice. Our analyses did not reveal any defects in development of T and B lymphocytes, granulocytes, macrophages and dendritic cells, and no defects in reactive oxygen species production and phagocytosis by neutrophils. Cellular and humoral responses against T-cell-dependent antigens were normal, as was dendritic cell maturation in response to microbial components and antigen uptake by dendritic cells. However, we demonstrated that dendritic cells from TRAF4-deficient mice exhibited reduced migration both in transwell experiments and in vivo. These results suggest that TRAF4 is not strictly required for immune development and functions but could participate in immune functions by facilitating immune cell migration.


Subject(s)
Dendritic Cells/immunology , TNF Receptor-Associated Factor 4/immunology , Animals , B-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cell Movement/immunology , Cell Proliferation , Cells, Cultured , Cytokines/biosynthesis , Immune System/growth & development , Immunity, Cellular , Immunoglobulin G/biosynthesis , Lymphocyte Activation/immunology , Mice , Mice, Knockout , Neutrophils/immunology , Ovalbumin/immunology , Shiga Toxins/immunology , T-Lymphocyte Subsets/immunology , TNF Receptor-Associated Factor 4/deficiency
20.
Int J Cancer ; 123(12): 2832-9, 2008 Dec 15.
Article in English | MEDLINE | ID: mdl-18798552

ABSTRACT

During melanoma progression, patients develop anti-tumor immunity including the production of anti-tumor antibodies. Although the strategies developed by malignant cells to escape anti-tumor cellular immunity have been extensively investigated, little is known about tumor resistance to humoral immunity. The main effect of IgG antibodies is to activate the immune response by binding to host Fc gamma receptors (FcgammaR) expressed by immune cells. We previously reported in a limited study that some human metastatic melanoma cells ectopically express the FcgammaRIIB1, an inhibitory isoform of FcgammaR. By analyzing a large panel of different types of human primary and metastatic solid tumors, we report herein that expression of FcgammaRIIB is restricted to melanoma and is acquired during tumor progression. We show that FcgammaRIIB expression prevents the lysis of human metastatic melanoma cells by NK cell-mediated antibody-dependent cellular cytotoxicity (ADCC) in vitro, independently of the intracytoplasmic region of FcgammaRIIB. Using experimental mouse models, we demonstrate that expression of FcgammaRIIB protects B16F0 melanoma tumors from the ADCC induced by monoclonal and polyclonal anti-tumor IgG in vivo. Thus, our results identify FcgammaRIIB as a marker of human metastatic melanoma that impairs the tumor susceptibility to FcgammaR-dependent innate effector responses.


Subject(s)
Biomarkers, Tumor/analysis , Immunoglobulin G/immunology , Melanoma/immunology , Receptors, IgG/analysis , Skin Neoplasms/immunology , Animals , Cell Line, Tumor , Disease Progression , Flow Cytometry , Gene Expression Regulation, Neoplastic , Humans , Immunohistochemistry , Melanoma/secondary , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, SCID , Skin Neoplasms/pathology
SELECTION OF CITATIONS
SEARCH DETAIL