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










Publication year range
1.
Sci Transl Med ; 15(687): eade0550, 2023 03 15.
Article in English | MEDLINE | ID: mdl-36921035

ABSTRACT

The diversity of vaccination modalities and infection history are both variables that have an impact on the immune memory of individuals vaccinated against SARS-CoV-2. To gain more accurate knowledge of how these parameters imprint on immune memory, we conducted a long-term follow-up of SARS-CoV-2 spike protein-specific immune memory in unvaccinated and vaccinated COVID-19 convalescent individuals as well as in infection-naïve vaccinated individuals. Here, we report that individuals from the convalescent vaccinated (hybrid immunity) group have the highest concentrations of spike protein-specific antibodies at 6 months after vaccination. As compared with infection-naïve vaccinated individuals, they also display increased frequencies of an atypical mucosa-targeted memory B cell subset. These individuals also exhibited enhanced TH1 polarization of their SARS-CoV-2 spike protein-specific follicular T helper cell pool. Together, our data suggest that prior SARS-CoV-2 infection increases the titers of SARS-CoV-2 spike protein-specific antibody responses elicited by subsequent vaccination and induces modifications in the composition of the spike protein-specific memory B cell pool that are compatible with enhanced functional protection at mucosal sites.


Subject(s)
COVID-19 , Humans , COVID-19/prevention & control , SARS-CoV-2 , Spike Glycoprotein, Coronavirus , Antibodies , Vaccination , Antibodies, Viral , Antibodies, Neutralizing
2.
Sci Transl Med ; 14(636): eabl6141, 2022 03 16.
Article in English | MEDLINE | ID: mdl-35103481

ABSTRACT

Transplant recipients, who receive therapeutic immunosuppression to prevent graft rejection, are characterized by high coronavirus disease 2019 (COVID-19)-related mortality and defective response to vaccines. We observed that previous infection with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), but not the standard two-dose regimen of vaccination, provided protection against symptomatic COVID-19 in kidney transplant recipients. We therefore compared the cellular and humoral immune responses of these two groups of patients. Neutralizing anti-receptor-binding domain (RBD) immunoglobulin G (IgG) antibodies were identified as the primary correlate of protection for transplant recipients. Analysis of virus-specific B and T cell responses suggested that the generation of neutralizing anti-RBD IgG may have depended on cognate T-B cell interactions that took place in germinal center, potentially acting as a limiting checkpoint. High-dose mycophenolate mofetil, an immunosuppressive drug, was associated with fewer antigen-specific B and T follicular helper (TFH) cells after vaccination; this was not observed in patients recently infected with SARS-CoV-2. Last, we observed that, in two independent prospective cohorts, administration of a third dose of SARS-CoV-2 mRNA vaccine restored neutralizing titers of anti-RBD IgG in about 40% of individuals who had not previously responded to two doses of vaccine. Together, these findings suggest that a third dose of SARS-CoV-2 mRNA vaccine improves the RBD-specific responses of transplant patients treated with immunosuppressive drugs.


Subject(s)
COVID-19 , Kidney Transplantation , Antibodies, Neutralizing , Antibodies, Viral , BNT162 Vaccine , COVID-19/prevention & control , COVID-19 Vaccines , Humans , Prospective Studies , SARS-CoV-2 , Transplant Recipients , Vaccines, Synthetic , mRNA Vaccines
3.
Nature ; 600(7890): 701-706, 2021 12.
Article in English | MEDLINE | ID: mdl-34673755

ABSTRACT

Following severe adverse reactions to the AstraZeneca ChAdOx1-S-nCoV-19 vaccine1,2, European health authorities recommended that patients under the age of 55 years who received one dose of ChAdOx1-S-nCoV-19 receive a second dose of the Pfizer BNT162b2 vaccine as a booster. However, the effectiveness and the immunogenicity of this vaccination regimen have not been formally tested. Here we show that the heterologous ChAdOx1-S-nCoV-19 and BNT162b2 combination confers better protection against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection than the homologous BNT162b2 and BNT162b2 combination in a real-world observational study of healthcare workers (n = 13,121). To understand the underlying mechanism, we conducted a longitudinal survey of the anti-spike immunity conferred by each vaccine combination. Both combinations induced strong anti-spike antibody responses, but sera from heterologous vaccinated individuals displayed a stronger neutralizing activity regardless of the SARS-CoV-2 variant. This enhanced neutralizing potential correlated with increased frequencies of switched and activated memory B cells that recognize the SARS-CoV-2 receptor binding domain. The ChAdOx1-S-nCoV-19 vaccine induced a weaker IgG response but a stronger T cell response than the BNT162b2 vaccine after the priming dose, which could explain the complementarity of both vaccines when used in combination. The heterologous vaccination regimen could therefore be particularly suitable for immunocompromised individuals.


Subject(s)
BNT162 Vaccine/administration & dosage , BNT162 Vaccine/immunology , COVID-19/immunology , COVID-19/prevention & control , ChAdOx1 nCoV-19/administration & dosage , ChAdOx1 nCoV-19/immunology , SARS-CoV-2/immunology , Vaccination/statistics & numerical data , Adult , Antibodies, Neutralizing/blood , Antibodies, Neutralizing/immunology , Female , France/epidemiology , Hospitals, University , Humans , Immunologic Memory/immunology , Incidence , Male , Memory B Cells/immunology , Memory T Cells/immunology , Middle Aged , Spike Glycoprotein, Coronavirus/immunology
4.
PLoS Pathog ; 13(9): e1006610, 2017 09.
Article in English | MEDLINE | ID: mdl-28957419

ABSTRACT

IFITMs are broad antiviral factors that block incoming virions in endosomal vesicles, protecting target cells from infection. In the case of HIV-1, we and others reported the existence of an additional antiviral mechanism through which IFITMs lead to the production of virions of reduced infectivity. However, whether this second mechanism of inhibition is unique to HIV or extends to other viruses is currently unknown. To address this question, we have analyzed the susceptibility of a broad spectrum of viruses to the negative imprinting of the virion particles infectivity by IFITMs. The results we have gathered indicate that this second antiviral property of IFITMs extends well beyond HIV and we were able to identify viruses susceptible to the three IFITMs altogether (HIV-1, SIV, MLV, MPMV, VSV, MeV, EBOV, WNV), as well as viruses that displayed a member-specific susceptibility (EBV, DUGV), or were resistant to all IFITMs (HCV, RVFV, MOPV, AAV). The swapping of genetic elements between resistant and susceptible viruses allowed us to point to specificities in the viral mode of assembly, rather than glycoproteins as dominant factors of susceptibility. However, we also show that, contrarily to X4-, R5-tropic HIV-1 envelopes confer resistance against IFITM3, suggesting that viral receptors add an additional layer of complexity in the IFITMs-HIV interplay. Lastly, we show that the overall antiviral effects ascribed to IFITMs during spreading infections, are the result of a bimodal inhibition in which IFITMs act both by protecting target cells from incoming viruses and in driving the production of virions of reduced infectivity. Overall, our study reports for the first time that the negative imprinting of the virion particles infectivity is a conserved antiviral property of IFITMs and establishes IFITMs as a paradigm of restriction factor capable of interfering with two distinct phases of a virus life cycle.


Subject(s)
Antigens, Differentiation/metabolism , Virion , Virus Replication , Cell Line , HIV-1/physiology , Host-Pathogen Interactions , Humans , Virus Internalization
5.
Viruses ; 7(12): 6233-40, 2015 Dec 01.
Article in English | MEDLINE | ID: mdl-26633464

ABSTRACT

The current outbreak of Ebola virus (EBOV) in West Africa has claimed the lives of more than 15,000 people and highlights an urgent need for therapeutics capable of preventing virus replication. In this study we screened known nucleoside analogues for their ability to interfere with EBOV replication. Among them, the cytidine analogue ß-d-N4-hydroxycytidine (NHC) demonstrated potent inhibitory activities against EBOV replication and spread at non-cytotoxic concentrations. Thus, NHC constitutes an interesting candidate for the development of a suitable drug treatment against EBOV.


Subject(s)
Antiviral Agents/pharmacology , Cytidine/analogs & derivatives , Ebolavirus/drug effects , Ebolavirus/physiology , Virus Replication/drug effects , Animals , Antiviral Agents/toxicity , Chlorocebus aethiops , Cytidine/pharmacology , Cytidine/toxicity , Vero Cells
6.
PLoS One ; 10(10): e0140561, 2015.
Article in English | MEDLINE | ID: mdl-26496699

ABSTRACT

Two cellular factors are currently known to modulate lentiviral infection specifically in myeloid cells: SAMHD1 and APOBEC3A (A3A). SAMHD1 is a deoxynucleoside triphosphohydrolase that interferes with viral infection mostly by limiting the intracellular concentrations of dNTPs, while A3A is a cytidine deaminase that has been described to edit incoming vDNA. The restrictive phenotype of myeloid cells can be alleviated through the direct degradation of SAMHD1 by the HIV-2/SIVSM Vpx protein or else, at least in the case of HIV-1, by the exogenous supplementation of nucleosides that artificially overcome the catabolic activity of SAMHD1 on dNTPs. Here, we have used Vpx and dNs to explore the relationship existing between vDNA cytidine deamination and SAMHD1 during HIV-1 or SIVMAC infection of primary dendritic cells. Our results reveal an interesting inverse correlation between conditions that promote efficient infection of DCs and the extent of vDNA editing that may reflect the different susceptibility of vDNA to cytoplasmic effectors during the infection of myeloid cells.


Subject(s)
Cytidine/genetics , DNA, Viral/genetics , Dendritic Cells/virology , HIV-1/physiology , Nucleosides/pharmacology , Simian Immunodeficiency Virus/physiology , Viral Regulatory and Accessory Proteins/pharmacology , Dendritic Cells/drug effects , HEK293 Cells , HIV-1/drug effects , HeLa Cells , Humans , Kinetics , Reverse Transcription/drug effects , Simian Immunodeficiency Virus/drug effects
7.
J Virol ; 89(7): 4030-4, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25609804

ABSTRACT

The block toward human immunodeficiency virus type 1 (HIV-1) infection of dendritic cells (DCs) can be relieved by Vpx (viral protein X), which degrades sterile alpha motif-hydroxylase domain 1 (SAMHD1) or by exogenously added deoxynucleosides (dNs), lending support to the hypothesis that SAMHD1 acts by limiting deoxynucleoside triphosphates (dNTPs). This notion has, however, been questioned. We show that while dNs and Vpx increase the infectivity of HIV-1, only the latter restores the infectivity of a simian immunodeficiency virus of macaques variant, SIVMACΔVpx virus. This distinct behavior seems to map to CA, suggesting that species-specific CA interactors modulate infection of DCs.


Subject(s)
Capsid Proteins/metabolism , Dendritic Cells/virology , HIV-1/physiology , Host-Pathogen Interactions , Nucleosides/metabolism , Simian Immunodeficiency Virus/physiology , Viral Regulatory and Accessory Proteins/metabolism , Animals , Cells, Cultured , HIV-1/growth & development , Humans , Macaca , Simian Immunodeficiency Virus/growth & development
8.
J Infect Dis ; 207(3): 469-78, 2013 Feb 01.
Article in English | MEDLINE | ID: mdl-23175762

ABSTRACT

Nipah virus (NiV) and Hendra virus (HeV) are closely related, recently emerged paramyxoviruses that are capable of causing considerable morbidity and mortality in several mammalian species, including humans. Henipavirus-specific vaccines are still commercially unavailable, and development of novel antiviral strategies to prevent lethal infections due to henipaviruses is highly desirable. Here we describe the development of adeno-associated virus (AAV) vaccines expressing the NiV G protein. Characterization of these vaccines in mice demonstrated that a single intramuscular AAV injection was sufficient to induce a potent and long-lasting antibody response. Translational studies in hamsters further demonstrated that all vaccinated animals were protected against lethal challenge with NiV. In addition, this vaccine induced a cross-protective immune response that was able to protect 50% of the animals against a challenge by HeV. This study presents a new efficient vaccination strategy against henipaviruses and opens novel perspectives on the use of AAV vectors as vaccines against emergent diseases.


Subject(s)
Dependovirus/genetics , Genetic Vectors/genetics , Henipavirus Infections/immunology , Henipavirus Infections/prevention & control , Vaccines, Synthetic/immunology , Viral Vaccines/immunology , Animals , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Cell Line, Tumor , Cricetinae , Disease Models, Animal , Henipavirus Infections/virology , Humans , Immunity, Humoral , Immunoglobulin G/immunology , Male , Mice , Vaccines, Synthetic/genetics , Viral Vaccines/genetics
9.
Virology ; 432(1): 1-9, 2012 Oct 10.
Article in English | MEDLINE | ID: mdl-22727829

ABSTRACT

The inability of Adeno-Associated Virus (AAV) to replicate on its own is a strong argument in favor of the use of recombinant AAV vectors for in vivo gene transfer. However, some previous studies suggested that AAV may become replication competent in cells exposed to a genotoxic stress even in the absence of co-infection with a helper virus. To comprehensively explore this phenomenon, we examined AAV genome replication in several human cell lines exposed to different genotoxic conditions. We found that all treatments induced only negligible levels of AAV replication never exceeding ten fold above background. Further investigation indicated that induction of helper-independent AAV replication relied on the synergistic contribution of several extrinsic factors linked to the origin of the cell line and the quality of the AAV preparation. These results further support the notion that helper independent AAV replication cannot occur at significant levels in vivo.


Subject(s)
DNA Damage , Dependovirus/physiology , Virus Replication , Cell Line , Helper Viruses/physiology , Humans
10.
Arterioscler Thromb Vasc Biol ; 32(3): 605-12, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22223728

ABSTRACT

OBJECTIVE: The immunoinflammatory response plays a critical role in the development and progression of atherosclerosis. Recent studies suggested an important role for regulatory T (Treg) cells in the inhibition of disease-related vascular inflammation. We hypothesized that induction of a specific Treg cell response to atherosclerosis-relevant antigens would be an attractive strategy to limit the development and progression of atherosclerosis through the promotion of immune tolerance. METHODS AND RESULTS: Young or old Apoe-/- mice were subcutaneously infused for 2 weeks with either a control ovalbumin (OVA) peptide or with apolipoprotein B100 (ApoB100)-derived peptides without adjuvant. Atherosclerosis development, progression and immunologic status were assessed at 8 weeks after the end of the infusion. Treatment with ApoB100 peptides led to significant reduction of lesion development in young Apoe-/- mice (P=0.001 versus OVA group) and abrogated atherosclerosis progression in old Apoe-/- mice with already established lesions (0% progression in ApoB100 versus 17% in OVA group, P<0.005). Limitation of plaque progression was associated with reduced vascular inflammation and increased collagen content, indicative of plaque stabilization. Infusion of ApoB100 peptides did not alterantibody production but promoted a specific Treg cell response, which was associated with a reduction of both T helper type 1-related and T helper type 2-related cytokines. Interestingly, depletion of CD4+CD25+ Treg cells abrogated ApoB100 peptides-dependent immune modulation and atheroprotection. CONCLUSION: Subcutaneous infusion of adjuvant-free ApoB100-derived peptides to Apoe-/- mice reduces atherosclerosis through the induction of a specific Treg cell response.


Subject(s)
Apolipoprotein B-100/administration & dosage , Atherosclerosis/prevention & control , Immune Tolerance , Immunotherapy/methods , Lymphocyte Activation , Peptides/administration & dosage , T-Lymphocytes, Regulatory/immunology , Animals , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Atherosclerosis/genetics , Atherosclerosis/immunology , Atherosclerosis/metabolism , Atherosclerosis/pathology , Cells, Cultured , Cytokines/metabolism , Disease Models, Animal , Disease Progression , Female , Humans , Infusions, Subcutaneous , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Ovalbumin/administration & dosage , T-Lymphocytes, Regulatory/metabolism , Time Factors
11.
Circ Res ; 103(7): 751-60, 2008 Sep 26.
Article in English | MEDLINE | ID: mdl-18723447

ABSTRACT

Cell-based therapy is a promising approach designed to enhance neovascularization and function of ischemic tissues. Interaction between endothelial and smooth muscle cells regulates vessels development and remodeling and is required for the formation of a mature and functional vascular network. Therefore, we assessed whether coadministration of endothelial progenitor cells (EPCs) and smooth muscle progenitor cells (SMPCs) can increase the efficiency of cell therapy. Unilateral hindlimb ischemia was surgically induced in athymic nude mice treated with or without intravenous injection of EPCs (0.5 x 10(6)), SMPCs (0.5 x 10(6)) and EPCs+SMPCs (0.25 x 10(6)+0.25 x 10(6)). Vessel density and foot perfusion were increased in mice treated with EPCs+SMPCs compared to animals receiving EPCs alone or SMPCs alone (P<0.001). In addition, capillary and arteriolar densities were enhanced in EPC+SMPC-treated mice compared to SMPC and EPC groups (P<0.01). We next examined the role of Ang-1/Tie2 signaling in the beneficial effect of EPC and SMPC coadministration. Small interfering RNA directed against Ang-1-producing SMPCs or Tie2-expressing EPCs blocked vascular network formation in Matrigel coculture assays, reduced the rate of incorporated EPCs within vascular structure, and abrogated the efficiency of cell therapy. Production of Ang-1 by SMPCs activates Tie2-expressing EPCs, resulting in increase of EPC survival and formation of a stable vascular network. Subsequently, the efficiency of EPC- and SMPC-based cotherapy is markedly increased. Therefore, coadministration of different types of vascular progenitor cells may constitute a novel therapeutic strategy for improving the treatment of ischemic diseases.


Subject(s)
Endothelial Cells/transplantation , Hindlimb/blood supply , Ischemia/therapy , Myocytes, Smooth Muscle/transplantation , Neovascularization, Physiologic , Stem Cell Transplantation , Stem Cells , Angiotensin I/metabolism , Animals , Endothelial Cells/metabolism , Humans , Ischemia/metabolism , Male , Mice , Mice, Nude , Myocytes, Smooth Muscle/cytology , Receptor, TIE-2/metabolism , Signal Transduction , Stem Cells/cytology , Stem Cells/metabolism
12.
Cardiovasc Res ; 77(3): 471-80, 2008 Feb 01.
Article in English | MEDLINE | ID: mdl-18006460

ABSTRACT

AIMS: We analysed the possible protective role of human endothelial (EPCs) and smooth muscle (SPCs) progenitor cells on atherosclerosis development in apoE(-/-)RAG2(-/-) mice. We determined plasma levels of SPCs in coronary patients. METHODS AND RESULTS: ApoE(-/-)RAG2(-/-) mice received four intravenous injections of saline, 5 x 10(5) SPCs, or 5 x 10(5) EPCs every other week, one (preventive approach) or 12(curative approach) weeks after starting a high fat diet. Derived-SPC levels were quantified from blood mononuclear cells of patients with stable angina (n = 10) and acute coronary syndromes (ACS, n = 9). SPCs reduced atherosclerosis development by 42% (P < 0.001), but had no effect on lesion progression. In the SPC group, collagen and smooth muscle cell content were increased (+80%, P < 0.001, +46%, P < 0.05, respectively), and macrophage content was decreased (-41%, P < 0.05). In the curative approach, macrophage content decreased by 40.5% (P < 0.05) after SPC injection. EPC injection had no effect on atherosclerosis development or progression. Peripheral blood-derived SPC levels were reduced in patients with ACS compared with stable angina patients (P < 0.05). CONCLUSION: We demonstrate that SPCs limit plaque development and promote changes in plaque composition towards a stable phenotype in mice. Our finding in patients suggests that reduced peripheral blood-derived SPC levels might represent a mechanism contributing to plaque destabilization.


Subject(s)
Atherosclerosis/etiology , Hematopoietic Stem Cells/physiology , Muscle, Smooth, Vascular/cytology , Aged , Animals , Apolipoproteins E/physiology , Atherosclerosis/metabolism , Atherosclerosis/pathology , Cell Separation , Cells, Cultured , DNA-Binding Proteins/physiology , Disease Progression , Endothelial Cells/cytology , Female , Fetal Blood/cytology , Humans , Male , Mice , Middle Aged
13.
Arterioscler Thromb Vasc Biol ; 27(12): 2691-8, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17690315

ABSTRACT

OBJECTIVE: Obesity is a major risk factor for atherosclerosis and is associated with increased cardiovascular morbidity and mortality. However, the precise molecular pathways responsible for this close association remain poorly understood. METHODS AND RESULTS: In this study, we report that leptin-deficiency (ob/ob) in low-density lipoprotein receptor knockout (ldlr(-/-)) mice induces an unexpected 2.2- to 6-fold reduction in atherosclerotic lesion development, compared with ldlr(-/-) mice having similar total cholesterol levels. Ldlr(-/-)/ob/ob mice show reduced T cell helper type 1 (Th1) response, enhanced expression of Foxp3, the specification transcription factor of regulatory T (Treg) cells, and improved Treg cell function. Leptin receptor-deficient (db/db) mice display marked increase in the number and suppressive function of Treg cells. Supplementation of Treg-deficient lymphocytes with Treg cells from db/db mice in an experimental model of atherosclerosis induces a significant reduction of lesion size and a marked inhibition of interferon (INF)-gamma production, compared with supplementation by Treg cells from wild-type mice. CONCLUSIONS: These results identify a critical role for leptin/leptin receptor pathway in the modulation of the regulatory immune response in atherosclerosis, and suggest that alteration in regulatory immunity may predispose obese individuals to atherosclerosis.


Subject(s)
Atherosclerosis/prevention & control , Immunity, Cellular , Leptin/metabolism , Obesity/complications , Receptors, Leptin/metabolism , Signal Transduction , T-Lymphocytes, Regulatory/metabolism , Th1 Cells/metabolism , Adoptive Transfer , Animals , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Apolipoproteins E/metabolism , Atherosclerosis/genetics , Atherosclerosis/immunology , Atherosclerosis/metabolism , Atherosclerosis/pathology , CD28 Antigens/genetics , CD28 Antigens/metabolism , Cell Proliferation , Cells, Cultured , Dietary Fats/administration & dosage , Disease Models, Animal , Female , Forkhead Transcription Factors/metabolism , Immunity, Cellular/genetics , Interferon-gamma/metabolism , Leptin/deficiency , Leptin/genetics , Lipids/blood , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Obese , Obesity/genetics , Obesity/immunology , Obesity/metabolism , Obesity/pathology , Receptors, Leptin/deficiency , Receptors, Leptin/genetics , Signal Transduction/genetics
14.
J Clin Invest ; 117(6): 1527-37, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17510705

ABSTRACT

Endothelial progenitor cell (EPC) transplantation has beneficial effects for therapeutic neovascularization; however, only a small proportion of injected cells home to the lesion and incorporate into the neocapillaries. Consequently, this type of cell therapy requires substantial improvement to be of clinical value. Erythropoietin-producing human hepatocellular carcinoma (Eph) receptors and their ephrin ligands are key regulators of vascular development. We postulated that activation of the EphB4/ephrin-B2 system may enhance EPC proangiogenic potential. In this report, we demonstrate in a nude mouse model of hind limb ischemia that EphB4 activation with an ephrin-B2-Fc chimeric protein increases the angiogenic potential of human EPCs. This effect was abolished by EphB4 siRNA, confirming that it is mediated by EphB4. EphB4 activation enhanced P selectin glycoprotein ligand-1 (PSGL-1) expression and EPC adhesion. Inhibition of PSGL-1 by siRNA reversed the proangiogenic and adhesive effects of EphB4 activation. Moreover, neutralizing antibodies to E selectin and P selectin blocked ephrin-B2-Fc-stimulated EPC adhesion properties. Thus, activation of EphB4 enhances EPC proangiogenic capacity through induction of PSGL-1 expression and adhesion to E selectin and P selectin. Therefore, activation of EphB4 is an innovative and potentially valuable therapeutic strategy for improving the recruitment of EPCs to sites of neovascularization and thereby the efficiency of cell-based proangiogenic therapy.


Subject(s)
Endothelial Cells/metabolism , Fetal Stem Cells/metabolism , Membrane Glycoproteins/metabolism , Neovascularization, Physiologic , Receptor, EphB4/metabolism , Animals , Base Sequence , Cell Adhesion , Cells, Cultured , DNA Primers/genetics , E-Selectin/metabolism , Endothelial Cells/cytology , Endothelial Cells/drug effects , Ephrin-B2/metabolism , Ephrin-B2/pharmacology , Fetal Blood/cytology , Fetal Stem Cells/cytology , Fetal Stem Cells/drug effects , Hindlimb/blood supply , Humans , In Vitro Techniques , Ischemia/metabolism , Ischemia/pathology , Ischemia/therapy , Male , Membrane Glycoproteins/antagonists & inhibitors , Membrane Glycoproteins/genetics , Mice , Mice, Nude , Neovascularization, Physiologic/drug effects , P-Selectin/metabolism , RNA Interference , RNA, Small Interfering/genetics , Receptor, EphB4/antagonists & inhibitors , Receptor, EphB4/genetics
15.
Circulation ; 115(16): 2168-77, 2007 Apr 24.
Article in English | MEDLINE | ID: mdl-17420351

ABSTRACT

BACKGROUND: Atherosclerosis is an immunoinflammatory disease; however, the key factors responsible for the maintenance of immune regulation in a proinflammatory milieu are poorly understood. METHODS AND RESULTS: Here, we show that milk fat globule-EGF factor 8 (Mfge8, also known as lactadherin) is expressed in normal and atherosclerotic human arteries and is involved in phagocytic clearance of apoptotic cells by peritoneal macrophages. Disruption of bone marrow-derived Mfge8 in a murine model of atherosclerosis leads to substantial accumulation of apoptotic debris both systemically and within the developing lipid lesions. The accumulation of apoptotic material is associated with a reduction in interleukin-10 in the spleen but an increase in interferon-gamma production in both the spleen and the atherosclerotic arteries. In addition, we report a dendritic cell-dependent alteration of natural regulatory T-cell function in the absence of Mfge8. These events are associated with a marked acceleration of atherosclerosis. CONCLUSIONS: Lack of Mfge8 in bone marrow-derived cells enhances the accumulation of apoptotic cell corpses in atherosclerosis and alters the protective immune response, which leads to an acceleration of plaque development.


Subject(s)
Antigens, Surface/physiology , Atherosclerosis/etiology , Carotid Stenosis/metabolism , Coronary Artery Disease/metabolism , Animals , Antigens, Surface/analysis , Antigens, Surface/genetics , Apoptosis/physiology , Atherosclerosis/genetics , Atherosclerosis/pathology , Bone Marrow Transplantation , Carotid Arteries/chemistry , Carotid Stenosis/pathology , Coronary Artery Disease/pathology , Coronary Vessels/chemistry , Diet, Atherogenic , Disease Progression , Endothelial Cells/metabolism , Endothelial Cells/pathology , Endothelium, Vascular/metabolism , Endothelium, Vascular/pathology , Humans , Interferon-gamma/biosynthesis , Interferon-gamma/genetics , Interleukin-10/biosynthesis , Interleukin-10/genetics , Macrophages, Peritoneal/physiology , Male , Mice , Mice, Knockout , Milk Proteins/analysis , Milk Proteins/genetics , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/pathology , Phagocytosis/physiology , Radiation Chimera , T-Lymphocytes, Regulatory/immunology
16.
Exp Cell Res ; 313(7): 1337-46, 2007 Apr 15.
Article in English | MEDLINE | ID: mdl-17362928

ABSTRACT

We described the ex vivo production of mature and functional human smooth muscle cells (SMCs) derived from skeletal myoblasts. Initially, myoblasts expressed all myogenic cell-related markers such as Myf5, MyoD and Myogenin and differentiate into myotubes. After culture in a medium containing vascular endothelial growth factor (VEGF), these cells were shown to have adopted a differentiated SMC identity as demonstrated by alphaSMA, SM22alpha, calponin and smooth muscle-myosin heavy chain expression. Moreover, the cells cultured in the presence of VEGF did not express MyoD anymore and were unable to fuse in multinucleated myotubes. We demonstrated that myoblasts-derived SMCs (MDSMCs) interacted with endothelial cells to form, in vitro, a capillary-like network in three-dimensional collagen culture and, in vivo, a functional vascular structure in a Matrigel implant in nonobese diabetic-severe combined immunodeficient mice. Based on the easily available tissue source and their differentiation into functional SMCs, these data argue that skeletal myoblasts might represent an important tool for SMCs-based cell therapy.


Subject(s)
Cell Differentiation , Collagen/pharmacology , Laminin/pharmacology , Myoblasts, Skeletal/physiology , Myocytes, Smooth Muscle/physiology , Proteoglycans/pharmacology , Tissue Culture Techniques , Animals , Biomarkers/analysis , Cells, Cultured , Drug Combinations , Epithelial Cells/physiology , Female , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Muscle Fibers, Skeletal/physiology , Myoblasts, Skeletal/drug effects , Myoblasts, Skeletal/metabolism , Myoblasts, Skeletal/transplantation , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/transplantation , Neovascularization, Physiologic , Transplantation, Heterologous , Vascular Endothelial Growth Factor A/pharmacology
17.
Circulation ; 114(4): 328-38, 2006 Jul 25.
Article in English | MEDLINE | ID: mdl-16847153

ABSTRACT

BACKGROUND: Proangiogenic cell therapy based on administration of bone marrow-derived mononuclear cells (BMCs) or endothelial progenitor cells (EPCs) is now under investigation in humans for the treatment of ischemic diseases. However, mechanisms leading to the beneficial effects of BMCs and EPCs remain unclear. METHODS AND RESULTS: BMC- and CD34+-derived progenitor cells interacted with ischemic femoral arteries through SDF-1 and CXCR4 signaling and released nitric oxide (NO) via an endothelial nitric oxide synthase (eNOS)-dependent pathway. BMC-induced NO production promoted a marked vasodilation and disrupted vascular endothelial-cadherin/beta-catenin complexes, leading to increased vascular permeability. NO-dependent vasodilation and hyperpermeability were critical for BMC infiltration in ischemic tissues and their proangiogenic potential in a model of hindlimb ischemia in mice. CONCLUSIONS: Our results propose a new concept that proangiogenic progenitor cell activity does not rely only on their ability to differentiate into endothelial cells but rather on their capacity to modulate the function of preexisting vessels.


Subject(s)
Capillary Permeability , Ischemia/therapy , Neovascularization, Physiologic , Stem Cell Transplantation/methods , Vasodilation , Animals , Arteries/pathology , Bone Marrow Cells , Endothelial Cells , Hindlimb , Mice , Mice, Knockout , Mice, Nude , Nitric Oxide/metabolism
18.
Arterioscler Thromb Vasc Biol ; 26(4): 773-9, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16410461

ABSTRACT

BACKGROUND: We investigated the putative proangiogenic activity and molecular pathway(s) of the tetrapeptide acetyl-N-Ser-Asp-Lys-Pro (AcSDKP) in a model of surgically induced hindlimb ischemia. METHODS AND RESULTS: Hindlimb ischemia was induced by femoral artery ligature and an osmotic minipump was implanted subcutaneously to deliver low (0.12 mg/kg per day) or high (1.2 mg/kg per day) doses of AcSDKP, for 7 or 21 days. Angiography scores, arteriole density, capillary number, and foot perfusion were increased at day 21 in the high-dose AcSDKP-treated mice (by 1.9-, 1.8-, 1.3-, and 1.6-fold, respectively) compared with control animals (P<0.05, P<0.01, P<0.01, respectively). AcSDKP treatment for 24 hours upregulated the monocyte chemoattractant protein-1 (MCP-1) mRNA and protein levels by 1.5-fold in cultured endothelial cells (P<0.01). In the ischemic hindlimb model, administration of AcSDKP also enhanced MCP-1 mRNA levels by 90-fold in ischemic leg (P<0.001) and MCP-1 plasma levels by 3-fold (P<0.001 versus untreated ischemic control mice). MCP-1 levels upregulation were associated with a 2.3-fold increase in the number of Mac3-positive cells in ischemic area of AcSDKP-treated mice (P<0.001 versus untreated animals). Interestingly, AcSDKP-induced monocyte/macrophage infiltration and postischemic neovascularization was fully blunted in MCP-1-deficient animals. CONCLUSIONS: AcSDKP stimulates postischemic neovascularization through activation of a proinflammatory MCP-1-related pathway.


Subject(s)
Chemokine CCL2/physiology , Hindlimb/blood supply , Ischemia/drug therapy , Neovascularization, Physiologic/drug effects , Oligopeptides/administration & dosage , Animals , Bone Marrow Cells/pathology , Cell Differentiation , Cell Line, Transformed , Chemokine CCL2/deficiency , Endothelium, Vascular/drug effects , Endothelium, Vascular/pathology , Femoral Artery/pathology , Ischemia/physiopathology , Mice , Mice, Inbred C57BL , Monocytes/pathology , Signal Transduction/drug effects
19.
Cardiovasc Res ; 62(1): 176-84, 2004 Apr 01.
Article in English | MEDLINE | ID: mdl-15023564

ABSTRACT

OBJECTIVES: Recent studies have provided increasing evidence that postnatal neovascularization does not rely exclusively on sprouting of preexisting vessels, but also involves bone marrow-derived circulating endothelial precursors (BM-EPCs). Animal studies revealed that neovascularization of ischemic tissue can be enhanced by BM-EPCs transplantation. But a possible limitation to the use of vascular precursors for therapeutic angiogenesis is the relatively low number of these cells. In this study, we demonstrate that ex vivo expanded differentiated endothelial cells (ECs) and smooth muscle cells (SMCs), may home to the tumor vasculature allowing targeting of transgene expression to the neoangiogenic site. METHODS: Mononuclear cells (MNCs) or CD34+ -enriched cells were purified from cord blood. We have defined culture conditions in which we observed two types of clones easily differentiated according to their morphology: cobblestone or spindle-shaped. Phenotypic characterization was assessed by immunocytochemistry, flow cytometry analysis and polymerase reaction with reverse transcription. Formation of capillary-like network in vitro was studied in three-dimensional collagen culture. And recruitment of these cells to a tumoral neoangiogenic site was assessed into tumor-bearing Severe Combined Immunodeficient (SCID) mouse model. RESULTS: The cobblestone cells uniformly positive for CD31, VE-cadherin, vWF, VEGF R1 and R2, ecNOS and incorporating acetylated LDL were ECs. Spindle-shaped cells expressed alpha-smooth muscle actin (alpha-SMA), Smooth Muscle Heavy Chain (SMHC), SM22 and calponin. They also displayed a carbachol-induced contractility in a medium containing IGF1. So we concluded that spindle-shaped cells were SMCs. ECs and SMCs interacted with each other to form a capillary like network in three-dimensional type I collagen culture. Moreover, these ex vivo differentiated cells are able to home to the tumor vasculature. CONCLUSION: We provide evidence that progenitors for ECs and SMCs circulate in human cord blood and differentiate into functional ECs and SMCs. These differentiated cells could provide a biomaterial for vascular cell therapy, because of their homing capacity to the neovascularization site.


Subject(s)
Endothelial Cells/cytology , Fetal Blood , Muscle, Smooth/cytology , Stem Cells/cytology , Animals , Blotting, Western/methods , Cell Line, Tumor , Cell Movement , Flow Cytometry , Humans , Mice , Mice, SCID , Neoplasm Transplantation , Neovascularization, Pathologic , Reverse Transcriptase Polymerase Chain Reaction , Vascular Endothelial Growth Factor Receptor-1/analysis
20.
Am J Pathol ; 164(2): 457-66, 2004 Feb.
Article in English | MEDLINE | ID: mdl-14742252

ABSTRACT

Mechanisms that hinder ischemia-induced neovascularization in diabetes remain poorly understood. We hypothesized that endogenous bone marrow mononuclear cell (BM-MNC) dysfunction may contribute to the abrogated postischemic revascularization reaction associated with diabetes. We first analyzed the effect of diabetes (streptozotocin, 40 mg/kg) on BM-MNC pro-angiogenic potential in a model of surgically induced hindlimb ischemia. In nondiabetic animals, transplantation of BM-MNCs isolated from nondiabetic animals raised the ischemic/nonischemic angiographic score, capillary number, and blood flow recovery by 1.8-, 2.7-, and 2.2-fold, respectively, over that of PBS-injected nondiabetic animals (P < 0.05). Administration of diabetic BM-MNCs also improved the neovascularization reaction in ischemic hindlimbs of nondiabetic mice but to a lesser extent from that observed with nondiabetic BM-MNC transplantation. In diabetic mice, injection of nondiabetic BM-MNCs was still more efficient than that of diabetic BM-MNCs. Such BM-MNC dysfunction was associated with the impairment of diabetic BM-MNC capacity to differentiate into endothelial progenitor cells (EPCs) in vitro and to participate in vascular-like structure formation in a subcutaneous Matrigel plug. Placenta growth factor (PlGF) administration improved by sixfold the number of EPCs differentiated from diabetic BM-MNCs in vitro and enhanced ischemic/nonischemic angiographic score, capillary number and blood flow recovery by 1.9-, 1.5- and 1.6-fold, respectively, over that of untreated diabetic animals (P < 0.01). Endogenous BM-MNC pro-angiogenic potential was affected in diabetes. Therapeutic strategy based on PlGF administration restored such defects and improved postischemic neovascularization in diabetic mice.


Subject(s)
Diabetes Mellitus, Experimental/physiopathology , Ischemia/pathology , Neovascularization, Physiologic , Pregnancy Proteins/pharmacology , Angiography , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/drug effects , Bone Marrow Transplantation , Cell Differentiation/drug effects , Cells, Cultured , Hindlimb/blood supply , Hindlimb/pathology , Hindlimb/surgery , Ischemia/drug therapy , Ischemia/surgery , Laser-Doppler Flowmetry , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/pathology , Mice , Neovascularization, Physiologic/drug effects , Placenta Growth Factor
SELECTION OF CITATIONS
SEARCH DETAIL
...