Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Jérôme Roncalli is active.

Publication


Featured researches published by Jérôme Roncalli.


Acta Biomaterialia | 2010

Development of Bioactive Peptide Amphiphiles for Therapeutic Cell Delivery

Matthew J. Webber; Jörn Tongers; Marie Ange Renault; Jérôme Roncalli; Douglas W. Losordo; Samuel I. Stupp

There is great clinical interest in cell-based therapies for ischemic tissue repair in cardiovascular disease. However, the regenerative potential of these therapies is limited due to poor cell viability and minimal retention following application. We report here the development of bioactive peptide amphiphile nanofibers displaying the fibronectin-derived RGDS cell adhesion epitope as a scaffold for therapeutic delivery of bone marrow derived stem and progenitor cells. When grown on flat substrates, a binary peptide amphiphile system consisting of 10 wt.% RGDS-containing molecules and 90wt.% negatively charged diluent molecules was found to promote optimal cell adhesion. This binary system enhanced adhesion 1.4-fold relative to substrates composed of only the non-bioactive diluent. Additionally, no enhancement was found upon scrambling the epitope and adhesion was no longer enhanced upon adding soluble RGDS to the cell media, indicating RGDS-specific adhesion. When encapsulated within self-assembled scaffolds of the binary RGDS nanofibers in vitro, cells were found to be viable and proliferative, increasing in number by 5.5 times after only 5 days, an effect again lost upon adding soluble RGDS. Cells encapsulated within a non-bioactive scaffold and those within a binary scaffold with scrambled epitope showed minimal viability and no proliferation. Cells encapsulated within this RGDS nanofiber gel also increase in endothelial character, evident by a decrease in the expression of CD34 paired with an increase in the expression of endothelial-specific markers VE-Cadherin, VEGFR2 and eNOS after 5 days. In an in vivo study, nanofibers and luciferase-expressing cells were co-injected subcutaneously in a mouse model. The binary RGDS material supported these cells in vivo, evident by a 3.2-fold increase in bioluminescent signal attributable to viable cells; this suggests the material has an anti-apoptotic and/or proliferative effect on the transplanted bone marrow cells. We conclude that the binary RGDS-presenting nanofibers developed here demonstrate enhanced viability, proliferation and adhesion of associated bone marrow derived stem and progenitor cells. This study suggests potential for this material as a scaffold to overcome current limitations of stem cell therapies for ischemic diseases.


Circulation | 2008

Therapeutic Angiogenesis for Critical Limb Ischemia Microvascular Therapies Coming of Age

Jörn Tongers; Jérôme Roncalli; Douglas W. Losordo

Despite progressive insights into the pathologies underlying coronary, cerebral, and peripheral artery atherosclerosis, these conditions continue to cause critical tissue ischemia and disability on an epidemic scale. For the past several decades, research and therapeutic development have focused on preventing or reversing occlusive disease in conduit vessels. The ultimate failure of macrovessel-targeted therapies is never more evident than in peripheral arterial disease, in which progressive disease leads to amputation at rates that have not changed significantly in 30 years. Despite modern therapy, up to 8 million Americans with peripheral arterial disease are devastated by immobility, intractable ischemia, ulceration, impaired wound healing, or amputation,1 and the lack of additional treatment options leaves many patients with little hope for relief. Article p 58 The concept of therapeutic angiogenesis evolved from pioneering work in the 1970s by Folkman,2 who observed that the development and maintenance of an adequate microvascular supply is essential for the growth of neoplastic tissue. His hypothesis that the inhibition of “tumor angiogenic factors” would be effective against solid tumors was met with widespread skepticism, but 30 years of persistent research led to the development and approval of antiangiogenic treatments that now constitute a significant portion of the anticancer armamentarium. Soon after the identification of angiogenic growth factors, cardiovascular investigators began testing the hypothesis that stimulating angiogenesis could improve perfusion and function in ischemic tissues independent of macrovessel manipulation.3 Abundant preclinical data supported the safety and clinical potential of therapeutic angiogenesis that used growth factors or cellular-based strategies.4,5 Ac


Microvascular Research | 2010

Role of endothelial progenitor cells during ischemia-induced vasculogenesis and collateral formation.

Jörn Tongers; Jérôme Roncalli; Douglas W. Losordo

Cell-based therapy has emerged as a promising therapeutic tool for treatment of ischemic cardiovascular disease. Both unselected bone marrow-derived mononuclear cells (BMNCs), which include stem/progenitor cells and several other cell types, and endothelial progenitor cells (EPCs), a subpopulation of BMNCs, display regenerative potential in ischemic tissue. Abundant evidence supports the involvement of EPCs in capillary growth, and EPCs also appear to participate in the formation of collateral vessels. Collectively, these effects have led to improved perfusion and functional recovery in animal models of myocardial and peripheral ischemia, and in early clinical trials, the therapeutic administration of EPCs to patients with myocardial infarction or chronic angina has been associated with positive trends in perfusion. EPCs also contribute to endothelial repair and may, consequently, impede the development or progression of arteriosclerosis. This review provides a brief summary of the preclinical and clinical evidence for the role of EPCs in blood-vessel formation and repair during ischemic cardiovascular disease.


Trends in Biotechnology | 2008

Endothelial progenitor cells in regenerative medicine and cancer: a decade of research

Jérôme Roncalli; Jörn Tongers; Marie-Ange Renault; Douglas W. Losordo

Endothelial progenitor cells (EPCs) are a heterogeneous subpopulation of bone marrow mononuclear cells that have an enhanced potential for differentiation within the endothelial cell lineage. In response to ischemic injury, EPCs are mobilized from the bone marrow to the peripheral circulation and home to the sites of new vessel growth, where they become incorporated into the growing vasculature. Thus, EPCs can be therapeutically useful for treating ischemic injury or for delivering anti-cancer agents to tumors.


Circulation | 2013

CXC-Chemokine Receptor 4 Antagonist AMD3100 Promotes Cardiac Functional Recovery After Ischemia/Reperfusion Injury via Endothelial Nitric Oxide Synthase–Dependent Mechanism

Kentaro Jujo; Masaaki; Haruki Sekiguchi; Ekaterina Klyachko; Sol Misener; Toshikazu Tanaka; Jörn Tongers; Jérôme Roncalli; Marie Ange Renault; Tina Thorne; Aiko Ito; Trevor Clarke; Christine Kamide; Yukio Tsurumi; Nobuhisa Hagiwara; Gangjian Qin; Michio Asahi; Douglas W. Losordo

Background— CXC-chemokine receptor 4 (CXCR4) regulates the retention of stem/progenitor cells in the bone marrow (BM), and the CXCR4 antagonist AMD3100 improves recovery from coronary ligation injury by mobilizing stem/progenitor cells from the BM to the peripheral blood. Thus, we investigated whether AMD3100 also improves recovery from ischemia/reperfusion injury, which more closely mimics myocardial infarction in patients, because blood flow is only temporarily obstructed. Methods and Results— Mice were treated with single subcutaneous injections of AMD3100 (5 mg/kg) or saline after ischemia/reperfusion injury. Three days later, histological measurements of the ratio of infarct area to area at risk were smaller in AMD3100-treated mice than in mice administered saline, and echocardiographic measurements of left ventricular function were greater in the AMD3100-treated mice at week 4. CXCR4+ cells were mobilized for just 1 day in both groups, but the mobilization of sca1+/flk1+ cells endured for 7 days in AMD3100-treated mice compared with just 1 day in the saline-treated mice. AMD3100 upregulated BM levels of endothelial nitric oxide synthase (eNOS) and 2 targets of eNOS signaling, matrix metalloproteinase-9 and soluble Kit ligand. Furthermore, the loss of BM eNOS expression abolished the benefit of AMD3100 on sca1+/flk1+ cell mobilization without altering the mobilization of CXCR4+ cells, and the cardioprotective effects of AMD3100 were retained in eNOS-knockout mice that had been transplanted with BM from wild-type mice but not in wild-type mice with eNOS-knockout BM. Conclusions— AMD3100 prolongs BM progenitor mobilization and improves recovery from ischemia/reperfusion injury, and these benefits appear to occur through a previously unidentified link between AMD3100 and BM eNOS expression.


Journal of the American College of Cardiology | 2011

Sonic-hedgehog–induced functional recovery after myocardial infarction is enhanced by AMD3100-mediated progenitor-cell mobilization

Jérôme Roncalli; Marie-Ange Renault; Jörn Tongers; Sol Misener; Tina Thorne; Christine Kamide; Kentaro Jujo; Toshikazu Tanaka; Masaaki; Ekaterina Klyachko; Douglas W. Losordo

OBJECTIVES This study was designed to compare the effectiveness of Sonic hedgehog (Shh) gene transfer, AMD3100-induced progenitor-cell mobilization, and Shh-AMD3100 combination therapy for treatment of surgically induced myocardial infarction (MI) in mice. BACKGROUND Shh gene transfer improves myocardial recovery by up-regulating angiogenic genes and enhancing the incorporation of bone marrow-derived progenitor cells (BMPCs) in infarcted myocardium. Here, we investigated whether the effectiveness of Shh gene therapy could be improved with AMD3100-induced progenitor-cell mobilization. METHODS Gene expression and cell function were evaluated in cells cultured with medium collected from fibroblasts transfected with plasmids encoding human Shh (phShh). MI was induced in wild-type mice, in matrix metalloproteinase (MMP)-9 knockout mice, and in mice transplanted with bone marrow that expressed green-fluorescent protein. Mice were treated with 100 μg of phShh (administered intramyocardially), 5 mg/kg of AMD3100 (administered subcutaneously), or both; cardiac function was evaluated echocardiographically, and fibrosis, capillary density, and BMPC incorporation were evaluated immunohistochemically. RESULTS phShh increased vascular endothelial growth factor and stromal cell-derived factor 1 expression in fibroblasts; the medium from phShh-transfected fibroblasts increased endothelial-cell migration and the migration, proliferation, and tube formation of BMPCs. Combination therapy enhanced cardiac functional recovery (i.e., left ventricular ejection fraction) in wild-type mice, but not in MMP-9 knockout mice, and was associated with less fibrosis, greater capillary density and smooth muscle-containing vessel density, and enhanced BMPC incorporation. CONCLUSIONS Combination therapy consisting of intramyocardial Shh gene transfer and AMD3100-induced progenitor-cell mobilization improves cardiac functional recovery after MI and is superior to either individual treatment for promoting therapeutic neovascularization.


Journal of Molecular and Cellular Cardiology | 2014

Enhanced Potency of Cell-based Therapy for Ischemic Tissue Repair Using an Injectable Bioactive Epitope-presenting Nanofiber Support Matrix

Jörn Tongers; Matthew J. Webber; Erin E Vaughan; Eduard Sleep; Marie Ange Renault; Jérôme Roncalli; Ekaterina Klyachko; Tina Thorne; Yang Yu; Katja Theres Marquardt; Christine Kamide; Aiko Ito; Sol Misener; Meredith Millay; Ting Liu; Kentaro Jujo; Gangjian Qin; Douglas W. Losordo; Samuel I. Stupp; Raj Kishore

The translation of cell-based therapies for ischemic tissue repair remains limited by several factors, including poor cell survival and limited target site retention. Advances in nanotechnology enable the development of specifically designed delivery matrices to address these limitations and thereby improve the efficacy of cell-based therapies. Given the relevance of integrin signaling for cellular homeostasis, we developed an injectable, bioactive peptide-based nanofiber matrix that presents an integrin-binding epitope derived from fibronectin, and evaluated its feasibility as a supportive artificial matrix for bone marrow-derived pro-angiogenic cells (BMPACs) used as a therapy in ischemic tissue repair. Incubation of BMPACs with these peptide nanofibers in vitro significantly attenuated apoptosis while enhancing proliferation and adhesion. Pro-angiogenic function was enhanced, as cells readily formed tubes. These effects were, in part, mediated via p38, and p44/p42 MAP kinases, which are downstream pathways of focal adhesion kinase. In a murine model of hind limb ischemia, an intramuscular injection of BMPACs within this bioactive peptide nanofiber matrix resulted in greater retention of cells, enhanced capillary density, increased limb perfusion, reduced necrosis/amputation, and preserved function of the ischemic limb compared to treatment with cells alone. This self-assembling, bioactive peptide nanofiber matrix presenting an integrin-binding domain of fibronectin improves regenerative efficacy of cell-based strategies in ischemic tissue by enhancing cell survival, retention, and reparative functions.


Expert Review of Cardiovascular Therapy | 2008

Biological approaches to ischemic tissue repair: gene- and cell-based strategies

Jérôme Roncalli; Jörn Tongers; Marie-Ange Renault; Douglas W. Losordo

Gene therapy is a potential therapeutic strategy for treatment of ischemic vascular diseases; however, the clinical application of gene therapy has met some anticipated challenges. Recent randomized, controlled trials suggest that patients with cardiovascular disease may also benefit from cell-based therapies, and the optimal treatment regimen may combine both approaches to take advantage of potential synergy between the underlying therapeutic mechanisms. This review discusses recent research into both gene and cell therapy and considers the potential application of a combined treatment approach for cardiovascular and cerebrovascular ischemic diseases.


Archives of Cardiovascular Diseases | 2010

Update on gene therapy for myocardial ischaemia and left ventricular systolic dysfunction or heart failure

Jérôme Roncalli; Jörn Tongers; Douglas W. Losordo

Despite considerable advances in pharmacological, surgical and technology-based cardiovascular therapy, left ventricular dysfunction and heart failure are increasingly prevalent health problems. Recent studies suggest that angiogenic gene therapy can restore perfusion in ischaemic myocardial tissue, and that the transfer of nonangiogenic genes may correct defects in calcium handling that contribute to abnormal contractile function in patients with heart failure; however, large clinical trials of gene therapy for treatment of left ventricular dysfunction and heart failure have yet to be completed, and only a small number of genes have been evaluated in patients. Researchers continue to investigate new genes, combinations of genes and approaches that combine gene and cell therapy, and to develop novel expression vectors and delivery systems; collectively, these refinements promise to improve both patient response and safety.


Acta Biomaterialia | 2015

Reprint of: Development of bioactive peptide amphiphiles for therapeutic cell delivery

Matthew J. Webber; Jörn Tongers; Marie Ange Renault; Jérôme Roncalli; Douglas W. Losordo; Samuel I. Stupp

There is great clinical interest in cell-based therapies for ischemic tissue repair in cardiovascular disease. However, the regenerative potential of these therapies is limited due to poor cell viability and minimal retention following application. We report here the development of bioactive peptide amphiphile nanofibers displaying the fibronectin-derived RGDS cell adhesion epitope as a scaffold for therapeutic delivery of bone marrow derived stem and progenitor cells. When grown on flat substrates, a binary peptide amphiphile system consisting of 10 wt.% RGDS-containing molecules and 90 wt.% negatively charged diluent molecules was found to promote optimal cell adhesion. This binary system enhanced adhesion 1.4-fold relative to substrates composed of only the non-bioactive diluent. Additionally, no enhancement was found upon scrambling the epitope and adhesion was no longer enhanced upon adding soluble RGDS to the cell media, indicating RGDS-specific adhesion. When encapsulated within self-assembled scaffolds of the binary RGDS nanofibers in vitro, cells were found to be viable and proliferative, increasing in number by 5.5 times after only 5 days, an effect again lost upon adding soluble RGDS. Cells encapsulated within a non-bioactive scaffold and those within a binary scaffold with scrambled epitope showed minimal viability and no proliferation. Cells encapsulated within this RGDS nanofiber gel also increase in endothelial character, evident by a decrease in the expression of CD34 paired with an increase in the expression of endothelial-specific markers VE-Cadherin, VEGFR2 and eNOS after 5days. In an in vivo study, nanofibers and luciferase-expressing cells were co-injected subcutaneously in a mouse model. The binary RGDS material supported these cells in vivo, evident by a 3.2-fold increase in bioluminescent signal attributable to viable cells; this suggests the material has an anti-apoptotic and/or proliferative effect on the transplanted bone marrow cells. We conclude that the binary RGDS-presenting nanofibers developed here demonstrate enhanced viability, proliferation and adhesion of associated bone marrow derived stem and progenitor cells. This study suggests potential for this material as a scaffold to overcome current limitations of stem cell therapies for ischemic diseases.

Collaboration


Dive into the Jérôme Roncalli's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Sol Misener

Northwestern University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Tina Thorne

Northwestern University

View shared research outputs
Top Co-Authors

Avatar

Gangjian Qin

Northwestern University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Aiko Ito

Northwestern University

View shared research outputs
Top Co-Authors

Avatar

Kentaro Jujo

Northwestern University

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge