Network


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

Hotspot


Dive into the research topics where Maria B. Arvelo is active.

Publication


Featured researches published by Maria B. Arvelo.


Journal of Immunology | 2003

Genetic Engineering of a Suboptimal Islet Graft with A20 Preserves β Cell Mass and Function

Shane T. Grey; Christopher R. Longo; Tala Shukri; Virendra I. Patel; Eva Csizmadia; Soizic Daniel; Maria B. Arvelo; Vaja Tchipashvili; Christiane Ferran

Transplantation of an excessive number of islets of Langerhans (two to four pancreata per recipient) into patients with type I diabetes is required to restore euglycemia. Hypoxia, nutrient deprivation, local inflammation, and the β cell inflammatory response (up-regulation of NF-κB-dependent genes such as inos) result in β cell destruction in the early post-transplantation period. Genetic engineering of islets with anti-inflammatory and antiapoptotic genes may prevent β cell loss and primary nonfunction. We have shown in vitro that A20 inhibits NF-κB activation in islets and protects from cytokine- and death receptor-mediated apoptosis. In vivo, protection of newly transplanted islets would reduce the number of islets required for successful transplantation. Transplantation of 500 B6/AF1 mouse islets into syngeneic, diabetic recipients resulted in a cure rate of 100% within 5 days. Transplantation of 250 islets resulted in a cure rate of only 20%. Transplantation of 250 islets overexpressing A20 resulted in a cure rate of 75% with a mean time to cure of 5.2 days, comparable to that achieved with 500 islets. A20-expressing islets preserve functional β cell mass and are protected from cell death. These data demonstrate that A20 is an ideal cytoprotective gene therapy candidate for islet transplantation.


The FASEB Journal | 2006

A20, a modulator of smooth muscle cell proliferation and apoptosis, prevents and induces regression of neointimal hyperplasia

Virendra I. Patel; Soizic Daniel; Christopher R. Longo; Gautam Shrikhande; Salvatore T. Scali; Eva Czismadia; Caroline M. Groft; Tala Shukri; Christina Motley-Dore; Haley Ramsey; Mark D. Fisher; Shane T. Grey; Maria B. Arvelo; Christiane Ferran

A20 is a NF‐κB‐dependent gene that has dual anti‐inflammatory and antiapoptotic functions in endothelial cells (EC). The function of A20 in smooth muscle cells (SMC) is unknown. We demonstrate that A20 is induced in SMC in response to inflammatory stimuli and serves an anti‐inflammatory function via blockade of NF‐κB and NF‐κB‐dependent proteins ICAM‐1 and MCP‐1. A20 inhibits SMC proliferation via increased expression of cyclin‐dependent kinase inhibitors p21waf1 and p27kip1. Surprisingly, A20 sensitizes SMC to cytokine‐ and Fas‐mediated apoptosis through a novel NO‐dependent mechanism. In vivo, adenoviral delivery of A20 to medial rat carotid artery SMC after balloon angioplasty prevents neointimal hyperplasia by blocking SMC proliferation and accelerating re‐endothelialization, without causing apoptosis. However, expression of A20 in established neointimal lesions leads to their regression through increased apoptosis. This is the first demonstration that A20 exerts two levels of control of vascular remodeling and healing. A20 prevents neointimal hyperplasia through combined anti‐inflammatory and antiproliferative functions in medial SMC. If SMC evade this first barrier and neointima is formed, A20 has a therapeutic potential by uniquely sensitizing neointimal SMC to apoptosis. A20‐based therapies hold promise for the prevention and treatment of neointimal disease.—Patel, V. I., Daniel, S., Longo, C. R., Shrikhande, G. V., Scali, S. T., Czismadia, E., Groft, C. M., Shukri, T., Motley‐Dore, C., Ramsey, H. E., Fisher, M. D., Grey, S. T., Arvelo, M. B., Ferran, C. A20, a modulator of smooth muscle cell proliferation and apoptosis, prevents and induces regression of neointimal hyperplasia. FASEB J. 20, 1418–1430 (2006)


Circulation | 2003

A20 Protects From CD40-CD40 Ligand-Mediated Endothelial Cell Activation and Apoptosis

Christopher R. Longo; Maria B. Arvelo; Virendra I. Patel; Soizic Daniel; Jerome Mahiou; Shane T. Grey; Christiane Ferran

Background—CD40/CD40 ligand (CD40L) signaling is a potent activator of endothelial cells (ECs) and promoter of atherosclerosis. In this study, we investigate whether A20 (a gene we have shown to be antiinflammatory and antiapoptotic in ECs) can protect from CD40/CD40L-mediated EC activation. Methods and Results—Overexpression of CD40, in a transient transfection system, activates the transcription factor NF-&kgr;B and upregulates I&kgr;B&agr;, E-selectin, and tissue factor (TF) reporter activity. Coexpression of A20 inhibits NF-&kgr;B and upregulation of I&kgr;B&agr; and E-Selectin but not TF, suggesting that CD40 induces TF in a non–NF-&kgr;B–dependent manner. In human coronary artery ECs (HCAECs), adenovirus-mediated overexpression of A20 blocks physiological, CD40-induced activation of NF-&kgr;B, upstream of I&kgr;B&agr; degradation (Western blot) and subsequently upregulation of ICAM-1, VCAM-1, and E-selectin (flow cytometry). Although A20 does not block TF transcription its expression in HCAECs inhibits TF induction (colorimetric assay and RT-PCR) by blunting CD40 upregulation. We demonstrate that CD40 signaling induces apoptosis in a proinflammatory microenvironment. A20 overexpression protects from CD40-mediated EC apoptosis (DNA content analysis and trypan blue exclusion). We also demonstrate that signaling through CD40L activates NF-&kgr;B and induces apoptosis in ECs, both of which are inhibited by A20 overexpression. Conclusion—A20 works at multiple levels to protect ECs from CD40/CD40L mediated activation and apoptosis. A20-based therapy could be beneficial for the treatment of vascular diseases such as atherosclerosis and transplant-associated vasculopathy.


Hepatology | 2005

A20 protects mice from lethal radical hepatectomy by promoting hepatocyte proliferation via a p21waf1‐dependent mechanism

Christopher R. Longo; Virendra I. Patel; Gautam Shrikhande; Salvatore T. Scali; Eva Csizmadia; Soizic Daniel; David W. Sun; Shane T. Grey; Maria B. Arvelo; Christiane Ferran

The liver has a remarkable regenerative capacity, allowing recovery following injury. Regeneration after injury is contingent on maintenance of healthy residual liver mass, otherwise fulminant hepatic failure (FHF) may arise. Understanding the protective mechanisms safeguarding hepatocytes and promoting their proliferation is critical for devising therapeutic strategies for FHF. We demonstrate that A20 is part of the physiological response of hepatocytes to injury. In particular, A20 is significantly upregulated in the liver following partial hepatectomy. A20 protects hepatocytes from apoptosis and ongoing inflammation by inhibiting NF‐κB. Hepatic expression of A20 in BALB/c mice dramatically improves survival following extended and radical lethal hepatectomy. A20 expression in the liver limits hepatocellular damage hence maintains bilirubin clearance and the liver synthetic function. In addition, A20 confers a proliferative advantage to hepatocytes via decreased expression of the cyclin‐dependent kinase inhibitor p21waf1. In conclusion, A20 provides a proliferative advantage to hepatocytes. By combining anti‐inflammatory, antiapoptotic and pro‐proliferative functions, A20‐based therapies could be beneficial in prevention and treatment of FHF. (HEPATOLOGY 2005;42:156–164.)


Biochimica et Biophysica Acta | 2013

The C-terminal domain of A1/Bfl-1 regulates its anti-inflammatory function in human endothelial cells

Renata Padilha Guedes; Eduardo Rocha; Jerome Mahiou; Herwig P. Moll; Maria B. Arvelo; Janis M. Taube; Clayton R. Peterson; Elzbieta Kaczmarek; Christopher R. Longo; Cleide G. da Silva; Christiane Ferran

A1/Bfl-1 is a NF-κB dependent, anti-apoptotic Bcl-2 family member that contains four Bcl-2 homology domains (BH) and an amphipathic C-terminal domain, and is expressed in endothelial cells (EC). Based on NF-κB reporter assays in bovine aortic EC, we have previously demonstrated that A1, like Bcl-2 and Bcl-xL, inhibits NF-κB activation. These results, however, do not fully translate when evaluating the cells own NF-κB machinery in human EC overexpressing A1 by means of recombinant adenovirus (rAd.) mediated gene transfer. Indeed, overexpression of full-length A1 in human umbilical vein EC (HUVEC), and human dermal microvascular EC (HDMEC) failed to inhibit NF-κB activation. However, overexpression of a mutant lacking the C-terminal domain of A1 (A1ΔC) demonstrated a potent NF-κB inhibitory effect in these cells. Disparate effects of A1 and A1ΔC on NF-κB inhibition in human EC correlated with mitochondrial (A1) versus non-mitochondrial (A1ΔC) localization. In contrast, both full-length A1 and A1ΔC protected EC from staurosporine (STS)-induced cell death, indicating that mitochondrial localization was not necessary for A1s cytoprotective function in human EC. In conclusion, our data uncover a regulatory role for the C-terminal domain of A1 in human EC: anchoring A1 to the mitochondrion, which conserves but is not necessary for its cytoprotective function, or by its absence freeing A1 from the mitochondrion and uncovering an additional anti-inflammatory effect.


Journal of Experimental Medicine | 1999

A20 Inhibits Cytokine-Induced Apoptosis and Nuclear Factor κB–Dependent Gene Activation in Islets

Shane T. Grey; Maria B. Arvelo; Wendy Hasenkamp; Fritz H. Bach; Christiane Ferran


Hepatology | 2002

A20 protects mice from D-galactosamine/lipopolysaccharide acute toxic lethal hepatitis☆

Maria B. Arvelo; Jeffrey T. Cooper; Christopher R. Longo; Soizic Daniel; Shane T. Grey; Jerome Mahiou; Eva Czismadia; Graziella Abu-Jawdeh; Christiane Ferran


Blood | 2004

A20 protects endothelial cells from TNF-, Fas-, and NK-mediated cell death by inhibiting caspase 8 activation

Soizic Daniel; Maria B. Arvelo; Virendra I. Patel; Christopher R. Longo; Gautam Shrikhande; Tala Shukri; Jerome Mahiou; David W. Sun; Christina Mottley; Shane T. Grey; Christiane Ferran


Transplantation | 2004

Depleting anti-CD4 monoclonal antibody cures new-onset diabetes, prevents recurrent autoimmune diabetes, and delays allograft rejection in nonobese diabetic mice

Leila Makhlouf; Shane T. Grey; Victor M. Dong; Eva Csizmadia; Maria B. Arvelo; Hugh Auchincloss; Christiane Ferran; Mohamed H. Sayegh


Kidney International | 2005

Combined expression of A1 and A20 achieves optimal protection of renal proximal tubular epithelial cells.

Uta Kunter; Soizic Daniel; Maria B. Arvelo; Jean Choi; Tala Shukri; Virendra I. Patel; Christopher R. Longo; Salvatore T. Scali; Gautam Shrikhande; Eduardo Rocha; Eva Czismadia; Christina Mottley; Shane T. Grey; Jürgen Floege; Christiane Ferran

Collaboration


Dive into the Maria B. Arvelo's collaboration.

Top Co-Authors

Avatar

Christiane Ferran

Beth Israel Deaconess Medical Center

View shared research outputs
Top Co-Authors

Avatar

Christopher R. Longo

Beth Israel Deaconess Medical Center

View shared research outputs
Top Co-Authors

Avatar

Soizic Daniel

Beth Israel Deaconess Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Virendra I. Patel

Columbia University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Gautam Shrikhande

Beth Israel Deaconess Medical Center

View shared research outputs
Top Co-Authors

Avatar

Jerome Mahiou

Beth Israel Deaconess Medical Center

View shared research outputs
Top Co-Authors

Avatar

Eva Csizmadia

Beth Israel Deaconess Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge