Network


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

Hotspot


Dive into the research topics where Arturo Bravo-Nuevo is active.

Publication


Featured researches published by Arturo Bravo-Nuevo.


Current Eye Research | 2004

Resistance of photoreceptors in the C57BL/6-c2J, C57BL/6J, and BALB/cJ mouse strains to oxygen stress: Evidence of an oxygen phenotype

Natalie Walsh; Arturo Bravo-Nuevo; Scott F. Geller; Jonathan Stone

Purpose. To assess the vulnerability of retinal photoreceptors in the BALB/cJ, C57BL/6J, and C57BL/6-c2J (c2J) mouse strains to hypoxic and hyperoxic stress. Methods. Mice were raised in dim cyclic light. Pups aged postnatal day 7 (P7) were exposed to hypoxia (11–12% oxygen) for periods up to 23 days. Adult mice were exposed to either hypoxia (12% oxygen) or to hyperoxia (75% oxygen) for up to 2 weeks. Using the TUNEL (terminal dUTP-mediated nick end labeling) technique retinas were examined for cell death. Results. In juvenile mice, hypoxia induced a robust increase in photoreceptor death in the C57BL/6J strain and a weaker increase in the C57BL/6-c2J strains. In the adult, hypoxia was associated with a small reduction in photoreceptor death in the C57BL/6-c2J strains. Hyperoxia caused substantial photoreceptor death in both the C57BL/6-c2J and C57BL/6J strains. The BALB/cJ strain was more resistant to oxygen stress than the C57BL strains. Conclusions. The difference in oxygen vulnerability between C57BL/6J and BALB/c strains may provide a useful starting point for the analysis of genetic regulation of this vulnerability. The resistance of the C57BL/6-c2J substrains to hypoxia may reflect their degenerative status.


Cancer Research | 2013

RhoB differentially controls Akt function in tumor cells and stromal endothelial cells during breast tumorigenesis

Shiva Kazerounian; Damien Gerald; Minzhou Huang; Y. Rebecca Chin; Durga Udayakumar; Ningning Zheng; Rebekah K. O'Donnell; Carole Perruzzi; Lee Mangiante; Jacob Pourat; Thuy L. Phung; Arturo Bravo-Nuevo; Sharon Shechter; Stephanie McNamara; James B. DuHadaway; Olivier Kocher; Lawrence F. Brown; Alex Toker; George C. Prendergast; Laura E. Benjamin

Tumors are composed of cancer cells but also a larger number of diverse stromal cells in the tumor microenvironment. Stromal cells provide essential supports to tumor pathophysiology but the distinct characteristics of their signaling networks are not usually considered in developing drugs to target tumors. This oversight potentially confounds proof-of-concept studies and increases drug development risks. Here, we show in established murine and human models of breast cancer how differential regulation of Akt by the small GTPase RhoB in cancer cells or stromal endothelial cells determines their dormancy versus outgrowth when angiogenesis becomes critical. In cancer cells in vitro or in vivo, RhoB functions as a tumor suppressor that restricts EGF receptor (EGFR) cell surface occupancy as well as Akt signaling. However, after activation of the angiogenic switch, RhoB functions as a tumor promoter by sustaining endothelial Akt signaling, growth, and survival of stromal endothelial cells that mediate tumor neoangiogenesis. Altogether, the positive impact of RhoB on angiogenesis and progression supercedes its negative impact in cancer cells themselves. Our findings elucidate the dominant positive role of RhoB in cancer. More generally, they illustrate how differential gene function effects on signaling pathways in the tumor stromal component can complicate the challenge of developing therapeutics to target cancer pathophysiology.


International Immunology | 2011

RhoB deficiency in thymic medullary epithelium leads to early thymic atrophy.

Arturo Bravo-Nuevo; Rebekah K. O'Donnell; Alexander Rosendahl; Jae Hoon Chung; Laura E. Benjamin; Chikako Odaka

RhoB, a member of the Rho subfamily of small GTPases, mediates diverse cellular functions, including cytoskeletal organization, cell transformation and vesicle trafficking. The thymus undergoes progressive decline in its structure and function after puberty. We found that RhoB was expressed in thymic medullary epithelium. To investigate a role of RhoB in the regulation of thymic epithelial organization or thymocyte development, we analyzed the thymi of RhoB-deficient mice. RhoB-deficient mice were found to display earlier thymic atrophy. RhoB deficiency showed significant reductions in thymus weight and cellularity, beginning as early as 5 weeks of age. The enhanced expression of TGF-β receptor type II (TGFβRII) in thymic medullary epithelium was observed in RhoB-null mice. In addition, the expression of fibronectin, which is shown to be regulated by TGF-β signaling, was accordingly increased in the mutant thymic medulla. Since there is no age-related change of RhoB expression in the thymus, it is unlikely that RhoB in thymic epithelium directly contributes to age-related thymic involution. Nevertheless, our findings strongly support a physiological role of RhoB in regulation of thymus development and maintenance through the inhibition of TGF-β signaling in thymic medullary epithelium.


Advances in Experimental Medicine and Biology | 2003

Mitochondrial Deletions in Normal and Degenerating Rat Retina

Arturo Bravo-Nuevo; Neal K. Williams; Scott F. Geller; Jonathan Stone

Photoreceptor death by apoptosis is the central pathology of most forms of retinal degeneration. Mitochondria play key roles in apoptosis, releasing both signals which induce apoptosis (cytochrome c, caspases) and signals which inhibit apoptosis (Bcl-2). Because mitochondria are the site of oxidative metabolism they are also a major site of formation of the toxic oxygen intermediates which form as oxygen is recruited into the oxidative phosphorylation pathway. Previous studies have shown that deletions in mtDNA accumulate in postmitotic tissues (central nervous, muscle) and that their accumulation is accelerated by oxidative stress (such as hypoxia) (Takeda et al. 1996; Lee et al. 1994; Merril et al. 1996; Englander et al. 1999). It seems possible therefore that mitochondria are a site at which oxidative stress induces the death of retinal neurones. This study investigates the accumulation of mtDNA deletions in the rat retina, in both normal (non-degenerative) and degenerative strains. Deletions were undetectable in Sprague-Dawley albino rats (24 months) but were detected at 15 months in the rapidly degenerating RCS strain. The appearance of deletions in the RCS strain, in which retinal oxygen tension is known to rise as the degeneration progresses, gives support to the ideas that oxidative stress is a factor in mtDNA deletions, and in the progress of the late stages of the degeneration.


EBioMedicine | 2016

IDO1 is an Integral Mediator of Inflammatory Neovascularization

Arpita Mondal; Courtney Smith; James B. DuHadaway; Erika Sutanto-Ward; George C. Prendergast; Arturo Bravo-Nuevo; Alexander J. Muller

The immune tolerogenic effects of IDO1 (indoleamine 2,3-dioxygenase 1) have been well documented and genetic studies in mice have clearly established the significance of IDO1 in tumor promotion. Dichotomously, the primary inducer of IDO1, the inflammatory cytokine IFNγ (interferon-γ), is a key mediator of immune-based tumor suppression. One means by which IFNγ can exert an anti-cancer effect is by decreasing tumor neovascularization. We speculated that IDO1 might contribute to cancer promotion by countering this anti-neovascular effect of IFNγ, possibly through IDO1-potentiated elevation of the pro-tumorigenic inflammatory cytokine IL6 (interleukin-6). In this study, we investigated how genetic loss of IDO1 affects neovascularization in mouse models of oxygen-induced retinopathy and lung metastasis. Neovascularization in both models was significantly reduced in mice lacking IDO1, was similarly reduced with loss of IL6, and was restored in both cases by concomitant loss of IFNγ. Likewise, the lack of IDO1 or IL6 resulted in reduced metastatic tumor burden and increased survival, which the concomitant loss of IFNγ abrogated. This insight into IDO1s involvement in pro-tumorigenic inflammatory neovascularization may have important ramifications for IDO1 inhibitor development, not only in cancer where clinical trials are currently ongoing, but in other disease indications associated with neovascularization as well.


American Journal of Pathology | 2011

RhoB Loss Prevents Streptozotocin-Induced Diabetes and Ameliorates Diabetic Complications in Mice

Arturo Bravo-Nuevo; Hikaru Sugimoto; Seema Iyer; Zachary Fallon; Jason M. Lucas; Shiva Kazerounian; George C. Prendergast; Raghu Kalluri; Nathan I. Shapiro; Laura E. Benjamin

RhoB is an early-response gene whose expression is elevated by multiple cellular stresses; this gene plays an important role in cancer, macrophage motility, and apoptosis. These factors are essential for the onset of type 1 diabetes mellitus and related complications. This study explores the role of RhoB in β-cell depletion and hyperglycemia-associated complications and tests whether the pleiotropic effect of statins on glycemic control is RhoB dependent. We induced β-cell depletion in RhoB(+/+), RhoB(+/-), and RhoB(-/-) mice with streptozotocin (STZ). Diabetic status was assessed by glucose tolerance and pancreatic islet loss. RhoB(-/-) mice showed a significant reduction in the severity of STZ-induced diabetes; only 13% of the STZ-treated RhoB-null animals became hyperglycemic, as opposed to 61% of the wild-type controls. Diabetes-related complications, such as wound healing rate and onset of nephropathy, were also assessed. Hyperglycemic RhoB(-/-) mice had fewer signs of nephropathy and showed faster wound healing than RhoB(+/+) animals. After assessing the diabetic status of mice treated simultaneously with STZ and simvastatin, we conclude that the effect of statins in improving glycemic control is RhoB independent. We propose that RhoB is a modifier of diabetes, important for the induction of β-cell loss. Suppression of RhoB expression may have potential application in the treatment of diabetes and associated complications.


PLOS ONE | 2014

Myo/Nog Cells: Targets for Preventing the Accumulation of Skeletal Muscle-Like Cells in the Human Lens

Jacquelyn Gerhart; Marvin Greenbaum; Victoria Scheinfeld; Paul B. Fitzgerald; Mitchell Crawford; Arturo Bravo-Nuevo; Meghan Pitts; Mindy George-Weinstein

Posterior capsule opacification (PCO) is a vision impairing condition that arises in some patients following cataract surgery. The fibrotic form of PCO is caused by myofibroblasts that may emerge in the lens years after surgery. In the chick embryo lens, myofibroblasts are derived from Myo/Nog cells that are identified by their expression of the skeletal muscle specific transcription factor MyoD, the bone morphogenetic protein inhibitor Noggin, and the epitope recognized by the G8 monoclonal antibody. The goal of this study was to test the hypothesis that depletion of Myo/Nog cells will prevent the accumulation of myofibroblasts in human lens tissue. Myo/Nog cells were present in anterior, equatorial and bow regions of the human lens, cornea and ciliary processes. In anterior lens tissue removed by capsulorhexis, Myo/Nog cells had synthesized myofibroblast and skeletal muscle proteins, including vimentin, MyoD and sarcomeric myosin. Alpha smooth muscle actin (α-SMA) was detected in a subpopulation of Myo/Nog cells. Areas of the capsule denuded of epithelial cells were surrounded by Myo/Nog cells. Some of these cell free areas contained a wrinkle in the capsule. Depletion of Myo/Nog cells eliminated cells expressing skeletal muscle proteins in 5-day cultures but did not affect cells immunoreactive for beaded filament proteins that accumulate in differentiating lens epithelial cells. Transforming growth factor-betas 1 and 2 that mediate an epithelial-mesenchymal transition, did not induce the expression of skeletal muscle proteins in lens cells following Myo/Nog cell depletion. This study demonstrates that Myo/Nog cells in anterior lens tissue removed from cataract patients have undergone a partial differentiation to skeletal muscle. Myo/Nog cells appear to be the source of skeletal muscle-like cells in explants of human lens tissue. Targeting Myo/Nog cells with the G8 antibody during cataract surgery may reduce the incidence of PCO.


Experimental Eye Research | 2016

Neuroprotective effect of Myo/Nog cells in the stressed retina

Arturo Bravo-Nuevo; Alice Brandli; Jacquelyn Gerhart; Jennifer Nichols; Meghan Pitts; Christopher K. Sutera; Sarah Assali; Victoria Scheinfeld; George C. Prendergast; Jonathan Stone; Mindy George-Weinstein

Myo/Nog cells are essential for eye development in the chick embryo and respond to injury in adult tissues. These cells express mRNA for the skeletal muscle specific transcription factor MyoD, the bone morphogenetic protein (BMP) inhibitor Noggin and the cell surface protein recognized by the G8 monoclonal antibody (mAb). In this study, we determined that Myo/Nog cells are present in low numbers in the retina of the mouse eye. G8-positive Myo/Nog cells were distinguished from neuronal, Müller and microglial cells that were identified with antibodies to calretinin, Chx10, glial fibrillary acidic protein and ionized calcium binding adaptor molecule 1, respectively. In the neonatal retina, the number of Myo/Nog cells increased in parallel with cell death induced by transient exposure to hyperoxia. In this model of retinopathy of prematurity, depletion of Myo/Nog cells by intravitreal injection of the G8 mAb and complement increased cell death. These findings demonstrate that Myo/Nog cells are a distinct population of cells, not previously described in the retina, which increases in response to retinal damage and mitigate hypoxia-induced cell death.


PLOS ONE | 2014

Meglumine exerts protective effects against features of metabolic syndrome and type II diabetes.

Arturo Bravo-Nuevo; Alice Marcy; Minzhou Huang; Frank Kappler; Jennifer Mulgrew; Lisa Laury-Kleintop; Melvin Reichman; Annette Tobia; George C. Prendergast

Metabolic syndrome, diabetes and diabetes complications pose a growing medical challenge worldwide, accentuating the need of safe and effective strategies for their clinical management. Here we present preclinical evidence that the sorbitol derivative meglumine (N-methyl-D-glucamine) can safely protect against several features of metabolic syndrome and diabetes, as well as elicit enhancement in muscle stamina. Meglumine is a compound routinely used as an approved excipient to improve drug absorption that has not been ascribed any direct biological effects in vivo. Normal mice (SV129) administered 18 mM meglumine orally for six weeks did not display any gastrointestinal or other observable adverse effects, but had a marked effect on enhancing muscle stamina and at longer times in limiting weight gain. In the established KK.Cg-Ay/J model of non-insulin dependent diabetes, oral administration of meglumine significantly improved glycemic control and significantly lowered levels of plasma and liver triglycerides. Compared to untreated control animals, meglumine reduced apparent diabetic nephropathy. Sorbitol can improve blood glucose uptake by liver and muscle in a manner associated with upregulation of the AMPK-related enzyme SNARK, but with undesirable gastrointestinal side effects not seen with meglumine. In murine myoblasts, we found that meglumine increased steady-state SNARK levels in a dose-dependent manner more potently than sorbitol. Taken together, these findings provide support for the clinical evaluation of meglumine as a low-cost, safe supplement offering the potential to improve muscle function, limit metabolic syndrome and reduce diabetic complications.


PLOS ONE | 2017

Role of Myo/Nog Cells in Neuroprotection: Evidence from the Light Damaged Retina.

Alice Brandli; Jacquelyn Gerhart; Christopher K. Sutera; Sivaraman Purushothuman; Mindy George-Weinstein; Jonathan Stone; Arturo Bravo-Nuevo

Purpose To identify Myo/Nog cells in the adult retina and test their role in protecting retinal photoreceptors from light damage. Methods Light damage was induced by exposing albino rats raised in dim cyclic light to 1000 lux light for 24 hours. In one group of rats, Myo/Nog cells were purified from rat brain tissue by magnetic cell sorting following binding of the G8 monoclonal antibody (mAb). These cells were injected into the vitreous humour of the eye within 2 hours following bright light exposure. Retinal function was assessed using full-field, flash electroretinogram (ERG) before and after treatment. The numbers of Myo/Nog cells, apoptotic photoreceptors, and the expression of glial fibrillary acidic protein (GFAP) in Muller cells were assessed by immunohistochemistry. Results Myo/Nog cells were present in the undamaged retina in low numbers. Light induced damage increased their numbers, particularly in the choroid, ganglion cell layer and outer plexiform layer. Intravitreal injection of G8-positive (G8+) cells harvested from brain mitigated all the effects of light damage examined, i.e. loss of retinal function (ERG), death of photoreceptors and the stress-induced expression of GFAP in Muller cells. Some of the transplanted G8+ cells were integrated into the retina from the vitreous. Conclusions Myo/Nog cells are a subpopulation of cells that are present in the adult retina. They increase in number in response to light induced stress. Intravitreal injection of Myo/Nog cells was protective to the retina, in part, by reducing retinal stress as measured by the Muller cell response. These results suggest that Myo/Nog cells, or the factors they produce, are neuroprotective and may be therapeutic in neurodegenerative retinal diseases.

Collaboration


Dive into the Arturo Bravo-Nuevo's collaboration.

Top Co-Authors

Avatar

George C. Prendergast

Lankenau Institute for Medical Research

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Alexander J. Muller

Lankenau Institute for Medical Research

View shared research outputs
Top Co-Authors

Avatar

Arpita Mondal

Lankenau Institute for Medical Research

View shared research outputs
Top Co-Authors

Avatar

Jacquelyn Gerhart

Lankenau Institute for Medical Research

View shared research outputs
Top Co-Authors

Avatar

James B. DuHadaway

Lankenau Institute for Medical Research

View shared research outputs
Top Co-Authors

Avatar

Mindy George-Weinstein

Lankenau Institute for Medical Research

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Courtney Smith

Lankenau Institute for Medical Research

View shared research outputs
Top Co-Authors

Avatar

Erika Sutanto-Ward

Lankenau Institute for Medical Research

View shared research outputs
Researchain Logo
Decentralizing Knowledge