Network


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

Hotspot


Dive into the research topics where Rena Feinman is active.

Publication


Featured researches published by Rena Feinman.


American Journal of Physiology-gastrointestinal and Liver Physiology | 2010

HIF-1 mediates pathogenic inflammatory responses to intestinal ischemia-reperfusion injury

Rena Feinman; Edwin A. Deitch; Anthony C. Watkins; Billy Abungu; Iriana Colorado; Kolenkode B. Kannan; Sharvil U. Sheth; Francis J. Caputo; Qi Lu; Madhuri Ramanathan; Shirhan Attan; Chirag D. Badami; Danielle R. Doucet; Dimitrios Barlos; Marta Bosch-Marce; Gregg L. Semenza; Da Zhong Xu

Acute lung injury (ALI) and the development of the multiple organ dysfunction syndrome (MODS) are major causes of death in trauma patients. Gut inflammation and loss of gut barrier function as a consequence of splanchnic ischemia-reperfusion (I/R) have been implicated as the initial triggering events that contribute to the development of the systemic inflammatory response, ALI, and MODS. Since hypoxia-inducible factor (HIF-1) is a key regulator of the physiological and pathophysiological response to hypoxia, we asked whether HIF-1 plays a proximal role in the induction of gut injury and subsequent lung injury. Utilizing partially HIF-1α-deficient mice in a global trauma hemorrhagic shock (T/HS) model, we found that HIF-1 activation was necessary for the development of gut injury and that the prevention of gut injury was associated with an abrogation of lung injury. Specifically, in vivo studies demonstrated that partial HIF-1α deficiency ameliorated T/HS-induced increases in intestinal permeability, bacterial translocation, and caspase-3 activation. Lastly, partial HIF-1α deficiency reduced TNF-α, IL-1β, cyclooxygenase-2, and inducible nitric oxide synthase levels in the ileal mucosa after T/HS whereas IL-1β mRNA levels were reduced in the lung after T/HS. This study indicates that prolonged intestinal HIF-1 activation is a proximal regulator of I/R-induced gut mucosal injury and gut-induced lung injury. Consequently, these results provide unique information on the initiating events in trauma-hemorrhagic shock-induced ALI and MODS as well as potential therapeutic insights.


American Journal of Physiology-gastrointestinal and Liver Physiology | 2011

Hypoxia-inducible factor plays a gut-injurious role in intestinal ischemia reperfusion injury

Kolenkode B. Kannan; Iriana Colorado; Diego Reino; David C. Palange; Qi Lu; Xiaofa Qin; Billy Abungu; Anthony C. Watkins; Francis J. Caputo; Da-Zhong Xu; Gregg L. Semenza; Edwin A. Deitch; Rena Feinman

Gut injury and loss of normal intestinal barrier function are key elements in the paradigm of gut-origin systemic inflammatory response syndrome, acute lung injury, and multiple organ dysfunction syndrome (MODS). As hypoxia-inducible factor (HIF-1) is a critical determinant of the physiological and pathophysiological response to hypoxia and ischemia, we asked whether HIF-1 plays a proximal role in the induction of gut injury and subsequent lung injury. Using partially HIF-1α-deficient mice in an isolated superior mesenteric artery occlusion (SMAO) intestinal ischemia reperfusion (I/R) injury model (45 min SMAO followed by 3 h of reperfusion), we showed a direct relationship between HIF-1 activation and intestinal I/R injury. Specifically, partial HIF-1α deficiency attenuated SMAO-induced increases in intestinal permeability, lipid peroxidation, mucosal caspase-3 activity, and IL-1β mRNA levels. Furthermore, partial HIF-1α deficiency prevented the induction of ileal mucosal inducible nitric oxide synthase (iNOS) protein levels after SMAO and iNOS deficiency ameliorated SMAO-induced villus injury. Resistance to SMAO-induced gut injury was also associated with resistance to lung injury, as reflected by decreased levels of myeloperoxidase, IL-6 and IL-10 in the lungs of HIF-1α(+/-) mice. In contrast, a short duration of SMAO (15 min) followed by 3 h of reperfusion neither induced mucosal HIF-1α protein levels nor caused significant gut and lung injury in wild-type or HIF-1α(+/-) mice. This study indicates that intestinal HIF-1 activation is a proximal regulator of I/R-induced gut mucosal injury and gut-induced lung injury. However, the duration and severity of the gut I/R insult dictate whether HIF-1 plays a gut-protective or deleterious role.


Shock | 2012

Activation of toll-like receptor 4 is necessary for trauma hemorrhagic shock-induced gut injury and polymorphonuclear neutrophil priming.

Diego Reino; David Palange; Elenora Feketeova; Robert P. Bonitz; Da Zhong Xu; Qi Lu; Sharvil U. Sheth; Geber Peña; Luis Ulloa; Antonio De Maio; Rena Feinman; Edwin A. Deitch

ABSTRACT Interactions of toll-like receptors (TLRs) with nonmicrobial factors play a major role in the pathogenesis of early trauma-hemorrhagic shock (T/HS)–induced organ injury and inflammation. Thus, we tested the hypothesis that TLR4 mutant (TLR4mut) mice would be more resistant to T/HS-induced gut injury and polymorphonuclear neutrophil (PMN) priming than their wild-type littermates and found that both were significantly reduced in the TLR4mut mice. In addition, the in vivo and ex vivo PMN priming effect of T/HS intestinal lymph observed in the wild-type mice was abrogated in TLR4mut mice as well the TRIFmut-deficient mice and partially attenuated in Myd88−/− mice, suggesting that TRIF activation played a more predominant role than MyD88 in T/HS lymph–induced PMN priming. Polymorphonuclear neutrophil depletion studies showed that T/HS lymph–induced acute lung injury was PMN dependent, because lung injury was totally abrogated in PMN-depleted animals. Because the lymph samples were sterile and devoid of endotoxin or bacterial DNA, we investigated whether the effects of T/HS lymph was related to endogenous nonmicrobial TLR4 ligands. High-mobility group box 1 protein 1, heat shock protein 70, heat shock protein 27, and hyaluronic acid all have been implicated in ischemia-reperfusion-induced tissue injury. None of these “danger” proteins appeared to be involved, because their levels were similar between the sham and shock lymph samples. In conclusion, TLR4 activation is important in T/HS-induced gut injury and in T/HS lymph–induced PMN priming and lung injury. However, the T/HS-associated effects of TLR4 on gut barrier dysfunction can be uncoupled from the T/HS lymph–associated effects of TLR4 on PMN priming.


Cancer Letters | 2001

Tributyrin, an oral butyrate analogue, induces apoptosis through the activation of caspase-3

Kevin Clarke; Rena Feinman; Lawrence E. Harrison

The purpose of this study was to investigate the anti-proliferative and pro-apoptotic effects of the butyrate analogues, tributyrin (TB) and phenylbutyrate (PB), in a colon cancer model. We demonstrate that HT-29 colon cancer cells exposed to PB and TB result in growth inhibition associated with an induction of apoptosis mediated through the activation of caspase-3 activity. A block in the G1/S cell cycle traverse associated with a decrease in CDK2 (cyclin dependent kinase) protein levels and retinoblastoma protein hypophosphorylation was also noted after PB and TB exposure. Importantly, TB proved to be the most potent agent in its ability to induce these phenotypic changes, and potentially may represent a novel therapy for patients with advanced colorectal cancer.


Cancer Chemotherapy and Pharmacology | 2002

Phenylbutyrate-induced apoptosis is associated with inactivation of NF-κB IN HT-29 colon cancer cells

Rena Feinman; Kevin Clarke; Lawrence E. Harrison

Abstract. Purpose: Cytotoxic chemotherapy has been used to treat patients with metastatic colorectal cancer with limited success. Therefore novel chemotherapeutic approaches are needed. Based on encouraging preclinical data, there has been an interest in developing derivatives of butyrate as clinically applicable agents. The purpose of this study was to investigate the effects of phenylbutyrate (PB), a butyrate analogue, on the cell growth and apoptosis in a colon cancer cell model. Methods: Growth curves, flow cytometric studies, Western blotting, DNA binding assays and transient transfection experiments were performed in vitro using the colon cancer cell line HT-29 after exposure to PB. Results: Exposure of HT-29 colon cancer cells to PB resulted in growth inhibition and induction of apoptosis as measured by annexin V staining. This increase in apoptosis was associated with a decrease in mitochondrial membrane potential, an increase in caspase-3 activity and a decrease in intact PARP protein levels. Since NF-κB plays a pivotal role in the regulation of apoptosis, we explored the effects of PB on the DNA binding and transcriptional activity of this transcription factor. After PB treatment, NF-κB-DNA binding was markedly decreased and specifically, this decreased DNA binding was observed in the p50:p65 heterodimer. The decreased NF-κB DNA binding was observed as early as 3xa0h after PB treatment, while no apparent changes in annexin V binding were detected until 12xa0h after PB treatment. Untreated HT-29 cells transfected with a κB-luciferase reporter plasmid demonstrated significant constitutive activity of the κB binding site, which was markedly decreased after treating the cells with PB. Conclusion: These results suggest that PB-induced apoptosis may be partly regulated through the inactivation of NF-κB. PB, an oral butyrate analogue, may have therapeutic potential in colon cancer.


Journal of Trauma-injury Infection and Critical Care | 2010

Estrogenic Hormone Modulation Abrogates Changes in Red Blood Cell Deformability and Neutrophil Activation in Trauma Hemorrhagic Shock

Danielle Doucet; R Paul Bonitz; Rena Feinman; Iriana Colorado; Mahdury Ramanathan; Eleanora Feketeova; Michael R. Condon; George W. Machiedo; Carl J. Hauser; Da-Zhong Xu; Edwin A. Deitch

BACKGROUNDnDecreased red blood cell (RBC) deformability and activation of neutrophils (polymorphonuclear leukocytes [PMN]) after trauma-hemorrhagic shock (T/HS) have been implicated in the development of multiple organ dysfunction. Experimentally, female animals seemed to be protected from the effects of T/HS, at least in part, because of elevated estrogen levels. Thus, we examined the relative role of estrogen receptor (ER)-alpha and -beta in this protective response.nnnMETHODSnTo accomplish this goal, RBC deformability and neutrophil respiratory burst activity were measured in several groups of hormonally intact or ovariectomized (OVX) female rats subjected to T/HS (laparotomy plus hemorrhage to an MAP of 30 mm Hg to 35 mm Hg for 90 minutes) or trauma-sham shock (T/SS) and 3 hours of reperfusion. These groups included rats receiving vehicle, estradiol, or either an ER-alpha agonist or an ER-beta agonist administered at the end of the shock period just before volume resuscitation.nnnRESULTSnRBC deformability and neutrophil activation were similar among all the T/SS groups and were not different from that observed in the non-OVX female rats subjected to T/HS. In contrast, RBC deformability was reduced and neutrophil activation was increased in the OVX, T/HS female rats as compared with the T/SS groups or the non-OVX, T/HS rats. The administration of estrogen to the T/HS, OVX rats returned RBC and neutrophil function to normal. Both the ER-alpha and -beta agonist partially, but not completely, protected the OVX rats from T/HS-induced loss of RBC deformability, whereas only the ER-beta agonist prevented the increase in neutrophil activation.nnnCONCLUSIONSnThe protective effects of estrogen on T/HS-induced RBC deformability are mediated, at least in part, via activation of both ER-alpha and -beta, whereas ER-beta activation is involved in limiting T/HS-induced neutrophil activation.


Shock | 2007

Molecular signatures of trauma-hemorrhagic shock-induced lung injury: hemorrhage- and injury-associated genes.

Rena Feinman; Edwin A. Deitch; Aris; Chu Hb; Billy Abungu; Francis J. Caputo; Galante A; Da Zhong Xu; Qi Lu; Colorado I; Streck D; Dermody J; Soteropoulos P

The etiology of trauma-hemorrhagic shock (T/HS)-induced acute lung injury has been difficult to elucidate because of, at least in part, the inability of in vivo studies to separate the noninjurious pulmonary effects of trauma-hemorrhage from the tissue-injurious ones. To circumvent this in vivo limitation, we used a model of T/HS in which T/HS lung injury was abrogated by dividing the mesenteric lymph duct. In this way, it was possible to separate the pulmonary injurious response from the noninjurious systemic response to T/HS by comparing the pulmonary molecular responses of rats subjected to T/HS, which did and did not develop lung injury, with those of nonshocked rats. Using high-density oligonucleotide arrays and treatment group comparisons of whole lung tissue collected at 3 h after the end of the shock or sham-shock period, 139 of 8,799 assessed genes were identified by significant analysis of microarrays. Hemorrhage without the secondary effects of lung injury modulated the expression of 21 genes such as interleukin 1&bgr;, metallothionein-2, and myeloctomatosis oncogene (c-myc). In response to injury, 42 genes were identified to be differentially expressed. Upregulated genes included the L1 retroposon and guanine deaminase, whereas downregulated genes included catalase and superoxide dismutase 1. Real-time polymerase chain reaction confirmed the differential expression for selected genes. PathwayAssist analysis identified interleukin 1&bgr; as a central regulator of two subpathways of stress response-related genes (c-myc and superoxide dismutase 1/catalase) as well as several unrelated genes such as lipoprotein lipase. Our model system provided a unique opportunity to distinguish the molecular changes associated with T/HS-induced acute lung injury from the systemic molecular response to T/HS.


Clinical Cancer Research | 2000

Changes in E2F Binding after Phenylbutyrate-induced Differentiation of Caco-2 Colon Cancer Cells

Qing Mei Wang; Rena Feinman; Fatah Kashanchi; Jean Marie Houghton; George P. Studzinski; Lawrence E. Harrison


Shock | 2006

PARTIAL HIF-1?? DEFICIENCY ATTENUATES ISCHEMIA REPERFUSION-MEDIATED GUT AND LUNG INJURY

Anthony C. Watkins; Billy Abungu; M. Sentil; Qi Lu; Dimitrios Barlos; L. Prescott; Francis J. Caputo; B.K. McCracken; Da Zhong Xu; M. Bosch-Marce; G.L. Semenza; Edwin A. Deitch; Rena Feinman


Shock | 2006

ROLE OF AIF IN TRAUMA HEMORRHAGIC SHOCK-INDUCED ENDOTHELIAL APOPTOSIS

Dimitrios Barlos; Anthony C. Watkins; Billy Abungu; Qi Lu; Francis J. Caputo; Da Zhong Xu; Rena Feinman; Edwin A. Deitch

Collaboration


Dive into the Rena Feinman's collaboration.

Top Co-Authors

Avatar

Edwin A. Deitch

University of Medicine and Dentistry of New Jersey

View shared research outputs
Top Co-Authors

Avatar

Qi Lu

University of Medicine and Dentistry of New Jersey

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Francis J. Caputo

University of Medicine and Dentistry of New Jersey

View shared research outputs
Top Co-Authors

Avatar

Anthony C. Watkins

University of Medicine and Dentistry of New Jersey

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge