Network


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

Hotspot


Dive into the research topics where Steven A. Garzon is active.

Publication


Featured researches published by Steven A. Garzon.


The FASEB Journal | 2013

Targeting sphingosine kinase 1 attenuates bleomycin-induced pulmonary fibrosis

Long Shuang Huang; Evgeny Berdyshev; Biji Mathew; Panfeng Fu; Irina Gorshkova; Donghong He; Wenli Ma; Imre Noth; Shwu Fan Ma; Srikanth Pendyala; Sekhar P. Reddy; Tong Zhou; Wei Zhang; Steven A. Garzon; Joe G. N. Garcia; Viswanathan Natarajan

Idiopathic pulmonary fibrosis (IPF) is a chronic and progressive interstitial lung disease, wherein transforming growth factor β (TGF‐β) and sphingosine‐1‐phosphate (S1P) contribute to the pathogenesis of fibrosis. However, the in vivo contribution of sphingosine kinase (SphK) in fibrotic processes has not been documented. Microarray analysis of blood mononuclear cells from patients with IPF and SphK1‐ or SphK2‐knockdown mice and SphK inhibitor were used to assess the role of SphKs in fibrogenesis. The expression of SphK1/2 negatively correlated with lung function and survival in patients with IPF. Also, the expression of SphK1 was increased in lung tissues from patients with IPF and bleomycin‐challenged mice. Knockdown of SphK1, but not SphK2, increased survival and resistance to pulmonary fibrosis in bleomycin‐challenged mice. Administration of SphK inhibitor reduced bleomycin‐induced mortality and pulmonary fibrosis in mice. Knockdown of SphK1 or treatment with SphK inhibitor attenuated S1P generation and TGF‐β secretion in a bleomycin‐induced lung fibrosis mouse model that was accompanied by reduced phosphorylation of Smad2 and MAPKs in lung tissue. In vitro, bleomycin‐induced expression of SphK1 in lung fibroblast was found to be TGF‐β dependent. Taken together, these data indicate that SphK1 plays a critical role in the pathology of lung fibrosis and is a novel therapeutic target.—Huang, L. S., Berdyshev, E., Mathew, B., Fu, P., Gorshkova, I. A., He, D., Ma, W., Noth, I., Ma, S.‐F., Pendyala, S., Reddy, S. P., Zhou, T., Zhang, W., Garzon, S. A., Garcia, J. G. N., Natarajan, V. Targeting sphingosine kinase 1 attenuates bleomycin‐induced pulmonary fibrosis. FASEB J. 27, 1749–1760 (2013). www.fasebj.org


American Journal of Physiology-gastrointestinal and Liver Physiology | 2012

Gut mucosal injury in neonates is marked by macrophage infiltration in contrast to pleomorphic infiltrates in adult: evidence from an animal model

Krishnan MohanKumar; Niroop Kaza; Ramasamy Jagadeeswaran; Steven A. Garzon; Anchal Bansal; Ashish Kurundkar; Kopperuncholan Namachivayam; Juan I. Remon; C. Rekha Bandepalli; Xu Feng; Joern Hendrik Weitkamp

Necrotizing enterocolitis (NEC) is an inflammatory bowel necrosis of premature infants. In tissue samples of NEC, we identified numerous macrophages and a few neutrophils but not many lymphocytes. We hypothesized that these pathoanatomic characteristics of NEC represent a common tissue injury response of the gastrointestinal tract to a variety of insults at a specific stage of gut development. To evaluate developmental changes in mucosal inflammatory response, we used trinitrobenzene sulfonic acid (TNBS)-induced inflammation as a nonspecific insult and compared mucosal injury in newborn vs. adult mice. Enterocolitis was induced in 10-day-old pups and adult mice (n = 25 animals per group) by administering TNBS by gavage and enema. Leukocyte populations were enumerated in human NEC and in murine TNBS-enterocolitis using quantitative immunofluorescence. Chemokine expression was measured using quantitative polymerase chain reaction, immunoblots, and immunohistochemistry. Macrophage recruitment was investigated ex vivo using intestinal tissue-conditioned media and bone marrow-derived macrophages in a microchemotaxis assay. Similar to human NEC, TNBS enterocolitis in pups was marked by a macrophage-rich leukocyte infiltrate in affected tissue. In contrast, TNBS-enterocolitis in adult mice was associated with pleomorphic leukocyte infiltrates. Macrophage precursors were recruited to murine neonatal gastrointestinal tract by the chemokine CXCL5, a known chemoattractant for myeloid cells. We also demonstrated increased expression of CXCL5 in surgically resected tissue samples of human NEC, indicating that a similar pathway was active in NEC. We concluded that gut mucosal injury in the murine neonate is marked by a macrophage-rich leukocyte infiltrate, which contrasts with the pleomorphic leukocyte infiltrates in adult mice. In murine neonatal enterocolitis, macrophages were recruited to the inflamed gut mucosa by the chemokine CXCL5, indicating that CXCL5 and its cognate receptor CXCR2 merit further investigation as potential therapeutic targets in NEC.


American Journal of Physiology-gastrointestinal and Liver Physiology | 2013

Smad7 inhibits autocrine expression of TGF-β2 in intestinal epithelial cells in baboon necrotizing enterocolitis

Kopperuncholan Namachivayam; Cynthia L. Blanco; Krishnan MohanKumar; Ramasamy Jagadeeswaran; Margarita M. Vasquez; Lisa L. McGill-Vargas; Steven A. Garzon; Sunil K. Jain; Ravinder K. Gill; Nancy E. Freitag; Jörn-Hendrik Weitkamp; Steven R. Seidner

Preterm infants may be at risk of necrotizing enterocolitis (NEC) due to deficiency of transforming growth factor-β 2 (TGF-β(2)) in the developing intestine. We hypothesized that low epithelial TGF-β(2) expression in preterm intestine and during NEC results from diminished autocrine induction of TGF-β(2) in these cells. Premature baboons delivered at 67% gestation were treated per current norms for human preterm infants. NEC was diagnosed by clinical and radiological findings. Inflammatory cytokines, TGF-β(2), Smad7, Ski, and strawberry notch N (SnoN)/Ski-like oncoprotein (SKIL) was measured using quantitative reverse transcriptase-polymerase chain reaction, immunoblots, and immunohistochemistry. Smad7 effects were examined in transfected IEC6 intestinal epithelial cells in vitro. Findings were validated in archived human tissue samples of NEC. NEC was recorded in seven premature baboons. Consistent with existing human data, premature baboon intestine expressed less TGF-β(2) than term intestine. TGF-β(2) expression was regulated in epithelial cells in an autocrine fashion, which was interrupted in the premature intestine and during NEC due to increased expression of Smad7. LPS increased Smad7 binding to the TGF-β(2) promoter and was associated with dimethylation of the lysine H3K9, a marker of transcriptional silencing, on the nucleosome of TGF-β(2). Increased Smad7 expression in preterm intestine was correlated with the deficiency of SnoN/SKIL, a repressor of the Smad7 promoter. Smad7 inhibits autocrine expression of TGF-β(2) in intestinal epithelial cells in the normal premature intestine and during NEC. Increased Smad7 expression in the developing intestine may be due to a developmental deficiency of the SnoN/SKIL oncoprotein.


American Journal of Physiology-gastrointestinal and Liver Physiology | 2013

Preterm human milk contains a large pool of latent TGF-β, which can be activated by exogenous neuraminidase

Kopperuncholan Namachivayam; Cynthia L. Blanco; Brandy L. Frost; Aaron A. Reeves; Ramasamy Jagadeeswaran; Krishnan MohanKumar; Azif Safarulla; Partha Mandal; Steven A. Garzon; J. Usha Raj

Human milk contains substantial amounts of transforming growth factor (TGF)-β, particularly the isoform TGF-β2. We previously showed in preclinical models that enterally administered TGF-β2 can protect against necrotizing enterocolitis (NEC), an inflammatory bowel necrosis of premature infants. In this study we hypothesized that premature infants remain at higher risk of NEC than full-term infants, even when they receive their own mothers milk, because preterm human milk contains less bioactive TGF-β than full-term milk. Our objective was to compare TGF-β bioactivity in preterm vs. full-term milk and identify factors that activate milk-borne TGF-β. Mothers who delivered between 23 0/7 and 31 6/7 wk or at ≥37 wk of gestation provided milk samples at serial time points. TGF-β bioactivity and NF-κB signaling were measured using specific reporter cells and in murine intestinal tissue explants. TGF-β1, TGF-β2, TGF-β3, and various TGF-β activators were measured by real-time PCR, enzyme immunoassays, or established enzymatic activity assays. Preterm human milk showed minimal TGF-β bioactivity in the native state but contained a large pool of latent TGF-β. TGF-β2 was the predominant isoform of TGF-β in preterm milk. Using a combination of several in vitro and ex vivo models, we show that neuraminidase is a key regulator of TGF-β bioactivity in human milk. Finally, we show that addition of bacterial neuraminidase to preterm human milk increased TGF-β bioactivity. Preterm milk contains large quantities of TGF-β, but most of it is in an inactive state. Addition of neuraminidase can increase TGF-β bioactivity in preterm milk and enhance its anti-inflammatory effects.


Pediatric Infectious Disease Journal | 2012

Absence of gastrointestinal pathogens in ileum tissue resected for necrotizing enterocolitis.

Tim Ullrich; Yi-Wei Tang; Hernan Correa; Steven A. Garzon; Melissa Hill; Pranathi Matta; Mohan K. Krishnan; Jörn-Hendrik Weitkamp

Necrotizing enterocolitis (NEC) is one of the most common gastrointestinal emergencies in premature infants and has been linked with viral antigens in as much as 40% of cases in single-center cohorts. We examined 28 tissue sections from surgically resected ileum from 27 preterm infants with NEC from 2 separate institutions for 15 common bacterial, viral, and parasitic gastrointestinal pathogens using multiplex reverse transcriptase polymerase chain reaction amplification and suspension array detection methods. We did not detect infectious enteritis pathogens in any of the NEC tissues and conclude that gastrointestinal pathogens are a rare cause of NEC.


Pediatric Research | 2016

Smad7 interrupts TGF-β signaling in intestinal macrophages and promotes inflammatory activation of these cells during necrotizing enterocolitis

Krishnan MohanKumar; Kopperuncholan Namachivayam; Kalyan C. Chapalamadugu; Steven A. Garzon; Muralidhar H. Premkumar; Srinivas M. Tipparaju

Background:Necrotizing enterocolitis (NEC) is an inflammatory bowel necrosis of premature infants. Based on our recent findings of increased Smad7 expression in surgically resected bowel affected by NEC, we hypothesized that NEC macrophages undergo inflammatory activation because increased Smad7 expression renders these cells resistant to normal, gut-specific, transforming growth factor (TGF)-β-mediated suppression of inflammatory pathways.Methods:We used surgically resected human NEC tissue, murine models of NEC-like injury, bone marrow-derived and intestinal macrophages, and RAW264.7 cells. Smad7 and IκB kinase-beta (IKK-β) were measured by quantitative PCR, western blots, and immunohistochemistry. Promoter activation was confirmed in luciferase reporter and chromatin immunoprecipitation assays.Results:NEC macrophages showed increased Smad7 expression, particularly in areas with severe tissue damage and high bacterial load. Lipopolysaccharide-induced Smad7 expression suppressed TGF-β signaling and augmented nuclear factor-kappa B (NF-κB) activation and cytokine production in macrophages. Smad7-mediated NF-κB activation was likely mediated via increased expression of IKK-β, which, further increased Smad7 expression in a feed-forward loop. We show that Smad7 induced IKK-β expression through direct binding to the IKK-β promoter and its transcriptional activation.Conclusion:Smad7 expression in NEC macrophages interrupts TGF-β signaling and promotes NF-κB-mediated inflammatory signaling in these cells through increased expression of IKK-β.


Journal of Perinatology | 2015

Depth of bacterial invasion in resected intestinal tissue predicts mortality in surgical necrotizing enterocolitis.

Juan I. Remon; Sachin C. Amin; Sangeeta R. Mehendale; Rakesh Rao; Angel A. Luciano; Steven A. Garzon

Objective:Up to a third of all infants who develop necrotizing enterocolitis (NEC) require surgical resection of necrotic bowel. We hypothesized that the histopathological findings in surgically resected bowel can predict the clinical outcome of these infants.Study Design:We reviewed the medical records and archived pathology specimens from all patients who underwent bowel resection/autopsy for NEC at a regional referral center over a 10-year period. Pathology specimens were graded for the depth and severity of necrosis, inflammation, bacteria invasion and pneumatosis, and histopathological findings were correlated with clinical outcomes.Result:We performed clinico-pathological analysis on 33 infants with confirmed NEC, of which 18 (54.5%) died. Depth of bacterial invasion in resected intestinal tissue predicted death from NEC (odds ratio 5.39 per unit change in the depth of bacterial invasion, 95% confidence interval 1.33 to 21.73). The presence of transmural necrosis and bacteria in the surgical margins of resected bowel was also associated with increased mortality.Conclusion:Depth of bacterial invasion in resected intestinal tissue predicts mortality in surgical NEC.


Ultrasound Quarterly | 2015

Ultrasound Imaging of Placenta Accreta with MR Correlation

Winnie Mar; Senta Berggruen; Ukamaka Atueyi; Simran Sekhon; Steven A. Garzon; M. Knuttinen; John P. McGahan

Abstract Placenta accreta is abnormal placental adherence or invasion of the myometrium or extrauterine structures. It is increasing in incidence because of increasing number of cesarean sections and is one of the main causes of excessive postpartum hemorrhage. Recognition of this entity is crucial because improved outcomes have been shown when the antenatal diagnosis of placenta accreta is made. Ultrasound is the first-line tool; magnetic resonance imaging (MRI) is complementary. Ultrasound and MRI features and MRI protocols will be reviewed.


Laboratory Investigation | 2014

Intestinal epithelial apoptosis initiates gut mucosal injury during extracorporeal membrane oxygenation in the newborn piglet

Krishnan MohanKumar; Cheryl R. Killingsworth; R. Britt McILwain; Joseph G. Timpa; Ramasamy Jagadeeswaran; Kopperuncholan Namachivayam; Ashish Kurundkar; David R. Kelly; Steven A. Garzon

Neonates and young infants exposed to extracorporeal circulation during extracorporeal membrane oxygenation (ECMO) and cardiopulmonary bypass are at risk of developing a systemic inflammatory response syndrome with multi-organ dysfunction. We used a piglet model of ECMO to investigate the hypothesis that epithelial apoptosis is an early event that precedes villous damage during ECMO-related bowel injury. Healthy 3-week-old piglets were subjected to ECMO for up to 8 h. Epithelial apoptosis was measured in histopathological analysis, nuclear imaging, and terminal deoxynucleotidyl transferase dUTP nick end labeling. Plasma intestinal fatty acid-binding protein (I-FABP) levels were measured by enzyme immunoassay. Intestinal mast cells were isolated by fluorescence-assisted cell sorting. Cleaved caspase-8, caspase-9, phospho-p38 MAPK, and fas ligand expression were investigated by immunohistochemistry, western blots, and reverse transcriptase-quantitative PCR. Piglet ECMO was associated with increased gut epithelial apoptosis. Extensive apoptotic changes were noted on villus tips and in scattered crypt cells after 2 h of ECMO. After 8 h, the villi were denuded and apoptotic changes were evident in a majority of crypt cells. Increased circulating I-FABP levels, a marker of gut epithelial injury, showed that epithelial injury occurred during ECMO. We detected increased cleaved caspase-8, but not cleaved caspase-9, in epithelial cells indicating that the extrinsic apoptotic pathway was active. ECMO was associated with increased fas ligand expression in intestinal mast cells, which was induced through activation of the p38 mitogen-activated protein kinase. We conclude that epithelial apoptosis is an early event that initiates gut mucosal injury in a piglet model of ECMO.


American Journal of Physiology-gastrointestinal and Liver Physiology | 2015

Transforming growth factor-β2 is sequestered in preterm human milk by chondroitin sulfate proteoglycans

Kopperuncholan Namachivayam; Hayley Coffing; Nehru Viji Sankaranarayanan; Yingzi Jin; Krishnan MohanKumar; Brandy L. Frost; Cynthia L. Blanco; Aloka L. Patel; Paula P. Meier; Steven A. Garzon; Umesh R. Desai

Human milk contains biologically important amounts of transforming growth factor-β2 isoform (TGF-β2), which is presumed to protect against inflammatory gut mucosal injury in the neonate. In preclinical models, enterally administered TGF-β2 can protect against experimental necrotizing enterocolitis, an inflammatory bowel necrosis of premature infants. In this study, we investigated whether TGF-β bioactivity in human preterm milk could be enhanced for therapeutic purposes by adding recombinant TGF-β2 (rTGF-β2) to milk prior to feeding. Milk-borne TGF-β bioactivity was measured by established luciferase reporter assays. Molecular interactions of TGF-β2 were investigated by nondenaturing gel electrophoresis and immunoblots, computational molecular modeling, and affinity capillary electrophoresis. Addition of rTGF-β2 (20-40 nM) to human preterm milk samples failed to increase TGF-β bioactivity in milk. Milk-borne TGF-β2 was bound to chondroitin sulfate (CS) containing proteoglycan(s) such as biglycan, which are expressed in high concentrations in milk. Chondroitinase treatment of milk increased the bioactivity of both endogenous and rTGF-β2, and consequently, enhanced the ability of preterm milk to suppress LPS-induced NF-κB activation in macrophages. These findings provide a mechanism for the normally low bioavailability of milk-borne TGF-β2 and identify chondroitinase digestion of milk as a potential therapeutic strategy to enhance the anti-inflammatory effects of preterm milk.

Collaboration


Dive into the Steven A. Garzon's collaboration.

Top Co-Authors

Avatar

Krishnan MohanKumar

University of Illinois at Chicago

View shared research outputs
Top Co-Authors

Avatar

Kopperuncholan Namachivayam

University of Illinois at Chicago

View shared research outputs
Top Co-Authors

Avatar

Cynthia L. Blanco

University of Texas Health Science Center at San Antonio

View shared research outputs
Top Co-Authors

Avatar

Ramasamy Jagadeeswaran

University of Illinois at Chicago

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Aloka L. Patel

Rush University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Ashish Kurundkar

University of Alabama at Birmingham

View shared research outputs
Top Co-Authors

Avatar

Elizabeth L. Wiley

University of Texas Southwestern Medical Center

View shared research outputs
Top Co-Authors

Avatar

Hayley Coffing

University of Illinois at Chicago

View shared research outputs
Top Co-Authors

Avatar

Juan I. Remon

University of Illinois at Chicago

View shared research outputs
Researchain Logo
Decentralizing Knowledge