Network


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

Hotspot


Dive into the research topics where Clayton R. Peterson is active.

Publication


Featured researches published by Clayton R. Peterson.


Hepatology | 2013

A20 promotes liver regeneration by decreasing SOCS3 expression to enhance IL‐6/STAT3 proliferative signals

Cleide G. da Silva; Peter Studer; Marco Skroch; Jerome Mahiou; Darlan C. Minussi; Clayton R. Peterson; Suzhuei W. Wilson; Virendra I. Patel; Averil Ma; Eva Csizmadia; Christiane Ferran

Liver regeneration is of major clinical importance in the setting of liver injury, resection, and transplantation. A20, a potent antiinflammatory and nuclear factor kappa B (NF‐κB) inhibitory protein, has established pro‐proliferative properties in hepatocytes, in part through decreasing expression of the cyclin dependent kinase inhibitor, p21. Both C‐terminal (7‐zinc fingers; 7Zn) and N‐terminal (Nter) domains of A20 were required to decrease p21 and inhibit NF‐κB. However, both independently increased hepatocyte proliferation, suggesting that additional mechanisms contributed to the pro‐proliferative function of A20 in hepatocytes. We ascribed one of A20′s pro‐proliferative mechanisms to increased and sustained interleukin (IL)‐6‐induced signal transducer and activator of transcription 3 (STAT3) phosphorylation, as a result of decreased hepatocyte expression of the negative regulator of IL‐6 signaling, suppressor of cytokine signaling 3 (SOCS3). This novel A20 function segregates with its 7Zn not Nter domain. Conversely, total and partial loss of A20 in hepatocytes increased SOCS3 expression, hampering IL‐6‐induced STAT3 phosphorylation. Following liver resection in mice pro‐proliferative targets downstream of IL‐6/STAT3 signaling were increased by A20 overexpression and decreased by A20 knockdown. In contrast, IL‐6/STAT3 proinflammatory targets were increased in A20‐deficient livers, and decreased or unchanged in A20 overexpressing livers. Upstream of SOCS3, levels of its microRNA regulator miR203 were significantly decreased in A20‐deficient livers. Conclusion: A20 enhances IL‐6/STAT3 pro‐proliferative signals in hepatocytes by down‐regulating SOCS3, likely through a miR203‐dependent manner. This finding together with A20 reducing the levels of the potent cell cycle brake p21 establishes its pro‐proliferative properties in hepatocytes and prompts the pursuit of A20‐based therapies to promote liver regeneration and repair. (HEPATOLOGY 2013)


PLOS ONE | 2010

O-Glycosylation Regulates Ubiquitination and Degradation of the Anti-Inflammatory Protein A20 to Accelerate Atherosclerosis in Diabetic ApoE-Null Mice

Gautam Shrikhande; Salvatore T. Scali; Cleide G. da Silva; Scott M. Damrauer; Eva Csizmadia; Prabhakar Putheti; Michaela Matthey; Roy Arjoon; Rakesh Patel; Jeffrey J. Siracuse; Elizabeth R. Maccariello; Nicholas D. Andersen; Thomas S. Monahan; Clayton R. Peterson; Sanah Essayagh; Peter Studer; Renata Padilha Guedes; Olivier Kocher; Anny Usheva; Aristidis Veves; Elzbieta Kaczmarek; Christiane Ferran

Background Accelerated atherosclerosis is the leading cause of morbidity and mortality in diabetic patients. Hyperglycemia is a recognized independent risk factor for heightened atherogenesis in diabetes mellitus (DM). However, our understanding of the mechanisms underlying glucose damage to the vasculature remains incomplete. Methodology/Principal Findings High glucose and hyperglycemia reduced upregulation of the NF-κB inhibitory and atheroprotective protein A20 in human coronary endothelial (EC) and smooth muscle cell (SMC) cultures challenged with Tumor Necrosis Factor alpha (TNF), aortae of diabetic mice following Lipopolysaccharide (LPS) injection used as an inflammatory insult and in failed vein-grafts of diabetic patients. Decreased vascular expression of A20 did not relate to defective transcription, as A20 mRNA levels were similar or even higher in EC/SMC cultured in high glucose, in vessels of diabetic C57BL/6 and FBV/N mice, and in failed vein grafts of diabetic patients, when compared to controls. Rather, decreased A20 expression correlated with post-translational O-Glucosamine-N-Acetylation (O-GlcNAcylation) and ubiquitination of A20, targeting it for proteasomal degradation. Restoring A20 levels by inhibiting O-GlcNAcylation, blocking proteasome activity, or overexpressing A20, blocked upregulation of the receptor for advanced glycation end-products (RAGE) and phosphorylation of PKCβII, two prime atherogenic signals triggered by high glucose in EC/SMC. A20 gene transfer to the aortic arch of diabetic ApoE null mice that develop accelerated atherosclerosis, attenuated vascular expression of RAGE and phospho-PKCβII, significantly reducing atherosclerosis. Conclusions High glucose/hyperglycemia regulate vascular A20 expression via O-GlcNAcylation-dependent ubiquitination and proteasomal degradation. This could be key to the pathogenesis of accelerated atherosclerosis in diabetes.


Transplantation | 2012

A20-mediated Modulation of Inflammatory and Immune Responses in Aortic Allografts and Development of Transplant Arteriosclerosis

Jeffrey J. Siracuse; Mark D. Fisher; Cleide G. da Silva; Clayton R. Peterson; Eva Csizmadia; Herwig P. Moll; Scott M. Damrauer; Peter Studer; Lynn E. Choi; Sanah Essayagh; Elzbieta Kaczmarek; Elizabeth R. Maccariello; Andy Lee; Soizic Daniel; Christiane Ferran

Background. Transplant arteriosclerosis (TA) is the pathognomonic feature of chronic rejection, the primary cause of allograft failure. We have shown that the NF-&kgr;B inhibitory protein A20 exerts vasculoprotective effects in endothelial and smooth muscle cells (SMC), and hence is a candidate to prevent TA. We sought direct proof for this hypothesis. Methods. Fully mismatched, C57BL/6 (H2b) into BALB/c (H2d), aorta to carotid allografts were preperfused with saline, recombinant A20 adenovirus (rAd.A20) or rAd.&bgr;-galactosidase (&bgr;-gal), implanted, harvested 4 weeks after transplantation, and analyzed by histology, immunohistochemistry, and immunofluorescence staining. We measured indoleamine 2,3-dioxygenase, interleukin-6, and transforming growth factor-&bgr; mRNA and protein levels in nontransduced, and rAd.A20 or rAd.&bgr;-gal-transduced human SMC cultures after cytokine treatment. Results. Vascular overexpression of A20 significantly reduced TA lesions. This correlated with decreased graft inflammation and increased apoptosis of neointimal SMC. Paradoxically, T-cell infiltrates increased in A20-expressing allografts, including the immunoprivileged media, which related to A20 preventing indoleamine 2,3-dioxygenase upregulation in SMC. However, infiltrating T cells were predominantly T-regulatory cells (CD25+/Forkhead Box P3 [FoxP3+]). This agrees with A20 inhibiting interleukin-6 and promoting transforming growth factor-&bgr; production by medial SMC and in SMC cultures exposed to cytokines, which favors differentiation of regulatory over pathogenic T cells. Conclusions. In summary, A20 prevents immune-mediated remodeling of vascular allografts, therefore reduces TA lesions by affecting apoptotic and inflammatory signals and modifying the local cytokine milieu to promote an immunoregulatory response within the vessel wall. This highlights a novel function for A20 in local immunosurveillance, which added to its vasculoprotective effects, supports its therapeutic promise in TA.


Journal of Cellular Physiology | 2012

Hepatocyte growth factor preferentially activates the anti‐inflammatory arm of NF‐κB signaling to induce A20 and protect renal proximal tubular epithelial cells from inflammation

Cleide G. da Silva; Elizabeth R. Maccariello; Szuhuei Wu Wilson; Prabhakar Putheti; Soizic Daniel; Scott M. Damrauer; Clayton R. Peterson; Jeffrey J. Siracuse; Elzbieta Kaczmarek; Christiane Ferran

Inflammation induces the NF‐κB dependent protein A20 in human renal proximal tubular epithelial cells (RPTEC), which secondarily contains inflammation by shutting down NF‐κB activation. We surmised that inducing A20 without engaging the pro‐inflammatory arm of NF‐κB could improve outcomes in kidney disease. We showed that hepatocyte growth factor (HGF) increases A20 mRNA and protein levels in RPTEC without causing inflammation. Upregulation of A20 by HGF was NF‐κB/RelA dependent as it was abolished by overexpressing IκBα or silencing p65/RelA. Unlike TNFα, HGF caused minimal IκBα and p65/RelA phosphorylation, with moderate IκBα degradation. Upstream, HGF led to robust and sustained AKT activation, which was required for p65 phosphorylation and A20 upregulation. While HGF treatment of RPTEC significantly increased A20 mRNA, it failed to induce NF‐κB dependent, pro‐inflammatory MCP‐1, VCAM‐1, and ICAM‐1 mRNA. This indicates that HGF preferentially upregulates protective (A20) over pro‐inflammatory NF‐κB dependent genes. Upregulation of A20 supported the anti‐inflammatory effects of HGF in RPTEC. HGF pretreatment significantly attenuated TNFα‐mediated increase of ICAM‐1, a finding partially reversed by silencing A20. In conclusion, this is the first demonstration that HGF activates an AKT‐p65/RelA pathway to preferentially induce A20 but not inflammatory molecules. This could be highly desirable in acute and chronic renal injury where A20‐based anti‐inflammatory therapies are beneficial. J. Cell. Physiol. 227: 1382–1390, 2012.


Cell Death & Differentiation | 2015

Significant lethality following liver resection in A20 heterozygous knockout mice uncovers a key role for A20 in liver regeneration.

Peter Studer; da Silva Cg; Revuelta Cervantes Jm; Alessandra Mele; Eva Csizmadia; Jeffrey J. Siracuse; Scott M. Damrauer; Clayton R. Peterson; D. Candinas; Stroka Dm; Averil Ma; Manoj Bhasin; Christiane Ferran

Hepatic expression of A20, including in hepatocytes, increases in response to injury, inflammation and resection. This increase likely serves a hepatoprotective purpose. The characteristic unfettered liver inflammation and necrosis in A20 knockout mice established physiologic upregulation of A20 as integral to the anti-inflammatory and anti-apoptotic armamentarium of hepatocytes. However, the implication of physiologic upregulation of A20 in modulating hepatocytes’ proliferative responses following liver resection remains controversial. To resolve the impact of A20 on hepatocyte proliferation and the liver’s regenerative capacity, we examined whether decreased A20 expression, as in A20 heterozygous knockout mice, affects outcome following two-third partial hepatectomy. A20 heterozygous mice do not demonstrate a striking liver phenotype, indicating that their A20 expression levels are still sufficient to contain inflammation and cell death at baseline. However, usually benign partial hepatectomy provoked a staggering lethality (>40%) in these mice, uncovering an unsuspected phenotype. Heightened lethality in A20 heterozygous mice following partial hepatectomy resulted from impaired hepatocyte proliferation due to heightened levels of cyclin-dependent kinase inhibitor, p21, and deficient upregulation of cyclins D1, E and A, in the context of worsened liver steatosis. A20 heterozygous knockout minimally affected baseline liver transcriptome, mostly circadian rhythm genes. Nevertheless, this caused differential expression of >1000 genes post hepatectomy, hindering lipid metabolism, bile acid biosynthesis, insulin signaling and cell cycle, all critical cellular processes for liver regeneration. These results demonstrate that mere reduction of A20 levels causes worse outcome post hepatectomy than full knockout of bona fide liver pro-regenerative players such as IL-6, clearly ascertaining A20’s primordial role in enabling liver regeneration. Clinical implications of these data are of utmost importance as they caution safety of extensive hepatectomy for donation or tumor in carriers of A20/TNFAIP3 single nucleotide polymorphisms alleles that decrease A20 expression or function, and prompt the development of A20-based liver pro-regenerative therapies.


Transplantation | 2016

A20 Haploinsufficiency Aggravates Transplant Arteriosclerosis in Mouse Vascular Allografts: Implications for Clinical Transplantation.

Herwig P. Moll; Andy Lee; Clayton R. Peterson; Jesus Revuelta Cervantes; Brandon M. Wojcik; Anshul Parulkar; Alessandra Mele; Philip J. LoGerfo; Jeffrey J. Siracuse; Eva Csizmadia; Cleide G. da Silva; Christiane Ferran

Background Inflammation is central to the pathogenesis of transplant arteriosclerosis (TA). We questioned whether physiologic levels of anti-inflammatory A20 influence TA severity. Methods We performed major histocompatibility complex mismatched aorta to carotid artery interposition grafts, using wild type (WT) or A20 heterozygote (HET) C57BL/6 (H-2b) donors and BALB/c (H-2d) recipients, and conversely BALB/c donors and WT/HET recipients. We analyzed aortic allografts by histology, immunohistochemistry, immunofluorescence, and gene profiling (quantitative real-time reverse-transcriptase polymerase chain reaction). We validated select in vivo A20 targets in human and mouse smooth muscle cell (SMC) cultures. Results We noted significantly greater intimal hyperplasia in HET versus WT allografts, indicating aggravated TA. Inadequate upregulation of A20 in HET allografts after transplantation was associated with excessive NF-кB activation, gauged by higher levels of IkB&agr;, p65, VCAM-1, ICAM-1, CXCL10, CCL2, TNF, and IL-6 (mostly localized to SMC). Correspondingly, cytokine-induced upregulation of TNF and IL-6 in human and mouse SMC cultures inversely correlated with A20 expression. Aggravated TA in HET versus WT allografts correlated with increased intimal SMC proliferation, and a higher number of infiltrating IFN&ggr;+ and Granzyme B+ CD4+ T cells and natural killer cells, and lower number of FoxP3+ regulatory T cells. A20 haploinsufficiency in allograft recipients did not influence TA. Conclusions A20 haploinsufficiency in vascular allografts aggravates lesions of TA by exacerbating inflammation, SMC proliferation, and infiltration of pathogenic T cells. A20 single nucleotide polymorphisms associating with lower A20 expression or function in donors of vascularized allografts may inform risk and severity of TA, highlighting the clinical implications of our findings.


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 The American College of Surgeons | 2010

Partial loss of A20 (Tnfaip3) promotes resistance to abdominal aortic aneurysms through altering smooth muscle cell differentiation

Scott M. Damrauer; Clayton R. Peterson; Csizmadia Eva; Peter Studer; Christiane Ferran


Transplantation | 2014

Partial Loss of A20 Aggravates Transplant Arteriosclerosis through De-Regulation of IFNβ/STAT1 Axis, Thereby Enhancing Pathologic IFNγ Signaling.: Abstract# 687

Herwig P. Moll; Clayton R. Peterson; Eva Csizmadia; Christiane Ferran


Journal of Vascular Surgery | 2013

Loss of Function Studies Highlight the Vasculoprotective Effect of A20 in Transplant Arteriosclerosis (TA)

Andy Lee; Clayton R. Peterson; Herwig P. Moll; Eva Csizmadia; Ashley Rogers; Alon B. Neidich; Victor Chien; Cleide G. da Silva; Christiane Ferran

Collaboration


Dive into the Clayton R. Peterson's collaboration.

Top Co-Authors

Avatar

Christiane Ferran

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

Cleide G. da Silva

Beth Israel Deaconess Medical Center

View shared research outputs
Top Co-Authors

Avatar

Herwig P. Moll

Beth Israel Deaconess Medical Center

View shared research outputs
Top Co-Authors

Avatar

Jeffrey J. Siracuse

Beth Israel Deaconess Medical Center

View shared research outputs
Top Co-Authors

Avatar

Peter Studer

Beth Israel Deaconess Medical Center

View shared research outputs
Top Co-Authors

Avatar

Scott M. Damrauer

Beth Israel Deaconess Medical Center

View shared research outputs
Top Co-Authors

Avatar

Andy Lee

Beth Israel Deaconess Medical Center

View shared research outputs
Top Co-Authors

Avatar

Elzbieta Kaczmarek

Beth Israel Deaconess Medical Center

View shared research outputs
Top Co-Authors

Avatar

Elizabeth R. Maccariello

Beth Israel Deaconess Medical Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge