Skip to Content

School of Medicine Columbia

  • Working together in a lab.

Center for Dietary Supplements and Inflammation

We're an innovative group that promotes junior faculty mentoring, entrepreneurship and collaborative and translational research to understand how regulating inflammation can have a positive effect on a number of autoimmune disorders. 

A Multi-Disciplinary Center

Our center is made up of faculty from six University of South Carolina colleges. Together, we're working to create a self-sustaining, nationally recognized, multi-disciplinary center for dietary supplement and inflammation research. 

Grant Opportunities (USC Faculty Only) 

Prakash Nagarkatti, Ph.D., and Mitzi Nagarkatti, Ph.D., have been recipients of $10 million in Phase II funding from National Institutes of Health (NIH) for the USC Center of Biomedical Research Excellence (COBRE) on Dietary Supplements and Inflammation to further enhance research in this area. 

This Center announces the availability of the following grants: 

Target Faculty Grant (Direct funding of $150,000, for a period of one year):

The COBRE (Center of Biomedical Research Excellence) will provide funding to perform research in inflammation and dietary supplements for junior Tenure track and nontenure track investigators (with preference given to tenure-track Assistant Professors) as Target faculty. A junior investigator is defined as an individual who does not have or has not previously had external, peer-reviewed Research Project Grant (RPG) such as R01 or Program Project Grant (PPG) support from either Federal or non-Federal sources for which the individual is named as the PD/PI. Those who have NIH R03, R21, or other similar types of grants are eligible to apply. Since the COBRE Target Faculty have to devote 50% effort towards this grant, those who currently have an NIH K award or equivalent with more than 50% effort on the Career Development Award are not eligible. Candidates who are now or have previously received support from another COBRE grant as Target Faculty are also not eligible to apply. 

Funded investigators will be expected to submit a Research Project Grant before the end of the funding year and demonstrate an effort to move toward independent research support. Upon receiving R01 type of funding, investigators will be considered “graduates” of the COBRE program. Details on the COBRE can be found on the following website:

https://www.nigms.nih.gov/Research/DRCB/IDeA/pages/COBRE.aspx

All successful proposals must be compliant with all NIH regulatory criteria such as human subjects, vertebrate animals, biohazards, etc., (if applicable) in order to receive final NIH approval.

 

Pilot Project Grants (Direct funding of $50,000, for a period of one year):

The NIH COBRE on Dietary Supplements and Inflammation will provide funding to support research in Dietary Supplements and Inflammation to Tenure-track, Tenured, and nontenure-track faculty who do not currently have R01 or equivalent grants as PI or who are not currently Target Faculty on any COBRE grants. Consideration will be given to the research productivity of the investigator and their ability to attract independent funding from NIH or other funding agencies. Tenured faculty interested in applying should demonstrate a desire to begin research outside their current field. 

All successful proposals must be compliant with all NIH regulatory criteria such as human subjects, vertebrate animals, biohazards, etc., (if applicable) in order to receive final NIH approval.

Areas of Interest

  • Study the role of a new Chinese herb-derived selective Toll-like receptor antagonist (Sparstolonin B) as an anti-inflammatory agent to treat atherosclerosis
  • Anti-inflammatory capabilities of plant polyphenols for the treatment of Alzheimer’s Disease
  • American Ginseng-mediated autophagy and suppression of inflammation in pathological cardiac remodeling and dysfunction
  • Macrophage-induced inflammation in high fat diet enhanced breast cancer: benefits of Quercetin

Research Projects

Phase 1. Target Faculty and Their Projects

Principal Investigator: Melissa Moss, Ph.D.

The global aim of our research is to use biomedical engineering and molecular biology tools to study the mechanism responsible for changes in protein folding, cell-protein interaction, and inflammatory immune response to misfolded proteins in Alzheimer’s Disease. Alzheimer’s Disease is characterized by deposits of aggregated amyloid-b protein (Ab) within the brain parenchyma and cerebrovasculature. This pathology is coupled with elevated inflammatory response. AD brains with Ab deposits co-localized with vessel-associated immune cells exhibit a compromised blood-brain barrier (BBB) integrity.

The goal of the current study is to expand on our preliminary
results that:
  1. Interaction of soluble Ab aggregates, but not monomeric Ab with cerebrovascular endothelial cells, is responsible for inflammatory responses such as increased endothelial expression of adhesion molecules, increased monocyte adhesion and reduced permeability when tested in vitro cultures.
  2. These inflammatory responses by aggregated Ab are mediated via NF-kB signaling, where reactive oxygen species (ROS) induced by aggregated Ab serve as second messengers.

We will test the hypothesis that plant polyphenols will reduce Ab-induced inflammatory responses in endothelial cells by interfering with both Ab aggregation and ROS second messengers. We will test if plant polyphenols act as aggregation inhibitors to attenuate Ab-induced vascular inflammatory responses. Combinations of polyphenols that are synergistic in action as demonstrated by their ability to reduce NF-kB signaling, exhibiting similar anti-oxidant capabilities and varying ability to inhibit Ab aggregation, will be identified. These studies will form the basis for future therapeutics development for the treatment of Alzheimer’s disease using plant polyphenols.

Principal Investigator: Susan K Wood

Stress exposure precipitates psychiatric disorders such as depression in susceptible individuals. Depression is not only the leading cause of disability in the U.S. but it increases one’s risk of cardiovascular disease. The long-term goal of our work is to identify neurobiological mechanisms that cause individuals with depression to be at greater risk of developing cardiovascular disease.

Recent data suggests that inflammation may be the link between depression and cardiovascular disease. Using a resident-intruder paradigm of social stress in rats, we previously identified a susceptible population of rats that developed behavioral and neuroendocrine endpoints related to depression and evidence of cardiovascular dysfunction. In these studies we will test the hypothesis that circulating cytokines and cytokines within stress-sensitive brain regions drive the vulnerability to depression and cardiovascular disease.

These studies will test the efficacy of the potent plant-based anti-inflammatory, resveratrol, to inhibit the effects of social stress on neuroinflammation, indices of cardiovascular disease and depressive-like behaviors in a stress-susceptible population.

  1. Aim 1 will utilize cardiovascular telemetry to examine the cardioprotective effects of resveratrol on stress-induced cardiac dysfunction.
  2. Because neuroinflammation is gaining recognition for its role in depression and cardiovascular disease, Aim 2 will test the notion that neuroinflammation within the stress-sensitive brain region, the locus coeruleus, is capable of altering neuronal activity and thereby drives the stress-induced depressive-like phenotype.
  3. Studies in Aim 3 will use resveratrol to establish a role for neuroinflammation in altered serotonin metabolism following social defeat in another stress-sensitive brain region, the dorsal raphe.

The implications of these studies seek to establish a therapeutic role for natural bioactive compounds with potent anti-inflammatory properties in treating stress-induced depression and cardiovascular disease in stress susceptible individuals.

Find Susan Woods on PubMed

The global aim of our research includes understanding the role of inflammation-associated molecules in the development and progression of prostate cancer and harnessing the power of immune system to improve its preventive and therapeutic efficacy against developing neoplastic cells and/or established cancer cells. 

The goal of the current study is to expand on our observation that Withaferin A (WFA) from the plant Withania somnifera inhibits expression of NLRP3 inflammasome (multi-protein complexes including NLRP3, IL-1β, IL-18) and therefore test the hypothesis that intake of dietary agent WFA will provide an anti-inflammatory environment in the prostate gland to prevent tumor development.

In this proposal we will:
  1. determine the mechanism of WFA-mediated regulation of inflammatory cytokines.
  2. determine WFA-targeted modulation of inflammation in vivo and inhibition of prostate cancer cell growth.

We will specifically test WFA-induced polarization (tumor promoting or tumor suppressive) of macrophages in the prostate gland and understand the role of macrophage inhibitory cytokine-1 (MIC-1) and  assess the WFA-induced functional activity of  NK cells in vitro and in immune competent mice.

Principal Investigator: Daping Fan

The global aim of our research is to promote the regression of atherosclerotic plaques through restoring macrophage cholesterol homeostasis and controlling macrophage inflammation.

The goal of the current study is to expand on our preliminary results that SsnB:
  • has potent anti-inflammatory effects on macrophages by blocking Toll-like receptor 2 (TLR2) and TLR4 signaling.
  • diminishes the ability of activated endothelial cells to attract monocyte for adhesion and decreases arterial smooth muscle cell migration.
  • effectively suppresses inflammatory response in mice.

We will test the hypothesis that SsnB can be developed as an anti-atherosclerosis agent by virtue of its selective inhibitory effects on TLR2 and TLR4 signaling.

To test this hypothesis, we propose three specific aims:
  1. SA1: To elucidate the molecular mechanism by which SsnB blocks TLR2 and TLR4 signaling. We will express and purify the Toll/IL-1 receptor (TIR) domains of TLRs, the adaptor proteins TIRAP/Mal and MyD88 and examine the binding of SsnB to these proteins.
  2. SA2: To examine the effects of SsnB on resident vascular cells. We will test the hypothesis that SsnB suppresses the inflammatory phenotype in arterial endothelial and smooth muscle cells by blocking TLR2 and TLR4 signaling.
  3. SA3: To test the hypothesis that SsnB attenuates atherogenesis in mice. LDL receptor (LDLR) deficient mice will be fed high fat diet to induce hypercholesterolemia and atherosclerosis. SsnB will be administrated to test if it attenuates atherogenesis in these mice.

Principal Investigator: Angela Murphy

The global aim of our research involves investigations of dietary and physical activity interventions to reduce macrophage-induced inflammation in cancer. The goal of the current study is to determine the effects of dietary quercetin on inflammation and subsequent tumor progression and overall survival in a mouse model of high-fat diet (HFD) enhanced breast cancer (BrCA).

We will test the hypothesis that the mechanism of action of quercetin on the regulation of MΦ-induced inflammation in HFD-enhanced BrCA is mediated through SIRT1. 

We will:
  • elucidate the stage-specific effects of quercetin on inflammation in HFD-enhanced BrCA.
  • evaluate whether MΦs are a target for the anti-inflammatory effects of quercetin in HFD-enhanced BrCA.
  • determine whether SIRT1 is a mediator of the effects of quercetin in the regulation of MΦ-induced inflammation in HFD-enhanced BrCA.

This investigation proposes to prevent incidence and progression of HFD-enhanced BrCA by using a dietary food component that targets inflammation, the mechanistic core of this disease.

Phase 1. Pilot Projects:

Principal Investigator: Jabbarzadeh, Ehsan

Project Goals:

Aim 1. Determine the in vitro potential of resveratrol to mediate osteogenic differentiation and inflammatory response in 3D scaffolds.

Hypothesis: We hypothesize that resveratrol-incorporated PLGA scaffolds will modulate the inflammation response of M1 macrophages and promote a phenotypic switch into wound-healing, anti-inflammatory M2 macrophages.

Aim 2. Determine the in vivo potential of resveratrol incorporated scaffolds to mediate inflammatory response and promote the neogenesis of bone and angiogenesis.

Hypothesis: We hypothesize that resveratrol-nanoparticle included PLGA sintered microsphere scaffolds can alleviate host immune response, and enhance vascular growth and ultimate bone healing.

Beyond the scope of this proposal, this approach enables efforts to prospectively engineer inflammatory response by “dialing” the appropriate degree of resveratrol release profile. The proposed studies are translational as they provide critical new insight into the principal mechanisms of directed angiogenesis and inflammation in porous biomaterials. The proposed approach is transformative as it tackles a confounding barrier in regenerative medicine with applicability to many other musculoskeletal tissue engineering approaches in addition to bone. Insufficient vascularization of implantable scaffolds is a profound barrier in regenerative medicine. Our results will impact the field as this strategy can work in different materials and is scalable to larger scaffolds.

Principal Investigator: Gower, Michael

Project Goals:

AIM 1: Employ biomaterial based gene delivery to promote brown adipose tissue (BAT) gene expression in white adipose tissue (WAT). The peritoneal fat is a depot of WAT that exhibits long-term transgene expression following implant of PLG scaffolds that release lentiviral vectors. We propose to implant scaffolds in the peritoneal fat that release vectors encoding for PRDM16 and PGC-1α, transcriptional coactivators required for expression of BAT genes within WAT. We will first investigate, in vitro, the ability PRDM16 and PGC-1α gene delivery to induce BAT gene programs in WAT cell lines and isolated white adipocytes. We will use our in vitro data to determine the number of viral particles and ratio of particles encoding for PRDM16 and PGC-1α to deliver in vivo on vector releasing scaffolds. Following implant we will characterize adipocytes within the implant site by morphology, cell surface markers, and gene expression.

AIM 2: Employ localized release of resveratrol to promote thermogenesis in engineered BAT. Resveratrol, a natural polyphenol found in red wine, modulates energy homeostasis by activation of SIRT1, a deacetylase that recruits coactivators PGC-1α and PRDM16 to the transcription factor PPARγ, leading to induction of BAT genes and repression of WAT genes. We propose to encapsulate resveratrol within scaffolds for BAT engineering and investigate its effect on thermogenesis, weight loss, insulin resistance, and hyperlipidemia when implanted in the peritoneal fat of mice fed a diabetogenic diet.

Principal Investigator: Chatterjee, Saurabh

Principal Investigator: Koh, Ho-Jin

Principal Investigator: Testerman, Traci

Principal Investigator: Colpitts, Tonya

Principal Investigator: Chanda, Anindya

Principal Investigator: Lizarraga, Sophia

Principal Investigator: Jarzynski, Mark

 Phase 2.  Target Faculty and Their Projects:

Principal Investigator: Gomez, Gregorio

Allergic disease is the fifth leading chronic disease in the United States. A major contributing factor to allergic inflammation including asthma is Prostaglandin D2 (PGD2) produced by mast cells, the cell type responsible for IgE-mediated immediate hypersensitivity reactions.  Therefore, targeting the arachidonic acid pathway in mast cells to inhibit PGD2 biosynthesis is one strategy to significantly limit allergic inflammation.  Recently, we discovered that Resveratrol, a natural plant-derived polyphenol, selectively inhibited IgE-dependent PGD2 production from human skin mast cells.  We further showed that Resveratrol inhibited FcεRI-induced expression of cyclooxygenase 2 (COX-2), a key enzyme in the arachidonic acid pathway that is directly involved in PGD2 biosynthesis. The central question of this study is: how does Resveratrol inhibit FcεRI-induced COX-2 expression and PGD2 production in mast cells?  Interestingly, miR-155 has been implicated as a positive regulator of COX-2 expression in different cancers, macrophages, airway smooth muscle, and in the severity of allergic asthma in mice.  Indeed, our miRNA array analysis and qRT-PCR validation studies revealed a positive correlation between miR-155 and COX-2 expression in human primary mast cells following FcεRI crosslinking.  Moreover, we found that Resveratrol significantly inhibited FcεRI-induced expression of miR-155 expression as well as COX-2.  Given that miRs negatively regulate target genes, these data suggest that miR-155 targets a repressor of COX-2.  Our in silico pathway analysis has identified several negative regulators of COX-2 such as ATF3, SOCS-1, SHIP-1, and PPARG, as potential targets of miR-155 in human and murine mast cells.  Thus, we hypothesize that Resveratrol inhibits allergic inflammation by regulating the expression of miR-155 in mast cells leading to increased induction of the repressors and consequently diminishing COX-2 expression and PGD2 biosynthesis.  Our study will (1) Characterize the effect of Resveratrol on the FcεRI-induced miRNA expression profile in situ-matured mast cells from human and mouse, (2) Identify the COX-2 repressor targeted by miR-155, and define the mechanism by which Resveratrol inhibits FcεRI-induced COX-2 expression and PGD2 biosynthesis in mast cells, and (3) Characterize the effects of dietary Resveratrol on airway remodeling and hyper-responsiveness in a mast cell dependent model of allergic asthma, and identify associated miRNAs.  To define the role of miR-155-5p in allergic asthma and the efficacy of Resveratrol in inhibiting disease development, we will also use miR-155-5p transgenic (Tg) and knockout (KO) mice in our model.  Overall, this study will shed new light on the role of miR-155 in the ability of Resveratrol to selectively inhibit FcεRI-induced COX-2 and PGD2 production in mast cells, thereby, attenuating allergic inflammation.

Principal Investigator: Testerman, Traci

Inflammatory bowel disease (IBD) afflicts over one million Americans, causing considerable suffering and lost work time.  The direct and indirect costs of IBD were estimated to be between $14.6 and $31.6 billion in 2014.  Furthermore, IBD greatly increases the risk of developing colorectal cancer.  Bacteria are now believed to be key players in both IBD and colorectal cancer.   A number of Helicobacter species infect the human colon and are known to cause colitis and colon cancer in colitis-prone mouse strains.  We have exciting data showing that H. muridarum exacerbates dextran sulfate sodium (DSS) induced colitis in wild-type mice.  There are no studies on the immune response triggered by H. muridarum.  Thus, this EHH species offers a unique experimental model to understand how colitis is triggered in an immunologically normal animal following a chemical insult.  Recent studies have shown that dietary indoles, such as Indole-3-carbinol (I3C), derived from cruciferous vegetables, have a number of anti-inflammatory and anti-carcinogenic properties.  Our preliminary studies showed that I3C attenuates H. muridarum+DSS-mediated exacerbation of colitis and inflammation in the colon. Furthermore, we noted that I3C treatment decreases the expression of miR-874, which targets FOXP3, and increases that of miR-30b which targets for RORC (RORγt) as well as increases miR-5112 that targets IL-17. Based on these data, in the current study, we will test the central hypothesis that I3C attenuates colitis and inflammation induced by H. muridarum through alterations in the expression of miRs that promote a switch in T cell differentiation from Th17 to Tregs.  The mechanisms of colitis exacerbation involving inflammation by H. muridarum are also not known.  Thus, it is critical to understand the nature of immune response against Helicobacter species in IBD.  To that end, we will simultaneously explore immunological and regulatory changes induced by these two agents.  First, we will examine the T cell responses occurring during DSS-mediated colitis with and without H. muridarum infection and with and without I3C treatment.  Our primary focus will be regulatory T cells (Treg), which are critical for intestinal homeostasis.  Next, we will determine whether H. muridarum can trigger colitis in Aryl hydrocarbon receptor (AhR)-deficient mice which fail to generate enough Tregs and are more susceptible to colitis.  These mice will also be used to test the efficacy of I3C, which has been known to act as an AhR ligand.  Finally, we will determine whether specific microRNA species induced by I3C contribute to the Treg response by changing FoxP3 expression both in vitro and in vivo.  Together, the insights gained from these experiments will be essential for understanding the mechanisms of action of I3C and could lead to additional highly targeted treatments.  This project will not only characterize the nature of immune response triggered by H. muridarum during DSS-induced colitis but also test the mode of action of I3C on H. muridarum associated colitis.  These data will support future explorations to investigate the role of other Helicobacter species in clinical IBD and the potential use of I3C in the treatment of IBD.

Principal Investigator:  Lizzarga, Sofia

Growing evidence strongly suggests a correlation between prenatal inflammation and autism; yet the mechanisms that underlie this correlation are largely unknown. The pathophysiology of autism is proposed to arise from defects in neuronal circuitry. Animal models of prenatal exposure to maternal immune activation (MIA) demonstrate that the offspring exhibit abnormal behaviors reminiscent of autistic human behaviors. The increase in autism-like behaviors in MIA animal models is mediated by the pro-inflammatory cytokine IL-17A. IL-17A is produced by CD4+T cells (TH17 cells) and activates the NF-κB signaling pathway in vitro. Resveratrol is a plant derived polyphenolic compound that suppresses IL-17A and has both anti-inflammatory and neuroprotective properties6-8. Interestingly, resveratrol has been shown to ameliorate autistic-like behaviors in a rodent model of ASD. Despite the available data on the role of inflammation in ASD etiology, there is a significant gap in knowledge regarding how inflammation affects the development of human neuronal circuitry. Here we will test the hypothesis that increased levels of IL-17A will impair the development of neuronal connectivity and that resveratrol can serve as a therapeutic modality through inhibition of IL-17A downstream signaling. The rationale for the proposed studies is that, determining the contribution of the different components of the Central Nervous system (CNS) and immune system will allow us to dissect the cellular mechanisms that underlie the role of neuronal inflammation in ASD pathology. The long-term goal of this project is to uncover the mecha- nisms underlying the pathology of autism associated with defects in neuronal connectivity that are induced by prenatal inflammation. The objectives of this application are: 1) to develop stem cell derived neural models and determine the effect of pro-inflammatory cytokines during neuronal development in vitro (Aim-1); 2) to investigate potential chromatin regulatory pathways that might underlie the effect of inflammation during neuronal develop- ment in vivo and in vitro (Aims 2-3); 3) to ascertain the therapeutic potential of plant supplements (i.e. resveratrol) in the pathology of autism (Aim1, 2 & 3). To address the objectives of this application, a combination of state of the art approaches including stem cell based and mouse models, imaging, transcriptome, epigenetic and bio- chemical technologies will be used. We expect that the objectives proposed in this application will provide: 1) clear mechanistic information on how neuronal inflammation might disrupt the normal development of human neuronal circuitry; 2) a testable pre-clinical model for dietary supplements such as resveratrol in the treatment of autism.

Principal Investigator: Chatterjee, Saurabh

Project Summary/Abstract: With obesity assuming pandemic proportions across the developed nations (USA, continental Europe) and emerging economies of India and China, it is estimated that 20-30% of this huge population will develop fatty liver disease (NAFLD). A sizable proportion (roughly 120 million) of those affected with NAFLD will have steatohepatitis and the disease progression is believed to be dependent on the built environment and diet. In spite of the enormous health risk, no suitable treatment regimen has been established so far due to the complications of the disease itself, following multiple risk factors of steatosis, metabolic disorder, inflammation and abnormal endocrine function. Andrographolide (ANDL) is a unique plant derivative which has shown profound caloric restriction, anti-inflammatory and metabolic signaling modulation properties. The use of this compound as a preventive and therapeutic agent in NAFLD is of immense importance to this very significant health risk. Importantly, identifying newer epigenetic modulating function of ANDL in NAFLD would go a long way in targeting effective therapy in this disease. In the current study, we will test the central hypothesis that oral administration of andrographolide (ANDL) attenuates NAFLD via its actions on miR-21-induced inflammatory checkpoints in sinusoidal endothelial dysfunction, stellate cell activation, TGF-beta signaling and defective macroautophagy. The long term objective of this project is to design a comprehensive experimental and preclinical evidence of a treatment regimen that derives from natural dietary supplements with proven anti- inflammatory potential against NAFLD. About seventy-five percent of obese subjects have hepatic steatosis, and about 20% of these individuals develop inflammatory liver disease marked by necroinflammation, a rise in inflammatory cytokines, and some degree of fibrosis. This advanced stage of the disease progression often leads to cirrhosis and autoimmune complications because of the highly inflammatory microenvironment. Because NAFLD has been shown to derive its progression and severity from an underlying condition of obesity and hepatic inflammation, it is imperative that Andrographolide, which has a potent anti-inflammatory effect, might restrict the progression of steatosis to steatohepatitis and thwart the development of more severe complications like hepatocellular carcinoma. The novel role of andrographolide as an epigenetic regulator in NAFLD therapy has never been explored. This project will aim to utilize the supplementation of andrographolide in steatotic mice to abrogate the progression of steatohepatitis following methionine choline deficient diet exposure by its effective regulation of miR21 via NF-kB inhibition leading to suppression of sinusoidal endothelial dysfunction, inflammation and defective autophagy. This project proposes to utilize the COBRE funds to generate sufficient evidence of andrographolide as a potential anti-inflammatory and epigenetic regulator as part of its therapeutic effect in NAFLD. 

Principal Investigator: Brandon Busbee

Colitis is an inflammatory bowel disorder (IBD) characterized by chronic inflammation of the large intestine or colon. This disease, affecting over 1 million people and costing over a billion dollars/year in the US alone, has a complex etiology and to date, there are few and effective treatment options to patients. In the current study, we demonstrate that in TNBS-induced colitis, a natural indole product found in numerous cruciferous vegetables (Indole-3-carbinol, or I3C) and ligand for the aryl hydrocarbon receptor (AhR) is effective at preventing symptoms of colitis. Most notably, I3C was able to prevent colitis-associated dysbiosis and increase colonic butyrate. Specifically, I3C prevented increases in colitis-associated gram-negative bacteria (e.g. Bacteroides acidifaciens), while also increasing butyrate producing Roseburia. IL-22 was found to be significantly increased after I3C treatment, and neutralization of this cytokine prevented I3C from reducing disease severity and altering the gut microbiome and metabolome. In the current proposal, the central hypothesis is I3C mediates its beneficial effects through regulation of cross talk immune cells and colonic epithelial cells (CECs) by activation of AhR and depends on IL-22 production to mediate its protective effects during colitis. The aims of this study will be as follows: 1.) Dependency of AhR in I3C-mediated protective effects will be investigated using conditional KO of AhR on immune cells (T cells and innate lymphoid type 3 cells/ILC3s) or CECs, both of which express AhR. These studies will determine if I3C-mediated effects are dependent on AhR expression in immune cells and/or CECs in preventing colitis and altering disease-associated microbial dysbiosis and the metabolomic profile,. Focus on the impact AhR plays in regulating I3C-mediated modulation of IL-22 will be performed to identify AhR-specific dioxin response elements (DREs) on IL-22 and IL-22 promoting genes. 2.) Additionally, studies will be conducted to determine the cell source of IL-22 production and investigate epigenetic modifications( microRNA/miRNA and DNA methlyation) affecting IL-22, which could be mediated by I3C treatment during colitis.

Principal Investigator: Jason Kubinak

Antibody deficiency is the most frequently diagnosed form of primary immunodeficiency in humans. Common variable immunodeficiency (CVID) is the most severe form of antibody deficiency and is characterized as hypogammaglobulinemia (low IgG) with an accompanying deficit in IgA and/or IgM titers. In both humans and laboratory mouse models, IgA deficiency has been associated with alterations to the composition and function of symbiotic microbial communities (a.k.a. the microbiota) in the gut, and emerging data from CVID patients indicate that a similar association exists. Up to 50% of CVID patients will develop gastrointestinal symptoms, and the major complication of CVID is CVID enteropathy. CVID enteropathy most often presents as chronic diarrhea and weight loss due to an underlying intestinal malabsorption. The pathophysiological mechanism driving CVID enteropathy is not known but pathological alterations to the microbiota ('dysbiosis') could be a key factor. Bile acids (BAs) are secreted into the gut where they play a crucial role in the emulsification of dietary lipids that facilitates their absorption. The microbiome plays a central role in shaping BA composition in the gut. Thus, dysbiosis caused by gut antibody deficiency may drive CVID enteropathy and associated metabolic disease by influencing BA metabolism in the gut. The objective of Specific Aim #1 is to test that intestinal malabsorption is an IgA-dependent phenotype using adoptive transfer models in antibody deficient recipients. The objective of Specific Aim #2 is to specifically test that bacterial bile salt hydrolase (bsh) activity results in enhanced BA deconjugation that drives malabsorption in antibody deficient mice. Mono-colonization experiments in germfree Ig-deficient mice using WT and bsh-null mutant strain of commensal bacteria will be used to address this hypothesis. The objective of Specific Aim #3 is to determine the impact of altered BA pools on host metabolism using a mixture of in vitro and in vivo models. Collectively, these experiments are the first to address the role of mucosal IgA deficiency in the context of CVID on the regulation of bacterial BA metabolism and its effect on host health. Several approaches will be utilized to assess the feasibility of treating malabsorption and chronic inflammation through dietary manipulation of the microbiome.

Principal Investigator: Jie Li

High fat diets (HFDs) alter both host inflammatory responses and gut microbial metabolites. While these metabolites have been hypothesized to mediate host intestinal inflammation, an existing gap is how to pinpoint the functional and responsible metabolites from an extremely complicated metabolites pool that contains numerous unknown chemicals. We seek to discover such functional metabolites and establish their role in modulating HFDs-induced intestinal inflammation. In our preliminary study, we first established a mouse model that displayed HFDsinduced intestinal inflammation. We next performed comparative metagenomic analysis of the gut microbiome collected from aforementioned mice, leading to identification of a genus, Alistipes, which was significantly increased during HFDs-induced inflammation. Alistipes is isolated primarily from clinical samples and shows emerging implications to inflammation, motivating us to investigate the potential links between Alistipes metabolites and the observed intestinal inflammation of our mouse model. Thus, we developed complementary metabolomics and genome mining approaches: metabolomic analysis of the mice fecal and serum samples directly displayed metabolic changes while genome mining revealed unique patterns of biosynthetic gene clusters that encode the metabolites of interests. Indeed, the cross-validation of these two approaches led to the discovery of a class of rare lipids, sulfonolipids (SLs) which were significantly increased in the HFDs-fed mice samples. The potential biosynthetic genes of these SLs were also accumulated in the HFDs-fed mice samples. The pure SLs were subsequently isolated, with the chemical structures elucidated by NMR. We then tested sulfobacin A, a major member of the isolated SLs, and it indeed induced macrophage RAW264.7 inflammatory responses by RT-PCR and ELISA analyses. All these preliminary data suggest that gut microbial metabolites SLs mediate HFDsinduced intestinal inflammation. Intriguingly, SLs structurally mimic human endogenous sphingolipids (SPs), with the latter known to mediate inflammation. In addition, a genus of gut microbiota, bacteroides, also produces SPs but not SLs. The bacteroidesderived SPs were recently shown to enter hosts’ metabolism and are critical for maintaining intestinal homeostasis and symbiosis. Taken together, this raises an interesting hypothesis that SLs may directly induce inflammation, but also may modulate inflammation by affecting intestinal homeostasis of SLs and SPs. Thus, we are now set up to unambiguously establish, both in vitro and in vivo, the role of SLs in mediating HFDsinduced intestinal inflammation, with an emphasis on the potential relationship between SLs and SPs. This goal will be achieved through completion of the following Specific Aims (SA). SA 1: Characterizing the HFDs-associated expression of microbial SLs, microbial SPs and host endogenous SPs. SA 2: Investigate the activities and relationship of SLs and SPs in mediating intestinal inflammation, using both invitro assays and in vivo germ-free mouse models. in vitro and in vivo, the role of SLs in mediating HFDs-induced intestinal inflammation, with an emphasis on the potential relationship between SLs and SPs. This goal will be achieved through completion of the following Specific Aims (SA).

Principal Investigator: Melissa Ellerman

Inflammatory bowel diseases (IBD) are chronic, relapsing, immune-mediated diseases influenced by host genetics, environmental factors and the gut microbiota. Intestinal inflammation alters gut microbiota composition and function to disrupt its symbiosis with the host (dysbiosis). Increased Escherichia coli is a common signature of gut dysbiosis in human IBD and murine colitis models and is thought to contribute to colitis development. The endocannabinoid (EC) system has emerged as a promising therapeutic target for human IBD because of its reported anti-inflammatory effects. ECs are lipid hormones that activate host cannabinoid receptors to modulate gut physiology and immunity. Host EC activity is regulated by biosynthetic and degradative enzymes that modulate tissue EC levels and by signaling at host cannabinoid receptors (e.g. CB1, CB2) . Inhibiting EC degradation, or agonism of CB1 or CB2, attenuates disease in chemically-induced colitis models. The host EC system also influences microbiota composition and directly modulates bacterial functions. However, it remains unknown whether cannabinoid modulation of the gut microbiota occurs in IBD and whether these interactions impact disease severity. Moreover, the therapeutic potential of cannabinoids in genetic models of IBD remains understudied. Using the Il10 KO mouse model of IBD, our initial studies demonstrate that inhibiting degradation of the EC 2-AG exacerbates colitis and and promotes dysbiosis as characterized by the expansion of intestinal E. coli. Moreover, we show that inhibiting 2-AG degradation increases gut E. coli in non-inflamed WT mice, which is counteracted with CB1 receptor blockade Our central hypothesis is that cannabinoid signaling at the host CB1 receptor promotes the outgrowth of intestinal E. coli, thus exacerbating inflammation in IBD-susceptible hosts. The objective of this proposal is to establish host cannabinoid signaling as a novel mediator of intestinal dysbiosis by completing the following Aims. Aim 1: Determine the contribution of the host CB1 and CB2 receptors in promoting the outgrowth of intestinal E. coli. Aim 2: Evaluate the effects of host cannabinoid signaling on intestinal dysbiosis and consequent inflammation in IBDsusceptible Il10 KO mice. Aim 3: Characterize the effects of host CB1 receptor signaling on the intestinal metabolome.

Principal Investigator: Kandy Velazquez

Unintentional body weight loss (cachexia) in chronic diseases, such as cancer can lead to the deterioration of quality of life, lack of response to treatment, and ultimately death. Cancer cachexia is characterized by systemic inflammation, muscle and fat loss, and reduced physical function. Tissue fibrosis or scarring has been observed in cancer cachectic patients. Muscle fibrosis is one of the most consistent findings among people with reduced muscle function due to myopathies, neuromuscular disorders, trauma, and sarcopenia. Blocking inflammation and fibrosis in the pre-cachectic and cachectic stages is critical to positively impact muscle health, locomotion, and performance. Silybin, one of the active compounds of the medicinal plant Silybum marianum has shown anti-inflammatory and anti-fibrotic properties in cardiac and liver disorders. Since there are no approved therapies for cancer cachexia and silybin has been associated to improve factors associated with cachexia, we propose to assess whether silybin reduces skeletal muscle fibrosis. Furthermore, we will determine if TGF-􀈕 is involved in initiating or maintaining loss of muscle mass and function. Our findings could lead to the discovery of a new mechanism and novel strategies to prevent and/or treat cachexia.

Principal Investigator: Carol Oskeritzian

The prevalence and incidence of atopic dermatitis (AD), most common type of eczema, have increased over the last several decades, as AD remains the most common chronic inflammatory skin disease. AD is characterized by widespread pruritic skin lesions, making it the skin disorder with the highest disease burden globally. The mechanisms leading to skin barrier disruption and overt lesions are ill defined. While some studies focus on T cell-derived cytokines as possible underlying effectors of disrupted skin, we reasoned that T cells may be preceded by mast cells as first-line effector cells as mast cells are skin resident immune cells facilitating T cell recruitment. Moreover, many AD studies are comparing features of lesional to neighboring non lesional skin samples, i.e., when the diseased state is already established, we used a preclinical human AD-like model to investigate the pathogenic events leading to overt lesions. We recently reported an early elevation of ceramide sphingolipids in skin samples treated with an AD-triggering antigen, compared to controls, that was driven by mast cells, enabling early cutaneous apoptosis through endoplasmic reticulum (ER) stress. In this proposal, we offer to investigate the contribution of ceramides and apoptosis in AD pathogenesis in another preclinical AD model and the effects of resveratrol, a natural compound, on restoring homeostatic ceramide metabolism to attenuate ER stress, apoptosis and subsequent loss of skin integrity. The objectives of this application are: To investigate the effects of resveratrol on early ceramide elevation and apoptosis in preclinical AD, and to study the impact of antigen exposure on human skin explants and the regulatory functions of resveratrol. We anticipate that our proposed studies will contribute to a better understanding of AD pathogenesis and the mechanisms of action of resveratrol. We are proposing that these studies will identify actionable pathogenic pathways preventing AD disease progression.

Phase 2.  Pilot Projects:

Principal Investigator: Kubinak, Jason

Principal Investigator: Xiao, Shuo

Principal Investigator: Reilly Enos

Principal Investigator: Jie Li

Principal Investigator: Allison Armstrong

Principal Investigator: Carole Oskeritzian

Principal Investigator: Joseph McQuail

Principal Investigator: Abbi Lane-Cordova

Principal Investigator: Mohamad Azhar

Principal Investigator: John Eberth

Principal Investigator: Xiaoming Yang

Principal Investigator: Cameron McCarthy

Principal Investigator: Rekha C. Patel 

Principal Investigator: Claudia Grillo

Principal Investigator: Norma Frizzell

Our Supporting Cores

Our center and researchers are support by various cores across campus. 

The Bioanalytical Core of the CDSI is located at Building 1 of the School of Medicine at the VA hospital campus. The major equipment in the Bioanalytical Core include: Illumina NextSeq 550, Qiagen Rotor-Gene Q 5plex HRM PCR , QIAcube HT/QIAxtractor Instr., Affymetrix GCS 3000 Fluidics Station, Illumina MiSeq, RS 2000 X-Ray Biological Irradiator, 10x Chromium, Qubit, and electrophoresis power supply. Data analysis is performed by GeneSpring GX (Agilent), Ingenuity Pathway Analysis, Cytoscape and Nephele.

The FCCA Core of the CDSI is located at Building 1 of the School of Medicine at the VA hospital campus. The major equipment in the FCCA Core include: BD FACS Celesta BVR, BD FACSymphony A5 SE, RS 2000 X-Ray Biological Irradiator, BD FACSymphony Sorter, Agilent Seahorse XFe96 Analyzer, Illumina NextSeq 550, 10x Chromium, Buxco for testing pulmonary function, and Vizgen Merscope Spatial Transcriptomics.

The Cores have additional equipment available which include laminar air flow hoods; CO2 incubators; Refrigerated centrifuges; Balances; Inverted/phase fluorescence microscopes with digital camera; Water baths; Electrophoresis apparatus and power; Refrigerators; -20°C and -80°C freezers; Liquid N2 tanks; Shakers; pH meters; Gel reader and densitometer; ELISA reader; Microfuges; Electroporator, Fume hoods, stomacher and Sysmex K21 for blood profile analysis, GentleMacs Dissociator (Miltenyi Biotech), DNA Thermal Cycler, 96- and 384-well Bio Rad PCR, and Digital qPCR machines, ECIS TEER24 to measure transendothelial/epithelial electric resistance, rodent pulse oximeter, etc.

Principal Investigators: Dr. Parkash Nagarkatti and Dr. Mitzi Nagarkatti

Contact:  prakash@mailbox.sc.edu and Mitzi.Nagarkatti@uscmed.sc.edu

The Administrative Core will build on the overwhelming success of Phase-1 COBRE by continuing to provide oversight on all aspects of the Center. The main focus of this Core is to continue to enhance the research quality and productivity of the Target faculty in the area of Dietary Supplements and Inflammation so that they will be successful in competing for independent, major extramural funding such as the NIH R01 grants. During COBRE Phase-1, 6 of our junior faculty ‘graduated’ successfully. Also, we were able to secure a P01/PPG Center involving 3 junior faculty who have graduated from the COBRE. Thus, the Phase-1 Administrative Core has been instrumental in overseeing the success of faculty and ensuring sustainability of the COBRE. The specific aims of the Administrative Core during Phase II are as follows: 1) To facilitate mentoring of target faculty, enhance use of research resources, and enable the transition of target faculty into independent, well-funded investigators. The Core will be responsible for organizing and promoting the scientific activities of the junior investigators through effective mentoring program. It will organize the Internal Advisory Committee (IAC) and External Advisory Committee (EAC) meetings that will provide input to the Center Director in assessment of research progress of the Target Faculty and Pilot Projects as well as Research Cores. 2) To recruit 5 new tenure-track junior faculty and provide them funds to initiate research on dietary supplements and inflammation. The Core will recruit an additional 5 tenure-track Assistant Professors at 4 different colleges in this area of Dietary Supplements and Inflammation to further expand multi-disciplinary research. Special consideration will be given to attracting women and underrepresented minorities which has also been a hallmark of our Phase I grant. 3) To provide oversight and sustain multi-disciplinary, collaborative research program in dietary supplements and inflammation: The Core will provide oversight in the day-to-day management of the Center and ensure that Research Cores evolve to meet the needs of current and future recruits to the COBRE. The junior investigators will be encouraged to be entrepreneurs and compete for NIH-funded SBIR/STTR grants. To sustain the COBRE research, we will apply for additional NIH P01 grants, institutional and individual pre- and post-doctoral training grants and career development awards. In summary, the overall objective of the Administrative Core is to further enhance the research infrastructure specifically in epigenetics, recruit additional junior faculty to increase the critical mass, build sustainable multi- disciplinary Center, and ensure successful transition of junior faculty into independent scientists.

Director: Dr. Narendra Singh

Contact:  Narendra.Singh@uscmed.sc.edu

The Flow Cytometry, Microscopy, and Imaging (FCMI) Core is a continuation from Phase-1 aimed at providing a wide range of biotechnology equipment and technical expertise to the COBRE junior faculty pursuing research on Dietary Supplements and Inflammation. The Core will provide state-of-the-art equipment to the Target and Pilot Project Faculty and their lab personnel for performing cutting-edge research in an accurate and timely manner that would lead to successful completion of their proposed projects. One of the focus of this Core is to provide multi-parameter flow cytometry and sorting services for phenotyping and isolation of specific cell types, determination of status on cell proliferation, differentiation, apoptosis as well as expression of signaling molecules and cytokines at the protein level in inflammation and following treatment with botanicals. The Core will also offer a range of imaging systems for live and fixed cells, tissues and organisms, including labeled and unlabeled samples. In addition, image visualization, data acquisition, processing and analysis that would meet the needs of the COBRE investigators are provided. Examination of structural changes that occur in response to inflammation, and how various dietary supplements induce alterations is critical to the understanding of the mechanisms of immune dysfunction, and development of therapeutic and preventive strategies. Specifically, the Core will provide imaging facilities for monitoring these changes at the whole animal to molecular level. This shared resource will lead to development of biomarkers for early detection and intermediate end points in the progression of the disease as well as response to therapeutic modalities. Furthermore, the instruments and expertise will provide specific and sensitive assay systems for determination of various molecular signaling pathways. At the whole animal level, ultrasound and fluorescence techniques are available to determine how inflammation and dietary supplements affect the experimental animals. The optical microscopy includes confocal and multiphoton microscopy for high resolution imaging of tissues to subcellular processes within tissues of interest. Finally, ultrastructural changes in cells will be imaged by electron microscopy to determine changes at the sub-micron level of resolution. A full range of image analysis hardware and software is also available to quantify the structural changes which occur during inflammation in the cells, tissues, and organs and how these changes are affected by the various dietary supplements. All equipment and technical expertise is available at USC SOM through outstanding institutional support. The Core faculty and staff will be available for assistance with experimental design, operation of equipment, and training on imaging instrumentation. In addition to individual interactions, didactic courses, seminars and workshops are regularly sponsored by the Instrumentation Resource Facility (IRF) at USC SOM, which will be made available to all members of the Target/Pilot Project Investigator laboratories.

Services Offered by this Core Include:

  • Electron Microscopy
  • Light microscopy and histology
  • Confocal imaging
  • Whole animal imaging
  • Flow cytometry and cell sorting
  • Seahorse extracellular flux (XF) analyzer to measure glycolysis and oxidative phosphorylation in cells
  • Single cell RNA-seq
  • Spatial transcriptomics and proteomics
  • Endoscopic video imaging of small animal intestines
  • Evaluation of small animal lung functions using non-invasive plethysmography

Director: Dr. Xiaoming Yang

Contact:  Xiaoming.Yang@uscmed.sc.edu

The Bioanalytical Core of CDSI will assist the COBRE Target and Pilot Project faculty and their lab personnel to pursue high-quality research including microRNA and epigenetic pathway analysis in immune cells following treatment with the dietary supplements in various models of inflammatory disease. Epigenetic studies being recent and cutting-edge, are more complex requiring state-of-the-art and expensive equipment, unique experimental parameters and data analytics experience, thereby requiring experienced operators dedicated to the technology. Thus, the Bioanalytical Core will serve as a technical expertise resource for the COBRE Target/Pilot Faculty and lab personnel. The Core will also assist the investigators with designing experiments, operating equipment, and trouble-shooting, aimed at helping advance research within the COBRE. The following specific aims will be pursued through this Core: 1) The Bioanalytical Core, will ensure purity and stability of any botanicals to be studied at the COBRE such as indole-3-carbinol (I3C), resveratrol (RES) and andrographolide (ANDR) currently being proposed in the COBRE as well as any future botanicals or plant extracts that will be used by the junior target faculty as well as those receiving pilot project grants. 2) To determine microRNA and mRNA expression profile using microarray-based approaches and perform pathway analysis to identify target genes following treatment with various botanicals. 3) To assist in performing genomic/epigenomic and microbiome research using next generation sequencing platforms, thereby helping to understand the mechanisms of inflammation and potential identification of biomarkers of disease. 4) To provide access to natural product library aimed at identifying novel anti-inflammatory compounds. 5) To provide statistical analysis for complex data generated using OMICS technology, which will be archived for analysis and sharing to enhance collaborations. In summary, the Bioanalytical Core will provide to the junior Target Faculty as well as Pilot Project Faculty, complete access to a wide range of state-of-the-art equipment, epigenetic assays, quality assurance for all botanicals tested at the CDSI, access to new natural products with potential anti-inflammatory properties, insights into the epigenetic and molecular basis of induction of disease, and help identify potential biomarkers and molecular signatures of pathogenesis. Together, this Core will be highly innovative value-added shared resource critical for advancing and sustaining the goals of the CDSI.

Services Offered by this Core Include:

  • miRNA analysis: The Core offers a microarray and two NGS sequencers, NextSeq 500 and MiSeq.  The Core uses Affymetrix GeneChip miRNA 3.0 array platform to profile miRNA expression.
  • Pathway analysis: Use of Pathway analysis such as Ingenuity, Panther, and DAVID.
  • RNA-Seq
  • Analysis of Histone Modifications
  • Chromatin immunoprecipitation (ChIP) to map DNA-binding proteins and histone modifications in a genome-wide manner.
  • MeDIP-seq: Used for measuring DNA methylation at genome-wide level.  
  • Bioinformatics analysis
  • Studies on Microbiome analysis including Fecal Transfer
  • Screening small molecule libraries for anti-inflammatory compounds
  • Analysis of the quality and integrity of natural compounds
  • In silico homology modeling, molecular docking and Molecular Dynamic Simulations

 

 


Challenge the conventional. Create the exceptional. No Limits.

©