Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
1.
Cancer Res ; 2024 Jun 11.
Article in English | MEDLINE | ID: mdl-38861367

ABSTRACT

Serine is critical for supporting cancer metabolism, and depriving malignant cells of this non-essential amino acid exerts anti-neoplastic effects, in large part, through disrupting metabolic pathways. Given the intricate relationship between cancer metabolism and the immune system, the metabolic defects imposed by serine deprivation might impact tumor-targeting immunity. Here, we demonstrated that restricting endogenous and exogenous sources of serine in colorectal cancer (CRC) cells results in mitochondrial dysfunction, leading to mitochondrial DNA (mtDNA) accumulation in the cytosol and consequent cGAS-STING1-driven type I interferon (IFN) secretion. Depleting mtDNA or blocking its release attenuated cGAS-STING1 activation during serine deprivation. In vivo studies revealed that serine deprivation limits tumor growth, accompanied by enhanced type I IFN signaling and intratumoral infiltration of immune effector cells. Notably, the tumor-suppressive and immune-enhancing effects of serine restriction were impaired by T cell depletion and IFN receptor blockade. Moreover, disrupting cGAS-STING1 signaling in CRC cells limited the immunostimulatory and tumor-suppressive effects of serine deprivation. Lastly, serine depletion increased the sensitivity of tumors to an immune checkpoint inhibitor targeting PD-1. Taken together, these findings reveal a role for serine as a suppressor of anti-tumor immunity, suggesting that serine deprivation may be employed to enhance tumor immunogenicity and improve responsiveness to immune checkpoint inhibitors.

2.
Am J Pathol ; 194(6): 927-940, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38417696

ABSTRACT

Inflammatory bowel diseases (IBD) are chronic inflammatory disorders of the gastrointestinal tract that are largely driven by immune cell activity, and mucosal healing is critical for remission. Serine is a nonessential amino acid that supports epithelial and immune cell metabolism and proliferation; however, whether these roles affect IBD pathogenesis is not well understood. Herein, the study showed that serine synthesis increased selectively in the epithelial cells of colons from patients with IBD and murine models of colitis. Inhibiting serine synthesis impaired colonic mucosal healing and increased susceptibility to acute injury in mice, effects associated with diminished epithelial cell proliferation. Dietary removal of serine similarly sensitized mice to acute chemically induced colitis but ameliorated inflammation in chronic colitis models. The anti-inflammatory effect of exogenous serine depletion in chronic colitis was associated with mitochondrial dysfunction of macrophages, resulting in impaired nucleotide production and proliferation. Collectively, these results suggest that serine plays an important role in both epithelial and immune cell biology in the colon and that modulating its availability could impact IBD pathogenesis.


Subject(s)
Cell Proliferation , Colitis , Epithelial Cells , Intestinal Mucosa , Serine , Animals , Colitis/immunology , Colitis/pathology , Colitis/chemically induced , Mice , Humans , Epithelial Cells/immunology , Epithelial Cells/metabolism , Epithelial Cells/pathology , Serine/metabolism , Intestinal Mucosa/immunology , Intestinal Mucosa/pathology , Intestinal Mucosa/metabolism , Macrophages/immunology , Macrophages/metabolism , Male , Mice, Inbred C57BL , Female , Colon/pathology , Colon/immunology , Colon/metabolism , Inflammatory Bowel Diseases/immunology , Inflammatory Bowel Diseases/pathology , Disease Models, Animal
3.
Nutr Cancer ; 75(7): 1499-1510, 2023.
Article in English | MEDLINE | ID: mdl-37254527

ABSTRACT

Dietary protein has been shown to impact physiology and pathophysiology, including inflammation and cancer, effects believed to occur through host and microbe-mediated mechanisms. However, the majority of studies investigating this concept have been conducted in animal models, with less information on the optimal approach, tolerability and biologic effects of modifying protein intake in humans. The current study presents a longitudinal controlled feeding trial carried out in healthy humans to acutely modulate protein intake using individualized diets. Adherence to study diets was monitored through subject-reported electronic picture-based assessments and global metabolomic analysis was performed on serum and stool, following each diet stage. Subjects exhibited strong adherence to study diets, with macronutrient intake meeting study goals during each stage. Metabolomic analysis revealed shifts in both serum and feces in association with modifying protein intake, including reciprocal changes in the abundance of amino acids and amino-acid related compounds, when comparing high to reduced protein stages. Additional fecal metabolite changes consisted of reduced microbial fermentation products following the reduced protein diet stage. Collectively, this study provides a robust method to precisely modify and monitor protein intake in humans, as well as assess corresponding metabolomic alterations.


Subject(s)
Diet , Metabolome , Animals , Humans , Feces/chemistry , Healthy Volunteers
4.
Mol Cell ; 83(2): 266-280.e6, 2023 Jan 19.
Article in English | MEDLINE | ID: mdl-36638783

ABSTRACT

Tumor suppression by TP53 involves cell-autonomous and non-cell-autonomous mechanisms. TP53 can suppress tumor growth by modulating immune system functions; however, the mechanistic basis for this activity is not well understood. We report that p53 promotes the degradation of the DNA exonuclease TREX1, resulting in cytosolic dsDNA accumulation. We demonstrate that p53 requires the ubiquitin ligase TRIM24 to induce TREX1 degradation. The cytosolic DNA accumulation resulting from TREX1 degradation activates the cytosolic DNA-sensing cGAS/STING pathway, resulting in induction of type I interferons. TREX1 overexpression sufficed to block p53 activation of the cGAS/STING pathway. p53-mediated induction of type I interferon (IFNB1) is suppressed by cGAS/STING knockout, and p53's tumor suppressor activities are compromised by the loss of signaling through the cGAS/STING pathway. Thus, our study reveals that p53 utilizes the cGAS/STING innate immune system pathway for both cell-intrinsic and cell-extrinsic tumor suppressor activities.


Subject(s)
Immunity, Innate , Interferon Type I , DNA/metabolism , Immunity, Innate/genetics , Interferon Type I/metabolism , Nucleotidyltransferases/genetics , Nucleotidyltransferases/metabolism , Signal Transduction/physiology , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Membrane Proteins/metabolism
5.
Int Rev Cell Mol Biol ; 373: 1-36, 2022.
Article in English | MEDLINE | ID: mdl-36283763

ABSTRACT

Limiting nutrient utilization by cancer cells in order to disrupt their metabolism and suppress their growth represents a promising approach for anti-cancer therapy. Recently, studies demonstrating the anti-neoplastic effects of lowering amino acid (AA) availability have opened up an exciting and quickly growing field of study. Although intracellular synthesis can often provide the AAs necessary to support cancer cells, diet and the tumor microenvironment can also be important sources. In fact, studies carried out in vitro and in animal tumor models have supported the anti-cancer potential of restricting exogenous sources of AAs. However the potential benefit of reducing AA intake in cancer patients requires further investigation. Furthermore, implementation of such an approach clinically, even if proven useful, could be challenging. In the enclosed review, we (1) summarize the pre-clinical studies showing the anti-tumorigenic effects of restricting exogenously available AAs, including through reducing dietary protein, (2) consider the role of microbiota in this process, (3) report on current recommendations for protein intake in cancer patients and studies that applied these guidelines, and (4) propose considerations for studies to test the potential therapeutic benefit of reducing protein/AA consumption in patients with cancer.


Subject(s)
Diet , Neoplasms , Animals , Dietary Proteins/metabolism , Amino Acids/metabolism , Neoplasms/drug therapy
7.
Cancer Prev Res (Phila) ; 15(12): 803-814, 2022 12 01.
Article in English | MEDLINE | ID: mdl-36049217

ABSTRACT

Bacteria are believed to play an important role in intestinal tumorigenesis and contribute to both gut luminal and circulating metabolites. Celecoxib, a selective cyclooxygenase-2 inhibitor, alters gut bacteria and metabolites in association with suppressing the development of intestinal polyps in mice. The current study sought to evaluate whether celecoxib exerts its chemopreventive effects, in part, through intestinal bacteria and metabolomic alterations. Using ApcMin/+ mice, we demonstrated that treatment with broad-spectrum antibiotics (ABx) reduced abundance of gut bacteria and attenuated the ability of celecoxib to suppress intestinal tumorigenesis. Use of ABx also impaired celecoxib's ability to shift microbial populations and gut luminal and circulating metabolites. Treatment with ABx alone markedly reduced tumor number and size in ApcMin/+ mice, in conjunction with profoundly altering the metabolite profiles of the intestinal lumen and blood. Many of the metabolite changes in the gut and circulation overlapped and included shifts in microbially derived metabolites. To complement these findings in mice, we evaluated the effects of ABx on circulating metabolites in patients with colon cancer. This showed that ABx treatment led to a shift in blood metabolites, including several that were of bacterial origin. Importantly, changes in metabolites in patients given ABx overlapped with alterations found in mice that also received ABx. Taken together, these findings suggest a potential role for bacterial metabolites in mediating both the chemopreventive effects of celecoxib and intestinal tumor growth. PREVENTION RELEVANCE: This study demonstrates novel mechanisms by which chemopreventive agents exert their effects and gut microbiota impact intestinal tumor development. These findings have the potential to lead to improved cancer prevention strategies by modulating microbes and their metabolites.


Subject(s)
Anticarcinogenic Agents , Gastrointestinal Microbiome , Mice , Animals , Celecoxib/pharmacology , Cyclooxygenase 2 Inhibitors/pharmacology , Metabolome , Anti-Bacterial Agents/pharmacology , Anticarcinogenic Agents/pharmacology , Bacteria , Carcinogenesis
8.
Methods Cell Biol ; 168: 1-17, 2022.
Article in English | MEDLINE | ID: mdl-35366977

ABSTRACT

Inflammatory bowel diseases (IBD) involve repetitive bouts of inflammation in the intestinal tract and can result in severe morbidity for patients. Moreover, long-standing IBD increases the risk for developing intestinal neoplasia. Although several factors including immune cell activity, microbiota and diet have been implicated in IBD pathogenesis, it is still considered a disease of idiopathic origin. Therefore, much work is needed to identify the critical mediators in IBD onset, severity and response to treatment. Mouse models are useful for identifying factors that contribute to IBD and the efficacy of therapy, which can then be tested in humans. There are currently multiple IBD models including the use of chemical induction, genetic manipulation and modulation of the immune response. The T cell transfer colitis model provides a quality mimic of human IBD that is T cell driven and results in inflammation in both the ileum and colon. Here, we have provided a detailed step-by-step protocol to induce inflammation and assess disease severity using this model. Such a detailed methodologic description will help to increase its utilization to advance IBD research.


Subject(s)
Colitis , Inflammatory Bowel Diseases , Animals , Colitis/pathology , Disease Models, Animal , Humans , Inflammation , Inflammatory Bowel Diseases/etiology , Inflammatory Bowel Diseases/pathology , Mice , T-Lymphocytes
9.
Cancer Res ; 82(8): 1464-1466, 2022 04 15.
Article in English | MEDLINE | ID: mdl-35425965

ABSTRACT

Dietary interventions including alterations in the amount or type of specific macronutrients have been shown to mediate antineoplastic effects in preclinical tumor models, but the underlying mechanisms are only partially understood. In this issue of Cancer Research, Wei and colleagues demonstrate that restoring ketogenesis in the colorectal cancer microenvironment decreases the KLF5-dependent synthesis of CXCL12 by cancer-associated fibroblasts, ultimately enhancing tumor infiltration by immune effector cells and increasing the therapeutic efficacy of an immune checkpoint inhibitor specific for PD-1. These findings provide a novel, therapeutically actionable link between suppressed ketogenesis and immunoevasion in the colorectal cancer microenvironment. See related article by Wei et al., p. 1575.


Subject(s)
Cancer-Associated Fibroblasts , Colorectal Neoplasms , Ketosis , Cancer-Associated Fibroblasts/pathology , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/pathology , Humans , Tumor Microenvironment/immunology
10.
Trends Cancer ; 7(8): 668-670, 2021 08.
Article in English | MEDLINE | ID: mdl-34219053

ABSTRACT

Several recent preclinical studies have demonstrated that simultaneously blocking exogenous and endogenous sources of serine in malignant cells mediates superior anticancer effects as compared with limiting either source alone. Here, we critically summarize key developments in targeting serine to treat cancer and discuss persisting challenges for implementing such a therapeutic approach in patients.


Subject(s)
Antimetabolites, Antineoplastic/pharmacology , Diet, Protein-Restricted , Neoplasms/therapy , Serine/antagonists & inhibitors , Antimetabolites, Antineoplastic/therapeutic use , Cell Line, Tumor , Combined Modality Therapy/methods , Dietary Proteins/adverse effects , Dietary Proteins/metabolism , Humans , Neoplasms/metabolism , Phosphoglycerate Dehydrogenase/antagonists & inhibitors , Phosphoglycerate Dehydrogenase/metabolism , Phosphoric Monoester Hydrolases/antagonists & inhibitors , Phosphoric Monoester Hydrolases/metabolism , Serine/biosynthesis , Transaminases/antagonists & inhibitors , Transaminases/metabolism , Xenograft Model Antitumor Assays
11.
Am J Physiol Gastrointest Liver Physiol ; 321(2): G232-G242, 2021 08 01.
Article in English | MEDLINE | ID: mdl-34133236

ABSTRACT

The Western diet has been suggested to contribute to the rising incidence of inflammatory bowel diseases. This has led to the hypothesis that fructose, a component of the Western diet, could play a role in the pathogenesis of inflammatory bowel diseases. A high-fructose diet is known to exacerbate experimental colitis. This study tested whether the expression of GLUT5, the fructose transporter, is a determinant of the severity of experimental colitis during elevated fructose consumption and whether ileal inflammation is associated with altered GLUT5 expression in Crohn's disease. Studies in genetically engineered mice showed that in comparison to Glut5+/+ mice, feeding a 15 kcal% fructose diet to Glut5-/- mice led to worse dextran sodium sulfate (DSS)-induced colitis. This effect was associated with elevated levels of colonic fructose and a shift in the fecal microbiota in Glut5-/- mice. Importantly, treatment with broad-spectrum antibiotics protected against the worsening of colitis mediated by dietary fructose in Glut5-/- mice. Gene expression analysis revealed that GLUT5 levels are reduced in the intestines of patients with ileal Crohn's disease. Moreover, levels of GLUT5 negatively correlated with expression of proinflammatory mediators in these samples. Collectively, these results demonstrate that dietary constituent (fructose)-host gene (GLUT5) interactions can shape the colonic microbiota, thereby impacting the severity of colitis.NEW & NOTEWORTHY This study provides the first evidence that reduced levels of GLUT5, the fructose transporter, worsen experimental colitis upon fructose feeding, an effect mediated by changes in the gut microbiota. Moreover, GLUT5 expression is reduced in Crohn's ileitis. Overall, these findings demonstrate the importance of interactions between dietary fructose and host GLUT5 as determinants of both the composition of colonic microbiota and severity of experimental colitis.


Subject(s)
Colitis, Ulcerative/metabolism , Crohn Disease/metabolism , Fructose/metabolism , Glucose Transporter Type 5/metabolism , Animals , Colitis, Ulcerative/etiology , Dietary Sugars/adverse effects , Dietary Sugars/metabolism , Fructose/adverse effects , Gastrointestinal Microbiome , Glucose Transporter Type 5/genetics , Humans , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Mice , Mice, Inbred C57BL , Sodium Dodecyl Sulfate/toxicity
12.
NPJ Breast Cancer ; 7(1): 58, 2021 May 24.
Article in English | MEDLINE | ID: mdl-34031428

ABSTRACT

ID proteins are helix-loop-helix (HLH) transcriptional regulators frequently overexpressed in cancer. ID proteins inhibit basic-HLH transcription factors often blocking differentiation and sustaining proliferation. A small-molecule, AGX51, targets ID proteins for degradation and impairs ocular neovascularization in mouse models. Here we show that AGX51 treatment of cancer cell lines impairs cell growth and viability that results from an increase in reactive oxygen species (ROS) production upon ID degradation. In mouse models, AGX51 treatment suppresses breast cancer colonization in the lung, regresses the growth of paclitaxel-resistant breast tumors when combined with paclitaxel and reduces tumor burden in sporadic colorectal neoplasia. Furthermore, in cells and mice, we fail to observe acquired resistance to AGX51 likely the result of the inability to mutate the binding pocket without loss of ID function and efficient degradation of the ID proteins. Thus, AGX51 is a first-in-class compound that antagonizes ID proteins, shows strong anti-tumor effects and may be further developed for the management of multiple cancers.

13.
Methods Cell Biol ; 163: 123-135, 2021.
Article in English | MEDLINE | ID: mdl-33785161

ABSTRACT

Long-standing inflammatory bowel diseases (IBD) increase the risk for the development of colorectal cancer (CRC). This increase is due in large part to chronic intestinal inflammation which exposes the epithelium to pro-carcinogenic factors. Moreover, enhanced mucosal proliferation associated with repetitive wound healing events following an inflammatory episode, further enhance this pro-tumorigenic environment. Although multiple factors involved in IBD pathogenesis and its associated neoplasia have been identified, more work is needed to develop and improve therapies to ameliorate disease and thus reduce CRC risk. Murine models have served as useful tools to identify factors involved in the pathogenesis of colitis-associated neoplasia and test therapies. These include both chemically-induced and genetic engineering approaches, resulting in chronic inflammation and tumor development. Here, we present a step-by-step method of inducing inflammation-associated colon neoplasia by combining administration of azoxymethane and dextran sodium sulfate in mice. A detailed description of this methodology will facilitate its use in the scientific community with the goals of further elucidating the mechanisms underlying colitis-associated tumorigenesis and developing risk reducing interventions.


Subject(s)
Colitis , Colonic Neoplasms , Colorectal Neoplasms , Animals , Azoxymethane/toxicity , Colitis/chemically induced , Colonic Neoplasms/chemically induced , Colorectal Neoplasms/chemically induced , Dextran Sulfate/toxicity , Disease Models, Animal , Mice , Mice, Inbred C57BL , Sulfates
14.
J Vis Exp ; (168)2021 02 05.
Article in English | MEDLINE | ID: mdl-33616099

ABSTRACT

Understanding the tissue and cellular changes that occur in the acute injury response as well as during the wound healing process is of paramount importance when studying diseases of the gastrointestinal (GI) tract. The murine colonic pinch biopsy model is a useful tool to define these processes. Additionally, the interplay between gut luminal content (e.g., microbes) and the colon can be studied. However, wound induction and the ability to track wound closure over time in a reliable manner can be challenging. Moreover, tissue preparation and orientation must be carried out in a standardized way to optimally interrogate histologic and molecular changes. Here, we present a detailed method describing biopsy-induced injury and the monitoring of wound closure through repeat colonoscopies. An approach is described that ensures consistent and reproducible measurements of wound size, the ability to collect the wound bed for molecular analyses as well as visualize the wound bed upon sectioning of tissues. The ability to successfully carry out these techniques allows for studies of the acute injury response, wound healing and luminal-host interactions within the colon.


Subject(s)
Colon/cytology , Colonoscopy/methods , Image-Guided Biopsy/methods , Wound Healing , Animals , Colon/pathology , Colon/surgery , Mice
15.
Cancer Res ; 81(9): 2275-2288, 2021 05 01.
Article in English | MEDLINE | ID: mdl-33526512

ABSTRACT

Serine is a nonessential amino acid generated by the sequential actions of phosphoglycerate dehydrogenase (PHGDH), phosphoserine aminotransferase (PSAT1), and phosphoserine phosphatase (PSPH). Increased serine biosynthesis occurs in several cancers and supports tumor growth. In addition, cancer cells can harness exogenous serine to enhance their metabolism and proliferation. Here we tested the relative contributions of exogenous and endogenous sources of serine on the biology of colorectal cancer. In murine tumors, Apc status was identified as a determinant of the expression of genes controlling serine synthesis. In patient samples, PSAT1 was overexpressed in both colorectal adenomas and adenocarcinomas. Combining genetic deletion of PSAT1 with exogenous serine deprivation maximally suppressed the proliferation of colorectal cancer cells and induced profound metabolic defects including diminished nucleotide production. Inhibition of serine synthesis enhanced the transcriptional changes following exogenous serine removal as well as alterations associated with DNA damage. Both loss of PSAT1 and removal of serine from the diet were necessary to suppress colorectal cancer xenograft growth and enhance the antitumor activity of 5-fluorouracil (5-FU). Restricting endogenous and exogenous serine in vitro augmented 5-FU-induced cell death, DNA damage, and metabolic perturbations, likely accounting for the observed antitumor effect. Collectively, our results suggest that both endogenous and exogenous sources of serine contribute to colorectal cancer growth and resistance to 5-FU. SIGNIFICANCE: These findings provide insights into the metabolic requirements of colorectal cancer and reveal a novel approach for its treatment. GRAPHICAL ABSTRACT: http://cancerres.aacrjournals.org/content/canres/81/9/2275/F1.large.jpg.


Subject(s)
Antimetabolites, Antineoplastic/administration & dosage , Colonic Neoplasms/diet therapy , Colonic Neoplasms/metabolism , Diet/methods , Drug Resistance, Neoplasm/drug effects , Fluorouracil/administration & dosage , Serine/deficiency , Aged , Animals , Colonic Neoplasms/genetics , Colonic Neoplasms/pathology , DNA Damage , Drug Resistance, Neoplasm/genetics , Female , Gene Expression , Gene Expression Regulation, Neoplastic , HCT116 Cells , Humans , Male , Mice , Mice, Nude , Mice, Transgenic , Middle Aged , Pregnancy , Serine/genetics , Transaminases/deficiency , Transaminases/genetics , Treatment Outcome , Tumor Burden/drug effects , Tumor Burden/genetics , Xenograft Model Antitumor Assays
16.
Carcinogenesis ; 42(6): 842-852, 2021 06 21.
Article in English | MEDLINE | ID: mdl-33513602

ABSTRACT

Diet is believed to be an important factor in the pathogenesis of inflammatory bowel disease. High consumption of dietary fructose has been shown to exacerbate experimental colitis, an effect mediated through the gut microbiota. This study evaluated whether dietary alterations could attenuate the detrimental effects of a high-fructose diet (HFrD) in experimental colitis. First, we determined whether the procolitic effects of a HFrD could be reversed by switching mice from a HFrD to a control diet. This diet change completely prevented HFrD-induced worsening of acute colitis, in association with a rapid normalization of the microbiota. Second, we tested the effects of dietary fiber, which demonstrated that psyllium was the most effective type of fiber for protecting against HFrD-induced worsening of acute colitis, compared with pectin, inulin, or cellulose. In fact, supplemental psyllium nearly completely prevented the detrimental effects of the HFrD, an effect associated with a shift in the gut microbiota. We next determined whether the protective effects of these interventions could be extended to chronic colitis and colitis-associated tumorigenesis. Using the azoxymethane/dextran sodium sulfate model, we first demonstrated that HFrD feeding exacerbated chronic colitis and increased colitis-associated tumorigenesis. Using the same dietary changes tested in the acute colitis setting, we also showed that mice were protected from HFrD-mediated enhanced chronic colitis and tumorigenesis, upon either diet switching or psyllium supplementation. Taken together, these findings suggest that high consumption of fructose may enhance colon tumorigenesis associated with long-standing colitis, an effect that could be reduced by dietary alterations.


Subject(s)
Colitis/complications , Colorectal Neoplasms/prevention & control , Dextran Sulfate/toxicity , Diet , Dietary Fiber/administration & dosage , Fructose/toxicity , Inflammation/prevention & control , Animals , Colitis/chemically induced , Colitis/pathology , Colorectal Neoplasms/etiology , Colorectal Neoplasms/pathology , Inflammation/etiology , Inflammation/pathology , Male , Mice , Mice, Inbred C57BL
17.
Cell Mol Gastroenterol Hepatol ; 11(2): 525-550, 2021.
Article in English | MEDLINE | ID: mdl-32961355

ABSTRACT

BACKGROUND & AIMS: The incidence of inflammatory bowel diseases has increased over the last half century, suggesting a role for dietary factors. Fructose consumption has increased in recent years. Recently, a high fructose diet (HFrD) was shown to enhance dextran sodium sulfate (DSS)-induced colitis in mice. The primary objectives of the current study were to elucidate the mechanism(s) underlying the pro-colitic effects of dietary fructose and to determine whether this effect occurs in both microbially driven and genetic models of colitis. METHODS: Antibiotics and germ-free mice were used to determine the relevance of microbes for HFrD-induced worsening of colitis. Mucus thickness and quality were determined by histologic analyses. 16S rRNA profiling, in situ hybridization, metatranscriptomic analyses, and fecal metabolomics were used to determine microbial composition, spatial distribution, and metabolism. The significance of HFrD on pathogen and genetic-driven models of colitis was determined by using Citrobacter rodentium infection and Il10-/- mice, respectively. RESULTS: Reducing or eliminating bacteria attenuated HFrD-mediated worsening of DSS-induced colitis. HFrD feeding enhanced access of gut luminal microbes to the colonic mucosa by reducing thickness and altering the quality of colonic mucus. Feeding a HFrD also altered gut microbial populations and metabolism including reduced protective commensal and bile salt hydrolase-expressing microbes and increased luminal conjugated bile acids. Administration of conjugated bile acids to mice worsened DSS-induced colitis. The HFrD also worsened colitis in Il10-/- mice and mice infected with C rodentium. CONCLUSIONS: Excess dietary fructose consumption has a pro-colitic effect that can be explained by changes in the composition, distribution, and metabolic function of resident enteric microbiota.


Subject(s)
Colitis/immunology , Dietary Sugars/adverse effects , Fructose/adverse effects , Gastrointestinal Microbiome/drug effects , Animals , Citrobacter rodentium/pathogenicity , Colitis/diagnosis , Colitis/genetics , Colitis/microbiology , Colon/immunology , Colon/microbiology , Colon/pathology , Dextran Sulfate/administration & dosage , Dextran Sulfate/toxicity , Disease Models, Animal , Feces/microbiology , Female , Gastrointestinal Microbiome/immunology , Humans , Interleukin-10/genetics , Intestinal Mucosa/immunology , Intestinal Mucosa/microbiology , Intestinal Mucosa/pathology , Male , Mice , Mice, Knockout , Severity of Illness Index
18.
Int Rev Cell Mol Biol ; 347: 1-26, 2019.
Article in English | MEDLINE | ID: mdl-31451211

ABSTRACT

As compared to their normal counterparts, neoplastic cells exhibit a variety of metabolic changes that reflect not only genetic and epigenetic defects underlying malignant transformation, but also the nutritional and immunobiological conditions of the tumor microenvironment. Such alterations, including the so-called Warburg effect (an increase in glucose uptake largely feeding anabolic and antioxidant metabolism), have attracted considerable attention as potential targets for the development of novel anticancer therapeutics. However, very few drugs specifically conceived to target bioenergetic cancer metabolism are currently approved by regulatory agencies for use in humans. This reflects the elevated degree of heterogeneity and redundancy in the metabolic circuitries exploited by neoplastic cells from different tumors (even of the same type), as well as the resemblance of such metabolic pathways to those employed by highly proliferating normal cells. Here, we summarize the major metabolic alterations that accompany oncogenesis, the potential of targeting bioenergetic metabolism for cancer therapy, and the obstacles that still prevent the clinical translation of such a promising therapeutic paradigm.


Subject(s)
Carbohydrate Metabolism , Carcinogenesis/metabolism , Lipid Metabolism , Metabolic Networks and Pathways/drug effects , Neoplasms , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Carbohydrate Metabolism/drug effects , Carcinogenesis/drug effects , Diet Therapy , Energy Metabolism/drug effects , Humans , Lipid Metabolism/drug effects , Molecular Targeted Therapy , Neoplasms/metabolism , Neoplasms/physiopathology , Neoplasms/therapy , Tumor Microenvironment/drug effects
19.
Am J Pathol ; 189(3): 492-501, 2019 03.
Article in English | MEDLINE | ID: mdl-30803496

ABSTRACT

The complex relationship between diet and metabolism is an important contributor to cellular metabolism and health. Over the past few decades, a central role for mammalian target of rapamycin (mTOR) in the regulation of multiple cellular processes, including the response to food intake, maintaining homeostasis, and the pathogenesis of disease, has been shown. Herein, we first review our current understanding of the biochemical functions of mTOR and its response to fluctuations in hormone levels, like insulin. Second, we highlight the role of mTOR in lipogenesis, adipogenesis, ß-oxidation of lipids, and ketosis of carbohydrates, lipids, and proteins. Special attention is paid to recent advances in mTOR signaling in white versus brown adipose tissues. Finally, we review how mTOR regulates cardiovascular health and disease. Together, these insights define a clearer picture of the connection between mTOR signaling, metabolic health, and disease.


Subject(s)
Adipose Tissue, Brown/metabolism , Cardiovascular Diseases/metabolism , Metabolic Diseases/metabolism , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , Adipogenesis , Adipose Tissue, Brown/pathology , Animals , Cardiovascular Diseases/pathology , Humans , Lipogenesis , Metabolic Diseases/pathology
20.
Obes Surg ; 29(5): 1593-1601, 2019 05.
Article in English | MEDLINE | ID: mdl-30623320

ABSTRACT

BACKGROUND AND AIMS: Bariatric surgery, such as vertical sleeve gastrectomy (VSG), is the most effective long-term treatment for obesity. However, there are conflicting reports on the effect of bariatric surgery on inflammatory bowel disease (IBD). Bariatric surgery increases bile acid concentrations, which can decrease inflammation by signaling through the bile acid receptor, TGR5. TGR5 signaling protects against chemically induced colitis in mice. VSG increases circulating bile acid concentrations to increase TGR5 signaling, which contributes to improved metabolic regulation after VSG. Therefore, we investigated the effect of VSG on chemically induced colitis development and the role of TGR5 in this context. METHODS: VSG or sham surgery was performed in high fat diet-fed male Tgr5+/+ and Tgr5-/- littermates. Sham-operated mice were food restricted to match their body weight to VSG-operated mice. Colitis was induced with 2.5% dextran sodium sulfate (DSS) in water post-operatively. Body weight, energy intake, fecal scoring, colon histopathology, colonic markers of inflammation, goblet cell counts, and colonic microRNA-21 levels were assessed. RESULTS: VSG decreased body weight independently of genotype. Consistent with previous work, genetic ablation of TGR5 increased the severity of DSS-induced colitis. Notably, despite the effect of VSG to decrease body weight and increase TGR5 signaling, VSG increased the severity of DSS-induced colitis. VSG-induced increases in colitis were associated with increased colonic expression of TNF-α, IL-6, MCP-1, and microRNA-21. CONCLUSIONS: While our data demonstrate that TGR5 protects against colitis, they also demonstrate that VSG potentiates chemically induced colitis in mice. These data suggest that individuals undergoing VSG may be at increased risk for developing colitis; however, further study is needed.


Subject(s)
Colitis/prevention & control , Gastrectomy/adverse effects , Obesity/surgery , Receptors, G-Protein-Coupled/metabolism , Animals , Bariatric Surgery/adverse effects , Bariatric Surgery/methods , Colitis/etiology , Colitis/metabolism , Colitis/pathology , Colon/pathology , Disease Models, Animal , Gastrectomy/methods , Inflammatory Bowel Diseases/etiology , Inflammatory Bowel Diseases/metabolism , Inflammatory Bowel Diseases/pathology , Inflammatory Bowel Diseases/prevention & control , Male , Mice , Mice, Inbred C57BL , Obesity/metabolism , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...