Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 78
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Cell ; 160(1-2): 269-84, 2015 Jan 15.
Article in English | MEDLINE | ID: mdl-25594183

ABSTRACT

The stem cells that maintain and repair the postnatal skeleton remain undefined. One model suggests that perisinusoidal mesenchymal stem cells (MSCs) give rise to osteoblasts, chondrocytes, marrow stromal cells, and adipocytes, although the existence of these cells has not been proven through fate-mapping experiments. We demonstrate here that expression of the bone morphogenetic protein (BMP) antagonist gremlin 1 defines a population of osteochondroreticular (OCR) stem cells in the bone marrow. OCR stem cells self-renew and generate osteoblasts, chondrocytes, and reticular marrow stromal cells, but not adipocytes. OCR stem cells are concentrated within the metaphysis of long bones not in the perisinusoidal space and are needed for bone development, bone remodeling, and fracture repair. Grem1 expression also identifies intestinal reticular stem cells (iRSCs) that are cells of origin for the periepithelial intestinal mesenchymal sheath. Grem1 expression identifies distinct connective tissue stem cells in both the bone (OCR stem cells) and the intestine (iRSCs).


Subject(s)
Bone and Bones/cytology , Intercellular Signaling Peptides and Proteins/metabolism , Intestine, Small/cytology , Mesenchymal Stem Cells/cytology , Animals , Cartilage/metabolism , Intestine, Small/metabolism , Mesenchymal Stem Cells/metabolism , Mice , Mice, Inbred C57BL
2.
J Nanobiotechnology ; 21(1): 133, 2023 Apr 24.
Article in English | MEDLINE | ID: mdl-37095500

ABSTRACT

Nanotechnology-enabled sensors or nanosensors are emerging as promising new tools for various in-vivo life science applications such as biosensing, components of delivery systems, and probes for spatial bioimaging. However, as with a wide range of synthetic biomaterials, tissue responses have been observed depending on cell types and various nanocomponent properties. The tissue response is critical for determining the acute and long term health of the organism and the functional lifetime of the material in-vivo. While nanomaterial properties can contribute significantly to the tissue response, it may be possible to circumvent adverse reactions by formulation of the encapsulation vehicle. In this study, five formulations of poly (ethylene glycol) diacrylate (PEGDA) hydrogel-encapsulated fluorescent nanosensors were implanted into SKH-1E mice, and the inflammatory responses were tracked in order to determine the favorable design rules for hydrogel encapsulation and minimization of such responses. Hydrogels with higher crosslinking density were found to allow faster resolution of acute inflammation. Five different immunocompromised mice lines were utilized for comparison across different inflammatory cell populations and responses. Degradation products of the gels were also characterized. Finally, the importance of the tissue response in determining functional lifetime was demonstrated by measuring the time-dependent nanosensor deactivation following implantation into animal models.


Subject(s)
Hydrogels , Polyethylene Glycols , Mice , Animals , Inflammation/chemically induced , Biocompatible Materials
3.
Gastroenterology ; 157(2): 492-506.e2, 2019 08.
Article in English | MEDLINE | ID: mdl-30998992

ABSTRACT

BACKGROUND & AIMS: Barrett's esophagus (BE) is a precursor to esophageal adenocarcinoma (EAC). Progression from BE to cancer is associated with obesity, possibly due to increased abdominal pressure and gastroesophageal reflux disease, although this pathogenic mechanism has not been proven. We investigated whether environmental or dietary factors associated with obesity contribute to the progression of BE to EAC in mice. METHODS: Tg(ED-L2-IL1RN/IL1B)#Tcw mice (a model of BE, called L2-IL1B mice) were fed a chow (control) or high-fat diet (HFD) or were crossbred with mice that express human interleukin (IL) 8 (L2-IL1B/IL8 mice). Esophageal tissues were collected and analyzed for gene expression profiles and by quantitative polymerase chain reaction, immunohistochemistry, and flow cytometry. Organoids were established from BE tissue of mice and cultured with serum from lean or obese individuals or with neutrophils from L2-IL1B mice. Feces from mice were analyzed by 16s ribosomal RNA sequencing and compared to 16s sequencing data from patients with dysplasia or BE. L2-IL1B were mice raised in germ-free conditions. RESULTS: L2-IL1B mice fed an HFD developed esophageal dysplasia and tumors more rapidly than mice fed the control diet; the speed of tumor development was independent of body weight. The acceleration of dysplasia by the HFD in the L2-IL1B mice was associated with a shift in the gut microbiota and an increased ratio of neutrophils to natural killer cells in esophageal tissues compared with mice fed a control diet. We observed similar differences in the microbiomes from patients with BE that progressed to EAC vs patients with BE that did not develop into cancer. Tissues from dysplasias of L2-IL1B mice fed the HFD contained increased levels of cytokines that are produced in response to CXCL1 (the functional mouse homolog of IL8, also called KC). Serum from obese patients caused organoids from L2-IL1B/IL8 mice to produce IL8. BE tissues from L2-IL1B mice fed the HFD and from L2-IL1B/IL8 mice contained increased numbers of myeloid cells and cells expressing Cxcr2 and Lgr5 messenger RNAs (epithelial progenitors) compared with mice fed control diets. BE tissues from L2-IL1B mice raised in germ-free housing had fewer progenitor cells and developed less dysplasia than in L2-IL1 mice raised under standard conditions; exposure of fecal microbiota from L2-IL1B mice fed the HFD to L2-IL1B mice fed the control diet accelerated tumor development. CONCLUSIONS: In a mouse model of BE, we found that an HFD promoted dysplasia by altering the esophageal microenvironment and gut microbiome, thereby inducing inflammation and stem cell expansion, independent of obesity.


Subject(s)
Adenocarcinoma/pathology , Barrett Esophagus/pathology , Esophageal Neoplasms/pathology , Gastrointestinal Microbiome/physiology , Interleukin-8/metabolism , Obesity/pathology , Adenocarcinoma/immunology , Adult , Aged , Animals , Barrett Esophagus/immunology , Carcinogenesis/immunology , Carcinogenesis/pathology , Diet, High-Fat/adverse effects , Disease Models, Animal , Disease Progression , Esophageal Neoplasms/immunology , Esophagus/immunology , Esophagus/pathology , Feces/microbiology , Female , Healthy Volunteers , Humans , Interleukin-8/genetics , Interleukin-8/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Middle Aged , Obesity/blood , Obesity/immunology , Organoids , Serum/immunology , Serum/metabolism , Time Factors , Tissue Culture Techniques
4.
Int J Mol Sci ; 21(23)2020 Nov 24.
Article in English | MEDLINE | ID: mdl-33255175

ABSTRACT

The prevalence of gastric Helicobacter pylori (Hp) infection is ~50% of the world population. However, how Hp infection influences inflammatory bowel disease in humans is not fully defined. In this study, we examined whether co-infection with Hp influenced Helicobacter hepaticus (Hh)-induced intestinal pathology in Rag2-/- mice. Rag2-/- mice of both sexes were infected with Hh, of which a subgroup was followed by infection with Hp two weeks later. Co-infected males, but not females, had significantly higher total colitis index scores in the colon at both 10 and 21 weeks post-Hh infection (WPI) and developed more severe dysplasia at 21 WPI compared with mono-Hh males. There were no significant differences in colonization levels of gastric Hp and colonic Hh between sexes or time-points. In addition, mRNA levels of colonic Il-1ß, Ifnγ, Tnfα, Il-17A, Il-17F, Il-18, and Il-23, which play important roles in the development and function of proinflammatory innate lymphoid cell groups 1 and 3, were significantly up-regulated in the dually infected males compared with mono-Hh males at 21 WPI. These data suggest that concomitant Hp infection enhances the inflammatory responses in the colon of-Hh-infected Rag2-/- males, which results in more severe colitis and dysplasia.


Subject(s)
Colitis/genetics , DNA-Binding Proteins/genetics , Helicobacter Infections/genetics , Sex Characteristics , Animals , Coinfection/genetics , Coinfection/microbiology , Colitis/microbiology , Colitis/pathology , Female , Helicobacter Infections/microbiology , Helicobacter Infections/pathology , Helicobacter hepaticus/pathogenicity , Helicobacter pylori/pathogenicity , Humans , Male , Mice , Mice, Knockout
5.
Can Vet J ; 61(4): 396-400, 2020 04.
Article in English | MEDLINE | ID: mdl-32255825

ABSTRACT

Gastric carcinoma is not commonly reported in dogs. There is an increased risk, however, in certain breeds such as the Belgian Tervuren. Review of the Veterinary Medical Database (VMDB) established an increase in risk for gastric carcinoma in the chow chow breed. In 106 chow chow dogs signs commenced, on average, 3 weeks before definitive diagnosis. The most common clinical signs were vomiting, loss of appetite, diarrhea, and melena. Most affected dogs were euthanized, without treatment, within 2 weeks of diagnosis. Two dogs which were treated aggressively (surgery and chemotherapy) survived a considerably longer time (12 and 36 months). Histologically, these chow chow dogs comprised a similar histologic type as familial gastric carcinoma in humans; diffuse-type carcinoma that was enriched in the signet ring and mucinous variants. Understanding the pathogenesis of diffuse gastric carcinoma in the chow chow dog may provide insight into the biology of this aggressive cancer in humans.


Risques et caractéristiques d'un carcinome gastrique chez le chien de race chow-chow. Le carcinome gastrique n'est par rapporté fréquemment chez les chiens. Il y a toutefois une augmentation du risque chez certaines espèces telle que le Tervuren belge. Une revue de la base de données Veterinary Medical Database (VMDB) a établi une augmentation dans le risque pour le carcinome gastrique chez la race chow chow. Chez 106 chiens chow chow les signes débutèrent, en moyenne, 3 semaines avant le diagnostic définitif. Les signes cliniques les plus fréquents étaient vomissement, perte d'appétit, diarrhée et méléna. La plupart des chiens affectés furent euthanasiés, sans traitement, à l'intérieur de 2 semaines du diagnostic. Deux chiens furent traités de manière agressive (chirurgie et chimiothérapie) ont survécu beaucoup plus longtemps (12 et 36 mois). Histologiquement, ces chiens chow chow comprennent un type histologique similaire aux carcinomes gastrique familiaux chez les humains; le carcinome de type-diffus qui s'est développé dans les variants de cellules en bague à chatons et mucineux. Comprendre la pathogénie du carcinome gastrique diffus chez le chien chow chow pourrait fournir des informations sur la biologie de ce cancer agressif chez l'humain.(Traduit par Dr Serge Messier).


Subject(s)
Adenocarcinoma/veterinary , Dog Diseases/genetics , Stomach Neoplasms/genetics , Stomach Neoplasms/veterinary , Animals , Breeding , Dogs
6.
J Toxicol Pathol ; 33(4): 297-302, 2020 Oct.
Article in English | MEDLINE | ID: mdl-33239848

ABSTRACT

Spontaneous nephroblastomas are uncommon tumors of laboratory rats. This report describes a spontaneous nephroblastoma with peritoneal metastasis in an 11-month-old, female Sprague Dawley rat. The rat was part of a breeding program and presented 15 days post parturition with clinical signs including tachypnea, dyspnea and abdominal distension. At necropsy, the right kidney was markedly enlarged by an expansile pale-tan to white multinodular mass with extension into the retroperitoneal space, with multifocal variably sized nodules involving the mesentery, and surface of pancreas, liver, uterus, and ovarian bursa. The rat also had severe bicavitary effusion. Histologically, the renal parenchyma of the affected kidney was replaced by a moderately cellular, poorly-demarcated, non-encapsulated, multilobulated mass that appeared to compress the adjacent renal outer medulla and cortex. Three distinct neoplastic cell populations were identified in this renal tumor: epithelial cells (convoluted and dilated tubules / rare primitive glomeruloid structures), mesenchymal (neoplastic spindle cells in connective tissue), and blastemal cells (primitive neoplastic cells). The extrarenal nodular masses were predominantly composed of neoplastic mesenchymal and pleomorphic blastemal cells. Immunohistochemically, neoplastic epithelial cells in the renal mass were positive for pancytokeratin, and blastemal cells in both renal and extrarenal masses were positive for Wilms' tumor 1 protein (WT1) and vimentin. Neoplastic mesenchymal elements in both renal and extrarenal masses were positive for vimentin. The neoplasm was negative for chromogranin A and S100. The tumor was classified as an anaplastic nephroblastoma with metastasis to the mesentery and peritoneal organs.

7.
Lab Invest ; 99(12): 1887-1905, 2019 12.
Article in English | MEDLINE | ID: mdl-31399638

ABSTRACT

Gastric cancer (GC) is the third leading cause of cancer-related deaths worldwide and is strongly associated with chronic Helicobacter pylori (Hp) infection. The ability of Hp to closely adhere to the gastric surface protective mucous layer containing mucins (MUC in humans and Muc in animals), primarily Muc5ac, is integral in the stepwise pathogenesis from gastritis to cancer. To probe the role of Muc5ac in Hp-induced gastric pathology, Muc5ac-/- and Muc5ac+/+ (WT) mice were experimentally infected with Hp Sydney strain (SS1). At 16 weeks and 32 weeks post infection (wpi), groups of mice were euthanized and evaluated for the following: gastric histopathological parameters, immunohistochemical expression of mucins (Muc5ac, Muc1, Muc2), Trefoil factor family proteins (Tff1 and Tff2), Griffonia (Bandeiraea) simplicifolia lectin II (GSL II) (mucous metaplasia marker) and Clusterin (Spasmolytic Polypeptide Expressing Metaplasia (SPEM) marker), Hp colonization density by qPCR and gastric cytokine mRNA levels. Our results demonstrate that Muc5ac-/- mice developed spontaneous antro-pyloric proliferation, adenomas and in one case with neuroendocrine differentiation; these findings were independent of Hp infection along with strong expression levels of Tff1, Tff2 and Muc1. Hp-infected Muc5ac-/- mice had significantly lowered gastric corpus mucous metaplasia at 16 wpi and 32 wpi (P = 0.0057 and P = 0.0016, respectively), with a slight reduction in overall gastric corpus pathology. GSII-positive mucous neck cells were decreased in Hp-infected Muc5ac-/- mice compared to WT mice and clusterin positivity was noted within metaplastic glands in both genotypes following Hp infection. Additionally, Hp colonization densities were significantly higher in Muc5ac-/- mice compared to WT at 16 wpi in both sexes (P = 0.05) along with a significant reduction in gastric Tnfα (16 wpi-males and females, P = 0.017 and P = 0.036, respectively and 32 wpi-males only, P = 0.025) and Il-17a (16 wpi-males) (P = 0.025). Taken together, our findings suggest a protective role for MUC5AC/Muc5ac in maintaining gastric antral equilibrium and inhibiting Hp colonization and associated inflammatory pathology.


Subject(s)
Adenoma/microbiology , Helicobacter Infections/complications , Mucin 5AC/physiology , Pyloric Antrum/pathology , Stomach Neoplasms/microbiology , Animals , Female , Gastric Mucosa/metabolism , Gastric Mucosa/pathology , Helicobacter Infections/pathology , Helicobacter pylori/physiology , Host-Pathogen Interactions , Hyperplasia , Male , Metaplasia , Mice, Inbred C57BL , Mucins/metabolism , Pyloric Antrum/metabolism , Trefoil Factors/metabolism
8.
Int J Cancer ; 145(4): 1042-1054, 2019 08 15.
Article in English | MEDLINE | ID: mdl-30977112

ABSTRACT

Inflammatory bowel disease and colonic tumors induced by Helicobacter hepaticus (Hh) infection in susceptible mouse strains are utilized to dissect the mechanisms underlying similar human diseases. In our study, infection with genotoxic cytolethal distending toxin-producing Hh in 129/SvEv Rag2-/- Il10-/- gpt delta (RagIl10gpt) mice of both sexes for 21 weeks induced significantly more severe cecal and colonic pathology compared to uninfected controls. The mutation frequencies in the infected RagIl10gpt males were 2.1-fold higher for the cecum and 1.7-fold higher for the colon than male RagIl10gpt controls. In addition, there was a 12.5-fold increase of G:C-to-T:A transversions in the colon of Hh-infected males compared to controls. In contrast, there was no statistical significance in mutation frequencies between infected female Rag2Il10gpt mice and controls. Moreover, Hh infection in RagIl10gpt males significantly up-regulated transcription of Tnfα and iNos, and decreased mRNA levels of cecal Atm compared to the infected females; there was no significant difference in mRNA levels of Il-22, Il-17A, Ifnγ and Atr between the infected males and females. Significantly higher levels of cecal and colonic iNos expression and γH2AX-positive epithelial cells (a biomarker for double-strand DNA breaks [DSB]) in Hh-infected Rag2Il10gpt males vs. Hh-infected females were noted. Finally, Hh infection and associated inflammation increased levels of intestinal mucosa-associated genotoxic colibactin-producing pks+ Escherichia coli. Elevated Tnfα and iNos responses and bacterial genotoxins, in concert with suppression of the DSB repair responses, may have promoted mutagenesis in the lower bowel mucosa of Hh-infected male RagIl10gpt mice.


Subject(s)
Colon/microbiology , DNA-Binding Proteins/genetics , Helicobacter Infections/genetics , Helicobacter hepaticus/pathogenicity , Interleukin-10/genetics , Intestinal Mucosa/microbiology , Mutagenesis/genetics , Animals , Epithelial Cells/microbiology , Female , Helicobacter Infections/microbiology , Inflammation/genetics , Inflammation/microbiology , Inflammatory Bowel Diseases/genetics , Inflammatory Bowel Diseases/microbiology , Interleukin-17/genetics , Male , Mice , Mutation/genetics , RNA, Messenger/genetics , Sex Factors , Tumor Necrosis Factor-alpha/genetics
9.
Gastroenterology ; 155(6): 1852-1867, 2018 12.
Article in English | MEDLINE | ID: mdl-30195448

ABSTRACT

BACKGROUND & AIMS: Loss of claudin 18 (CLDN18), a membrane-spanning tight junction protein, occurs during early stages of development of gastric cancer and associates with shorter survival times of patients. We investigated whether loss of CLDN18 occurs in mice that develop intraepithelial neoplasia with invasive glands due to infection with Helicobacter pylori, and whether loss is sufficient to promote the development of similar lesions in mice with or without H pylori infection. METHODS: We performed immunohistochemical analyses in levels of CLDN18 in archived tissues from B6:129 mice infected with H pylori for 6 to 15 months. We analyzed gastric tissues from B6:129S5-Cldn18tm1Lex/Mmucd mice, in which the CLDN18 gene was disrupted in gastric tissues (CLDN18-knockout mice), or from control mice with a full-length CLDN18 gene (CLDN18+/+; B6:129S5/SvEvBrd) or heterozygous disruption of CLDN18 (CLDN18+/-; B6:129S5/SvEvBrd) that were infected with H pylori SS1 or PMSS1 at 6 weeks of age and tissues collected for analysis at 20 and 30 weeks after infection. Tissues from CLDN18-knockout mice and control mice with full-length CLDN18 gene expression were also analyzed without infection at 7 weeks and 2 years after birth. Tissues from control and CLDN18-knockout mice were analyzed by electron microscopy, stained by conventional methods and analyzed for histopathology, prepared by laser capture microdissection and analyzed by RNAseq, and immunostained for lineage markers, proliferation markers, and stem cell markers and analyzed by super-resolution or conventional confocal microscopy. RESULTS: CLDN18 had a basolateral rather than apical tight junction localization in gastric epithelial cells. B6:129 mice infected with H pylori, which developed intraepithelial neoplasia with invasive glands, had increasing levels of CLDN18 loss over time compared with uninfected mice. In B6:129 mice infected with H pylori compared with uninfected mice, CLDN18 was first lost from most gastric glands followed by disrupted and reduced expression in the gastric neck and in surface cells. Gastric tissues from CLDN18-knockout mice had low levels of inflammation but increased cell proliferation, expressed markers of intestinalized proliferative spasmolytic polypeptide-expressing metaplasia, and had defects in signal transduction pathways including p53 and STAT signaling by 7 weeks after birth compared with full-length CLDN18 gene control mice. By 20 to 30 weeks after birth, gastric tissues from uninfected CLDN18-knockout mice developed intraepithelial neoplasia that invaded the submucosa; by 2 years, gastric tissues contained large and focally dysplastic polypoid tumors with invasive glands that invaded the serosa. CONCLUSIONS: H pylori infection of B6:129 mice reduced the expression of CLDN18 early in gastric cancer progression, similar to previous observations from human gastric tissues. CLDN18 regulates cell lineage differentiation and cellular signaling in mouse stomach; CLDN18-knockout mice develop intraepithelial neoplasia and then large and focally dysplastic polypoid tumors in the absence of H pylori infection.


Subject(s)
Carcinoma in Situ/metabolism , Claudins/metabolism , Helicobacter Infections/metabolism , Stomach Neoplasms/metabolism , Animals , Carcinoma in Situ/etiology , Carcinoma in Situ/microbiology , Carcinoma in Situ/pathology , Cell Differentiation , Cell Lineage , Disease Progression , Female , Helicobacter Infections/complications , Helicobacter pylori , Hyperplasia/genetics , Hyperplasia/microbiology , Male , Mice , Mice, Knockout , Signal Transduction , Stomach/microbiology , Stomach/pathology , Stomach Neoplasms/etiology , Stomach Neoplasms/microbiology , Stomach Neoplasms/pathology
10.
Cell Microbiol ; 19(7)2017 07.
Article in English | MEDLINE | ID: mdl-28111881

ABSTRACT

Multiple pathogenic Gram-negative bacteria produce the cytolethal distending toxin (CDT) with activity of DNase I; CDT can induce DNA double-strand breaks (DSBs), G2/M cell cycle arrest, and apoptosis in cultured mammalian cells. However, the link of CDT to in vivo tumorigenesis is not fully understood. In this study, 129/SvEv Rag2-/- mice were gavaged with wild-type Helicobacter hepatics 3B1(Hh) and its isogenic cdtB mutant HhcdtBm7, followed by infection for 10 and 20 weeks (WPI). HhCDT deficiency did not affect cecal colonization levels of HhcdtBm7, but attenuated severity of cecal pathology in HhcdtBm7-infected mice. Of importance, preneoplasic dysplasia was progressed to cancer from 10 to 20 WPI in the Hh-infected mice but not in the HhcdtBm7-infected mice. In addition, the loss of HhCDT significantly dampened transcriptional upregulation of cecal Tnfα and Il-6, but elevated Il-10 mRNA levels when compared to Hh at 10 WPI. Furthermore, the presence of HhCDT increased numbers of lower bowel intestinal γH2AX-positive epithelial cells (a marker of DSBs) at both 10 and 20 WPI and augmented phospho-Stat3 foci+ intestinal crypts (activation of Stat3) at 20 WPI. Our findings suggest that CDT promoted Hh carcinogenesis by enhancing DSBs and activation of the Tnfα/Il-6-Stat3 signaling pathway.


Subject(s)
Bacterial Toxins/metabolism , Carcinogenesis/pathology , DNA Breaks, Double-Stranded , Helicobacter hepaticus/pathogenicity , Interleukin-6/metabolism , Intestines/pathology , STAT3 Transcription Factor/metabolism , Tumor Necrosis Factor-alpha/metabolism , Animals , Apoptosis , Bacterial Toxins/genetics , Cecum/microbiology , Cecum/pathology , Female , G2 Phase Cell Cycle Checkpoints , Helicobacter hepaticus/genetics , Histones/metabolism , Interleukin-10/genetics , Male , Mice , Mice, Transgenic , Neoplasms/microbiology , Neoplasms/pathology , RNA, Messenger/biosynthesis , Signal Transduction/physiology
11.
PLoS Genet ; 11(2): e1004901, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25647331

ABSTRACT

Mutations are a critical driver of cancer initiation. While extensive studies have focused on exposure-induced mutations, few studies have explored the importance of tissue physiology as a modulator of mutation susceptibility in vivo. Of particular interest is inflammation, a known cancer risk factor relevant to chronic inflammatory diseases and pathogen-induced inflammation. Here, we used the fluorescent yellow direct repeat (FYDR) mice that harbor a reporter to detect misalignments during homologous recombination (HR), an important class of mutations. FYDR mice were exposed to cerulein, a potent inducer of pancreatic inflammation. We show that inflammation induces DSBs (γH2AX foci) and that several days later there is an increase in cell proliferation. While isolated bouts of inflammation did not induce HR, overlap between inflammation-induced DNA damage and inflammation-induced cell proliferation induced HR significantly. To study exogenously-induced DNA damage, animals were exposed to methylnitrosourea, a model alkylating agent that creates DNA lesions relevant to both environmental exposures and cancer chemotherapy. We found that exposure to alkylation damage induces HR, and importantly, that inflammation-induced cell proliferation and alkylation induce HR in a synergistic fashion. Taken together, these results show that, during an acute bout of inflammation, there is a kinetic barrier separating DNA damage from cell proliferation that protects against mutations, and that inflammation-induced cell proliferation greatly potentiates exposure-induced mutations. These studies demonstrate a fundamental mechanism by which inflammation can act synergistically with DNA damage to induce mutations that drive cancer and cancer recurrence.


Subject(s)
Cell Proliferation/genetics , DNA Damage/genetics , Inflammation/genetics , Neoplasms/genetics , Animals , DNA Repair/genetics , Histones/genetics , Homologous Recombination , Humans , Inflammation/complications , Inflammation/pathology , Mice , Mutation , Neoplasms/etiology , Neoplasms/pathology , Pancreas/pathology , Risk Factors
12.
Proc Natl Acad Sci U S A ; 111(45): E4878-86, 2014 Nov 11.
Article in English | MEDLINE | ID: mdl-25349415

ABSTRACT

Inflammation is accompanied by the release of highly reactive oxygen and nitrogen species (RONS) that damage DNA, among other cellular molecules. Base excision repair (BER) is initiated by DNA glycosylases and is crucial in repairing RONS-induced DNA damage; the alkyladenine DNA glycosylase (Aag/Mpg) excises several DNA base lesions induced by the inflammation-associated RONS release that accompanies ischemia reperfusion (I/R). Using mouse I/R models we demonstrate that Aag(-/-) mice are significantly protected against, rather than sensitized to, I/R injury, and that such protection is observed across three different organs. Following I/R in liver, kidney, and brain, Aag(-/-) mice display decreased hepatocyte death, cerebral infarction, and renal injury relative to wild-type. We infer that in wild-type mice, Aag excises damaged DNA bases to generate potentially toxic abasic sites that in turn generate highly toxic DNA strand breaks that trigger poly(ADP-ribose) polymerase (Parp) hyperactivation, cellular bioenergetics failure, and necrosis; indeed, steady-state levels of abasic sites and nuclear PAR polymers were significantly more elevated in wild-type vs. Aag(-/-) liver after I/R. This increase in PAR polymers was accompanied by depletion of intracellular NAD and ATP levels plus the translocation and extracellular release of the high-mobility group box 1 (Hmgb1) nuclear protein, activating the sterile inflammatory response. We thus demonstrate the detrimental effects of Aag-initiated BER during I/R and sterile inflammation, and present a novel target for controlling I/R-induced injury.


Subject(s)
Brain/enzymology , DNA Glycosylases/metabolism , DNA Repair , Kidney/enzymology , Liver/enzymology , Reperfusion Injury/enzymology , Acute Kidney Injury/enzymology , Acute Kidney Injury/genetics , Acute Kidney Injury/pathology , Animals , Brain/pathology , Brain Infarction/enzymology , Brain Infarction/genetics , Brain Infarction/pathology , Cell Death , DNA Damage , DNA Glycosylases/genetics , Enzyme Induction/genetics , HMGB1 Protein/genetics , HMGB1 Protein/metabolism , Hepatocytes/enzymology , Hepatocytes/pathology , Inflammation/enzymology , Inflammation/genetics , Inflammation/pathology , Kidney/pathology , Liver/pathology , Mice , Mice, Knockout , Poly(ADP-ribose) Polymerases/genetics , Poly(ADP-ribose) Polymerases/metabolism , Reactive Nitrogen Species/metabolism , Reactive Oxygen Species/metabolism , Reperfusion Injury/genetics , Reperfusion Injury/pathology
13.
Carcinogenesis ; 37(12): 1190-1198, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27655833

ABSTRACT

A novel Helicobacter species Helicobacter japonicum was isolated from the stomach and intestines of clinically normal mice received from three institutes from Japan. The novel Helicobacter sp. was microaerobic, grew at 37°C and 42°C, was catalase and oxidase positive, but urease negative. It is most closely related to the 16S rRNA gene of H.muridarum (98.6%); to the 23S rRNA gene of H.hepaticus (97.9%); to the hsp60 gene of H.typhlonius (87%). The novel Helicobacter sp. has in vitro cytolethal distending toxin (CDT) activity; its cdtB gene sequence has 83.8% identity with that of H.hepaticus The whole genome sequence of H.japonicum MIT 01-6451 has a 2.06-Mb genome length with a 37.5% G + C content. When the organism was inoculated into C57BL/129 IL10-/- mice, it was cultured from the stomach, colon and cecum of infected mice at 6 and 10 weeks post-infection. The cecum had the highest H.japonicum colonization levels by quantitative PCR. The histopathology of the lower bowel was characterized by moderate to severe inflammation, mild edema, epithelial defects, mild to severe hyperplasia, dysplasia and carcinoma. Inflammatory cytokines IFNγ, TNFα and IL17a, as well as iNOS were significantly upregulated in the cecal tissue of infected mice. These results demonstrate that the novel H.japonicum can induce inflammatory bowel disease and carcinoma in IL10-/- mice and highlights the importance of identifying novel Helicobacter spp. especially when they are introduced from outside mouse colonies from different geographic locations.


Subject(s)
Carcinoma/microbiology , Helicobacter/pathogenicity , Inflammatory Bowel Diseases/microbiology , Intestines/microbiology , Animals , Carcinoma/pathology , Helicobacter/genetics , Helicobacter/isolation & purification , Helicobacter Infections/microbiology , Helicobacter Infections/pathology , Inflammatory Bowel Diseases/pathology , Interferon-gamma/biosynthesis , Interleukin-10/genetics , Interleukin-17/biosynthesis , Intestines/pathology , Japan , Mice , Mice, Inbred C57BL , Mice, Knockout , Nitric Oxide Synthase Type II/biosynthesis , Tumor Necrosis Factor-alpha/biosynthesis , Typhlitis/microbiology , Typhlitis/pathology
14.
Helicobacter ; 21(3): 201-17, 2016 Jun.
Article in English | MEDLINE | ID: mdl-26348390

ABSTRACT

BACKGROUND: Aged hamsters naturally infected with novel Helicobacter spp. classified in the H. bilis cluster develop hepatobiliary lesions and typhlocolitis. METHODS: To determine whether enterohepatic H. spp. contribute to disease, Helicobacter-free hamsters were experimentally infected with H. spp. after suppression of intestinal bacteria by tetracycline treatment of dams and pups. After antibiotic withdrawal, weanlings were gavaged with four H. bilis-like Helicobacter spp. isolated from hamsters or H. bilis ATCC 43879 isolated from human feces and compared to controls (n = 7 per group). RESULTS: Helicobacter bilis 43879-dosed hamsters were necropsied at 33 weeks postinfection (WPI) due to the lack of detectable infection by fecal PCR; at necropsy, 5 of 7 were weakly PCR positive but lacked intestinal lesions. The remaining hamsters were maintained for ~95 WPI; chronic H. spp. infection in hamsters (6/7) was confirmed by PCR, bacterial culture, fluorescent in situ hybridization, and ELISA. Hamsters had mild-to-moderate typhlitis, and three of the male H. spp.-infected hamsters developed small intestinal lymphoma, in contrast to one control. Of the three lymphomas in H. spp.-infected hamsters, one was a focal ileal mucosa-associated lymphoid tissue (MALT) B-cell lymphoma, while the other two were multicentric small intestinal large B-cell lymphomas involving both the MALT and extra-MALT mucosal sites with lymphoepithelial lesions. The lymphoma in the control hamster was a diffuse small intestinal lymphoma with a mixed population of T and B cells. CONCLUSIONS: Results suggest persistent H. spp. infection may augment risk for gastrointestinal MALT origin lymphomas. This model is consistent with H. pylori/heilmannii-associated MALT lymphoma in humans and could be further utilized to investigate the mechanisms of intestinal lymphoma development.


Subject(s)
Gastrointestinal Diseases/microbiology , Helicobacter Infections/complications , Helicobacter/pathogenicity , Lymphoma, B-Cell, Marginal Zone/microbiology , Age Factors , Animals , Anti-Bacterial Agents/administration & dosage , Antibodies, Bacterial/biosynthesis , Antibodies, Bacterial/blood , Cecum/pathology , Colon/pathology , Cricetinae , Disease Models, Animal , Female , Helicobacter/immunology , Helicobacter Infections/microbiology , Humans , In Situ Hybridization, Fluorescence , Liver/pathology , Male , Mesocricetus , Specific Pathogen-Free Organisms
15.
Gut ; 64(4): 544-53, 2015 Apr.
Article in English | MEDLINE | ID: mdl-24951258

ABSTRACT

OBJECTIVE: Progastrin is the incompletely cleaved precursor of gastrin that is secreted by G-cells in the gastric antrum. Both gastrin and progastrin bind to the CCK2 receptor (Cckbr or CCK2R) expressed on a subset of gastric epithelial cells. Little is known about how gastrin peptides and CCK2R regulate gastric stem cells and carcinogenesis. Interconversion among progenitors in the intestine is documented, but the mechanisms by which this occurs are poorly defined. DESIGN: We generated CCK2R-CreERT mice and performed inducible lineage tracing experiments. CCK2R+ antral cells and Lgr5+ antral stem cells were cultured in a three-dimensional in vitro system. We crossed progastrin-overexpressing mice with Lgr5-GFP-CreERT mice and examined the role of progastrin and CCK2R in Lgr5+ stem cells during MNU-induced carcinogenesis. RESULTS: Through lineage tracing experiments, we found that CCK2R defines antral stem cells at position +4, which overlapped with an Lgr5(neg or low) cell population but was distinct from typical antral Lgr5(high) stem cells. Treatment with progastrin interconverts Lgr5(neg or low) CCK2R+ cells into Lgr5(high) cells, increases CCK2R+ cell numbers and promotes gland fission and carcinogenesis in response to the chemical carcinogen MNU. Pharmacological inhibition or genetic ablation of CCK2R attenuated progastrin-dependent stem cell expansion and carcinogenesis. CONCLUSIONS: CCK2R labels +4 antral stem cells that can be activated and expanded by progastrin, thus identifying one hormonal trigger for gastric stem cell interconversion and a potential target for gastric cancer chemoprevention and therapy.


Subject(s)
Carcinogenesis , Pyloric Antrum/cytology , Receptor, Cholecystokinin B/physiology , Stem Cells/physiology , Animals , Cells, Cultured , Gastrins/physiology , Mice , Protein Precursors/physiology
16.
Am J Physiol Gastrointest Liver Physiol ; 308(4): G335-49, 2015 Feb 15.
Article in English | MEDLINE | ID: mdl-25477375

ABSTRACT

"Black" pigment gallstones form in sterile gallbladder bile in the presence of excess bilirubin conjugates ("hyperbilirubinbilia") from ineffective erythropoiesis, hemolysis, or induced enterohepatic cycling (EHC) of unconjugated bilirubin. Impaired gallbladder motility is a less well-studied risk factor. We evaluated the spontaneous occurrence of gallstones in adult germfree (GF) and conventionally housed specific pathogen-free (SPF) Swiss Webster (SW) mice. GF SW mice were more likely to have gallstones than SPF SW mice, with 75% and 23% prevalence, respectively. In GF SW mice, gallstones were observed predominately in heavier, older females. Gallbladders of GF SW mice were markedly enlarged, contained sterile black gallstones composed of calcium bilirubinate and <1% cholesterol, and had low-grade inflammation, edema, and epithelial hyperplasia. Hemograms were normal, but serum cholesterol was elevated in GF compared with SPF SW mice, and serum glucose levels were positively related to increasing age. Aged GF and SPF SW mice had deficits in gallbladder smooth muscle activity. In response to cholecystokinin (CCK), gallbladders of fasted GF SW mice showed impaired emptying (females: 29%; males: 1% emptying), whereas SPF SW females and males emptied 89% and 53% of volume, respectively. Bilirubin secretion rates of GF SW mice were not greater than SPF SW mice, repudiating an induced EHC. Gallstones likely developed in GF SW mice because of gallbladder hypomotility, enabled by features of GF physiology, including decreased intestinal CCK concentration and delayed intestinal transit, as well as an apparent genetic predisposition of the SW stock. GF SW mice may provide a valuable model to study gallbladder stasis as a cause of black pigment gallstones.


Subject(s)
Bile Pigments/metabolism , Cholecystokinin/metabolism , Gallbladder/metabolism , Gallstones/etiology , Muscle Contraction , Muscle, Smooth/metabolism , Age Factors , Animals , Body Weight , Calcium/metabolism , Female , Gallbladder/pathology , Gallbladder/physiopathology , Gallstones/genetics , Gallstones/metabolism , Gallstones/pathology , Gallstones/physiopathology , Genetic Predisposition to Disease , Germ-Free Life , Hydrogen-Ion Concentration , Logistic Models , Male , Mice , Muscle, Smooth/pathology , Muscle, Smooth/physiopathology , Risk Factors , Sex Factors , Species Specificity , Time Factors
17.
Proc Natl Acad Sci U S A ; 109(27): E1820-9, 2012 Jul 03.
Article in English | MEDLINE | ID: mdl-22689960

ABSTRACT

Helicobacter hepaticus-infected Rag2(-/-) mice emulate many aspects of human inflammatory bowel disease, including the development of colitis and colon cancer. To elucidate mechanisms of inflammation-induced carcinogenesis, we undertook a comprehensive analysis of histopathology, molecular damage, and gene expression changes during disease progression in these mice. Infected mice developed severe colitis and hepatitis by 10 wk post-infection, progressing into colon carcinoma by 20 wk post-infection, with pronounced pathology in the cecum and proximal colon marked by infiltration of neutrophils and macrophages. Transcriptional profiling revealed decreased expression of DNA repair and oxidative stress response genes in colon, but not in liver. Mass spectrometric analysis revealed higher levels of DNA and RNA damage products in liver compared to colon and infection-induced increases in 5-chlorocytosine in DNA and RNA and hypoxanthine in DNA. Paradoxically, infection was associated with decreased levels of DNA etheno adducts. Levels of nucleic acid damage from the same chemical class were strongly correlated in both liver and colon. The results support a model of inflammation-mediated carcinogenesis involving infiltration of phagocytes and generation of reactive species that cause local molecular damage leading to cell dysfunction, mutation, and cell death. There are strong correlations among histopathology, phagocyte infiltration, and damage chemistry that suggest a major role for neutrophils in inflammation-associated cancer progression. Further, paradoxical changes in nucleic acid damage were observed in tissue- and chemistry-specific patterns. The results also reveal features of cell stress response that point to microbial pathophysiology and mechanisms of cell senescence as important mechanistic links to cancer.


Subject(s)
Colitis/microbiology , Colonic Neoplasms/microbiology , DNA Damage/immunology , Helicobacter Infections/complications , Helicobacter Infections/immunology , Helicobacter hepaticus/immunology , Animals , Biomarkers , Chronic Disease , Colitis/immunology , Colonic Neoplasms/genetics , Colonic Neoplasms/immunology , DNA-Binding Proteins/genetics , Disease Models, Animal , Gene Expression/immunology , Hepatitis/immunology , Hepatitis/microbiology , Macrophages/immunology , Mass Spectrometry , Mice , Mice, 129 Strain , Mice, Mutant Strains , Neutrophils/immunology , Oxidative Stress/immunology , RNA/genetics
18.
Gut ; 63(1): 54-63, 2014 Jan.
Article in English | MEDLINE | ID: mdl-23812323

ABSTRACT

OBJECTIVES: Gastric colonisation with intestinal flora (IF) has been shown to promote Helicobacter pylori (Hp)-associated gastric cancer. However, it is unknown if the mechanism involves colonisation with specific or diverse microbiota secondary to gastric atrophy. DESIGN: Gastric colonisation with Altered Schaedler's flora (ASF) and Hp were correlated with pathology, immune responses and mRNA expression for proinflammatory and cancer-related genes in germ-free (GF), Hp monoassociated (mHp), restricted ASF (rASF; 3 species), and specific pathogen-free (complex IF), hypergastrinemic INS-GAS mice 7 months postinfection. RESULTS: Male mice cocolonised with rASFHp or IFHp developed the most severe pathology. IFHp males had the highest inflammatory responses, and 40% developed invasive gastrointestinal intraepithelial neoplasia (GIN). Notably, rASFHp colonisation was highest in males and 23% developed invasive GIN with elevated expression of inflammatory biomarkers. Lesions were less severe in females and none developed GIN. Gastritis in male rASFHp mice was accompanied by decreased Clostridum species ASF356 and Bacteroides species ASF519 colonisation and an overgrowth of Lactobacillus murinus ASF361, supporting that inflammation-driven atrophy alters the gastric niche for GI commensals. Hp colonisation also elevated expression of IL-11 and cancer-related genes, Ptger4 and Tgf-ß, further supporting that Hp infection accelerates gastric cancer development in INS-GAS mice. CONCLUSIONS: rASFHp colonisation was sufficient for GIN development in males, and lower GIN incidence in females was associated with lower inflammatory responses and gastric commensal and Hp colonisation. Colonisation efficiency of commensals appears more important than microbial diversity and lessens the probability that specific gastrointestinal pathogens are contributing to cancer risk.


Subject(s)
Adenocarcinoma/microbiology , Gastric Mucosa/microbiology , Gastritis, Atrophic/microbiology , Helicobacter Infections/complications , Helicobacter pylori , Stomach Neoplasms/microbiology , Symbiosis , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Animals , Bacteroides , Biomarkers/metabolism , Biomarkers, Tumor/metabolism , Carcinogenesis , Clostridium , Cytokines/metabolism , Female , Gastric Mucosa/metabolism , Gastric Mucosa/pathology , Gastritis, Atrophic/complications , Gastritis, Atrophic/metabolism , Gastritis, Atrophic/pathology , Helicobacter Infections/metabolism , Helicobacter Infections/pathology , Intestine, Large/microbiology , Lactobacillus , Male , Mice , Specific Pathogen-Free Organisms , Stomach Neoplasms/metabolism , Stomach Neoplasms/pathology
19.
Gut ; 63(3): 385-94, 2014 Mar.
Article in English | MEDLINE | ID: mdl-23729675

ABSTRACT

OBJECTIVE: To investigate the role of obesity-associated inflammation and immune modulation in gastric carcinogenesis during Helicobacter-induced chronic gastric inflammation. DESIGN: C57BL/6 male mice were infected with H felis and placed on a high-fat diet (45% calories from fat). Study animals were analysed for gastric and adipose pathology, inflammatory markers in serum, stomach and adipose tissue, and immune responses in blood, spleen, stomach and adipose tissue. RESULTS: H felis-induced gastric carcinogenesis was accelerated in diet-induced obese mice compared with lean controls. Obesity increased bone marrow-derived immature myeloid cells in blood and gastric tissue of H felis-infected mice. Obesity also led to elevations in CD4 T cells, IL-17A, granulocyte macrophage colony-stimulating factor, phosphorylated STAT3 and prosurvival gene expression in gastric tissue of H felis-infected mice. Conversely, in adipose tissue of obese mice, H felis infection increased macrophage accumulation and expression of IL-6, C-C motif ligand 7 (CCL7) and leptin. Finally, the combination of obesity and gastric inflammation synergistically increased serum proinflammatory cytokines, including IL-6. CONCLUSIONS: Here, we have established a model to study the molecular mechanism by which obesity predisposes individuals to gastric cancer. In H felis-infected mice, obesity increased proinflammatory immune responses and accelerated gastric carcinogenesis. Interestingly, gastric inflammation augmented obesity-induced adipose inflammation and production of adipose-derived factors in obese, but not lean, mice. Our findings suggest that obesity accelerates Helicobacter-associated gastric cancer through cytokine-mediated cross-talk between inflamed gastric and adipose tissues, augmenting immune responses at both tissue sites, and thereby contributing to a protumorigenic gastric microenvironment.


Subject(s)
Gastritis/complications , Helicobacter Infections/complications , Helicobacter felis , Myeloid Progenitor Cells/physiology , Obesity/complications , Stomach Neoplasms/etiology , Th17 Cells/physiology , Animals , Biomarkers/metabolism , Cell Movement , Cytokines/metabolism , Diet, High-Fat , Flow Cytometry , Gastritis/diagnosis , Gastritis/metabolism , Gastritis/microbiology , Helicobacter Infections/immunology , Inflammation/complications , Inflammation/metabolism , Inflammation/microbiology , Male , Mice , Mice, Inbred C57BL , Obesity/immunology , STAT3 Transcription Factor/metabolism , Stomach Neoplasms/immunology , Stomach Neoplasms/microbiology
20.
Gastroenterology ; 144(1): 155-66, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23041326

ABSTRACT

BACKGROUND & AIMS: Interleukin (IL)-8 has an important role in initiating inflammation in humans, attracting immune cells such as neutrophils through their receptors CXCR1 and CXCR2. IL-8 has been proposed to contribute to chronic inflammation and cancer. However, mice do not have the IL-8 gene, so human cancer cell lines and xenograft studies have been used to study the role of IL-8 in colon and gastric carcinogenesis. We generated mice that carry a bacterial artificial chromosome that encompasses the entire human IL-8 gene, including its regulatory elements (IL-8Tg mice). METHODS: We studied the effects of IL-8 expression in APCmin(+/-) mice and IL-8Tg mice given azoxymethane and dextran sodium sulfate (DSS). We also examined the effects of IL-8 expression in gastric cancer in INS-GAS mice that overexpress gastrin and IL-8Tg mice infected with Helicobacter felis. RESULTS: In IL-8Tg mice, expression of human IL-8 was controlled by its own regulatory elements, with virtually no messenger RNA or protein detectable under basal conditions. IL-8 was strongly up-regulated on systemic or local inflammatory stimulation, increasing mobilization of immature CD11b(+)Gr-1(+) myeloid cells (IMCs) with thioglycolate-induced peritonitis, DSS-induced colitis, and H. felis-induced gastritis. IL-8 was increased in colorectal tumors from patients and IL-8Tg mice compared with nontumor tissues. IL-8Tg mice developed more tumors than wild-type mice following administration of azoxymethane and DSS. Expression of IL-8 increased tumorigenesis in APCmin(+/-) mice compared with APCmin(+/-) mice that lack IL-8; this was associated with increased numbers of IMCs and angiogenesis in the tumors. CONCLUSIONS: IL-8 contributes to gastrointestinal carcinogenesis by mobilizing IMCs and might be a therapeutic target for gastrointestinal cancers.


Subject(s)
Cell Movement/drug effects , Cell Transformation, Neoplastic/metabolism , Colitis/metabolism , Colonic Neoplasms/metabolism , Gastritis/metabolism , Interleukin-8/metabolism , Myeloid Cells/drug effects , RNA, Messenger/metabolism , Animals , Azoxymethane , Cell Line, Tumor , Colitis/chemically induced , Colonic Neoplasms/chemically induced , Colonic Neoplasms/pathology , Dendritic Cells/metabolism , Dextran Sulfate , Gastritis/microbiology , Helicobacter Infections/complications , Helicobacter felis , Humans , Interleukin-8/genetics , Interleukin-8/pharmacology , Lipopolysaccharides/pharmacology , Macrophages/metabolism , Mice , Mice, Transgenic , Myeloid Cells/metabolism , Primary Cell Culture , Tumor Burden , Up-Regulation/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL