Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
Add more filters











Publication year range
1.
Crohns Colitis 360 ; 6(2): otae031, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38800569

ABSTRACT

Background: Cannabis is used by patients with Crohn's disease (CD) and ulcerative colitis (UC) as an alternative to, or in combination with, conventional therapies to treat symptoms such as abdominal pain, poor sleep, and reduced appetite. The clinical efficacy of cannabis for these disorders is controversial, with some studies showing harmful outcomes associated with its use. Previous studies suggest that cannabis is used by ~12% of patients with UC and ~16% of patients with CD in the USA despite legal prohibition. Methods: We conducted a prospective cohort study of adult patients with inflammatory bowel diseases (IBD) followed in a Canadian tertiary care center. Patients completed an online 40-question survey that included demographics, IBD disease history, cannabis use, and the Short Inflammatory Bowel Disease Questionnaire (SIBDQ). Results: Completed surveys were obtained from 254 participants (148 with CD, 90 with UC, and 16 with indeterminate colitis). Recent cannabis use was reported by 41% of CD and 31% of UC participants. Interestingly, only 46% of participants who used cannabis discussed their use with their physician. Participants who recently used cannabis reported more abdominal pain, poor appetite, and flatulence, and importantly this was associated with lower SIBDQ scores (recent use 37 vs non-recent use 40). Conclusions: Cannabis use among patients with IBD has more than doubled since its legalization. Cannabis use is associated with worse abdominal symptoms and quality of life. Physicians should inquire about cannabis use and optimize symptom control with evidence-based therapies.

2.
EMBO J ; 42(24): e114462, 2023 Dec 11.
Article in English | MEDLINE | ID: mdl-37934086

ABSTRACT

Mammalian cells repress expression of repetitive genomic sequences by forming heterochromatin. However, the consequences of ectopic repeat expression remain unclear. Here we demonstrate that inhibitors of EZH2, the catalytic subunit of the Polycomb repressive complex 2 (PRC2), stimulate repeat misexpression and cell death in resting splenic B cells. B cells are uniquely sensitive to these agents because they exhibit high levels of histone H3 lysine 27 trimethylation (H3K27me3) and correspondingly low DNA methylation at repeat elements. We generated a pattern recognition receptor loss-of-function mouse model, called RIC, with mutations in Rigi (encoding for RIG-I), Ifih1 (MDA5), and Cgas. In both wildtype and RIC mutant B cells, EZH2 inhibition caused loss of H3K27me3 at repetitive elements and upregulated their expression. However, NF-κB-dependent expression of inflammatory chemokines and subsequent cell death was suppressed by the RIC mutations. We further show that inhibition of EZH2 in cancer cells requires the same pattern recognition receptors to activate an interferon response. Together, the results reveal chemokine expression induced by EZH2 inhibitors in B cells as a novel inflammatory response to genomic repeat expression. Given the overlap of genes induced by EZH2 inhibitors and Epstein-Barr virus infection, this response can be described as a form of viral mimicry.


Subject(s)
B-Lymphocytes , Enhancer of Zeste Homolog 2 Protein , Epstein-Barr Virus Infections , Animals , Mice , B-Lymphocytes/drug effects , B-Lymphocytes/metabolism , DNA Methylation , Enhancer of Zeste Homolog 2 Protein/antagonists & inhibitors , Enhancer of Zeste Homolog 2 Protein/genetics , Enhancer of Zeste Homolog 2 Protein/metabolism , Epstein-Barr Virus Infections/genetics , Herpesvirus 4, Human/genetics , Histones/metabolism , Repetitive Sequences, Nucleic Acid
3.
Cell Stem Cell ; 30(8): 1091-1109.e7, 2023 08 03.
Article in English | MEDLINE | ID: mdl-37541213

ABSTRACT

While adult pancreatic stem cells are thought not to exist, it is now appreciated that the acinar compartment harbors progenitors, including tissue-repairing facultative progenitors (FPs). Here, we study a pancreatic acinar population marked by trefoil factor 2 (Tff2) expression. Long-term lineage tracing and single-cell RNA sequencing (scRNA-seq) analysis of Tff2-DTR-CreERT2-targeted cells defines a transit-amplifying progenitor (TAP) population that contributes to normal homeostasis. Following acute and chronic injury, Tff2+ cells, distinct from FPs, undergo depopulation but are eventually replenished. At baseline, oncogenic KrasG12D-targeted Tff2+ cells are resistant to PDAC initiation. However, KrasG12D activation in Tff2+ cells leads to survival and clonal expansion following pancreatitis and a cancer stem/progenitor cell-like state. Selective ablation of Tff2+ cells prior to KrasG12D activation in Mist1+ acinar or Dclk1+ FP cells results in enhanced tumorigenesis, which can be partially rescued by adenoviral Tff2 treatment. Together, Tff2 defines a pancreatic TAP population that protects against Kras-driven carcinogenesis.


Subject(s)
Carcinoma, Pancreatic Ductal , Pancreatic Neoplasms , Humans , Pancreatic Neoplasms/genetics , Trefoil Factor-2/metabolism , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/metabolism , Pancreas/metabolism , Acinar Cells/metabolism , Carcinogenesis/genetics , Carcinogenesis/metabolism
4.
Gastroenterology ; 164(4): 593-609.e13, 2023 04.
Article in English | MEDLINE | ID: mdl-36634827

ABSTRACT

BACKGROUND & AIMS: Colorectal cancer is a leading cause of cancer death, and a major risk factor is chronic inflammation. Despite the link between colitis and cancer, the mechanism by which inflammation leads to colorectal cancer is not well understood. METHODS: To investigate whether different forms of inflammation pose the same risk of cancer, we compared several murine models of colitis (dextran sodium sulfate [DSS], 2,4,6-trinitrobenzene sulfonic acid, 4-ethoxylmethylene-2-phenyloxazol-5-one, Citrobacter rodentium, Fusobacterium nucleatum, and doxorubicin) with respect to their ability to lead to colonic tumorigenesis. We attempted to correlate the severity of colitis and inflammatory profile with the risk of tumorigenesis in both azoxymethane-dependent and Dclk1/APCfl/fl murine models of colitis-associated cancer. RESULTS: DSS colitis reproducibly led to colonic tumors in both mouse models of colitis-associated cancer. In contrast, all other forms of colitis did not lead to cancer. When compared with the colitis not associated with tumorigenesis, DSS colitis was characterized by significantly increased CD11b+F4/80+Ly6Chigh macrophages and CD11b+Ly6G+ neutrophils. Interestingly, depletion of the CD11b+F4/80+Ly6Chigh macrophages inhibited tumorigenesis, whereas depletion of CD11b+Ly6G+ neutrophils had no effect on tumorigenesis. Furthermore, the macrophage-derived cytokines interleukin-1ß, tumor necrosis factor-α, and interleukin-6 were significantly increased in DSS colitis and promoted stemness of Dclk1+ tuft cells that serve as the cellular origin of cancer. CONCLUSIONS: We have identified CD11b+F4/80+Ly6Chigh macrophages as key mediators of cancer initiation in colitis-associated cancer. Development of new therapies that target these cells may provide an effective preventative strategy for colitis-associated cancer.


Subject(s)
Colitis-Associated Neoplasms , Colitis , Animals , Mice , Azoxymethane , Carcinogenesis/metabolism , Cell Plasticity , Colitis/chemically induced , Colitis/complications , Colitis/metabolism , Colitis-Associated Neoplasms/metabolism , Dextran Sulfate/toxicity , Disease Models, Animal , Inflammation/metabolism , Macrophages/metabolism , Mice, Inbred C57BL
5.
Gastroenterology ; 162(3): 890-906, 2022 03.
Article in English | MEDLINE | ID: mdl-34883119

ABSTRACT

BACKGROUND & AIMS: Cancer-associated fibroblasts (CAFs) play an important role in colorectal cancer (CRC) progression and predict poor prognosis in CRC patients. However, the cellular origins of CAFs remain unknown, making it challenging to therapeutically target these cells. Here, we aimed to identify the origins and contribution of colorectal CAFs associated with poor prognosis. METHODS: To elucidate CAF origins, we used a colitis-associated CRC mouse model in 5 different fate-mapping mouse lines with 5-bromodeoxyuridine dosing. RNA sequencing of fluorescence-activated cell sorting-purified CRC CAFs was performed to identify a potential therapeutic target in CAFs. To examine the prognostic significance of the stromal target, CRC patient RNA sequencing data and tissue microarray were used. CRC organoids were injected into the colons of knockout mice to assess the mechanism by which the stromal gene contributes to colorectal tumorigenesis. RESULTS: Our lineage-tracing studies revealed that in CRC, many ACTA2+ CAFs emerge through proliferation from intestinal pericryptal leptin receptor (Lepr)+ cells. These Lepr-lineage CAFs, in turn, express melanoma cell adhesion molecule (MCAM), a CRC stroma-specific marker that we identified with the use of RNA sequencing. High MCAM expression induced by transforming growth factor ß was inversely associated with patient survival in human CRC. In mice, stromal Mcam knockout attenuated orthotopically injected colorectal tumoroid growth and improved survival through decreased tumor-associated macrophage recruitment. Mechanistically, fibroblast MCAM interacted with interleukin-1 receptor 1 to augment nuclear factor κB-IL34/CCL8 signaling that promotes macrophage chemotaxis. CONCLUSIONS: In colorectal carcinogenesis, pericryptal Lepr-lineage cells proliferate to generate MCAM+ CAFs that shape the tumor-promoting immune microenvironment. Preventing the expansion/differentiation of Lepr-lineage CAFs or inhibiting MCAM activity could be effective therapeutic approaches for CRC.


Subject(s)
Cancer-Associated Fibroblasts/pathology , Cancer-Associated Fibroblasts/physiology , Carcinogenesis/pathology , Cell Lineage , Colorectal Neoplasms/pathology , Mesenchymal Stem Cells/physiology , Actins/genetics , Actins/metabolism , Adult , Aged , Aged, 80 and over , Animals , CD146 Antigen/genetics , CD146 Antigen/metabolism , Carcinogenesis/genetics , Carcinogenesis/metabolism , Cell Differentiation , Cell Proliferation , Colorectal Neoplasms/metabolism , Disease Models, Animal , Female , Humans , Intestinal Mucosa/pathology , Ki-67 Antigen/metabolism , Male , Mice , Mice, Transgenic , Middle Aged , Organoids/pathology , Organoids/physiology , Prognosis , Receptors, Leptin/genetics , Receptors, Leptin/metabolism , Sequence Analysis, RNA , Survival Rate , Tumor Microenvironment
6.
EMBO J ; 38(4)2019 02 15.
Article in English | MEDLINE | ID: mdl-30635334

ABSTRACT

During homeostasis, the colonic epithelium is replenished every 3-5 days by rapidly cycling Lgr5+ stem cells. However, various insults can lead to depletion of Lgr5+ stem cells, and colonic epithelium can be regenerated from Lgr5-negative cells. While studies in the small intestine have addressed the lineage identity of the Lgr5-negative regenerative cell population, in the colon this question has remained unanswered. Here, we set out to identify which cell(s) contribute to colonic regeneration by performing genetic fate-mapping studies of progenitor populations in mice. First, using keratin-19 (Krt19) to mark a heterogeneous population of cells, we found that Lgr5-negative cells can regenerate colonic crypts and give rise to Lgr5+ stem cells. Notch1+ absorptive progenitor cells did not contribute to epithelial repair after injury, whereas Atoh1+ secretory progenitors did contribute to this process. Additionally, while colonic Atoh1+ cells contributed minimally to other lineages during homeostasis, they displayed plasticity and contributed to epithelial repair during injury, independent of Lgr5+ cells. Our findings suggest that promotion of secretory progenitor plasticity could enable gut healing in colitis.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Colitis/prevention & control , Colon/cytology , Intestine, Small/cytology , Receptors, G-Protein-Coupled/metabolism , Regeneration , Stem Cells/cytology , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Cells, Cultured , Colitis/chemically induced , Colitis/pathology , Colon/physiology , Homeostasis , Intestine, Small/physiology , Keratin-19/genetics , Keratin-19/metabolism , Mice , Receptor, Notch1/genetics , Receptor, Notch1/metabolism , Receptors, G-Protein-Coupled/genetics , Stem Cells/physiology
7.
Mol Cell ; 73(1): 1-2, 2019 01 03.
Article in English | MEDLINE | ID: mdl-30609388

ABSTRACT

PD-L1 plays a central role in immune recognition of cancer cells. In this issue of Molecular Cell, Jin et al. (2019) report that a phosphorylated retinoblastoma protein contacts the DNA-binding domain of p65 NF-κB, thereby blocking transcription of PD-L1.


Subject(s)
B7-H1 Antigen , Transcription Factor RelA/genetics , Gene Expression Regulation , NF-kappa B/genetics , Retinoblastoma Protein , Signal Transduction
8.
Gastroenterology ; 156(4): 1066-1081.e16, 2019 03.
Article in English | MEDLINE | ID: mdl-30448068

ABSTRACT

BACKGROUND & AIMS: The intestinal epithelium is maintained by long-lived intestinal stem cells (ISCs) that reside near the crypt base. Above the ISC zone, there are short-lived progenitors that normally give rise to lineage-specific differentiated cell types but can dedifferentiate into ISCs in certain circumstances. However, the role of epithelial dedifferentiation in cancer development has not been fully elucidated. METHODS: We performed studies with Bhlha15-CreERT, Lgr5-DTR-GFP, Apcflox/flox, LSL-Notch (IC), and R26-reporter strains of mice. Some mice were given diphtheria toxin to ablate Lgr5-positive cells, were irradiated, or were given 5-fluorouracil, hydroxyurea, doxorubicin, or dextran sodium sulfate to induce intestinal or colonic tissue injury. In intestinal tissues, we analyzed the fate of progeny that expressed Bhlha15. We used microarrays and reverse-transcription PCR to analyze gene expression patterns in healthy and injured intestinal tissues and in tumors. We analyzed gene expression patterns in human colorectal tumors using The Cancer Genome Atlas data set. RESULTS: Bhlha15 identified Paneth cells and short-lived secretory precursors (including pre-Paneth label-retaining cells) located just above the ISC zone in the intestinal epithelium. Bhlha15+ cells had no plasticity after loss of Lgr5-positive cells or irradiation. However, Bhlha15+ secretory precursors started to supply the enterocyte lineage after doxorubicin-induced epithelial injury in a Notch-dependent manner. Sustained activation of Notch converts Bhlha15+ secretory precursors to long-lived enterocyte progenitors. Administration of doxorubicin and expression of an activated form of Notch resulted in a gene expression pattern associated with enterocyte progenitors, whereas only sustained activation of Notch altered gene expression patterns in Bhlha15+ precursors toward those of ISCs. Bhlha15+ enterocyte progenitors with sustained activation of Notch formed intestinal tumors with serrated features in mice with disruption of Apc. In the colon, Bhlha15 marked secretory precursors that became stem-like, cancer-initiating cells after dextran sodium sulfate-induced injury, via activation of Src and YAP signaling. In analyses of human colorectal tumors, we associated activation of Notch with chromosome instability-type tumors with serrated features in the left colon. CONCLUSIONS: In mice, we found that short-lived precursors can undergo permanent reprogramming by activation of Notch and YAP signaling. These cells could mediate tumor formation in addition to traditional ISCs.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Colonic Neoplasms/genetics , Enterocytes/pathology , Intestinal Mucosa/metabolism , Receptors, Notch/metabolism , Stem Cells/metabolism , Transcriptome , Adaptor Proteins, Signal Transducing/metabolism , Animals , Antibiotics, Antineoplastic/pharmacology , Antineoplastic Agents, Hormonal/pharmacology , Basic Helix-Loop-Helix Transcription Factors/genetics , CD24 Antigen/metabolism , Calcium-Binding Proteins , Cell Cycle Proteins , Cell Plasticity , Chromogranin A/genetics , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Doxorubicin/pharmacology , Enterocytes/metabolism , Gene Expression , Gene Expression Profiling , Intercellular Signaling Peptides and Proteins/genetics , Intestinal Mucosa/drug effects , Intestinal Mucosa/pathology , Intestine, Small/cytology , Intestine, Small/metabolism , Mice , Neoplasm Proteins/genetics , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Pancreatitis-Associated Proteins , Paneth Cells , Phosphoproteins/metabolism , Proto-Oncogene Proteins c-kit/metabolism , Receptors, G-Protein-Coupled/metabolism , Signal Transduction , Stem Cells/drug effects , Stem Cells/physiology , Stem Cells/radiation effects , Tamoxifen/pharmacology , YAP-Signaling Proteins , src-Family Kinases/metabolism
9.
Nat Commun ; 9(1): 5293, 2018 12 13.
Article in English | MEDLINE | ID: mdl-30546048

ABSTRACT

We used allogeneic bone marrow transplantation (BMT) and a mouse multistage cutaneous carcinogenesis model to probe recruitment of bone marrow-derived epithelial cells (BMDECs) in skin tumors initiated with the carcinogen, dimethylbenz[a]anthracene (DMBA), and promoted with 12-O-tetradecanolyphorbol-13-acetate (TPA). BMDECs clustered in the lesional epithelium, expressed cytokeratins, proliferated, and stratified. We detected cytokeratin induction in plastic-adherent bone marrow cells (BMCs) cultured in the presence of filter-separated keratinocytes (KCs) and bone morphogenetic protein 5 (BMP5). Lineage-depleted BMCs migrated towards High Mobility Group Box 1 (HMGB1) protein and epidermal KCs in ex vivo invasion assays. Naive female mice receiving BMTs from DMBA-treated donors developed benign and malignant lesions after TPA promotion alone. We conclude that BMDECs contribute to the development of papillomas and dysplasia, demonstrating a systemic contribution to these lesions. Furthermore, carcinogen-exposed BMCs can initiate benign and malignant lesions upon tumor promotion. Ultimately, these findings may suggest targets for treatment of non-melanoma skin cancers.


Subject(s)
Bone Marrow Cells/pathology , Cell Transformation, Neoplastic/chemically induced , Cell Transformation, Neoplastic/pathology , Epithelial Cells/pathology , Skin Neoplasms/pathology , 9,10-Dimethyl-1,2-benzanthracene/toxicity , Animals , Bone Marrow Cells/cytology , Bone Marrow Transplantation , Bone Morphogenetic Protein 5/metabolism , Cell Movement , Cell Plasticity/physiology , Coculture Techniques , Epithelial Cells/cytology , Female , HMGB1 Protein/metabolism , Hair Follicle/cytology , Keratinocytes/pathology , Keratins/metabolism , Male , Mice , Mice, Inbred C57BL , Neoplasm Invasiveness/pathology , Papilloma/pathology , Stem Cells/cytology , Stem Cells/pathology , Tetradecanoylphorbol Acetate/toxicity , Tumor Cells, Cultured
10.
Cell Stem Cell ; 21(6): 747-760.e7, 2017 Dec 07.
Article in English | MEDLINE | ID: mdl-29198940

ABSTRACT

Myeloid-biased hematopoietic stem cells (MB-HSCs) play critical roles in recovery from injury, but little is known about how they are regulated within the bone marrow niche. Here we describe an auto-/paracrine physiologic circuit that controls quiescence of MB-HSCs and hematopoietic progenitors marked by histidine decarboxylase (Hdc). Committed Hdc+ myeloid cells lie in close anatomical proximity to MB-HSCs and produce histamine, which activates the H2 receptor on MB-HSCs to promote their quiescence and self-renewal. Depleting histamine-producing cells enforces cell cycle entry, induces loss of serial transplant capacity, and sensitizes animals to chemotherapeutic injury. Increasing demand for myeloid cells via lipopolysaccharide (LPS) treatment specifically recruits MB-HSCs and progenitors into the cell cycle; cycling MB-HSCs fail to revert into quiescence in the absence of histamine feedback, leading to their depletion, while an H2 agonist protects MB-HSCs from depletion after sepsis. Thus, histamine couples lineage-specific physiological demands to intrinsically primed MB-HSCs to enforce homeostasis.


Subject(s)
Bone Marrow/metabolism , Hematopoietic Stem Cells/metabolism , Histamine/metabolism , Myeloid Cells/metabolism , Animals , Bone Marrow/drug effects , Bone Marrow Transplantation , Flow Cytometry , Hematopoietic Stem Cells/drug effects , Lipopolysaccharides/pharmacology , Mice , Myeloid Cells/drug effects
11.
Endosc Int Open ; 5(7): E675-E682, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28691053

ABSTRACT

BACKGROUND AND STUDY AIMS: Adenoma detection rate (ADR) is an important measure of colonoscopy quality, as are polyp, advanced ADR, and adenocarcinoma detection rates. We investigated whether performance report cards improved these outcome measures. PATIENTS AND METHODS: Endoscopists were given report cards comparing their detection rates to the institutional mean on an annual basis. Detection rates were evaluated at baseline, 1 year after report cards (Year 1), and 2 years after report cards (Year 2). Endoscopists were unaware of the study and received no other interventions. The primary outcome was ADR and secondary outcomes were polyp detection rate (PDR), advanced ADR, and adenocarcinoma detection rate. Multivariate regression was performed to adjust for temporal trends in patient, endoscopists, and procedural factors. RESULTS: Seventeen physicians performed 3,118 screening colonoscopies in patients with positive FOBT or family history of colon cancer. The ADR increased from 34.5 % (baseline) to 39.4 % (Year 1) and 41.2 % (Year 2) ( P  = 0.0037). The PDR increased from 45 % (baseline) to 48.8 % (Year 1) and 51.8 % (Year 2) ( P  = 0.011). There was no significant improvement in advanced ADR or adenocarcinoma detection rates. On multivariate analysis, the ADR increased by 22 % in Year 1 ( P  = 0.03) and 30 % in Year 2 ( P  = 0.008). Among physicians with a baseline ADR < 25 %, improvement in ADR was even greater, increasing 2.2 times by the end of the study ( P  = 0.004). Improvements in ADR were not correlated with specialty although gastroenterologists were 52 % more likely to find an adenoma than general surgeons. CONCLUSIONS: Annual performance report cards increased adenoma detection rates, especially among physicians with low ADR < 25 %.

12.
Cancer Cell ; 31(1): 21-34, 2017 01 09.
Article in English | MEDLINE | ID: mdl-27989802

ABSTRACT

Within the gastrointestinal stem cell niche, nerves help to regulate both normal and neoplastic stem cell dynamics. Here, we reveal the mechanisms underlying the cancer-nerve partnership. We find that Dclk1+ tuft cells and nerves are the main sources of acetylcholine (ACh) within the gastric mucosa. Cholinergic stimulation of the gastric epithelium induced nerve growth factor (NGF) expression, and in turn NGF overexpression within gastric epithelium expanded enteric nerves and promoted carcinogenesis. Ablation of Dclk1+ cells or blockade of NGF/Trk signaling inhibited epithelial proliferation and tumorigenesis in an ACh muscarinic receptor-3 (M3R)-dependent manner, in part through suppression of yes-associated protein (YAP) function. This feedforward ACh-NGF axis activates the gastric cancer niche and offers a compelling target for tumor treatment and prevention.


Subject(s)
Acetylcholine/physiology , Nerve Growth Factor/physiology , Signal Transduction/physiology , Stomach Neoplasms/etiology , Adaptor Proteins, Signal Transducing/physiology , Animals , Cell Cycle Proteins , Doublecortin-Like Kinases , Gastric Mucosa/innervation , Mice , Mice, Inbred C57BL , Phosphoproteins/physiology , Protein Serine-Threonine Kinases/analysis , Receptor, Muscarinic M3/physiology , YAP-Signaling Proteins
13.
Oncotarget ; 8(67): 111012-111025, 2017 Dec 19.
Article in English | MEDLINE | ID: mdl-29340033

ABSTRACT

Mist1 was recently shown to identify a discrete population of stem cells within the isthmus of the oxyntic gland within the gastric corpus. Chief cells at the base of the gastric corpus also express Mist1. The relevance of Mist1 expression as a marker of specific cell populations within the antral glands of the distal stomach, however, is unknown. Using Mist1-CreERT mice, we revealed that Mist1+ antral cells, distinct from the Mist1+ population in the corpus, comprise long-lived progenitors that reside within the antral isthmus above Lgr5+ or CCK2R+ cells. Mist1+ antral progenitors can serve as an origin of antral tumors induced by loss of Apc or MNU treatment. Mist1+ antral progenitors, as well as other antral stem/progenitor population, express Cxcr4, and are located in close proximity to Cxcl12 (the Cxcr4 ligand)-expressing endothelium. During antral carcinogenesis, there is an expansion of Cxcr4+ epithelial cells as well as the Cxcl12+ perivascular niche. Deletion of Cxcl12 in endothelial cells or pharmacological blockade of Cxcr4 inhibits antral tumor growth. Cxcl12/Cxcr4 signaling may be a potential therapeutic target.

14.
Nat Commun ; 7: 13096, 2016 10 13.
Article in English | MEDLINE | ID: mdl-27734833

ABSTRACT

WNT/ß-catenin signalling is crucial for intestinal homoeostasis. The intestinal epithelium and stroma are the major source of WNT ligands but their origin and role in intestinal stem cell (ISC) and epithelial repair remains unknown. Macrophages are a major constituent of the intestinal stroma. Here, we analyse the role of macrophage-derived WNT in intestinal repair in mice by inhibiting their release using a macrophage-restricted ablation of Porcupine, a gene essential for WNT synthesis. Such Porcn-depleted mice have normal intestinal morphology but are hypersensitive to radiation injury in the intestine compared with wild-type (WT) littermates. Porcn-null mice are rescued from radiation lethality by treatment with WT but not Porcn-null bone marrow macrophage-conditioned medium (CM). Depletion of extracellular vesicles (EV) from the macrophage CM removes WNT function and its ability to rescue ISCs from radiation lethality. Therefore macrophage-derived EV-packaged WNTs are essential for regenerative response of intestine against radiation.


Subject(s)
Extracellular Vesicles/metabolism , Macrophages/metabolism , Radiation Injuries/metabolism , Stem Cells/metabolism , Wnt Proteins/metabolism , Wnt Signaling Pathway , Acyltransferases/genetics , Acyltransferases/metabolism , Animals , Cells, Cultured , Female , Intestinal Mucosa/cytology , Intestinal Mucosa/metabolism , Kaplan-Meier Estimate , Male , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice, Inbred ICR , Mice, Knockout , Mice, Transgenic , Radiation Injuries/genetics
15.
Cell Stem Cell ; 18(4): 441-55, 2016 Apr 07.
Article in English | MEDLINE | ID: mdl-27058937

ABSTRACT

The existence of adult pancreatic progenitor cells has been debated. While some favor the concept of facultative progenitors involved in homeostasis and repair, neither a location nor markers for such cells have been defined. Using genetic lineage tracing, we show that Doublecortin-like kinase-1 (Dclk1) labels a rare population of long-lived, quiescent pancreatic cells. In vitro, Dclk1+ cells proliferate readily and sustain pancreatic organoid growth. In vivo, Dclk1+ cells are necessary for pancreatic regeneration following injury and chronic inflammation. Accordingly, their loss has detrimental effects after cerulein-induced pancreatitis. Expression of mutant Kras in Dclk1+ cells does not affect their quiescence or longevity. However, experimental pancreatitis converts Kras mutant Dclk1+ cells into potent cancer-initiating cells. As a potential effector of Kras, Dclk1 contributes functionally to the pathogenesis of pancreatic cancer. Taken together, these observations indicate that Dclk1 marks quiescent pancreatic progenitors that are candidates for the origin of pancreatic cancer.


Subject(s)
Carcinogenesis/metabolism , Carcinoma, Pancreatic Ductal/metabolism , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Pancreatitis/metabolism , Pancreatitis/pathology , Protein Serine-Threonine Kinases/metabolism , Administration, Oral , Animals , Carcinogenesis/pathology , Carcinoma, Pancreatic Ductal/chemically induced , Carcinoma, Pancreatic Ductal/etiology , Carcinoma, Pancreatic Ductal/pathology , Cell Proliferation , Doublecortin-Like Kinases , Mice , Organoids/cytology , Organoids/growth & development , Pancreatic Neoplasms/chemically induced , Pancreatic Neoplasms/etiology , Pancreatitis/chemically induced , Pancreatitis/complications , Protein Serine-Threonine Kinases/genetics , Tamoxifen/administration & dosage
16.
Nat Commun ; 7: 10517, 2016 Feb 04.
Article in English | MEDLINE | ID: mdl-26841680

ABSTRACT

CD11b(+)Gr-1(+) myeloid-derived suppressor cells (MDSCs) expand in the spleen during cancer and promote progression through suppression of cytotoxic T cells. An anti-inflammatory reflex arc involving the vagus nerve and memory T cells is necessary for resolution of acute inflammation. Failure of this neural circuit could promote procarcinogenic inflammation and altered tumour immunity. Here we show that splenic TFF2, a secreted anti-inflammatory peptide, is released by vagally modulated memory T cells to suppress the expansion of MDSCs through CXCR4. Splenic denervation interrupts the anti-inflammatory neural arc, resulting in the expansion of MDSCs and colorectal cancer. Deletion of Tff2 recapitulates splenic denervation to promote carcinogenesis. Colorectal carcinogenesis could be suppressed through transgenic overexpression of TFF2, adenoviral transfer of TFF2 or transplantation of TFF2-expressing bone marrow. TFF2 is important to the anti-inflammatory reflex arc and plays an essential role in arresting MDSC proliferation. TFF2 offers a potential approach to prevent and to treat cancer.


Subject(s)
Cell Proliferation/genetics , Colitis/genetics , Colorectal Neoplasms/genetics , Mucins/genetics , Muscle Proteins/genetics , Myeloid Cells/immunology , Peptides/genetics , Receptors, CXCR4/immunology , Spleen/immunology , T-Lymphocytes, Cytotoxic/immunology , Vagus Nerve , Adoptive Transfer , Animals , Blotting, Western , Bone Marrow Transplantation , Colitis/immunology , Colon/metabolism , Colon/pathology , Colorectal Neoplasms/immunology , Cytokines/immunology , Denervation , Disease Progression , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Immunohistochemistry , Inflammation , Mice , Mice, Inbred C57BL , Mice, Knockout , Mucins/immunology , Mucins/metabolism , Muscle Proteins/immunology , Muscle Proteins/metabolism , Neoplasms/genetics , Neoplasms/immunology , Peptides/immunology , Peptides/metabolism , Permeability , Reverse Transcriptase Polymerase Chain Reaction , Spleen/innervation , T-Lymphocyte Subsets/immunology , Trefoil Factor-2 , Vagotomy, Truncal , Vagus Nerve Stimulation
17.
Curr Opin Pharmacol ; 25: 62-6, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26654865

ABSTRACT

The intestinal epithelium is renewed every 3-5 days from at least two principal stem cell pools. Actively cycling crypt based columnar (CBC) Lgr5(+) cells and slower cycling Bmi1-expressing or Krt19-expressing cells maintain the small intestinal and colonic epithelium in homeostasis and injury. Following acute epithelial damage, Lgr5+ stem cells are susceptible to injury and a reserve stem cell or progenitor pool is responsible for regeneration of the epithelium. Current data suggests that intestinal stem cells respond to inflammatory signals to modulate their expansion during epithelial regeneration. Here, we review how inflammation and injury affect intestinal and colonic stem cells.


Subject(s)
Inflammation/physiopathology , Intestinal Mucosa/physiology , Regeneration/physiology , Stem Cells/physiology , Humans , Inflammation/pathology , Intestinal Mucosa/cytology , Intestinal Mucosa/pathology , Stem Cells/cytology
18.
Cancer Cell ; 28(6): 800-814, 2015 Dec 14.
Article in English | MEDLINE | ID: mdl-26585400

ABSTRACT

The regulation and stem cell origin of normal and neoplastic gastric glands are uncertain. Here, we show that Mist1 expression marks quiescent stem cells in the gastric corpus isthmus. Mist1(+) stem cells serve as a cell-of-origin for intestinal-type cancer with the combination of Kras and Apc mutation and for diffuse-type cancer with the loss of E-cadherin. Diffuse-type cancer development is dependent on inflammation mediated by Cxcl12(+) endothelial cells and Cxcr4(+) gastric innate lymphoid cells (ILCs). These cells form the perivascular gastric stem cell niche, and Wnt5a produced from ILCs activates RhoA to inhibit anoikis in the E-cadherin-depleted cells. Targeting Cxcr4, ILCs, or Wnt5a inhibits diffuse-type gastric carcinogenesis, providing targets within the neoplastic gastric stem cell niche.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Epithelial Cells/metabolism , Gastric Mucosa/metabolism , Neoplastic Stem Cells/metabolism , Stem Cell Niche , Stomach Neoplasms/metabolism , Tumor Microenvironment , Animals , Anoikis , Antineoplastic Agents/pharmacology , Basic Helix-Loop-Helix Transcription Factors/genetics , Bone Marrow Transplantation , Cadherins/metabolism , Cell Communication , Cell Line, Tumor , Cell Lineage , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Cellular Senescence , Chemokine CXCL12/metabolism , Endothelial Cells/metabolism , Endothelial Cells/pathology , Epithelial Cells/drug effects , Epithelial Cells/pathology , Gastric Mucosa/drug effects , Gastric Mucosa/pathology , Humans , Lymphocytes/metabolism , Lymphocytes/pathology , Male , Mice , Mice, Transgenic , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/pathology , Receptors, CXCR4/metabolism , Signal Transduction , Stomach Neoplasms/drug therapy , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , Time Factors , Wnt Proteins/metabolism , Wnt Signaling Pathway , Wnt-5a Protein , rho GTP-Binding Proteins/metabolism , rhoA GTP-Binding Protein
19.
Am J Physiol Gastrointest Liver Physiol ; 309(12): G975-87, 2015 Dec 15.
Article in English | MEDLINE | ID: mdl-26492922

ABSTRACT

There are two major stem cell populations in the intestinal crypt region that express either Bmi1 or Lgr5; however, it has been shown that other populations in the crypt can regain stemness. In this study, we demonstrate that the transcription factor NK2 homeobox 2 (Nkx2.2) is expressed in enteroendocrine cells located in the villus and crypt of the intestinal epithelium and is coexpressed with the stem cell markers Bmi1 and Lgr5 in a subset of crypt cells. To determine whether Nkx2.2-expressing enteroendocrine cells display cellular plasticity and stem cell potential, we performed genetic lineage tracing of the Nkx2.2-expressing population using Nkx2.2(Cre/+);R26RTomato mice. These studies demonstrated that Nkx2.2+ cells are able to give rise to all intestinal epithelial cell types in basal conditions. The proliferative capacity of Nkx2.2-expressing cells was also demonstrated in vitro using crypt organoid cultures. Injuring the intestine with irradiation, systemic inflammation, and colitis did not enhance the lineage potential of Nkx2.2-expressing cells. These findings demonstrate that a rare mature enteroendocrine cell subpopulation that is demarcated by Nkx2.2 expression display stem cell properties during normal intestinal epithelial homeostasis, but is not easily activated upon injury.


Subject(s)
Cell Differentiation , Cell Lineage , Cell Proliferation , Enteroendocrine Cells/metabolism , Homeodomain Proteins/metabolism , Intestinal Mucosa/metabolism , Pluripotent Stem Cells/metabolism , Transcription Factors/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Biomarkers/metabolism , Cells, Cultured , Enteroendocrine Cells/pathology , Enteroendocrine Cells/radiation effects , Genotype , Homeobox Protein Nkx-2.2 , Homeodomain Proteins/genetics , Intestinal Mucosa/pathology , Intestinal Mucosa/radiation effects , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Mice, Inbred C57BL , Mice, Transgenic , Nerve Tissue Proteins/metabolism , Phenotype , Polycomb Repressive Complex 1/metabolism , Proto-Oncogene Proteins/metabolism , Receptors, G-Protein-Coupled/metabolism , Transcription Factors/genetics , Whole-Body Irradiation , Zebrafish Proteins , Red Fluorescent Protein
20.
Cell Stem Cell ; 16(6): 627-38, 2015 Jun 04.
Article in English | MEDLINE | ID: mdl-26046762

ABSTRACT

Epithelium of the colon and intestine are renewed every 3 days. In the intestine there are at least two principal stem cell pools. The first contains rapid cycling crypt-based columnar (CBC) Lgr5(+) cells, and the second is composed of slower cycling Bmi1-expressing cells at the +4 position above the crypt base. In the colon, however, the identification of Lgr5(-) stem cell pools has proven more challenging. Here, we demonstrate that the intermediate filament keratin-19 (Krt19) marks long-lived, radiation-resistant cells above the crypt base that generate Lgr5(+) CBCs in the colon and intestine. In colorectal cancer models, Krt19(+) cancer-initiating cells are also radioresistant, while Lgr5(+) stem cells are radiosensitive. Moreover, Lgr5(+) stem cells are dispensable in both the normal and neoplastic colonic epithelium, as ablation of Lgr5(+) stem cells results in their regeneration from Krt19-expressing cells. Thus, Krt19(+) stem cells are a discrete target relevant for cancer therapy.


Subject(s)
Colon/pathology , Intestines/pathology , Keratin-19/metabolism , Neoplastic Stem Cells/metabolism , Radiation Tolerance , Receptors, G-Protein-Coupled/metabolism , Animals , Keratin-19/genetics , Mice, Transgenic , Neoplastic Stem Cells/pathology , RNA Transport , RNA, Messenger/genetics , RNA, Messenger/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL