Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cell Stem Cell ; 30(8): 1091-1109.e7, 2023 08 03.
Artigo em Inglês | MEDLINE | ID: mdl-37541213

RESUMO

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.


Assuntos
Carcinoma Ductal Pancreático , Neoplasias Pancreáticas , Humanos , Neoplasias Pancreáticas/genética , Fator Trefoil-2/metabolismo , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/metabolismo , Pâncreas/metabolismo , Células Acinares/metabolismo , Carcinogênese/genética , Carcinogênese/metabolismo
2.
Gut ; 70(2): 330-341, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-32393543

RESUMO

OBJECTIVE: Long-standing chronic pancreatitis is an established risk factor for pancreatic ductal adenocarcinoma (PDAC). Interleukin-1ß (IL-1ß) has been associated in PDAC with shorter survival. We employed murine models to investigate the mechanisms by which IL-1ß and chronic pancreatitis might contribute to PDAC progression. DESIGN: We crossed LSL-Kras+/G12D;Pdx1-Cre (KC) mice with transgenic mice overexpressing IL-1ß to generate KC-IL1ß mice, and followed them longitudinally. We used pancreatic 3D in vitro culture to assess acinar-to-ductal metaplasia formation. Immune cells were analysed by flow cytometry and immunohistochemical staining. B lymphocytes were adoptively transferred or depleted in Kras-mutant mice. B-cell infiltration was analysed in human PDAC samples. RESULTS: KC-IL1ß mice developed PDAC with liver metastases. IL-1ß treatment increased Kras+/G12D pancreatic spheroid formation. CXCL13 expression and B lymphocyte infiltration were increased in KC-IL1ß pancreata. Adoptive transfer of B lymphocytes from KC-IL1ß mice promoted tumour formation, while depletion of B cells prevented tumour progression in KC-IL1ß mice. B cells isolated from KC-IL1ß mice had much higher expression of PD-L1, more regulatory B cells, impaired CD8+ T cell activity and promoted tumorigenesis. IL-35 was increased in the KC-IL1ß pancreata, and depletion of IL-35 decreased the number of PD-L1+ B cells. Finally, in human PDAC samples, patients with PDAC with higher B-cell infiltration within tumours showed significantly shorter survival. CONCLUSION: We show here that IL-1ß promotes tumorigenesis in part by inducing an expansion of immune-suppressive B cells. These findings point to the growing significance of B suppressor cells in pancreatic tumorigenesis.


Assuntos
Linfócitos B/imunologia , Carcinoma Ductal Pancreático/etiologia , Tolerância Imunológica/imunologia , Neoplasias Pancreáticas/etiologia , Pancreatite/complicações , Animais , Linfócitos T CD8-Positivos/imunologia , Carcinoma Ductal Pancreático/imunologia , Citometria de Fluxo , Interleucina-1beta/efeitos adversos , Camundongos , Camundongos Transgênicos , Neoplasias Pancreáticas/imunologia , Pancreatite/etiologia , Pancreatite/imunologia
3.
Nat Commun ; 11(1): 111, 2020 01 08.
Artigo em Inglês | MEDLINE | ID: mdl-31913277

RESUMO

The enteric neurotransmitter acetylcholine governs important intestinal epithelial secretory and immune functions through its actions on epithelial muscarinic Gq-coupled receptors such as M3R. Its role in the regulation of intestinal stem cell function and differentiation, however, has not been clarified. Here, we find that nonselective muscarinic receptor antagonism in mice as well as epithelial-specific ablation of M3R induces a selective expansion of DCLK1-positive tuft cells, suggesting a model of feedback inhibition. Cholinergic blockade reduces Lgr5-positive intestinal stem cell tracing and cell number. In contrast, Prox1-positive endocrine cells appear as primary sensors of cholinergic blockade inducing the expansion of tuft cells, which adopt an enteroendocrine phenotype and contribute to increased mucosal levels of acetylcholine. This compensatory mechanism is lost with acute irradiation injury, resulting in a paucity of tuft cells and acetylcholine production. Thus, enteroendocrine tuft cells appear essential to maintain epithelial homeostasis following modifications of the cholinergic intestinal niche.


Assuntos
Acetilcolina/metabolismo , Proteínas de Homeodomínio/metabolismo , Mucosa Intestinal/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Animais , Quinases Semelhantes a Duplacortina , Células Enteroendócrinas/metabolismo , Feminino , Proteínas de Homeodomínio/genética , Mucosa Intestinal/citologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neurotransmissores/metabolismo , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Proteínas Supressoras de Tumor/genética
4.
ACS Nano ; 13(2): 1354-1364, 2019 02 26.
Artigo em Inglês | MEDLINE | ID: mdl-30624916

RESUMO

Cancers of the gastrointestinal (GI) tract are among the most frequent and most lethal cancers worldwide. An important reason for this high mortality is that early disease is typically asymptomatic, and patients often present with advanced, incurable disease. Even in high-risk patients who routinely undergo endoscopic screening, lesions can be missed due to their small size or subtle appearance. Thus, current imaging approaches lack the sensitivity and specificity to accurately detect incipient GI tract cancers. Here we report our finding that a single dose of a high-sensitivity surface-enhanced resonance Raman scattering nanoparticle (SERRS-NP) enables reliable detection of precancerous GI lesions in animal models that closely mimic disease development in humans. Some of these animal models have not been used previously to evaluate imaging probes for early cancer detection. The studies were performed using a commercial Raman imaging system, a newly developed mouse Raman endoscope, and finally a clinically applicable Raman endoscope for larger animal studies. We show that this SERRS-NP-based approach enables robust detection of small, premalignant lesions in animal models that faithfully recapitulate human esophageal, gastric, and colorectal tumorigenesis. This method holds promise for much earlier detection of GI cancers than currently possible and could lead therefore to marked reduction of morbidity and mortality of these tumor types.


Assuntos
Endoscopia/métodos , Neoplasias Gastrointestinais/diagnóstico , Trato Gastrointestinal/metabolismo , Nanopartículas/química , Análise Espectral Raman/métodos , Animais , Modelos Animais de Doenças , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout
5.
Gastroenterology ; 156(4): 1066-1081.e16, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30448068

RESUMO

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.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Neoplasias do Colo/genética , Enterócitos/patologia , Mucosa Intestinal/metabolismo , Receptores Notch/metabolismo , Células-Tronco/metabolismo , Transcriptoma , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Antibióticos Antineoplásicos/farmacologia , Antineoplásicos Hormonais/farmacologia , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Antígeno CD24/metabolismo , Proteínas de Ligação ao Cálcio , Proteínas de Ciclo Celular , Plasticidade Celular , Cromogranina A/genética , Neoplasias do Colo/metabolismo , Neoplasias do Colo/patologia , Doxorrubicina/farmacologia , Enterócitos/metabolismo , Expressão Gênica , Perfilação da Expressão Gênica , Peptídeos e Proteínas de Sinalização Intercelular/genética , Mucosa Intestinal/efeitos dos fármacos , Mucosa Intestinal/patologia , Intestino Delgado/citologia , Intestino Delgado/metabolismo , Camundongos , Proteínas de Neoplasias/genética , Células-Tronco Neoplásicas/efeitos dos fármacos , Células-Tronco Neoplásicas/metabolismo , Proteínas Associadas a Pancreatite , Celulas de Paneth , Fosfoproteínas/metabolismo , Proteínas Proto-Oncogênicas c-kit/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Transdução de Sinais , Células-Tronco/efeitos dos fármacos , Células-Tronco/fisiologia , Células-Tronco/efeitos da radiação , Tamoxifeno/farmacologia , Proteínas de Sinalização YAP , Quinases da Família src/metabolismo
6.
Cancer Gene Ther ; 26(1-2): 48-57, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30042499

RESUMO

TFF2 is a small, secreted protein with anti-inflammatory properties. We previously have shown that TFF2 gene delivery via adenovirus (Ad-Tff2) suppresses colon tumor growth in colitis associated cancer. Therefore, systemic administration of TFF2 peptide could potentially provide a similar benefit. Because TFF2 shows a poor pharmacokinetic, we sought to modify the TFF2 peptide in a manner that would lower its clearance rate but retain bioactivity. Given the absence of a sequence-based prediction of TFF2 functionality, we chose to genetically fuse the C-terminus of TFF2 with the carboxyl-terminal peptide of human chorionic gonadotropin ß subunit, and inserted into adenoviral vector that expresses Flag. The resulting Ad-Tff2-CTP-Flag construct translates into a TFF2 fused with two CTP and three Flag motifs. Administered Ad-Tff2-CTP-Flag decreased tumorigenesis and suppressed the expansion of myeloid cells in vivo. The fusion peptide TFF2-CTP-Flag delivered by adenovirus Ad-Tff2-CTP-Flag as well purified recombinant fusion TFF2-CTP-Flag was retained in the blood longer compared with wild-type TFF2 delivered by Ad-Tff2 or recombinant TFF2. Consistently, purified recombinant fusion TFF2-CTP-Flag suppressed expansion of myeloid cells by down-regulating cyclin D1 mRNA in vitro. Here, we demonstrate for the very first time the retained bioactivity and possible pharmacokinetic advantages of TFF2 with a modified C-terminus.


Assuntos
Adenoviridae/genética , Neoplasias Colorretais/terapia , Terapia Genética , Vetores Genéticos , Fator Trefoil-2/genética , Animais , Gonadotropina Coriônica/genética , Colite/induzido quimicamente , Colite/complicações , Neoplasias Colorretais/etiologia , Camundongos , Oligopeptídeos/genética , Fragmentos de Peptídeos/genética , Transgenes
8.
Cancer Discov ; 8(11): 1458-1473, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30185628

RESUMO

In many solid tumors, parasympathetic input is provided by the vagus nerve, which has been shown to modulate tumor growth. However, whether cholinergic signaling directly regulates progression of pancreatic ductal adenocarcinoma (PDAC) has not been defined. Here, we found that subdiaphragmatic vagotomy in LSL-Kras +/G12D;Pdx1-Cre (KC) mice accelerated PDAC development, whereas treatment with the systemic muscarinic agonist bethanechol restored the normal KC phenotype, thereby suppressing the accelerated tumorigenesis caused by vagotomy. In LSL-Kras +/G12D;LSL-Trp53 +/R172H;Pdx1-Cre mice with established PDAC, bethanechol significantly extended survival. These effects were mediated in part through CHRM1, which inhibited downstream MAPK/EGFR and PI3K/AKT pathways in PDAC cells. Enhanced cholinergic signaling led to a suppression of the cancer stem cell (CSC) compartment, CD11b+ myeloid cells, TNFα levels, and metastatic growth in the liver. Therefore, these data suggest that cholinergic signaling directly and indirectly suppresses growth of PDAC cells, and therapies that stimulate muscarinic receptors may be useful in the treatment of PDAC.Significance: Subdiaphragmatic vagotomy or Chrm1 knockout accelerates pancreatic tumorigenesis, in part via expansion of the CSC compartment. Systemic administration of a muscarinic agonist suppresses tumorigenesis through MAPK and PI3K/AKT signaling, in early stages of tumor growth and in more advanced, metastatic disease. Therefore, CHRM1 may represent a potentially attractive therapeutic target. Cancer Discov; 8(11); 1458-73. ©2018 AACR. This article is highlighted in the In This Issue feature, p. 1333.


Assuntos
Carcinoma Ductal Pancreático/prevenção & controle , Transformação Celular Neoplásica/efeitos dos fármacos , Colinérgicos/farmacologia , Células-Tronco Neoplásicas/efeitos dos fármacos , Neoplasias Pancreáticas/prevenção & controle , Receptor Muscarínico M1/fisiologia , Animais , Carcinoma Ductal Pancreático/metabolismo , Carcinoma Ductal Pancreático/patologia , Transformação Celular Neoplásica/metabolismo , Transformação Celular Neoplásica/patologia , Genes ras , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos NOD , Camundongos Knockout , Camundongos SCID , Células-Tronco Neoplásicas/metabolismo , Células-Tronco Neoplásicas/patologia , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patologia , Transdução de Sinais , Neoplasias Pancreáticas
9.
Cancer Cell ; 33(1): 75-90.e7, 2018 01 08.
Artigo em Inglês | MEDLINE | ID: mdl-29249692

RESUMO

Catecholamines stimulate epithelial proliferation, but the role of sympathetic nerve signaling in pancreatic ductal adenocarcinoma (PDAC) is poorly understood. Catecholamines promoted ADRB2-dependent PDAC development, nerve growth factor (NGF) secretion, and pancreatic nerve density. Pancreatic Ngf overexpression accelerated tumor development in LSL-Kras+/G12D;Pdx1-Cre (KC) mice. ADRB2 blockade together with gemcitabine reduced NGF expression and nerve density, and increased survival of LSL-Kras+/G12D;LSL-Trp53+/R172H;Pdx1-Cre (KPC) mice. Therapy with a Trk inhibitor together with gemcitabine also increased survival of KPC mice. Analysis of PDAC patient cohorts revealed a correlation between brain-derived neurotrophic factor (BDNF) expression, nerve density, and increased survival of patients on nonselective ß-blockers. These findings suggest that catecholamines drive a feedforward loop, whereby upregulation of neurotrophins increases sympathetic innervation and local norepinephrine accumulation.


Assuntos
Catecolaminas/farmacologia , Fatores de Crescimento Neural/metabolismo , Neoplasias Pancreáticas/tratamento farmacológico , Adrenérgicos/farmacologia , Animais , Carcinoma in Situ/metabolismo , Linhagem Celular Tumoral , Desoxicitidina/análogos & derivados , Desoxicitidina/farmacologia , Camundongos Transgênicos , Neoplasias Pancreáticas/patologia , Gencitabina , Neoplasias Pancreáticas
10.
Cell Stem Cell ; 21(6): 747-760.e7, 2017 Dec 07.
Artigo em Inglês | MEDLINE | ID: mdl-29198940

RESUMO

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.


Assuntos
Medula Óssea/metabolismo , Células-Tronco Hematopoéticas/metabolismo , Histamina/metabolismo , Células Mieloides/metabolismo , Animais , Medula Óssea/efeitos dos fármacos , Transplante de Medula Óssea , Citometria de Fluxo , Células-Tronco Hematopoéticas/efeitos dos fármacos , Lipopolissacarídeos/farmacologia , Camundongos , Células Mieloides/efeitos dos fármacos
11.
Oncotarget ; 8(25): 40606-40619, 2017 Jun 20.
Artigo em Inglês | MEDLINE | ID: mdl-28380450

RESUMO

Overexpression of human progastrin increases colonic mucosal proliferation and colorectal cancer progression in mice. The G-protein coupled receptor 56 (GPR56) is known to regulate cell adhesion, migration, proliferation and stem cell biology, but its expression in the gut has not been studied. We hypothesized that the promotion of colorectal cancer by progastrin may be mediated in part through GPR56. Here, we found that GPR56 expresses in rare colonic crypt cells that lineage trace colonic glands consistent with GPR56 marking long-lived colonic stem-progenitor cells. GPR56 was upregulated in transgenic mice overexpressing human progastrin. While recombinant human progastrin promoted the growth and survival of wild-type colonic organoids in vitro, colonic organoids cultured from GPR56-/- mice were resistant to progastrin. We found that progastrin directly bound to, and increased the proliferation of, GPR56-expressing colon cancer cells in vitro, and proliferation was increased in cells that expressed both GPR56 and the cholecystokinin-2 receptor (CCK2R). In vivo, deletion of GPR56 in the mouse germline abrogated progastrin-dependent colonic mucosal proliferation and increased apoptosis. Loss of GPR56 also inhibited progastrin-dependent colonic crypt fission and colorectal carcinogenesis in the azoxymethane (AOM) mouse model of colorectal cancer. Overall, we found that progastrin binds to GPR56 expressing colonic stem cells, which in turn promotes their expansion, and that this GPR56-dependent pathway is an important driver and potential new target in colorectal carcinogenesis.


Assuntos
Neoplasias do Colo/metabolismo , Gastrinas/metabolismo , Precursores de Proteínas/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Células-Tronco/metabolismo , Animais , Apoptose/efeitos dos fármacos , Apoptose/genética , Carcinogênese/efeitos dos fármacos , Carcinogênese/genética , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/genética , Colo/metabolismo , Neoplasias do Colo/genética , Neoplasias do Colo/patologia , Gastrinas/genética , Gastrinas/farmacologia , Humanos , Mucosa Intestinal/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Ligação Proteica , Precursores de Proteínas/genética , Precursores de Proteínas/farmacologia , Receptor de Colecistocinina B/genética , Receptor de Colecistocinina B/metabolismo , Receptores Acoplados a Proteínas G/genética
12.
Oncoimmunology ; 6(3): e1290034, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28405523

RESUMO

The colorectal tumor microenvironment contains a diverse population of myeloid cells that are recruited and converted to immunosuppressive cells, thus facilitating tumor escape from immunoediting. We have identified a genetically and functionally distinct subset of dynamic bone marrow myeloid cells that are characterized by histidine decarboxylase (HDC) expression. Lineage tracing in Hdc-CreERT2;R26-LSL-tdTomato mice revealed that in homeostasis, there is a strong bias by HDC+ myeloid cells toward the CD11b+Ly6Ghi granulocytic lineage, which was accelerated during azoxymethane/dextran sodium sulfate (AOM/DSS)-induced colonic carcinogenesis. More importantly, HDC+ myeloid cells strongly promoted colonic tumorigenesis, and colon tumor progression was profoundly suppressed by diphtheria toxin A (DTA)-mediated depletion of HDC+ granulocytic myeloid cells. In addition, tumor infiltration by Foxp3+ regulatory T cells (Tregs) was markedly impaired following HDC+ myeloid cell depletion. We identified an HDC+ myeloid-derived Cxcl13/Cxcr5 axis that mediated Foxp3 expression and Treg proliferation. Ablation of HDC+ myeloid cells or disruption of the Cxcl13/Cxcr5 axis by gene knockdown impaired the production and recruitment of Tregs. Cxcl13 induction of Foxp3 expression in Tregs during tumorigenesis was associated with Stat3 phosphorylation. Overall, HDC+ granulocytic myeloid cells affect CD8+ T cells directly and indirectly through the modulation of Tregs and thus appear to play key roles in suppressing tumoricidal immunity.

13.
Oncotarget ; 8(67): 111012-111025, 2017 Dec 19.
Artigo em Inglês | MEDLINE | ID: mdl-29340033

RESUMO

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.
Cancer Cell ; 31(1): 21-34, 2017 01 09.
Artigo em Inglês | MEDLINE | ID: mdl-27989802

RESUMO

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.


Assuntos
Acetilcolina/fisiologia , Fator de Crescimento Neural/fisiologia , Transdução de Sinais/fisiologia , Neoplasias Gástricas/etiologia , Proteínas Adaptadoras de Transdução de Sinal/fisiologia , Animais , Proteínas de Ciclo Celular , Quinases Semelhantes a Duplacortina , Mucosa Gástrica/inervação , Camundongos , Camundongos Endogâmicos C57BL , Fosfoproteínas/fisiologia , Proteínas Serina-Treonina Quinases/análise , Receptor Muscarínico M3/fisiologia , Proteínas de Sinalização YAP
15.
Nat Commun ; 7: 13096, 2016 10 13.
Artigo em Inglês | MEDLINE | ID: mdl-27734833

RESUMO

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.


Assuntos
Vesículas Extracelulares/metabolismo , Macrófagos/metabolismo , Lesões por Radiação/metabolismo , Células-Tronco/metabolismo , Proteínas Wnt/metabolismo , Via de Sinalização Wnt , Aciltransferases/genética , Aciltransferases/metabolismo , Animais , Células Cultivadas , Feminino , Mucosa Intestinal/citologia , Mucosa Intestinal/metabolismo , Estimativa de Kaplan-Meier , Masculino , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos Endogâmicos ICR , Camundongos Knockout , Camundongos Transgênicos , Lesões por Radiação/genética
16.
Cell Stem Cell ; 18(4): 441-55, 2016 Apr 07.
Artigo em Inglês | MEDLINE | ID: mdl-27058937

RESUMO

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.


Assuntos
Carcinogênese/metabolismo , Carcinoma Ductal Pancreático/metabolismo , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patologia , Pancreatite/metabolismo , Pancreatite/patologia , Proteínas Serina-Treonina Quinases/metabolismo , Administração Oral , Animais , Carcinogênese/patologia , Carcinoma Ductal Pancreático/induzido quimicamente , Carcinoma Ductal Pancreático/etiologia , Carcinoma Ductal Pancreático/patologia , Proliferação de Células , Quinases Semelhantes a Duplacortina , Camundongos , Organoides/citologia , Organoides/crescimento & desenvolvimento , Neoplasias Pancreáticas/induzido quimicamente , Neoplasias Pancreáticas/etiologia , Pancreatite/induzido quimicamente , Pancreatite/complicações , Proteínas Serina-Treonina Quinases/genética , Tamoxifeno/administração & dosagem
17.
Nat Commun ; 7: 10517, 2016 Feb 04.
Artigo em Inglês | MEDLINE | ID: mdl-26841680

RESUMO

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.


Assuntos
Proliferação de Células/genética , Colite/genética , Neoplasias Colorretais/genética , Mucinas/genética , Proteínas Musculares/genética , Células Mieloides/imunologia , Peptídeos/genética , Receptores CXCR4/imunologia , Baço/imunologia , Linfócitos T Citotóxicos/imunologia , Nervo Vago , Transferência Adotiva , Animais , Western Blotting , Transplante de Medula Óssea , Colite/imunologia , Colo/metabolismo , Colo/patologia , Neoplasias Colorretais/imunologia , Citocinas/imunologia , Denervação , Progressão da Doença , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Imuno-Histoquímica , Inflamação , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mucinas/imunologia , Mucinas/metabolismo , Proteínas Musculares/imunologia , Proteínas Musculares/metabolismo , Neoplasias/genética , Neoplasias/imunologia , Peptídeos/imunologia , Peptídeos/metabolismo , Permeabilidade , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Baço/inervação , Subpopulações de Linfócitos T/imunologia , Fator Trefoil-2 , Vagotomia Troncular , Estimulação do Nervo Vago
18.
Cancer Cell ; 28(6): 800-814, 2015 Dec 14.
Artigo em Inglês | MEDLINE | ID: mdl-26585400

RESUMO

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.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Células Epiteliais/metabolismo , Mucosa Gástrica/metabolismo , Células-Tronco Neoplásicas/metabolismo , Nicho de Células-Tronco , Neoplasias Gástricas/metabolismo , Microambiente Tumoral , Animais , Anoikis , Antineoplásicos/farmacologia , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Transplante de Medula Óssea , Caderinas/metabolismo , Comunicação Celular , Linhagem Celular Tumoral , Linhagem da Célula , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/metabolismo , Transformação Celular Neoplásica/patologia , Senescência Celular , Quimiocina CXCL12/metabolismo , Células Endoteliais/metabolismo , Células Endoteliais/patologia , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/patologia , Mucosa Gástrica/efeitos dos fármacos , Mucosa Gástrica/patologia , Humanos , Linfócitos/metabolismo , Linfócitos/patologia , Masculino , Camundongos , Camundongos Transgênicos , Células-Tronco Neoplásicas/efeitos dos fármacos , Células-Tronco Neoplásicas/patologia , Receptores CXCR4/metabolismo , Transdução de Sinais , Neoplasias Gástricas/tratamento farmacológico , Neoplasias Gástricas/genética , Neoplasias Gástricas/patologia , Fatores de Tempo , Proteínas Wnt/metabolismo , Via de Sinalização Wnt , Proteína Wnt-5a , Proteínas rho de Ligação ao GTP/metabolismo , Proteína rhoA de Ligação ao GTP
19.
Oncotarget ; 6(32): 32966-79, 2015 Oct 20.
Artigo em Inglês | MEDLINE | ID: mdl-26429861

RESUMO

Food allergy can influence the development of colorectal cancer, although the underlying mechanisms are unclear. While mast cells (MC) store and secrete histamine, immature myeloid cells (IMC) are the major site of histidine decarboxylase (HDC) expression, the enzyme responsible for histamine production. From our earlier work, we hypothesized that histamine is central to the association between allergy and colorectal carcinogenesis through its influence on the MC-MDSC axis. Here, we show that in wild type (WT) mice, ovalbumin (OVA) immunization elicits a typical TH2 response. In contrast, in HDC-/- mice, the response to OVA allergy is skewed towards infiltration by IL-17 expressing MCs. This response is inhibited by histamine treatment. The HDC-/- allergic IL-17-expressing MCs promote MDSC proliferation and upregulation of Cox-2 and Arg-1. OVA allergy in HDC-/- mice increases the growth of colon tumor cells in both the MC38 tumor cell implantation model and the AOM/DSS carcinogenesis model. Taken together, our results show that histamine represses IL-17-expressing MCs and their subsequent activation of MDSCs, attenuating the risk of colorectal cancer in the setting of food allergy. Targeting the MC-MDSC axis may be useful for cancer prevention and treatment in patients, particularly in those with food allergy.


Assuntos
Neoplasias Colorretais/imunologia , Hipersensibilidade/imunologia , Interleucina-17/biossíntese , Mastócitos/imunologia , Células Mieloides/imunologia , Sequência de Aminoácidos , Animais , Neoplasias Colorretais/patologia , Modelos Animais de Doenças , Feminino , Histamina/farmacologia , Interleucina-17/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Dados de Sequência Molecular , Células Mieloides/patologia , Ovalbumina/imunologia , Ovalbumina/farmacologia , Fragmentos de Peptídeos/imunologia , Fragmentos de Peptídeos/farmacologia
20.
Cell ; 160(1-2): 269-84, 2015 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-25594183

RESUMO

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).


Assuntos
Osso e Ossos/citologia , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Intestino Delgado/citologia , Células-Tronco Mesenquimais/citologia , Animais , Cartilagem/metabolismo , Intestino Delgado/metabolismo , Células-Tronco Mesenquimais/metabolismo , Camundongos , Camundongos Endogâmicos C57BL
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...