Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
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.
Cells ; 12(13)2023 06 21.
Artigo em Inglês | MEDLINE | ID: mdl-37443709

RESUMO

Oral cancer patients suffer pain at the site of the cancer. Calcitonin gene related polypeptide (CGRP), a neuropeptide expressed by a subset of primary afferent neurons, promotes oral cancer growth. CGRP also mediates trigeminal pain (migraine) and neurogenic inflammation. The contribution of CGRP to oral cancer pain is investigated in the present study. The findings demonstrate that CGRP-immunoreactive (-ir) neurons and neurites innervate orthotopic oral cancer xenograft tumors in mice. Cancer increases anterograde transport of CGRP in axons innervating the tumor, supporting neurogenic secretion as the source of CGRP in the oral cancer microenvironment. CGRP antagonism reverses oral cancer nociception in preclinical oral cancer pain models. Single-cell RNA-sequencing is used to identify cell types in the cancer microenvironment expressing the CGRP receptor components, receptor activity modifying protein 1 Ramp1 and calcitonin receptor like receptor (CLR, encoded by Calcrl). Ramp1 and Calcrl transcripts are detected in cells expressing marker genes for Schwann cells, endothelial cells, fibroblasts and immune cells. Ramp1 and Calcrl transcripts are more frequently detected in cells expressing fibroblast and immune cell markers. This work identifies CGRP as mediator of oral cancer pain and suggests the antagonism of CGRP to alleviate oral cancer pain.


Assuntos
Dor do Câncer , Neoplasias Bucais , Hormônios Peptídicos , Humanos , Camundongos , Animais , Peptídeo Relacionado com Gene de Calcitonina/metabolismo , Calcitonina , Pró-Calcitonina , Receptores de Peptídeo Relacionado com o Gene de Calcitonina/genética , Receptores de Peptídeo Relacionado com o Gene de Calcitonina/metabolismo , Dor do Câncer/tratamento farmacológico , Células Endoteliais/metabolismo , Neoplasias Bucais/tratamento farmacológico , Microambiente Tumoral
3.
Adv Biol (Weinh) ; 6(9): e2200073, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-35802912

RESUMO

Oral cancer pain is attributed to the release from cancers of mediators that sensitize and activate sensory neurons. Intraplantar injection of conditioned media (CM) from human tongue cancer cell line HSC-3 or OSC-20 evokes nociceptive behavior. By contrast, CM from noncancer cell lines, DOK, and HaCaT are non-nociceptive. Pain mediators are carried by extracellular vesicles (EVs) released from cancer cells. Depletion of EVs from cancer cell line CM reverses mechanical allodynia and thermal hyperalgesia. CM from non-nociceptive cell lines become nociceptive when reconstituted with HSC-3 EVs. Two miRNAs (hsa-miR-21-5p and hsa-miR-221-3p) are identified that are present in increased abundance in EVs from HSC-3 and OSC-20 CM compared to HaCaT CM. The miRNA target genes suggest potential involvement in oral cancer pain of the toll like receptor 7 (TLR7) and 8 (TLR8) pathways, as well as signaling through interleukin 6 cytokine family signal transducer receptor (gp130, encoded by IL6ST) and colony stimulating factor receptor (G-CSFR, encoded by CSF3R), Janus kinase and signal transducer and activator of transcription 3 (JAK/STAT3). These studies confirm the recent discovery of the role of cancer EVs in pain and add to the repertoire of algesic and analgesic cancer pain mediators and pathways that contribute to oral cancer pain.


Assuntos
Dor do Câncer , Vesículas Extracelulares , MicroRNAs , Neoplasias Bucais , Dor do Câncer/metabolismo , Vesículas Extracelulares/metabolismo , Humanos , Hiperalgesia/metabolismo , MicroRNAs/genética , Neoplasias Bucais/metabolismo , Dor/metabolismo
4.
Sci Rep ; 11(1): 1840, 2021 01 19.
Artigo em Inglês | MEDLINE | ID: mdl-33469141

RESUMO

Oral cancer is very painful and impairs a patient's ability to eat, talk, and drink. Mediators secreted from oral cancer can excite and sensitize sensory neurons inducing pain. Cancer mediators can also activate Schwann cells, the peripheral glia that regulates neuronal function and repair. The contribution of Schwann cells to oral cancer pain is unclear. We hypothesize that the oral cancer mediator TNFα activates Schwann cells, which further promotes cancer progression and pain. We demonstrate that TNFα is overexpressed in human oral cancer tissues and correlates with increased self-reported pain in patients. Antagonizing TNFα reduces oral cancer proliferation, cytokine production, and nociception in mice with oral cancer. Oral cancer or TNFα alone increases Schwann cell activation (measured by Schwann cell proliferation, migration, and activation markers), which can be inhibited by neutralizing TNFα. Cancer- or TNFα-activated Schwann cells release pro-nociceptive mediators such as TNFα and nerve growth factor (NGF). Activated Schwann cells induce nociceptive behaviors in mice, which is alleviated by blocking TNFα. Our study suggests that TNFα promotes cancer proliferation, progression, and nociception at least partially by activating Schwann cells. Inhibiting TNFα or Schwann cell activation might serve as therapeutic approaches for the treatment of oral cancer and associated pain.


Assuntos
Dor do Câncer/fisiopatologia , Proliferação de Células/fisiologia , Neoplasias Bucais/patologia , Células de Schwann/patologia , Fator de Necrose Tumoral alfa/fisiologia , Animais , Progressão da Doença , Humanos , Camundongos , Neoplasias Bucais/complicações , Fator de Crescimento Neural/metabolismo , Medição da Dor , Células de Schwann/metabolismo , Fator de Necrose Tumoral alfa/antagonistas & inibidores , Fator de Necrose Tumoral alfa/metabolismo
5.
Sci Rep ; 10(1): 14724, 2020 09 07.
Artigo em Inglês | MEDLINE | ID: mdl-32895418

RESUMO

Oral cancer patients experience pain at the site of the primary cancer. Patients with metastatic oral cancers report greater pain. Lack of pain identifies patients at low risk of metastasis with sensitivity = 0.94 and negative predictive value = 0.89. In the same cohort, sensitivity and negative predictive value of depth of invasion, currently the best predictor, were 0.95 and 0.92, respectively. Cancer pain is attributed to cancer-derived mediators that sensitize neurons and is associated with increased neuronal density. We hypothesized that pain mediators would be overexpressed in metastatic cancers from patients reporting high pain. We identified 40 genes overexpressed in metastatic cancers from patients reporting high pain (n = 5) compared to N0 cancers (n = 10) and normal tissue (n = 5). The genes are enriched for functions in extracellular matrix organization and angiogenesis. They have oncogenic and neuronal functions and are reported in exosomes. Hierarchical clustering according to expression of neurotrophic and axon guidance genes also separated cancers according to pain and nodal status. Depletion of exosomes from cancer cell line supernatant reduced nociceptive behavior in a paw withdrawal assay, supporting a role for exosomes in cancer pain. The identified genes and exosomes are potential therapeutic targets for stopping cancer and attenuating pain.


Assuntos
Dor do Câncer/genética , Exossomos/genética , Neoplasias Bucais/genética , Oncogenes/genética , Idoso , Carcinogênese/genética , Linhagem Celular Tumoral , Matriz Extracelular/genética , Feminino , Humanos , Masculino , Pacientes
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
7.
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
8.
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
9.
Am J Physiol Gastrointest Liver Physiol ; 308(1): G12-24, 2015 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-25324506

RESUMO

The trefoil factor TFF2 is a member of a tripartite family of small proteins that is produced by the stomach and the colon. Recombinant TFF2, when applied intrarectally in a rodent model of hapten colitis, hastens mucosal healing and reduces inflammatory indexes. Additionally, TFF2 is expressed in immune organs, supporting a potential immunomodulatory and reparative role in the bowel. In this study we confirm that TFF2 is expressed in the colon and is specifically enriched in epithelial cells relative to colonic leukocytes. TFF2-deficient, but not TFF1-deficient, mice exhibit a more severe response to acute or chronic dextran sulfate (DSS)-induced colitis that correlates with a 50% loss of expression of TFF3, the principal colonic trefoil. In addition, the response to acute colitis is associated with altered expression of IL-6 and IL-33, but not other inflammatory cytokines. While TFF2 can reduce macrophage responsiveness and block inflammatory cell recruitment to the colon, the major role in limiting the susceptibility to acute colitis appears to be maintenance of barrier function. Bone marrow transfer experiments demonstrate that leukocyte expression of TFF2 is not sufficient for prevention of colitis induction but, rather, that the gastrointestinal epithelium is the primary source of TFF2. Together, these findings illustrate that epithelial TFF2 is an important endogenous regulator of gut mucosal homeostasis that can modulate immune and epithelial compartments. Because of its extreme stability, even in the corrosive gut lumen, TFF2 is an attractive candidate as an oral therapeutic scaffold for future drug development in the treatment of inflammatory bowel disease.


Assuntos
Transplante de Medula Óssea , Colite/metabolismo , Colo/metabolismo , Citocinas/metabolismo , Sulfato de Dextrana , Células Epiteliais/metabolismo , Mediadores da Inflamação/metabolismo , Mucinas/deficiência , Proteínas Musculares/deficiência , Peptídeos/deficiência , Redução de Peso , Animais , Células Cultivadas , Colite/induzido quimicamente , Colite/genética , Colite/imunologia , Colite/patologia , Colite/prevenção & controle , Colo/imunologia , Colo/patologia , Modelos Animais de Doenças , Células Epiteliais/imunologia , Células Epiteliais/patologia , Feminino , Interleucina-33 , Interleucina-6/metabolismo , Interleucinas/metabolismo , Leucócitos/imunologia , Leucócitos/metabolismo , Macrófagos/imunologia , Macrófagos/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mucinas/genética , Mucinas/metabolismo , Proteínas Musculares/genética , Peptídeos/genética , Peptídeos/metabolismo , Índice de Gravidade de Doença , Fatores de Tempo , Fator Trefoil-1 , Fator Trefoil-2 , Fator Trefoil-3
10.
Gut ; 64(4): 544-53, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24951258

RESUMO

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.


Assuntos
Carcinogênese , Antro Pilórico/citologia , Receptor de Colecistocinina B/fisiologia , Células-Tronco/fisiologia , Animais , Células Cultivadas , Gastrinas/fisiologia , Camundongos , Precursores de Proteínas/fisiologia
11.
Gastroenterology ; 144(1): 155-66, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23041326

RESUMO

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.


Assuntos
Movimento Celular/efeitos dos fármacos , Transformação Celular Neoplásica/metabolismo , Colite/metabolismo , Neoplasias do Colo/metabolismo , Gastrite/metabolismo , Interleucina-8/metabolismo , Células Mieloides/efeitos dos fármacos , RNA Mensageiro/metabolismo , Animais , Azoximetano , Linhagem Celular Tumoral , Colite/induzido quimicamente , Neoplasias do Colo/induzido quimicamente , Neoplasias do Colo/patologia , Células Dendríticas/metabolismo , Sulfato de Dextrana , Gastrite/microbiologia , Infecções por Helicobacter/complicações , Helicobacter felis , Humanos , Interleucina-8/genética , Interleucina-8/farmacologia , Lipopolissacarídeos/farmacologia , Macrófagos/metabolismo , Camundongos , Camundongos Transgênicos , Células Mieloides/metabolismo , Cultura Primária de Células , Carga Tumoral , Regulação para Cima/efeitos dos fármacos
12.
Gut ; 62(2): 192-200, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22362916

RESUMO

OBJECTIVE: Stromal cell-derived factor-1 (SDF-1/CXCL12), the main ligand for CXCR4, is overexpressed in human cancer. This study addressed the precise contribution of SDF-1 to gastric carcinogenesis. DESIGN: SDF-1 transgenic mice were created and a Helicobacter-induced gastric cancer model was used in combination with H/K-ATPase-IL-1ß mice. Gastric tissue was analysed by histopathology and cells isolated from the stomach were analysed by molecular biological methods. RESULTS: Analysis of the H/K-ATPase/SDF-1 transgenic (SDF-Tg) mice showed that SDF-1 overexpression results in significant gastric epithelial hyperproliferation, mucous neck cell hyperplasia and spontaneous gastric dysplasia (wild-type mice 0/15 (0%) vs SDF-Tg mice 4/14 (28.6%), p=0.042, Fisher exact test) but has minimal effects on inflammation. SDF-Tg mice also showed a dramatic expansion of α-smooth muscle actin-positive myofibroblasts and CXCR4-expressing gastric epithelial cells in the progenitor zone, both of which preceded the development of significant gastritis or dysplasia. Gremlin 1-expressing mesenchymal stem cells, the putative precursors of myofibroblasts, were also increased within the dysplastic stomachs of SDF-Tg mice and showed chemotaxis in response to SDF-1 stimulation. SDF-1 overexpression alone resulted in minimal recruitment of haematopoietic cells to the gastric mucosa, although macrophages were increased late in the disease. When SDF-Tg mice were crossed with H/K-ATPase-IL-1ß mice or infected with Helicobacter felis, however, there were dramatic synergistic effects on recruitment of bone marrow-derived cells and progression to preneoplasia. CONCLUSION: Activation of the SDF-1/CXCR4 axis can contribute to early stages of carcinogenesis primarily through recruitment of stromal cells and modulation of the progenitor niche.


Assuntos
Quimiocina CXCL12/genética , Regulação da Expressão Gênica/fisiologia , Células-Tronco Mesenquimais/patologia , Miofibroblastos/patologia , Lesões Pré-Cancerosas/genética , Neoplasias Gástricas/genética , Animais , Modelos Animais de Doenças , Ensaio de Imunoadsorção Enzimática , Mucosa Gástrica/patologia , Imuno-Histoquímica , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Células-Tronco Mesenquimais/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Miofibroblastos/metabolismo , Lesões Pré-Cancerosas/patologia , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Receptores CXCR4/metabolismo , Neoplasias Gástricas/patologia
13.
Cancer Cell ; 21(1): 36-51, 2012 Jan 17.
Artigo em Inglês | MEDLINE | ID: mdl-22264787

RESUMO

Esophageal adenocarcinoma (EAC) arises from Barrett esophagus (BE), intestinal-like columnar metaplasia linked to reflux esophagitis. In a transgenic mouse model of BE, esophageal overexpression of interleukin-1ß phenocopies human pathology with evolution of esophagitis, Barrett-like metaplasia and EAC. Histopathology and gene signatures closely resembled human BE, with upregulation of TFF2, Bmp4, Cdx2, Notch1, and IL-6. The development of BE and EAC was accelerated by exposure to bile acids and/or nitrosamines, and inhibited by IL-6 deficiency. Lgr5(+) gastric cardia stem cells present in BE were able to lineage trace the early BE lesion. Our data suggest that BE and EAC arise from gastric progenitors due to a tumor-promoting IL-1ß-IL-6 signaling cascade and Dll1-dependent Notch signaling.


Assuntos
Esôfago de Barrett/patologia , Ácidos e Sais Biliares/farmacologia , Cárdia/patologia , Esofagite/patologia , Células-Tronco/patologia , Adenocarcinoma/patologia , Animais , Esôfago de Barrett/etiologia , Proteína Morfogenética Óssea 4/genética , Proteína Morfogenética Óssea 4/metabolismo , Fator de Transcrição CDX2 , Neoplasias Esofágicas/patologia , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Interleucina-6/genética , Interleucina-6/metabolismo , Metaplasia/patologia , Camundongos , Camundongos Transgênicos , Mucinas/genética , Mucinas/metabolismo , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Peptídeos/genética , Peptídeos/metabolismo , Receptor Notch1/genética , Receptor Notch1/metabolismo , Transdução de Sinais , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Fator Trefoil-2 , Regulação para Cima
14.
Am J Physiol Gastrointest Liver Physiol ; 300(2): G334-44, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21051525

RESUMO

Gastrin is secreted from a subset of neuroendocrine cells residing in the gastric antrum known as G cells, but low levels are also expressed in fetal pancreas and intestine and in many solid malignancies. Although past studies have suggested that antral gastrin is transcriptionally regulated by inflammation, gastric pH, somatostatin, and neoplastic transformation, the transcriptional regulation of gastrin has not previously been demonstrated in vivo. Here, we describe the creation of an enhanced green fluorescent protein reporter (mGAS-EGFP) mouse using a bacterial artificial chromosome that contains the entire mouse gastrin gene. Three founder lines expressed GFP signals in the gastric antrum and the transitional zone to the corpus. In addition, GFP(+) cells could be detected in the fetal pancreatic islets and small intestinal villi, but not in these organs of the adult mice. The administration of acid-suppressive reagents such as proton pump inhibitor omeprazole and gastrin/CCK-2 receptor antagonist YF476 significantly increased GFP signal intensity and GFP(+) cell numbers in the antrum, whereas these parameters were decreased by overnight fasting, octreotide (long-lasting somatostatin ortholog) infusion, and Helicobacter felis infection. GFP(+) cells were also detected in the anterior lobe of the pituitary gland and importantly in the colonic tumor cells induced by administration with azoxymethane and dextran sulfate sodium salt. This transgenic mouse provides a useful tool to study the regulation of mouse gastrin gene in vivo, thus contributing to our understanding of the mechanisms involved in transcriptional control of the gastrin gene.


Assuntos
Cromossomos Artificiais Bacterianos/genética , Células Secretoras de Gastrina/metabolismo , Gastrinas/genética , Proteínas de Fluorescência Verde/genética , Helicobacter felis/genética , Envelhecimento/metabolismo , Animais , Azoximetano , Carcinógenos , Neoplasias do Colo/induzido quimicamente , Neoplasias do Colo/metabolismo , Neoplasias do Colo/patologia , Sulfato de Dextrana , Regulação para Baixo , Jejum , Feto/metabolismo , Ácido Gástrico/metabolismo , Gastrinas/deficiência , Regulação da Expressão Gênica no Desenvolvimento , Genes Reporter , Proteínas de Fluorescência Verde/metabolismo , Infecções por Helicobacter/genética , Infecções por Helicobacter/metabolismo , Camundongos , Camundongos Transgênicos , Antro Pilórico/metabolismo , Antro Pilórico/patologia , Somatostatina/administração & dosagem , Distribuição Tecidual , Transcrição Gênica , Transgenes , Regulação para Cima
15.
Am J Pathol ; 175(1): 365-75, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19556515

RESUMO

We have previously described a synergistic interaction between hypergastrinemia and Helicobacter felis infection on gastric corpus carcinogenesis in FVB/N mice housed under specific-pathogen-free conditions. However, gastrin-deficient (GAS-KO) mice on a mixed C57BL/6/129Sv genetic background maintained in conventional housing were reported to develop spontaneous gastric antral tumors. Therefore, we investigated the role of gastrin in Helicobacter-associated gastric carcinogenesis in H. felis-infected mice on a uniform C57BL/6 background housed in specific-pathogen-free conditions. Hypergastrinemic transgenic (INS-GAS) mice, GAS-KO mice, and C57BL/6 wild-type mice were infected with H. felis for either 12 or 18 months. At 12 months postinfection, INS-GAS mice had mild corpus dysplasia, while B6 wild-type mice had either severe gastritis or metaplasia, and GAS-KO mice had only mild to moderate gastritis. At 18 months postinfection, both INS-GAS and B6 wild-type mice had both severe atrophic gastritis and corpus dysplasia, while GAS-KO mice had severe gastritis with mild gastric atrophy, but no corpus dysplasia. In contrast, both GAS-KO and B6 wild-type mice had mild to moderate antral dysplasia, while INS-GAS mice did not. H. felis antral colonization remained stable over time among the three groups of mice. These results point to a distinct effect of gastrin on carcinogenesis of both the gastric corpus and antrum, suggesting that gastrin is an essential cofactor for gastric corpus carcinogenesis in C57BL/6 mice.


Assuntos
Gastrinas/metabolismo , Infecções por Helicobacter/metabolismo , Lesões Pré-Cancerosas/metabolismo , Neoplasias Gástricas/metabolismo , Animais , Citocinas/biossíntese , Citocinas/imunologia , Feminino , Gastrinas/genética , Gastrite/genética , Gastrite/imunologia , Gastrite/metabolismo , Infecções por Helicobacter/genética , Infecções por Helicobacter/imunologia , Helicobacter felis , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Lesões Pré-Cancerosas/genética , Lesões Pré-Cancerosas/patologia , RNA Mensageiro/análise , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Neoplasias Gástricas/genética , Neoplasias Gástricas/patologia , Linfócitos T Auxiliares-Indutores/imunologia
16.
J Biol Chem ; 284(6): 3650-62, 2009 Feb 06.
Artigo em Inglês | MEDLINE | ID: mdl-19064997

RESUMO

The secreted trefoil factor family 2 (TFF2) protein contributes to the protection of the gastrointestinal mucosa from injury by strengthening and stabilizing mucin gels, stimulating epithelial restitution, and restraining the associated inflammation. Although trefoil factors have been shown to activate signaling pathways, no cell surface receptor has been directly linked to trefoil peptide signaling. Here we demonstrate the ability of TFF2 peptide to activate signaling via the CXCR4 chemokine receptor in cancer cell lines. We found that both mouse and human TFF2 proteins (at approximately 0.5 microm) activate Ca2+ signaling in lymphoblastic Jurkat cells that could be abrogated by receptor desensitization (with SDF-1alpha) or pretreatment with the specific antagonist AMD3100 or an anti-CXCR4 antibody. TFF2 pretreatment of Jurkat cells decreased Ca2+ rise and chemotactic response to SDF-1alpha. In addition, the CXCR4-negative gastric epithelial cell line AGS became highly responsive to TFF2 treatment upon expression of the CXCR4 receptor. TFF2-induced activation of mitogen-activated protein kinases in gastric and pancreatic cancer cells, KATO III and AsPC-1, respectively, was also dependent on the presence of the CXCR4 receptor. Finally we demonstrate a distinct proliferative effect of TFF2 protein on an AGS gastric cancer cell line that expresses CXCR4. Overall these data identify CXCR4 as a bona fide signaling receptor for TFF2 and suggest a mechanism through which TFF2 may modulate immune and tumorigenic responses in vivo.


Assuntos
Mucinas/metabolismo , Proteínas Musculares/metabolismo , Proteínas de Neoplasias/metabolismo , Neoplasias Epiteliais e Glandulares/metabolismo , Peptídeos/metabolismo , Leucemia-Linfoma Linfoblástico de Células Precursoras/metabolismo , Receptores CXCR4/metabolismo , Animais , Fármacos Anti-HIV/farmacologia , Benzilaminas , Sinalização do Cálcio/efeitos dos fármacos , Quimiocina CXCL12/farmacologia , Ciclamos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Compostos Heterocíclicos/farmacologia , Humanos , Células Jurkat , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Camundongos , Proteínas Quinases Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Mucinas/farmacologia , Proteínas Musculares/farmacologia , Células NIH 3T3 , Proteínas de Neoplasias/agonistas , Proteínas de Neoplasias/farmacologia , Neoplasias Epiteliais e Glandulares/patologia , Peptídeos/farmacologia , Leucemia-Linfoma Linfoblástico de Células Precursoras/patologia , Receptores CXCR4/agonistas , Fator Trefoil-2
17.
Regul Pept ; 151(1-3): 106-14, 2008 Nov 29.
Artigo em Inglês | MEDLINE | ID: mdl-18674570

RESUMO

The unprocessed gastrin precursor, progastrin (PG), is often overexpressed in colon cancer and other malignancies where it appears to stimulate colonic growth. Overexpression of progastrin also stimulates proliferation of normal colonic mucosa, but the receptors mediating these effects have not been identified. Here we report the development of a non-radioactive assay for assessment of PG binding to normal and transformed cells. Progastrin was labeled using biotinylation, and binding of biotinylated PG to cells was assessed using flow cytometry. Using this approach, we show strong and specific binding of PG to some cell lines (IEC-6, IEC-18, HT-29, COLO320) and minimal binding to others (HeLa, DC2.4, Jurkat). We also found PG binding to several non-gut epithelial lines, such as CHO-K1, COS-6 and HEK293 cells. The specificity of binding was confirmed by competition with cold, unlabeled PG but not with glycine-extended gastrin or amidated gastrin-17. Binding was not influenced by the presence of the classical CCK-2 receptor, but was partially dependent on the charged glycosaminoglycans (GAG). The analysis of primary colonic tissues isolated from wild type C57BL/6 mouse, revealed a small epithelial subpopulation of non-hematopoietic (CD45-negative) cells that strongly interacted with PG. Surprisingly, this population was greatly expanded in gastrin knockout mice. This non-radioactive, FACS-based assay should prove useful for further characterization of cells expressing the progastrin receptor.


Assuntos
Gastrinas/metabolismo , Trato Gastrointestinal/metabolismo , Precursores de Proteínas/metabolismo , Animais , Anexina A2/metabolismo , Células CHO , Células COS , Linhagem Celular , Chlorocebus aethiops , Colo/citologia , Colo/metabolismo , Cricetinae , Cricetulus , Células Epiteliais/metabolismo , Citometria de Fluxo , Gastrinas/deficiência , Gastrinas/genética , Trato Gastrointestinal/citologia , Glicosaminoglicanos/metabolismo , Células HeLa , Humanos , Técnicas In Vitro , Células Jurkat , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptor de Colecistocinina B/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA