Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
1.
Immunity ; 49(1): 107-119.e4, 2018 07 17.
Artículo en Inglés | MEDLINE | ID: mdl-29958798

RESUMEN

Intestinal macrophages are critical for gastrointestinal (GI) homeostasis, but our understanding of their role in regulating intestinal motility is incomplete. Here, we report that CX3C chemokine receptor 1-expressing muscularis macrophages (MMs) were required to maintain normal GI motility. MMs expressed the transient receptor potential vanilloid 4 (TRPV4) channel, which senses thermal, mechanical, and chemical cues. Selective pharmacologic inhibition of TRPV4 or conditional deletion of TRPV4 from macrophages decreased intestinal motility and was sufficient to reverse the GI hypermotility that is associated with chemotherapy treatment. Mechanistically, stimulation of MMs via TRPV4 promoted the release of prostaglandin E2 and elicited colon contraction in a paracrine manner via prostaglandin E receptor signaling in intestinal smooth muscle cells without input from the enteric nervous system. Collectively, our data identify TRPV4-expressing MMs as an essential component required for maintaining normal GI motility and provide potential drug targets for GI motility disorders.


Asunto(s)
Colon/fisiología , Motilidad Gastrointestinal , Macrófagos/metabolismo , Miocitos del Músculo Liso/metabolismo , Transducción de Señal , Canales Catiónicos TRPV/metabolismo , Animales , Receptor 1 de Quimiocinas CX3C/metabolismo , Colon/fisiopatología , Ciclooxigenasa 1/deficiencia , Ciclooxigenasa 1/metabolismo , Dinoprostona/análisis , Dinoprostona/metabolismo , Femenino , Mucosa Gástrica/citología , Expresión Génica , Masculino , Proteínas de la Membrana/antagonistas & inhibidores , Proteínas de la Membrana/deficiencia , Proteínas de la Membrana/metabolismo , Ratones , Ratones Noqueados , Contracción Muscular , Receptores de Prostaglandina E/antagonistas & inhibidores , Receptores de Prostaglandina E/metabolismo , Canales Catiónicos TRPV/antagonistas & inhibidores , Canales Catiónicos TRPV/deficiencia , Canales Catiónicos TRPV/genética
2.
Am J Physiol Gastrointest Liver Physiol ; 319(1): G63-G73, 2020 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-32538139

RESUMEN

Hyaluronic acid (HA), a glycosaminoglycan in the extracellular matrix, binds to CD44 and Toll-like receptor 4 (TLR4). We previously demonstrated that both CD44 and TLR4, but predominately TLR4, mediated HA stimulation of Lgr5+ stem cell proliferation, crypt fission, and intestinal growth in postnatal mice. Here we address the questions of which cell type expresses the relevant TLR4 in driving intestinal growth and what are the downstream events from TLR4 activation. Studies were done in 14-day-old mice: wild type (WT), mice deficient in cyclooxygenase 2 (COX2), mice deficient in myeloid cell TLR4, and mice deficient in epithelial cell epidermal growth factor receptor (EGFR). Biological end points included crypt fission and Lgr5 cell proliferation. In WT mice, treatment with NS-398 (a COX2 inhibitor), clodronate (a macrophage-depleting agent), or tyrphostin (an EGFR inhibitor) resulted in 30% reductions in crypt fission and Lgr5+ stem cell proliferation compared with control mice. Mice deficient in COX2 or myeloid TLR4 or epithelial cell EGFR all had 30% reductions in crypt fission and Lgr5+ stem cell proliferation compared with WT mice. Administration of dimethyl PGE2, a stable PGE2 analog, increased crypt fission and Lgr5+ stem cell proliferation. Administration of dimethyl PGE2 reversed the effects of NS-398, clodronate, COX2 deficiency, and myeloid TLR4 deficiency but had no effect on mice treated with tyrphostin or mice deficient in epithelial cell EGFR. We conclude that, in postnatal mice, ~30% of intestinal growth as manifested by crypt fission and Lgr5+ stem cell proliferation is driven by a novel pathway: Extracellular HA binds TLR4 on pericryptal macrophages, inducing the production of PGE2 through COX2. PGE2 transactivates EGFR in Lgr5+ epithelial stem cells, resulting in Lgr5+ stem cell proliferation and crypt fission.NEW & NOTEWORTHY This study, in newborn mice, describes a novel molecular pathway regulating Lgr5+ epithelial stem cell proliferation and normal intestinal elongation, as assessed by crypt fission. In this pathway, endogenous extracellular hyaluronic acid binds to Toll-like receptor 4 on pericryptal macrophages releasing PGE2 which binds to epidermal growth factor receptor on Lgr5+ stem cells resulting in proliferation. Lgr5+ stem cell proliferation leads to crypt fission and intestinal elongation. The demonstration that normal growth requires microbial-independent Toll-like receptor activation is novel.


Asunto(s)
Dinoprostona/metabolismo , Receptores ErbB/efectos de los fármacos , Ácido Hialurónico/farmacología , Receptor Toll-Like 4/efectos de los fármacos , Animales , Proliferación Celular/efectos de los fármacos , Ciclooxigenasa 2/metabolismo , Receptores ErbB/metabolismo , Ácido Hialurónico/antagonistas & inhibidores , Intestinos/efectos de los fármacos , Ratones Noqueados , Receptor Toll-Like 4/metabolismo , Activación Transcripcional/efectos de los fármacos
3.
Gut ; 68(6): 1003-1013, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-29934438

RESUMEN

OBJECTIVE: Lactobacillus rhamnosus GG (LGG), a probiotic, given by gavage is radioprotective of the mouse intestine. LGG-induced radioprotection is toll-like receptor 2 (TLR2) and cyclooxygenase-2 (COX-2)-dependent and is associated with the migration of COX-2+mesenchymal stem cells (MSCs) from the lamina propria of the villus to the lamina propria near the crypt epithelial stem cells. Our goals were to define the mechanism of LGG radioprotection including identification of the TLR2 agonist, and the mechanism of the MSC migration and to determine the safety and efficacy of this approach in models relevant to clinical radiation therapy. DESIGN: Intestinal radioprotection was modelled in vitro with cell lines and enteroids as well as in vivo by assaying clinical outcomes and crypt survival. Fractionated abdominal and single dose radiation were used along with syngeneic CT26 colon tumour grafts to assess tumour radioprotection. RESULTS: LGG with a mutation in the processing of lipoteichoic acid (LTA), a TLR2 agonist, was not radioprotective, while LTA agonist and native LGG were. An agonist of CXCR4 blocked LGG-induced MSC migration and LGG-induced radioprotection. LGG given by gavage induced expression of CXCL12, a CXCR4 agonist, in pericryptal macrophages and depletion of macrophages by clodronate liposomes blocked LGG-induced MSC migration and radioprotection. LTA effectively protected the normal intestinal crypt, but not tumours in fractionated radiation regimens. CONCLUSIONS: LGG acts as a 'time-release capsule' releasing radioprotective LTA. LTA then primes the epithelial stem cell niche to protect epithelial stem cells by triggering a multicellular, adaptive immune signalling cascade involving macrophages and PGE2 secreting MSCs. TRIAL REGISTRATION NUMBER: NCT01790035; Pre-results.


Asunto(s)
Mucosa Intestinal/metabolismo , Lacticaseibacillus rhamnosus , Lipopolisacáridos/metabolismo , Probióticos/farmacología , Traumatismos por Radiación/prevención & control , Ácidos Teicoicos/metabolismo , Animales , Movimiento Celular/efectos de la radiación , Células Cultivadas , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática , Inmunohistoquímica , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/efectos de la radiación , Activación de Macrófagos/efectos de la radiación , Células Madre Mesenquimatosas/citología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Protectores contra Radiación , Valores de Referencia , Sensibilidad y Especificidad
4.
Am J Physiol Gastrointest Liver Physiol ; 309(11): G874-87, 2015 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-26505972

RESUMEN

Hyaluronic acid, a glycosaminoglycan in the extracellular matrix, binds to CD44 and Toll-like receptor 4 (TLR4). We previously addressed the role of hyaluronic acid in small intestinal and colonic growth in mice. We addressed the role of exogenous hyaluronic acid by giving hyaluronic acid intraperitoneally and the role of endogenous hyaluronic acid by giving PEP-1, a peptide that blocks hyaluronic acid binding to its receptors. Exogenous hyaluronic acid increased epithelial proliferation but had no effect on intestinal length. PEP-1 resulted in a shortened small intestine and colon and diminished epithelial proliferation. In the current study, we sought to determine whether the effects of hyaluronic acid on growth were mediated by signaling through CD44 or TLR4 by giving exogenous hyaluronic acid or PEP-1 twice a week from 3-8 wk of age to wild-type, CD44(-/-), and TLR4(-/-) mice. These studies demonstrated that signaling through both CD44 and TLR4 were important in mediating the effects of hyaluronic acid on growth in the small intestine and colon. Extending our studies to early postnatal life, we assessed the effects of exogenous hyaluronic acid and PEP-1 on Lgr5(+) stem cell proliferation and crypt fission. Administration of PEP-1 to Lgr5(+) reporter mice from postnatal day 7 to day 14 decreased Lgr5(+) cell proliferation and decreased crypt fission. These studies indicate that endogenous hyaluronic acid increases Lgr5(+) stem cell proliferation, crypt fission, and intestinal lengthening and that these effects are dependent on signaling through CD44 and TLR4.


Asunto(s)
Proliferación Celular , Colon/metabolismo , Receptores de Hialuranos/metabolismo , Ácido Hialurónico/metabolismo , Mucosa Intestinal/metabolismo , Intestino Delgado/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Células Madre/efectos de los fármacos , Receptor Toll-Like 4/metabolismo , Animales , Biomarcadores/metabolismo , Proliferación Celular/efectos de los fármacos , Colon/efectos de los fármacos , Colon/crecimiento & desarrollo , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Genotipo , Receptores de Hialuranos/genética , Ácido Hialurónico/administración & dosificación , Ácido Hialurónico/antagonistas & inhibidores , Inyecciones Intraperitoneales , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/crecimiento & desarrollo , Intestino Delgado/efectos de los fármacos , Intestino Delgado/crecimiento & desarrollo , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Péptidos/farmacología , Fenotipo , Transducción de Señal/efectos de los fármacos , Células Madre/metabolismo , Receptor Toll-Like 4/deficiencia , Receptor Toll-Like 4/genética
5.
J Biol Chem ; 288(22): 16085-97, 2013 May 31.
Artículo en Inglés | MEDLINE | ID: mdl-23589310

RESUMEN

The stem cell in the isthmus of gastric units continually replenishes the epithelium. Atrophy of acid-secreting parietal cells (PCs) frequently occurs during infection with Helicobacter pylori, predisposing patients to cancer. Atrophy causes increased proliferation of stem cells, yet little is known about how this process is regulated. Here we show that CD44 labels a population of small, undifferentiated cells in the gastric unit isthmus where stem cells are known to reside. Loss of CD44 in vivo results in decreased proliferation of the gastric epithelium. When we induce PC atrophy by Helicobacter infection or tamoxifen treatment, this CD44(+) population expands from the isthmus toward the base of the unit. CD44 blockade during PC atrophy abrogates the expansion. We find that CD44 binds STAT3, and inhibition of either CD44 or STAT3 signaling causes decreased proliferation. Atrophy-induced CD44 expansion depends on pERK, which labels isthmal cells in mice and humans. Our studies delineate an in vivo signaling pathway, ERK → CD44 → STAT3, that regulates normal and atrophy-induced gastric stem/progenitor-cell proliferation. We further show that we can intervene pharmacologically at each signaling step in vivo to modulate proliferation.


Asunto(s)
Proliferación Celular , Infecciones por Helicobacter/metabolismo , Helicobacter pylori/metabolismo , Receptores de Hialuranos/metabolismo , Células Parietales Gástricas/metabolismo , Células Madre/metabolismo , Animales , Femenino , Infecciones por Helicobacter/genética , Infecciones por Helicobacter/patología , Humanos , Receptores de Hialuranos/genética , Sistema de Señalización de MAP Quinasas/genética , Masculino , Metaplasia/genética , Metaplasia/metabolismo , Metaplasia/patología , Ratones , Células Parietales Gástricas/patología , Factor de Transcripción STAT3/genética , Factor de Transcripción STAT3/metabolismo , Células Madre/patología
6.
FASEB J ; 27(3): 1191-202, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23233532

RESUMEN

Genetic variants in the fatty acid (FA) translocase FAT/CD36 associate with abnormal postprandial lipids and influence risk for the metabolic syndrome. CD36 is abundant on apical enterocyte membranes in the proximal small intestine, where it facilitates FA uptake and FA-initiated signaling. We explored whether CD36 signaling influences FA-mediated secretion of cholecystokinin (CCK) and secretin, peptides released by enteroendocrine cells (EECs) in the duodenum/jejunum, which regulate events important for fat digestion and homeostasis. CD36 was immunodetected on apical membranes of secretin- and CCK-positive EECs and colocalized with cytosolic granules. Intragastric lipid administration to CD36 mice released less secretin (-60%) and CCK (-50%) compared with wild-type mice. Likewise, diminished secretin and CCK responses to FA were observed with CD36 intestinal segments in vitro, arguing against influence of alterations in fat absorption. Signaling mechanisms underlying peptide release were examined in STC-1 cells stably expressing human CD36 or a signaling-impaired mutant (CD36K/A). FA stimulation of cells expressing CD36 (vs. vector or CD36K/A) released more secretin (3.5- to 4-fold) and CCK (2- to 3-fold), generated more cAMP (2- to 2.5-fold), and enhanced protein kinase A activation. Protein kinase A inhibition (H-89) blunted secretin (80%) but not CCK release, which was reduced (50%) by blocking of calmodulin kinase II (KN-62). Coculture of STC-1 cells with Caco-2 cells stably expressing CD36 did not alter secretin or CCK release, consistent with a minimal effect of adjacent enterocytes. In summary, CD36 is a major mediator of FA-induced release of CCK and secretin. These peptides contribute to the role of CD36 in fat absorption and to its pleiotropic metabolic effects.


Asunto(s)
Antígenos CD36/metabolismo , Colecistoquinina/metabolismo , Duodeno/metabolismo , Células Enteroendocrinas/metabolismo , Ácidos Grasos/metabolismo , Yeyuno/metabolismo , Secretina/metabolismo , Animales , Antígenos CD36/genética , Células CACO-2 , Colecistoquinina/genética , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Duodeno/citología , Células Enteroendocrinas/citología , Activación Enzimática , Ácidos Grasos/genética , Humanos , Yeyuno/citología , Ratones , Ratones Noqueados , Secretina/genética , Transducción de Señal/fisiología
7.
Gut ; 61(6): 829-38, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22027478

RESUMEN

BACKGROUND: The small intestinal epithelium is highly sensitive to radiation and is a major site of injury during radiation therapy and environmental overexposure. OBJECTIVE: To examine probiotic bacteria as potential radioprotective agents in the intestine. METHODS: 8-week-old C57BL/6 wild-type or knockout mice were administered probiotic by gavage for 3 days before 12 Gy whole body radiation. The intestine was evaluated for cell-positional apoptosis (6 h) and crypt survival (84 h). RESULTS: Gavage of 5×107 Lactobacillus rhamnosus GG (LGG) improved crypt survival about twofold (p<0.01); the effect was observed when administered before, but not after, radiation. Conditioned medium (CM) from LGG improved crypt survival (1.95-fold, p<0.01), and both LGG and LGG-CM reduced epithelial apoptosis particularly at the crypt base (33% to 18%, p<0.01). LGG was detected in the distal ileal contents after the gavage cycle, but did not lead to a detectable shift in bacterial family composition. The reduction in epithelial apoptosis and improved crypt survival offered by LGG was lost in MyD88⁻/⁻, TLR-2⁻/⁻ and cyclo-oxygenase-2⁻/⁻ (COX-2) mice but not TLR-4⁻/⁻ mice. LGG administration did not lead to increased jejunal COX-2 mRNA or prostaglandin E2 levels or a change in number of COX-2-expressing cells. However, a location shift was observed in constitutively COX-2-expressing cells of the lamina propria from the villi to a position near the crypt base (villi to crypt ratio 80:20 for control and 62:38 for LGG; p<0.001). Co-staining revealed these COX-2-expressing small intestinal lamina propria cells to be mesenchymal stem cells. CONCLUSIONS: LGG or its CM reduce radiation-induced epithelial injury and improve crypt survival. A TLR-2/MyD88 signalling mechanism leading to repositioning of constitutive COX-2-expressing mesenchymal stem cells to the crypt base is invoked.


Asunto(s)
Ciclooxigenasa 2/fisiología , Mucosa Intestinal/efectos de la radiación , Lacticaseibacillus rhamnosus/metabolismo , Probióticos/uso terapéutico , Traumatismos Experimentales por Radiación/prevención & control , Receptor Toll-Like 2/fisiología , Irradiación Corporal Total/efectos adversos , Animales , Apoptosis/efectos de la radiación , Femenino , Mucosa Intestinal/microbiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Factor 88 de Diferenciación Mieloide/fisiología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
8.
Am J Physiol Gastrointest Liver Physiol ; 303(3): G377-88, 2012 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-22556141

RESUMEN

Hyaluronic acid (HA), a component of the extracellular matrix, affects gastrointestinal epithelial proliferation in injury models, but its role in normal growth is unknown. We sought to determine the effects of exogenous HA on intestinal and colonic growth by intraperitoneal injection of HA twice a week into C57BL/6 mice from 3 to 8 wk of age. Similarly, to determine the effects of endogenous HA on intestinal and colonic growth, we administered PEP-1, a peptide that blocks the binding of HA to its receptors, on the same schedule. In mice treated with exogenous HA, villus height and crypt depth in the intestine, crypt depth in the colon, and epithelial proliferation in the intestine and colon were increased. In mice treated with PEP-1, intestinal and colonic length were markedly decreased and crypt depth and villus height in the intestine, crypt depth in the colon, and epithelial proliferation in the intestine and colon were decreased. Administration of HA was associated with increased levels of EGF (intestine) and IGF-I (colon), whereas administration of PEP-1 was associated with decreased levels of IGF-I (intestine) and epiregulin (colon). Exogenous HA increases intestinal and colonic epithelial proliferation, resulting in hyperplasia. Blocking the binding of endogenous HA to its receptors results in decreased intestinal and colonic length and a mucosal picture of hypoplasia, suggesting that endogenous HA contributes to the regulation of normal intestinal and colonic growth.


Asunto(s)
Colon/crecimiento & desarrollo , Ácido Hialurónico/farmacología , Intestinos/crecimiento & desarrollo , Animales , Proliferación Celular/efectos de los fármacos , Colon/efectos de los fármacos , Factor de Crecimiento Epidérmico/metabolismo , Epirregulina , Células Epiteliales/citología , Glucuronosiltransferasa/biosíntesis , Receptores de Hialuranos/efectos de los fármacos , Hialuronano Sintasas , Hialuronoglucosaminidasa/biosíntesis , Factor I del Crecimiento Similar a la Insulina/metabolismo , Intestinos/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Péptidos/farmacología , ARN Mensajero/metabolismo
9.
Am J Physiol Gastrointest Liver Physiol ; 302(3): G309-16, 2012 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-22038822

RESUMEN

The intestinal epithelium is sensitive to radiation injury. Damage to the intestinal epithelium is dose limiting in radiation therapy of abdominal cancers. There is a need for agents that can be given before radiation therapy to protect the intestinal epithelium. C57BL6 mice were subjected to 12 Gy of total body radiation. Some mice received intraperitoneal hyaluronic acid (HA) before radiation. Mice were killed 6 h after radiation to assess radiation-induced apoptosis in the intestine; other mice were killed at 84 h to assess crypt survival. Total body radiation (12 Gy) resulted in increased expression of HA synthases and HA in the intestine and increased plasma HA (5-fold). Intraperitoneal injection of HA (30 mg/kg) before radiation resulted in a 1.8-fold increase in intestinal crypt survival and a decrease in radiation-induced apoptosis. The radioprotective effects of HA were not seen in Toll-like receptor 4 (TLR4)- or cyclooxygenase-2 (COX-2)-deficient mice. Intraperitoneal injection of HA induced a 1.5-fold increase in intestinal COX-2 expression, a 1.5-fold increase in intestinal PGE2, and the migration of COX-2-expressing mesenchymal stem cells from the lamina propria in the villi to the lamina propria near the crypt. We conclude that 1) radiation induces increased HA expression through inducing HA synthases, 2) intraperitoneal HA given before radiation reduces radiation-induced apoptosis and increases crypt survival, and 3) these radioprotective effects are mediated through TLR4, COX-2, and the migration of COX-2-expressing mesenchymal stem cells.


Asunto(s)
Ciclooxigenasa 2/metabolismo , Ácido Hialurónico/farmacología , Intestinos/efectos de los fármacos , Intestinos/efectos de la radiación , Protectores contra Radiación/farmacología , Receptor Toll-Like 4/metabolismo , Animales , Antígenos CD/metabolismo , Apoptosis/efectos de los fármacos , Apoptosis/efectos de la radiación , Movimiento Celular/efectos de los fármacos , Ciclooxigenasa 2/genética , Dinoprostona/metabolismo , Expresión Génica/genética , Expresión Génica/efectos de la radiación , Glucuronosiltransferasa/genética , Receptores de Hialuranos/genética , Hialuronano Sintasas , Ácido Hialurónico/antagonistas & inhibidores , Ácido Hialurónico/sangre , Ácido Hialurónico/metabolismo , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patología , Mucosa Intestinal/efectos de la radiación , Intestinos/patología , Yeyuno/efectos de los fármacos , Yeyuno/metabolismo , Yeyuno/patología , Yeyuno/efectos de la radiación , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Protectores contra Radiación/metabolismo , Receptor Toll-Like 4/agonistas , Receptor Toll-Like 4/genética
10.
J Biol Chem ; 285(7): 5026-39, 2010 Feb 12.
Artículo en Inglés | MEDLINE | ID: mdl-20018844

RESUMEN

We previously found that a population of colonic stromal cells that constitutively express high levels of prostaglandin-endoperoxide synthase 2 (Ptgs2, also known as Cox-2) altered their location in the lamina propria in response to injury in a Myd88-dependent manner (Brown, S. L., Riehl, T. E., Walker, M. R., Geske, M. J., Doherty, J. M., Stenson, W. F., and Stappenbeck, T. S. (2007) J. Clin. Invest. 117, 258-269). At the time of this study, the identity of these cells and the mechanism by which they expressed high levels of Ptgs2 were unknown. Here we found that these colonic stromal cells were mesenchymal stem cells (MSCs). These colonic MSCs expressed high Ptgs2 levels not through interaction with bacterial products but instead as a consequence of mRNA stabilization downstream of Fgf9 (fibroblast growth factor 9), a growth factor that is constitutively expressed by the intestinal epithelium. This stabilization was mediated partially through a mechanism involving endogenous CUG-binding protein 2 (CUGbp2). These studies suggest that Fgf9 is an important factor in the regulation of Ptgs2 in colonic MSCs and may be a factor involved in its constitutive expression in vivo.


Asunto(s)
Colon/citología , Ciclooxigenasa 2/metabolismo , Células Madre Mesenquimatosas/metabolismo , Animales , Proteínas CELF , Línea Celular , Células Cultivadas , Ciclooxigenasa 2/genética , Factor 9 de Crecimiento de Fibroblastos/farmacología , Flavonoides/farmacología , Citometría de Flujo , Humanos , Immunoblotting , Inmunohistoquímica , Lipopolisacáridos/farmacología , Células Madre Mesenquimatosas/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Modelos Biológicos , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Células del Estroma/metabolismo , Receptor Toll-Like 4/genética , Receptor Toll-Like 4/metabolismo
11.
J Clin Invest ; 117(1): 258-69, 2007 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17200722

RESUMEN

We identified cellular and molecular mechanisms within the stem cell niche that control the activity of colonic epithelial progenitors (ColEPs) during injury. Here, we show that while WT mice maintained ColEP proliferation in the rectum following injury with dextran sodium sulfate, similarly treated Myd88(-/-) (TLR signaling-deficient) and prostaglandin-endoperoxide synthase 2(-/-) (Ptgs2(-/-)) mice exhibited a profound inhibition of epithelial proliferation and cellular organization within rectal crypts. Exogenous addition of 16,16-dimethyl PGE(2) (dmPGE(2)) rescued the effects of this injury in both knockout mouse strains, indicating that Myd88 signaling is upstream of Ptgs2 and PGE(2). In WT and Myd88(-/-) mice, Ptgs2 was expressed in scattered mesenchymal cells. Surprisingly, Ptgs2 expression was not regulated by injury. Rather, in WT mice, the combination of injury and Myd88 signaling led to the repositioning of a subset of the Ptgs2-expressing stromal cells from the mesenchyme surrounding the middle and upper crypts to an area surrounding the crypt base adjacent to ColEPs. These findings demonstrate that Myd88 and prostaglandin signaling pathways interact to preserve epithelial proliferation during injury using what we believe to be a previously undescribed mechanism requiring proper cellular mobilization within the crypt niche.


Asunto(s)
Ciclooxigenasa 2/genética , Mucosa Intestinal/patología , Factor 88 de Diferenciación Mieloide/fisiología , Células del Estroma/fisiología , Animales , División Celular , Ciclooxigenasa 2/deficiencia , Mucosa Intestinal/fisiopatología , Ratones , Ratones Noqueados , Modelos Genéticos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células del Estroma/enzimología , Transcripción Genética , Heridas y Lesiones/fisiopatología
12.
Gastroenterology ; 137(6): 2041-51, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19732774

RESUMEN

BACKGROUND & AIMS: The protective component of the host response to dextran sodium sulfate (DSS)-induced colitis in the mouse is mediated through the activation of Toll-like receptor (TLR) 4, the induction of cyclooxygenase (COX)-2, and prostaglandin E(2) production. TLR4 ligands include bacterial lipopolysaccharide and hyaluronic acid, a component of the extracellular matrix. Our hypothesis is that hyaluronic acid, through TLRs, plays a protective role in the host response to DSS-induced colitis. METHODS: DSS (2.5%) was administered for 7 days in wild-type and MyD88(-/-) mice. The mice also received intraperitoneal hyaluronic acid. The expression of hyaluronic acid, COX-2, and macrophage inflammatory protein (MIP)-2 was evaluated by immunohistochemistry. RESULTS: DSS induced a marked increase in hyaluronic acid in the lamina propria of wild-type but not MyD88(-/-) mice. Treatment with DSS also induced the MyD88-dependent expression of hyaluronic acid synthases 2 and 3, enzymes involved in hyaluronic acid synthesis, in lamina propria macrophages. Exogenous hyaluronic acid induced the expression of tumor necrosis factor alpha, MIP-2, and COX-2 in the colon in a MyD88-dependent manner. In wild-type but not MyD88(-/-), TLR4(-/-), COX-2(-/-) mice, hyaluronic acid was protective against DSS-induced colitis. In wild-type mice, hyaluronic acid was therapeutic in established DSS-induced colitis. CONCLUSIONS: Endogenous hyaluronic acid expression is markedly increased in DSS-induced colitis and preserves the epithelium through TLR activation and COX-2 expression. Furthermore, exogenous hyaluronic acid, through the activation of TLRs and the production of prostaglandin E(2) through COX-2, has protective effects in DSS-induced colitis.


Asunto(s)
Antiinflamatorios/metabolismo , Proliferación Celular , Colitis/patología , Colon/patología , Ácido Hialurónico/metabolismo , Mucosa Intestinal/patología , Receptor Toll-Like 4/metabolismo , Animales , Antiinflamatorios/administración & dosificación , Quimiocina CXCL2/metabolismo , Colitis/inducido químicamente , Colitis/inmunología , Colitis/metabolismo , Colitis/prevención & control , Colon/inmunología , Colon/metabolismo , Ciclooxigenasa 2/genética , Ciclooxigenasa 2/metabolismo , Sulfato de Dextran , Modelos Animales de Enfermedad , Femenino , Glucuronosiltransferasa/metabolismo , Receptores de Hialuranos/genética , Receptores de Hialuranos/metabolismo , Hialuronano Sintasas , Ácido Hialurónico/administración & dosificación , Inyecciones Intraperitoneales , Mucosa Intestinal/inmunología , Mucosa Intestinal/metabolismo , Ligandos , Macrófagos Peritoneales/inmunología , Macrófagos Peritoneales/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Factor 88 de Diferenciación Mieloide/genética , Factor 88 de Diferenciación Mieloide/metabolismo , Regeneración , Factores de Tiempo , Receptor Toll-Like 4/deficiencia , Receptor Toll-Like 4/genética , Factor de Necrosis Tumoral alfa/metabolismo , Regulación hacia Arriba
13.
Stem Cells ; 27(10): 2571-9, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19676123

RESUMEN

It is thought that small intestinal epithelia (IE) undergo continuous self-renewal primarily due to their population of undifferentiated stem cells. These stem cells give rise to transit amplifying (daughter/progenitor) cells, which can differentiate into all mature cell types required for normal gut function. Identification of stem cells in IE is paramount to fully understanding this renewal process. One major obstacle in gastrointestinal stem cell biology has been the lack of definitive markers that identify small intestinal stem cells (ISCs). Here we demonstrate that the novel putative ISC marker doublecortin and CaM kinase-like-1 (DCAMKL-1) is predominantly expressed in quiescent cells in the lower two-thirds of intestinal crypt epithelium and in occasional crypt-based columnar cells (CBCs). In contrast, the novel putative stem cell marker leucine-rich-repeat-containing G-protein-coupled receptor (LGR5) is observed in rapidly cycling CBCs and in occasional crypt epithelial cells. Furthermore, functionally quiescent DCAMKL-1+ crypt epithelial cells retain bromo-deoxyuridine in a modified label retention assay. Moreover, we demonstrate that DCAMKL-1 is a cell surface expressing protein; DCAMKL-1+ cells, isolated from the adult mouse small intestine by fluorescence activated cell sorting, self-renew and ultimately form spheroids in suspension culture. These spheroids formed glandular epithelial structures in the flanks of athymic nude mice, which expressed multiple markers of gut epithelial lineage. Thus, DCAMKL-1 is a marker of quiescent ISCs and can be distinguished from the cycling stem/progenitors (LGR5+). Moreover, DCAMKL-1 can be used to isolate normal small intestinal stem cells and represents a novel research tool for regenerative medicine and cancer therapy.


Asunto(s)
Mucosa Intestinal/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Células Madre/metabolismo , Animales , Antígenos de Superficie/análisis , Antígenos de Superficie/metabolismo , Biomarcadores/análisis , Biomarcadores/metabolismo , Bromodesoxiuridina , Ciclo Celular/fisiología , División Celular/fisiología , Línea Celular Tumoral , Linaje de la Célula/fisiología , Proliferación Celular , Quinasas Similares a Doblecortina , Citometría de Flujo , Humanos , Mucosa Intestinal/citología , Ratones , Ratones Endogámicos C57BL , Ratones Desnudos , Proteínas Serina-Treonina Quinasas/análisis , Receptores Acoplados a Proteínas G/análisis , Esferoides Celulares , Células Madre/citología
14.
Stem Cells ; 26(3): 630-7, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18055444

RESUMEN

In the gut, tumorigenesis arises from intestinal or colonic crypt stem cells. Currently, no definitive markers exist that reliably identify gut stem cells. Here, we used the putative stem cell marker doublecortin and CaM kinase-like-1 (DCAMKL-1) to examine radiation-induced stem cell apoptosis and adenomatous polyposis coli (APC)/multiple intestinal neoplasia (min) mice to determine the effects of APC mutation on DCAMKL-1 expression. Immunoreactive DCAMKL-1 staining was demonstrated in the intestinal stem cell zone. Furthermore, we observed apoptosis of the cells negative for DCAMKL-1 at 6 hours. We found DNA damage in all the cells in the crypt region, including the DCAMKL-1-positive cells. We also observed stem cell apoptosis and mitotic DCAMKL-1-expressing cells 24 hours after irradiation. Moreover, in APC/min mice, DCAMKL-1-expressing cells were negative for proliferating cell nuclear antigen and nuclear beta-catenin in normal-appearing intestine. However, beta-catenin was nuclear in DCAMKL-1-positive cells in adenomas. Thus, nuclear translocation of beta-catenin distinguishes normal and adenoma stem cells. Targeting DCAMKL-1 may represent a strategy for developing novel chemotherapeutic agents.


Asunto(s)
Poliposis Adenomatosa del Colon/metabolismo , Biomarcadores de Tumor/metabolismo , Tracto Gastrointestinal/citología , Neoplasias Intestinales/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Células Madre/citología , Animales , Quinasas Similares a Doblecortina , Tracto Gastrointestinal/efectos de la radiación , Mucosa Intestinal/metabolismo , Intestinos/citología , Intestinos/patología , Intestinos/efectos de la radiación , Ratones , Proteínas del Tejido Nervioso/metabolismo , Transporte de Proteínas/efectos de la radiación , Proteínas de Unión al ARN/metabolismo , Radiación Ionizante , Regeneración/efectos de la radiación , Células Madre/efectos de la radiación , beta Catenina/metabolismo
15.
Mol Cancer Ther ; 18(12): 2446-2456, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31484704

RESUMEN

Hyaluronic acid (HA), a constituent of the extracellular matrix, promotes colorectal cancer growth. CD44 is a relevant HA receptor in this context. However, HA is also a ligand for TLR4, a receptor of significance in colorectal cancer. In this study, we examine the relative contribution of HA interactions with CD44 and TLR4 in colon tumorigenesis. Colorectal cancer models included ApcMin/+ mice, azoxymethane/dextran sodium sulfate (AOM-DSS), and CT26 tumor isografts. We used knockout mice and CT26 colorectal cancer cells with CRISPR knockdown of CD44 and TLR4. HA activity was modulated by PEP1 (a 12-mer peptide that blocks HA from binding its receptors), hyaluronidase (which promotes HA degradation), or 4-MU (HA synthesis inhibitor). Blockade of HA binding via PEP1 decreased growth in all colorectal cancer models and in cell culture. The effects were significant in WT and with CD44 deletion, but not with TLR4 deletion. In the AOM-DSS model, mice deficient in CD44 or TLR4 had fewer tumors. CD44- and TLR4-deficient CT26 isografts grew more slowly, exhibiting decreased tumor cell proliferation and increased apoptosis. In vitro, endogenous HA blocked LPS binding to TLR4 suggesting that HA is a relevant TLR4 ligand in colon cancer. Finally, PEP1 enhanced tumor radiation sensitivity in the isograft model. Together, these results indicate that HA binding to TLR4, as well as CD44, plays a key role in colon tumorigenesis. These findings also raise the possibility that an agent that blocks HA binding, such as PEP1, may be useful as an adjuvant therapy in colon cancer.


Asunto(s)
Neoplasias del Colon/tratamiento farmacológico , Neoplasias del Colon/genética , Ácido Hialurónico/uso terapéutico , Receptor Toll-Like 4/efectos de los fármacos , Animales , Apoptosis , Línea Celular Tumoral , Neoplasias del Colon/patología , Humanos , Ácido Hialurónico/farmacología , Ratones
16.
J Clin Invest ; 114(11): 1676-85, 2004 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-15578100

RESUMEN

Prostaglandin E2 (PGE2) synthesis modulates the response to radiation injury in the mouse intestinal epithelium through effects on crypt survival and apoptosis; however, the downstream signaling events have not been elucidated. WT mice receiving 16,16-dimethyl PGE2 (dmPGE2) had fewer apoptotic cells per crypt than untreated mice. Apoptosis in Bax(-/-) mice receiving 12 Gy was approximately 50% less than in WT mice, and the ability of dmPGE2 to attenuate apoptosis was lost in Bax(-/-) mice. Positional analysis revealed that apoptosis in the Bax(-/-) mice was diminished only in the bax-expressing cells of the lower crypts and that in WT mice, dmPGE2 decreased apoptosis only in the bax-expressing cells. The HCT-116 intestinal cell line and Bax(-/-) HCT-116 recapitulated the apoptotic response of the mouse small intestine with regard to irradiation and dmPGE2. Irradiation of HCT-116 cells resulted in phosphorylation of AKT that was enhanced by dmPGE2 through transactivation of the EGFR. Inhibition of AKT phosphorylation prevented the reduction of apoptosis by dmPGE2 following radiation. Transfection of HCT-116 cells with a constitutively active AKT reduced apoptosis in irradiated cells to the same extent as in nontransfected cells treated with dmPGE2. Treatment with dmPGE2 did not alter bax or bcl-x expression but suppressed bax translocation to the mitochondrial membrane. Our in vivo studies indicate that there are bax-dependent and bax-independent radiation-induced apoptosis in the intestine but that only the bax-dependent apoptosis is reduced by dmPGE2. The in vitro studies indicate that dmPGE2, most likely by signaling through the E prostaglandin receptor EP2, reduces radiation-induced apoptosis through transactivation of the EGFR and enhanced activation of AKT and that this results in reduced bax translocation to the mitochondria.


Asunto(s)
Apoptosis/fisiología , Dinoprostona/farmacología , Células Epiteliales/efectos de los fármacos , Células Epiteliales/efectos de la radiación , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Animales , Línea Celular , Dinoprostona/administración & dosificación , Dinoprostona/análogos & derivados , Activación Enzimática , Células Epiteliales/metabolismo , Humanos , Mucosa Intestinal/citología , Mucosa Intestinal/efectos de la radiación , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Proteínas Proto-Oncogénicas c-akt , Transducción de Señal/fisiología , Proteína X Asociada a bcl-2 , Proteína bcl-X
17.
Shock ; 23(4): 311-8, 2005 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15803053

RESUMEN

Tumor necrosis factor (TNF)-alpha, a cardinal molecule in the cascade of sepsis-induced host injury, binds to two distinct receptors: tumor necrosis factor receptor (TNFR) 1 and TNFR2. We used the cecal ligation and puncture model of polymicrobial sepsis to elucidate the role of these receptors in sepsis pathogenesis. Mice lacking TNFR1 had prolonged survival with less hypothermia, whereas mice lacking TNFR2-/- had shortened survival and more profound hypothermia than wild-type mice. TNFR1-/- and TNFR2-/- mice had increased serum concentrations of interleukin (IL) 1beta and total TNF-alpha (free plus receptor bound) compared with wild-type mice, but there were no differences in IL6 or IL10 concentrations. Furthermore, free TNF-alpha was markedly elevated in the serum and peritoneal fluid of mice lacking TNFR2, supporting a role for this receptor in regulating the concentration of TNF-alpha. Lastly, apoptosis of ileal crypt epithelial cells was increased in mice lacking TNFR1, but there were no differences in lymphocyte apoptosis. These data suggest that in sepsis, TNFR1 mediates much of the TNF-alpha-induced pathology, whereas TNFR2 mediates protective effects.


Asunto(s)
Ciego/cirugía , Íleon/patología , Receptores Tipo II del Factor de Necrosis Tumoral/fisiología , Receptores Tipo I de Factores de Necrosis Tumoral/fisiología , Animales , Apoptosis , Líquido Ascítico/metabolismo , Temperatura Corporal , Ciego/metabolismo , Proliferación Celular , Supervivencia Celular , Citocinas/metabolismo , Células Epiteliales/citología , Femenino , Citometría de Flujo , Íleon/metabolismo , Interleucina-1/sangre , Interleucina-1/metabolismo , Interleucina-10/metabolismo , Interleucina-6/metabolismo , Linfocitos/patología , Masculino , Ratones , Ratones Transgénicos , Peritoneo/metabolismo , Sepsis , Factores de Tiempo , Factor de Necrosis Tumoral alfa/biosíntesis , Factor de Necrosis Tumoral alfa/metabolismo
18.
Oecologia ; 54(2): 193-199, 1982 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28311428

RESUMEN

The relationship between the distribution and growth of Salicornia europaea and soil conditions was studied on an inland saline marsh. Data were collected concerning plant growth, ion content, and water potential of S. europaea throughout the growing season to determine how these responses were related to soil-ionic content, electrical conductivity, and water potential. Soil salinity concentration was the factor most highly correlated with plant growth, survival and ionic content of organs of S. europaea.

19.
EBioMedicine ; 1(1): 46-57, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26125048

RESUMEN

The spread of multidrug-resistant microorganisms globally has created an urgent need for novel therapeutic strategies to combat urinary tract infections (UTIs). Immunomodulatory therapy may provide benefit, as treatment of mice with dexamethasone during acute UTI improved outcome by reducing the development of chronic cystitis, which predisposes to recurrent infection. Here we discovered soluble biomarkers engaged in myeloid cell development and chemotaxis that were predictive of future UTI recurrence when elevated in the sera of young women with UTI. Translation of these findings revealed that temperance of the neutrophil response early during UTI, and specifically disruption of bladder epithelial transmigration of neutrophils by inhibition of cyclooxygenase-2, protected mice against chronic and recurrent cystitis. Further, proteomics identified bladder epithelial remodeling consequent to chronic infection that enhances sensitivity to neutrophil damage. Thus, cyclooxygenase-2 expression during acute UTI is a critical molecular trigger determining disease outcome and drugs targeting cyclooxygenase-2 could prevent recurrent UTI.

20.
Inflamm Bowel Dis ; 18(7): 1214-20, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21823214

RESUMEN

BACKGROUND: Indoleamine 2,3 dioxygenase-1 (IDO1) is a tryptophan catabolizing enzyme with immunotolerance-promoting functions. We sought to determine if increased gut expression of IDO1 in Crohn's disease (CD) would result in detectable changes in serum levels of tryptophan and the initial IDO1 pathway catabolite, kynurenine. METHODS: Individuals were prospectively enrolled through the Washington University Digestive Diseases Research Center. The Montreal Classification was used for disease phenotyping. Disease severity was categorized by the Physician's Global Assessment. Serum tryptophan and kynurenine were measured by high-pressure liquid chromatography. IDO1 immunohistochemical staining was performed on formalin-fixed tissue blocks. RESULTS: In all, 25 CD patients and 11 controls were enrolled. Eight CD patients had serum collected at two different timepoints and levels of disease activity compared. Strong IDO1 expression exists in both the lamina propria and epithelium during active CD compared to controls. Suppressed serum tryptophan levels and an elevated kynurenine/tryptophan (K/T) ratio were found in individuals with active CD as compared to those in remission or the control population. K/T ratios correlated positively with disease activity as well as with C-reactive protein and erythrocyte sedimentation rate. In the subgroup of CD patients with two serum measurements, tryptophan levels were elevated while kynurenine levels and the K/T ratio lowered as the disease activity lessened. CONCLUSIONS: IDO1 expression in CD is associated with lower serum tryptophan and an elevated K/T ratio. These levels may serve as a reasonable objective marker of gut mucosal immune activation and as a surrogate for CD activity.


Asunto(s)
Biomarcadores/sangre , Enfermedad de Crohn/sangre , Indolamina-Pirrol 2,3,-Dioxigenasa/metabolismo , Quinurenina/sangre , Triptófano/sangre , Adulto , Estudios de Casos y Controles , Cromatografía Líquida de Alta Presión , Enfermedad de Crohn/diagnóstico , Enfermedad de Crohn/enzimología , Femenino , Humanos , Técnicas para Inmunoenzimas , Masculino , Persona de Mediana Edad , Pronóstico , Estudios Prospectivos , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA