Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Neoplasia ; 51: 100984, 2024 05.
Artículo en Inglés | MEDLINE | ID: mdl-38467087

RESUMEN

INTRODUCTION: Colorectal cancer is the third most common cause of cancer death. Rectal cancer makes up a third of all colorectal cases. Treatment for locally advanced rectal cancer includes chemoradiation followed by surgery. We have previously identified ST6GAL1 as a cause of resistance to chemoradiation in vitro and hypothesized that it would be correlated with poor response in human derived models and human tissues. METHODS: Five organoid models were created from primary human rectal cancers and ST6GAL1 was knocked down via lentivirus transduction in one model. ST6GAL1 and Cleaved Caspase-3 (CC3) were assessed after chemoradiation via immunostaining. A tissue microarray (TMA) was created from twenty-six patients who underwent chemoradiation and had pre- and post-treatment specimens of rectal adenocarcinoma available at our institution. Immunohistochemistry was performed for ST6GAL1 and percent positive cancer cell staining was assessed and correlation with pathological grade of response was measured. RESULTS: Organoid models were treated with chemoradiation and both ST6GAL1 mRNA and protein significantly increased after treatment. The organoid model targeted with ST6GAL1 knockdown was found to have increased CC3 after treatment. In the tissue microarray, 42 percent of patient samples had an increase in percent tumor cell staining for ST6GAL1 after treatment. Post-treatment percent staining was associated with a worse grade of treatment response (p = 0.01) and increased staining post-treatment compared to pre-treatment was also associated with a worse response (p = 0.01). CONCLUSION: ST6GAL1 is associated with resistance to treatment in human rectal cancer and knockdown in an organoid model abrogated resistance to apoptosis caused by chemoradiation.


Asunto(s)
Quimioradioterapia , Neoplasias del Recto , beta-D-Galactósido alfa 2-6-Sialiltransferasa , Humanos , Antígenos CD , beta-D-Galactósido alfa 2-6-Sialiltransferasa/efectos de los fármacos , beta-D-Galactósido alfa 2-6-Sialiltransferasa/metabolismo , beta-D-Galactósido alfa 2-6-Sialiltransferasa/efectos de la radiación , Estadificación de Neoplasias , Neoplasias del Recto/tratamiento farmacológico , Neoplasias del Recto/genética , Neoplasias del Recto/radioterapia
2.
Neoplasia ; 25: 53-61, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-35168148

RESUMEN

Treatment of locally advanced rectal cancer includes chemoradiation and surgery, but patient response to treatment is variable. Patients who have a complete response have improved outcomes; therefore, there is a critical need to identify mechanisms of resistance to circumvent them. DNA-PK is involved in the repair of DNA double-strand breaks caused by radiation, which we found to be increased in rectal cancer after treatment. We hypothesized that inhibiting this complex with a DNA-PK inhibitor, Peposertib (M3814), would improve treatment response. We assessed pDNA-PK in a rectal cancer cell line and mouse model utilizing western blotting, viability assays, γH2AX staining, and treatment response. The three treatment groups were: standard of care (SOC) (5-fluorouracil (5FU) with radiation), M3814 with radiation, and M3814 with SOC. SOC treatment of rectal cancer cells increased pDNA-PK protein and increased γH2AX foci, but this was abrogated by the addition of M3814. Mice with CT26 tumors treated with M3814 with SOC did not differ in average tumor size but individual tumor response varied. The clinical complete response rate improved significantly with the addition of M3814 but pathological complete response did not. We investigated alterations in DNA repair and found that Kap1 and pATM are increased after M3814 addition suggesting this may mediate resistance. When the DNA-PK inhibitor, M3814, is combined with SOC treatment, response improved in some rectal cancer models but an increase in other repair mechanisms likely diminishes the effect. A clinical trial is ongoing to further explore the role of DNA-PK inhibition in rectal cancer treatment.


Asunto(s)
Terapia Neoadyuvante , Neoplasias del Recto , Animales , Quimioradioterapia , ADN , Humanos , Ratones , Piridazinas , Quinazolinas/farmacología , Neoplasias del Recto/genética , Neoplasias del Recto/patología , Neoplasias del Recto/terapia , Resultado del Tratamiento
3.
J Biol Chem ; 298(3): 101594, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-35041825

RESUMEN

Locally advanced rectal cancer is typically treated with chemoradiotherapy followed by surgery. Most patients do not display a complete response to chemoradiotherapy, but resistance mechanisms are poorly understood. ST6GAL-1 is a sialyltransferase that adds the negatively charged sugar, sialic acid (Sia), to cell surface proteins in the Golgi, altering their function. We therefore hypothesized that ST6GAL-1 could mediate resistance to chemoradiation in rectal cancer by inhibiting apoptosis. Patient-derived xenograft and organoid models of rectal cancer and rectal cancer cell lines were assessed for ST6GAL-1 protein with and without chemoradiation treatment. ST6GAL-1 mRNA was assessed in untreated human rectal adenocarcinoma by PCR assays. Samples were further assessed by Western blotting, Caspase-Glo apoptosis assays, and colony formation assays. The presence of functional ST6GAL-1 was assessed via flow cytometry using the Sambucus nigra lectin, which specifically binds cell surface α2,6-linked Sia, and via lectin precipitation. In patient-derived xenograft models of rectal cancer, we found that ST6GAL-1 protein was increased after chemoradiation in a subset of samples. Rectal cancer cell lines demonstrated increased ST6GAL-1 protein and cell surface Sia after chemoradiation. ST6GAL-1 was also increased in rectal cancer organoids after treatment. ST6GAL-1 knockdown in rectal cancer cell lines resulted in increased apoptosis and decreased survival after treatment. We concluded that ST6GAL-1 promotes resistance to chemoradiotherapy by inhibiting apoptosis in rectal cancer cell lines. More research will be needed to further elucidate the importance and mechanism of ST6GAL-1-mediated resistance.


Asunto(s)
Antígenos CD , Neoplasias del Recto , Sialiltransferasas , Antígenos CD/metabolismo , Apoptosis/efectos de los fármacos , Apoptosis/efectos de la radiación , Quimioradioterapia , Resistencia a Antineoplásicos , Humanos , Ácido N-Acetilneuramínico/metabolismo , Tolerancia a Radiación , Neoplasias del Recto/tratamiento farmacológico , Neoplasias del Recto/metabolismo , Neoplasias del Recto/patología , Neoplasias del Recto/radioterapia , Sialiltransferasas/genética , Sialiltransferasas/metabolismo , beta-D-Galactósido alfa 2-6-Sialiltransferasa
4.
Neoplasia ; 21(10): 1051-1062, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31521947

RESUMEN

Treatment of locally advanced rectal cancer includes chemotherapy, radiation, and surgery but patient responses to neoadjuvant treatment are variable. We have shown that rectal tumors are comprised of multiple genetically distinct sub-clones. Unique sub-clones within tumors may harbor mutations which contribute to inter-patient variation in response to neoadjuvant chemoradiotherapy (nCRT). Analysis of the influence of nCRT on the extent and nature of intra-tumoral genetic heterogeneity in rectal cancer may provide insights into mechanisms of resistance. Locally advanced rectal cancer patients underwent pre-treatment biopsies. At the time of surgery, tissue from the treated tumor was obtained and analyzed. Pre- and post-treatment specimens were subjected to whole exome and confirmatory deep sequencing for somatic mutations. Copy number variation was assessed using OncoScan SNP arrays. Genomic data were analyzed using PyClone to identify sub-clonal tumor population following nCRT. Alterations that persisted or were enriched in the post-treatment tumor specimen following nCRT were defined for each patient. Thirty-two samples were obtained from ten patients. PyClone identified 2 to 10 genetic sub-clones per tumor. Substantial changes in the proportions of individual sub-clones in pre- versus post-treatment tumor material were found in all patients. Resistant sub-clones recurrently contained mutations in TP53, APC, ABCA13, MUC16, and THSD4. Recurrent copy number variation was observed across multiple chromosome regions after nCRT. Pathway analysis including variant alleles and copy number changes associated with resistant sub-clones revealed significantly altered pathways, especially those linked to the APC and TP53 genes, which were the two most frequently mutated genes. Intra-tumoral heterogeneity is evident in pre-treatment rectal cancer. Following treatment, sub-clonal populations are selectively modified and enrichment of a subset of pre-treatment sub-clones is seen. Further studies are needed to define recurrent alterations at diagnosis that may contribute to resistance to nCRT.


Asunto(s)
Antineoplásicos/farmacología , Evolución Clonal/efectos de los fármacos , Evolución Clonal/genética , Heterogeneidad Genética , Neoplasias del Recto/genética , Adulto , Anciano , Antineoplásicos/uso terapéutico , Biomarcadores de Tumor , Quimioradioterapia , Resistencia a Antineoplásicos , Femenino , Humanos , Masculino , Persona de Mediana Edad , Mutación , Terapia Neoadyuvante , Clasificación del Tumor , Estadificación de Neoplasias , Neoplasias del Recto/metabolismo , Neoplasias del Recto/patología , Neoplasias del Recto/terapia , Transducción de Señal , Resultado del Tratamiento , Secuenciación del Exoma
6.
Bone ; 118: 20-31, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-29604350

RESUMEN

High fat diets can have detrimental effects on the skeleton as well as cause intestinal dysbiosis. Exercise prevents high fat (HF) diet-induced obesity and also improves bone density and prevents the intestinal dysbiosis that promotes energy storage. Previous studies indicate a link between intestinal microbial balance and bone health. Therefore, we examined whether exercise could prevent HF-induced bone pathology in male mice and determined whether benefits correlate to changes in host intestinal microbiota. Male C57Bl/6 mice were fed either a low fat diet (LF; 10 kcal% fat) or a HF diet (60 kcal% fat) and put under sedentary or voluntary exercise conditions for 14 weeks. Our results indicated that HF diet reduced trabecular bone volume, when corrected for differences in body weight, of both the tibia (40% reduction) and vertebrae (25% reduction) as well and increased marrow adiposity (44% increase). More importantly, these effects were prevented by exercise. Exercise also had a significant effect on several cortical bone parameters and enhanced bone mechanical properties in LF but not HF fed mice. Microbiome analyses indicated that exercise altered the HF induced changes in microbial composition by reducing the Firmicutes/Bacteriodetes ratio. This ratio negatively correlated with bone volume as did levels of Clostridia and Lachnospiraceae. In contrast, the abundance of several Actinobacteria phylum members (i.e., Bifidobacteriaceae) were positively correlated with bone volume. Taken together, exercise can prevent many of the negative effects of a high fat diet on male skeletal health. Exercise induced changes in microbiota composition could represent a novel mechanism that contributes to exercise induced benefits to bone health.


Asunto(s)
Adiposidad , Médula Ósea/patología , Resorción Ósea/prevención & control , Dieta Alta en Grasa/efectos adversos , Disbiosis/prevención & control , Condicionamiento Físico Animal , Animales , Biomarcadores/sangre , Resorción Ósea/sangre , Resorción Ósea/complicaciones , Resorción Ósea/fisiopatología , Hueso Esponjoso/patología , Hueso Esponjoso/fisiopatología , Hueso Cortical/patología , Hueso Cortical/fisiopatología , Disbiosis/sangre , Disbiosis/complicaciones , Microbioma Gastrointestinal , Masculino , Ratones Endogámicos C57BL , Obesidad/prevención & control , Osteoblastos/metabolismo , Osteoclastos/metabolismo , Osteogénesis
7.
Adv Exp Med Biol ; 1033: 225-247, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29101658

RESUMEN

The intestinal environment is linked to an array of conditions and diseases, including osteoporosis. Human and animal studies indicate that probiotics can benefit intestinal health and may provide a useful therapeutic to prevent and/or treat bone loss. Probiotics are defined as live microorganisms that when administered in adequate amounts will confer a health benefit on the host. In this review, we will focus on (1) probiotics (definition, history, nomenclature, types), (2) the effects of probiotics on bone health, and (3) mechanisms of probiotic prevention of bone pathologies.


Asunto(s)
Huesos/fisiología , Tracto Gastrointestinal/fisiología , Probióticos/uso terapéutico , Transducción de Señal/efectos de los fármacos , Animales , Enfermedades Óseas/fisiopatología , Enfermedades Óseas/prevención & control , Humanos , Osteoblastos/efectos de los fármacos , Osteoblastos/fisiología , Osteogénesis/efectos de los fármacos , Osteogénesis/fisiología , Probióticos/administración & dosificación
8.
Sci Rep ; 7: 42505, 2017 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-28198415

RESUMEN

We have developed a straightforward method that uses paraffin-embedded bone for undemineralized thin sectioning, which is amenable to subsequent dynamic bone formation measurements. Bone has stiffer material properties than paraffin, and therefore has hereforto usually been embedded in plastic blocks, cured and sectioned with a tungsten carbide knife to obtain mineralized bone sections for dynamic bone formation measures. This process is expensive and requires special equipment, experienced personnel, and time for the plastic to penetrate the bone and cure. Our method utilizes a novel way to prepare mineralized bone that increases its compliance so that it can be embedded and easily section in paraffin blocks. The approach is simple, quick, and costs less than 10% of the price for plastic embedded bone sections. While not effective for static bone measures, this method allows dynamic bone analyses to be readily performed in laboratories worldwide which might not otherwise have access to traditional (plastic) equipment and expertise.


Asunto(s)
Huesos/citología , Huesos/metabolismo , Histocitoquímica , Osteogénesis , Animales , Biomarcadores , Técnica del Anticuerpo Fluorescente , Histocitoquímica/métodos , Ratones
9.
Am J Physiol Regul Integr Comp Physiol ; 311(6): R1149-R1157, 2016 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-27733383

RESUMEN

Increasing evidence indicates a strong link between intestinal health and bone health. For example, inflammatory bowel disease can cause systemic inflammation, weight loss, and extra-intestinal manifestations, such as decreased bone growth and density. However, the effects of moderate intestinal inflammation without weight loss on bone health have never been directly examined; yet this condition is relevant not only to IBD but to conditions of increased intestinal permeability and inflammation, as seen with ingestion of high-fat diets, intestinal dysbiosis, irritable bowel syndrome, metabolic syndrome, and food allergies. Here, we induced moderate intestinal inflammation without weight loss in young male mice by treating with a low dose of dextran sodium sulfate (1%) for 15 days. The mice displayed systemic changes marked by significant bone loss and a redistribution of fat from subcutaneous to visceral fat pad stores. Bone loss was caused by reduced osteoblast activity, characterized by decreased expression of osteoblast markers (runx2, osteocalcin), histomorphometry, and dynamic measures of bone formation. In addition, we observed a reduction in growth plate thickness and hypertrophic chondrocyte matrix components (collagen X). Correlation analyses indicate a link between gut inflammation and disease score, but more importantly, we observed that bone density measures negatively correlated with intestinal disease score, as well as colon and bone TNF-α levels. These studies demonstrate that colitis-induced bone loss is not dependent upon weight loss and support a role for inflammation in the link between gut and bone health, an important area for future therapeutic development.


Asunto(s)
Tejido Adiposo/fisiopatología , Densidad Ósea , Desarrollo Óseo , Colitis/fisiopatología , Osteoporosis/fisiopatología , Pérdida de Peso , Animales , Colitis/complicaciones , Masculino , Ratones , Ratones Endogámicos C57BL , Osteoporosis/etiología , Tibia/fisiopatología
10.
PLoS One ; 11(4): e0153180, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27058036

RESUMEN

BACKGROUND & AIMS: We previously demonstrated that short-term oral administration of the probiotic Lactobacillus reuteri 6475 enhanced bone density in male but not female mice. We also established that L. reuteri 6475 enhanced bone health and prevented bone loss in estrogen-deficient female mice. In this study, we tested whether a mild inflammatory state and/or a long-term treatment with the probiotic was required to promote a positive bone effect in estrogen-sufficient female mice. METHODS: A mild inflammatory state was induced in female mice by dorsal surgical incision (DSI). Following DSI animals were orally supplemented with L. reuteri or vehicle control for a period of 8 weeks. Gene expression was measured in the intestine and bone marrow by qPCR. Distal femoral bone density and architecture was analyzed by micro-CT. RESULTS: We report that 8 weeks after DSI there is a significant increase in the weight of spleen, thymus and visceral (retroperitoneal) fat pads. Expression of intestinal cytokines and tight junction proteins are also altered 8 weeks post-DSI. Interestingly, L. reuteri treatment was found to display both intestinal region- and inflammation-dependent effects. Unexpectedly we identified that 1) L. reuteri treatment increased bone density in females but only in those that underwent DSI and 2) DSI benefited cortical bone parameters. In the bone marrow, dorsal surgery induced CD4+ T cell numbers, a response that was unaffected by L. reuteri treatment, whereas expression of RANKL, OPG and IL-10 were significantly affected by L. reuteri treatment. CONCLUSION: Our data reveals a previously unappreciated effect of a mild surgical procedure causing a long-lasting effect on inflammatory gene expression in the gut and the bone. Additionally, we demonstrate that in intact female mice, the beneficial effect of L. reuteri on bone requires an elevated inflammatory status.


Asunto(s)
Densidad Ósea , Inflamación/terapia , Limosilactobacillus reuteri/fisiología , Probióticos/uso terapéutico , Animales , Células de la Médula Ósea/inmunología , Células de la Médula Ósea/metabolismo , Remodelación Ósea/fisiología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Citocinas/genética , Estrógenos/metabolismo , Femenino , Expresión Génica , Inflamación/genética , Inflamación/metabolismo , Mucosa Intestinal/metabolismo , Intestinos/inmunología , Masculino , Ratones , Ratones Endogámicos BALB C , Probióticos/administración & dosificación , Microtomografía por Rayos X
11.
Endocrinology ; 156(9): 3169-82, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26135835

RESUMEN

Type 1 diabetes (T1D)-induced osteoporosis is characterized by a predominant suppression of osteoblast number and activity, as well as increased bone marrow adiposity but no change in osteoclast activity. The fundamental mechanisms and alternative anabolic treatments (with few side effects) for T1D bone loss remain undetermined. Recent studies by our laboratory and others indicate that probiotics can benefit bone health. Here, we demonstrate that Lactobacillus reuteri, a probiotic with anti-inflammatory and bone health properties, prevents T1D-induced bone loss and marrow adiposity in mice. We further found that L. reuteri treatment prevented the suppression of Wnt10b in T1D bone. Consistent with a role for attenuated bone Wnt10b expression in T1D osteoporosis, we observed that bone-specific Wnt10b transgenic mice are protected from T1D bone loss. To examine the mechanisms of this protection, we focused on TNF-α, a cytokine up-regulated in T1D that causes suppression of osteoblast Wnt10b expression in vitro. Addition of L. reuteri prevented TNF-α-mediated suppression of Wnt10b and osteoblast maturation markers. Taken together, our findings reveal a mechanism by which T1D causes bone loss and open new avenues for use of probiotics to benefit the bone.


Asunto(s)
Diabetes Mellitus Tipo 1/complicaciones , Limosilactobacillus reuteri , Osteoporosis/prevención & control , Probióticos/uso terapéutico , Proteínas Wnt/metabolismo , Animales , Diabetes Mellitus Tipo 1/metabolismo , Masculino , Ratones Endogámicos C57BL , Ratones Transgénicos , Osteoblastos/efectos de los fármacos , Osteoblastos/metabolismo , Osteoporosis/etiología , Osteoporosis/metabolismo , Probióticos/farmacología
12.
Mater Sci Eng C Mater Biol Appl ; 44: 191-200, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25280696

RESUMEN

Microcracks are present in bone and can result from fatigue damage due to repeated, cyclically applied stresses. From a mechanical point, microcracks can dissipate strain energy at the advancing tip of a crack to improve overall bone toughness. Physiologically, microcracks are thought to trigger bone remodeling. Here, we examine the effect of microcracks specifically on osteoblasts, which are bone-forming cells, by comparing cell responses on microcracked versus non-microcracked hydroxyapatite (HA) specimens. Osteoblast attachment was found to be greater on microcracked HA specimens (p<0.05). More importantly, we identified the preferential alignment of osteoblasts in the direction of the microcracks on HA. Cells also displayed a preferential attachment that was 75 to 90 µm away from the microcrack indent. After 21 days of culture, osteoblast maturation was notably enhanced on the HA with microcracks, as indicated by increased alkaline phosphatase activity and gene expression. Furthermore, examination of bone deposition by confocal laser scanning microscopy indicated preferential mineralization at microcrack indentation sites. Dissolution studies indicate that the microcracks increase calcium release, which could contribute to osteoblast responses. Our findings suggest that microcracks signal osteoblast attachment and bone formation/healing.


Asunto(s)
Osteoblastos/efectos de los fármacos , Osteogénesis/efectos de los fármacos , Células 3T3-L1 , Animales , Remodelación Ósea/efectos de los fármacos , Huesos/efectos de los fármacos , Huesos/fisiología , Diferenciación Celular/efectos de los fármacos , Durapatita/farmacología , Ratones , Osteoblastos/citología , Tamaño de la Partícula , Manejo de Especímenes , Propiedades de Superficie , Andamios del Tejido/química , Difracción de Rayos X
13.
J Cell Physiol ; 229(11): 1822-30, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24677054

RESUMEN

Estrogen deficiency is a major risk factor for osteoporosis that is associated with bone inflammation and resorption. Half of women over the age of 50 will experience an osteoporosis related fracture in their lifetime, thus novel therapies are needed to combat post-menopausal bone loss. Recent studies suggest an important role for gut-bone signaling pathways and the microbiota in regulating bone health. Given that the bacterium Lactobacillus reuteri ATCC PTA 6475 (L. reuteri) secretes beneficial immunomodulatory factors, we examined if this candidate probiotic could reduce bone loss associated with estrogen deficiency in an ovariectomized (Ovx) mouse menopausal model. Strikingly, L. reuteri treatment significantly protected Ovx mice from bone loss. Osteoclast bone resorption markers and activators (Trap5 and RANKL) as well as osteoclastogenesis are significantly decreased in L. reuteri-treated mice. Consistent with this, L. reuteri suppressed Ovx-induced increases in bone marrow CD4+ T-lymphocytes (which promote osteoclastogenesis) and directly suppressed osteoclastogenesis in vitro. We also identified that L. reuteri treatment modifies microbial communities in the Ovx mouse gut. Together, our studies demonstrate that L. reuteri treatment suppresses bone resorption and loss associated with estrogen deficiency. Thus, L. reuteri treatment may be a straightforward and cost-effective approach to reduce post-menopausal bone loss.


Asunto(s)
Resorción Ósea/tratamiento farmacológico , Resorción Ósea/prevención & control , Ovariectomía , Probióticos/uso terapéutico , Animales , Biomarcadores/metabolismo , Células de la Médula Ósea/metabolismo , Células de la Médula Ósea/patología , Diferenciación Celular , Línea Celular , Modelos Animales de Enfermedad , Femenino , Fémur/patología , Citometría de Flujo , Lactobacillus , Menopausia , Ratones , Ratones Endogámicos BALB C , Tamaño de los Órganos , Osteoblastos/metabolismo , Osteoblastos/patología , Osteoclastos/metabolismo , Osteoclastos/patología , Análisis de Componente Principal , Ligando RANK/metabolismo , Columna Vertebral/patología
14.
Inflamm Bowel Dis ; 19(8): 1586-97, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23702805

RESUMEN

BACKGROUND: Patients with inflammatory bowel disease (IBD) are at increase risk for bone loss and fractures. Therefore, in the present study, we examined the effect of experimental IBD on bone health. METHODS: We used a murine model of colitis, Helicobacter hepaticus-infected interleukin-10-deficient animals. Molecular and histological properties of bone and intestine were examined to identify the immunopathological consequences of colitis in male and female mice. RESULTS: At 6 weeks postinfection, we observed significant trabecular bone loss in male mice but surprisingly not in female mice. This was true for both distal femur and vertebral locations. In addition, H. hepaticus infection suppressed osteoblast markers only in male mice. Consistent with effects on bone health, male mice with H. hepaticus infection had more severe colitis as determined by histology and elevated levels of inflammatory cytokines in the colon. Although H. hepaticus levels in the stool appeared similar in male and female mice 1 week after infection, by 6 weeks, H. hepaticus levels were greater in male mice, indicating that H. hepaticus survival and virulence within the gastrointestinal tract could be gender dependent. CONCLUSION: In summary, H. hepaticus-induced colitis severity and associated bone loss is gender regulated, possibly as a result of gender-specific effects on H. hepaticus colonization in the mouse gastrointestinal tract and the consequent immunopathological responses.


Asunto(s)
Enfermedades Óseas/etiología , Colitis/complicaciones , Infecciones por Helicobacter/complicaciones , Helicobacter hepaticus/patogenicidad , Inflamación/etiología , Interleucina-10/fisiología , Intestinos/patología , Animales , Western Blotting , Densidad Ósea , Enfermedades Óseas/patología , Colitis/microbiología , Colitis/patología , Citocinas/metabolismo , Ensayo de Inmunoadsorción Enzimática , Femenino , Infecciones por Helicobacter/microbiología , Infecciones por Helicobacter/patología , Inflamación/patología , Intestinos/microbiología , Masculino , Ratones , Ratones Noqueados , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores Sexuales
15.
Am J Pathol ; 182(4): 1114-23, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23395087

RESUMEN

ß-Arrestins are intracellular scaffolding proteins that modulate specific cell signaling pathways. Recent studies, in both cell culture and in vivo models, have demonstrated an important role for ß-arrestin-1 in inflammation. However, the role of ß-arrestin-1 in the pathogenesis of inflammatory bowel disease (IBD) is not known. Our goal was to investigate the role of ß-arrestin-1 in IBD using mouse models of colitis. To this end, we subjected wild-type (WT) and ß-arrestin-1 knockout (ß-arr-1(-/-)) mice to colitis induced by trinitrobenzenesulfonic acid or dextran sulfate sodium and examined the clinical signs, gross pathology, and histopathology of the colon, as well as inflammatory components. The ß-arr-1(-/-) mice displayed significantly attenuated colitis, compared with WT mice, in both models. Consistent with the phenotypic observations, histological examination of the colon revealed attenuated disease pathology in the ß-arr-1(-/-) mice. Our results further demonstrate that ß-arr-1(-/-) mice are deficient in IL-6 expression in the colon, but have higher expression of the anti-inflammatory IL-10 family of cytokines. Our results also demonstrate diminished ERK and NFκB pathways in the colons of ß-arr-1(-/-) mice, compared with WT mice. Taken together, our results demonstrate that decreased IL-6 production and enhanced IL-10 and IL-22 production in ß-arrestin-1-deficient mice likely lead to attenuated gut inflammation.


Asunto(s)
Arrestinas/deficiencia , Colitis/patología , Colitis/prevención & control , Animales , Arrestinas/metabolismo , Colitis/sangre , Colitis/enzimología , Colon/patología , Sulfato de Dextran , Activación Enzimática , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Inflamación/patología , Interleucina-10/sangre , Interleucina-6/sangre , Ratones , Ratones Noqueados , FN-kappa B/metabolismo , Transducción de Señal , Ácido Trinitrobencenosulfónico , Pérdida de Peso , beta-Arrestina 1 , beta-Arrestinas
16.
J Cell Physiol ; 228(8): 1793-8, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23389860

RESUMEN

Osteoporosis can result from intestinal inflammation, as is seen with inflammatory bowel disease. Probiotics, microorganisms that provide a health benefit to the host when ingested in adequate amounts, can have anti-inflammatory properties and are currently being examined to treat inflammatory bowel disease. Here, we examined if treating healthy male mice with Lactobacillus reuteri ATCC PTA 6475 (a candidate probiotic with anti-TNFα activity) could affect intestinal TNFα levels and enhance bone density. Adult male mice were given L. reuteri 6475 orally by gavage for 3×/week for 4 weeks. Examination of jejunal and ileal RNA profiles indicates that L. reuteri suppressed basal TNFα mRNA levels in the jejunum and ileum in male mice, but surprisingly not in female mice. Next, we examined bone responses. Micro-computed tomography demonstrated that L. reuteri 6475 treatment increased male trabecular bone parameters (mineral density, bone volume fraction, trabecular number, and trabecular thickness) in the distal femur metaphyseal region as well as in the lumbar vertebrae. Cortical bone parameters were unaffected. Dynamic and static histomorphometry and serum remodeling parameters indicate that L. reuteri ingestion increases osteoblast serum markers and dynamic measures of bone formation in male mice. In contrast to male mice, L. reuteri had no effect on bone parameters in female mice. Taken together our studies indicate that femoral and vertebral bone formation increases in response to oral probiotic use, leading to increased trabecular bone volume in male mice.


Asunto(s)
Enfermedades Inflamatorias del Intestino/prevención & control , Probióticos/farmacología , Animales , Densidad Ósea/fisiología , Femenino , Enfermedades Inflamatorias del Intestino/complicaciones , Enfermedades Inflamatorias del Intestino/patología , Limosilactobacillus reuteri , Masculino , Ratones , Ratones Endogámicos C57BL , Osteoporosis/etiología , Osteoporosis/patología , Osteoporosis/prevención & control , Caracteres Sexuales
17.
Proc Natl Acad Sci U S A ; 109(41): 16594-9, 2012 Oct 09.
Artículo en Inglés | MEDLINE | ID: mdl-23012474

RESUMEN

The bone marrow (BM) is a large, highly active, and responsive tissue. Interestingly, little is known about the impact of colitis on hematopoietic functions. Using dextran sodium sulfate (DSS) to induce colitis in mice, we identified significant changes in the BM. Specifically, cells of the monocytic and granulocytic lineages increased nearly 60% and 80%, respectively. This change would support and promote the large infiltration of the gut with neutrophils and monocytes that are the primary cause of inflammation and tissue damage during colitis. Conversely, the early lineages of B and T cells declined in the marrow and thymus with particularly large losses observed among pre-B and pre-T cells with heightened levels of apoptosis noted among CD4(+)CD8(+) thymocytes from DSS-treated mice. Also noteworthy was the 40% decline in cells of the erythrocytic lineages in the marrow of colitis mice, which undoubtedly contributed to the anemia observed in these mice. The peripheral blood reflected the marrow changes as demonstrated by a 2.6-fold increase in neutrophils, a 60% increase in monocytes, and a decline in the lymphocyte population. Thus, colitis changed the BM in profound ways that parallel the general outcomes of colitis including infiltration of the gut with monocytes and neutrophils, inflammation, and anemia. The data provide important understandings of the full impact of colitis that may lead to unique treatments and therapies.


Asunto(s)
Linaje de la Célula/inmunología , Colitis/inmunología , Granulocitos/inmunología , Monocitos/inmunología , Animales , Linfocitos B/inmunología , Linfocitos B/patología , Células de la Médula Ósea/inmunología , Células de la Médula Ósea/patología , Antígenos CD4/inmunología , Antígenos CD8/inmunología , Colitis/inducido químicamente , Colitis/patología , Sulfato de Dextran , Citometría de Flujo , Granulocitos/patología , Ratones , Ratones Endogámicos C57BL , Monocitos/patología , Neutrófilos/inmunología , Neutrófilos/patología , Linfocitos T/inmunología , Linfocitos T/patología , Timocitos/inmunología , Timocitos/patología , Timo/inmunología , Timo/patología , Factores de Tiempo
18.
J Cell Physiol ; 226(2): 477-83, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20677222

RESUMEN

Type I diabetes increases an individual's risk for bone loss and fracture, predominantly through suppression of osteoblast activity (bone formation). During diabetes onset, levels of blood glucose and pro-inflammatory cytokines (including tumor necrosis factor α (TNFα)) increased. At the same time, levels of osteoblast markers are rapidly decreased and stay decreased chronically (i.e., 40 days later) at which point bone loss is clearly evident. We hypothesized that early bone marrow inflammation can promote osteoblast death and hence reduced osteoblast markers. Indeed, examination of type I diabetic mouse bones demonstrates a greater than twofold increase in osteoblast TUNEL staining and increased expression of pro-apoptotic factors. Osteoblast death was amplified in both pharmacologic and spontaneous diabetic mouse models. Given the known signaling and inter-relationships between marrow cells and osteoblasts, we examined the role of diabetic marrow in causing the osteoblast death. Co-culture studies demonstrate that compared to control marrow cells, diabetic bone marrow cells increase osteoblast (MC3T3 and bone marrow derived) caspase 3 activity and the ratio of Bax/Bcl-2 expression. Mouse blood glucose levels positively correlated with bone marrow induced osteoblast death and negatively correlated with osteocalcin expression in bone, suggesting a relationship between type I diabetes, bone marrow and osteoblast death. TNF expression was elevated in diabetic marrow (but not co-cultured osteoblasts); therefore, we treated co-cultures with TNFα neutralizing antibodies. The antibody protected osteoblasts from bone marrow induced death. Taken together, our findings implicate the bone marrow microenvironment and TNFα in mediating osteoblast death and contributing to type I diabetic bone loss.


Asunto(s)
Médula Ósea/metabolismo , Diabetes Mellitus Tipo 1/patología , Osteoblastos/patología , Células 3T3 , Animales , Glucemia/metabolismo , Células de la Médula Ósea/citología , Células de la Médula Ósea/metabolismo , Huesos/metabolismo , Huesos/patología , Células Cultivadas , Técnicas de Cocultivo , Diabetes Mellitus Tipo 1/metabolismo , Etiquetado Corte-Fin in Situ , Masculino , Ratones , Ratones Endogámicos C57BL , Osteoblastos/citología , Osteoblastos/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
19.
J Cell Physiol ; 218(3): 575-83, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19006181

RESUMEN

Type I diabetes is associated with bone loss and marrow adiposity. To identify early events involved in the etiology of diabetic bone loss, diabetes was induced in mice by multiple low dose streptozotocin injections. Serum markers of bone metabolism and inflammation as well as tibial gene expression were examined between 1 and 17 days post-injection (dpi). At 3 dpi, when blood glucose levels were significantly elevated, body, fat pad and muscle mass were decreased. Serum markers of bone resorption and formation significantly decreased at 5 dpi in diabetic mice and remained suppressed throughout the time course. An osteoclast gene, TRAP5 mRNA, was suppressed at early and late time points. Suppression of osteogenic genes (runx2 and osteocalcin) and induction of adipogenic genes (PPARgamma2 and aP2) were evident as early as 5 dpi. These changes were associated with an elevation of serum cytokines, but more importantly we observed an increase in the expression of cytokines in bone, supporting the idea that bone, itself, exhibits an inflammatory response during diabetes induction. This inflammation could in turn contribute to diabetic bone pathology. IFN-gamma (one of the key cytokines elevated in bone and known to be involved in bone regulation) deficiency did not prevent diabetic bone pathology. Taken together, our findings indicate that bone becomes inflamed with the onset of T1-diabetes and during this time bone phenotype markers become altered. However, inhibition of one cytokine, IFN-gamma was not sufficient to prevent the rapid bone phenotype changes.


Asunto(s)
Diabetes Mellitus Experimental/complicaciones , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Tipo 1/complicaciones , Diabetes Mellitus Tipo 1/genética , Regulación de la Expresión Génica , Osteítis/complicaciones , Osteítis/genética , Animales , Biomarcadores/metabolismo , Resorción Ósea/complicaciones , Resorción Ósea/prevención & control , Citocinas/sangre , Citocinas/genética , Diabetes Mellitus Experimental/sangre , Diabetes Mellitus Tipo 1/sangre , Femenino , Ratones , Ratones Endogámicos BALB C , Osteítis/sangre , Osteoclastos/metabolismo , Fenotipo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Factores de Tiempo
20.
Endocrinology ; 147(12): 5760-7, 2006 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16973725

RESUMEN

Type I diabetes is characterized by little or no insulin production and hyperglycemic conditions. It is also associated with significant bone loss and increased bone marrow adiposity. To examine the role of reduced insulin signaling in type I diabetic bone loss without inducing hyperglycemia, we used genetically reconstituted insulin receptor knockout mice (IRKO-L1) that are euglycemic as a result of human insulin receptor transgene expression in the pancreas, liver, and brain. RT-PCR analyses demonstrated undetectable levels of insulin receptor expression in IRKO-L1 bone, yet IRKO-L1 bones exhibit similar (and trend toward greater) bone density compared with wild-type animals as determined by microcomputed tomography. More detailed bone analyses indicated that cortical bone area was increased in tibias of IRKO-L1 mice. Osteoblast markers (osteocalcin and runx2 mRNA levels) and resorption markers (serum pyridinoline levels) were similar in wild-type and IRKO-L1 bones. When marrow adiposity was examined, we noticed a decrease in adipocyte number and fatty-acid-binding protein 2 expression in IRKO-L1 mice compared with wild-type mice. Bone marrow stromal cell cultures obtained from wild-type and IRKO-L1 mice demonstrated similar adipogenic and osteogenic potentials, indicating that systemic factors likely contribute to differences in marrow adiposity in vivo. Interestingly, IGF-I receptor mRNA levels were elevated in IRKO-L1 bones, suggesting (in combination with hyperinsulinemic conditions) that increased IGF-I receptor signaling may represent a compensatory response and contribute to the changes in cortical bone. Taken together, these results suggest that reduced insulin receptor signaling in bone is not a major factor contributing to bone loss in type I diabetes.


Asunto(s)
Densidad Ósea/fisiología , Huesos/metabolismo , Receptor de Insulina/genética , Receptor de Insulina/metabolismo , Adipocitos Blancos/citología , Animales , Médula Ósea/metabolismo , Proteínas de Unión a Ácidos Grasos/metabolismo , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Osteocalcina/metabolismo , Receptor IGF Tipo 1/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...