Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
1.
Asian J Endosc Surg ; 17(1): e13274, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38212269

RESUMEN

BACKGROUND: Pelvic lymph node dissection is a procedure performed in gastroenterological surgery, urology, and gynecology. However, due to discrepancies in the understanding of pelvic anatomy among these departments, cross-disciplinary discussions have not been easy. Recently, with the rapid spread of robotic surgery, the importance of visual information in understanding pelvic anatomy has become even more significant. In this project, we attempted to clarify a shared understanding of pelvic anatomy through cross-disciplinary discussions. METHOD: From May 2020 to November 2021, a total of 11 discussions were held entirely online with 5 colorectal surgery specialists, 4 urologists, and 4 gynecologists. The discussions focused on evidence from each specialty and surgical videos, aiming to create a universally understandable pelvic anatomical illustration. RESULTS: The common area of dissection recognized across the three departments was identified as the obturator lymph nodes. A dynamic illustration of pelvic anatomy was created. In addition to a bird's-eye view of the pelvis, a pelvic half view was developed to enhance understanding of the deeper pelvic anatomy. The following insights were incorporated into the illustration: (1) the cardinal ligament in gynecology partly overlaps with the vesicohypogastric fascia in colorectal surgery; (2) the obturator lymph nodes continue cephalad into the fossa of Marcille in urology; and (3) the deep uterine vein in gynecology corresponds to the inferior vesical vein in colorectal surgery. CONCLUSION: Based on the dynamic illustration of pelvic anatomy from cross-disciplinary discussions, we anticipate advancements in pelvic lymph node dissection aiming for curative and safe outcomes.


Asunto(s)
Cirugía Colorrectal , Ginecología , Robótica , Urología , Humanos , Anatomía Regional , Escisión del Ganglio Linfático/métodos , Ganglios Linfáticos/patología , Estándares de Referencia
3.
Cell Mol Gastroenterol Hepatol ; 9(2): 277-293, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-31622786

RESUMEN

BACKGROUND & AIMS: Ral guanosine triphosphatase-activating protein α2 (RalGAPα2) is the major catalytic subunit of the negative regulators of the small guanosine triphosphatase Ral, a member of the Ras subfamily. Ral regulates tumorigenesis and invasion/metastasis of some cancers; however, the role of Ral in colitis-associated cancer (CAC) has not been investigated. We aimed to elucidate the role of Ral in the mechanism of CAC. METHODS: We used wild-type (WT) mice and RalGAPα2 knockout (KO) mice that showed Ral activation, and bone marrow chimeric mice were generated as follows: WT to WT, WT to RalGAPα2 KO, RalGAPα2 KO to WT, and RalGAPα2 KO to RalGAPα2 KO mice. CAC was induced in these mice by intraperitoneal injection of azoxymethane followed by dextran sulfate sodium intake. Intestinal epithelial cells were isolated from colon tissues, and we performed complementary DNA microarray analysis. Cytokine expression in normal colon tissues and CAC was analyzed by quantitative polymerase chain reaction. RESULTS: Bone marrow chimeric mice showed that immune cell function between WT mice and RalGAPα2 KO mice was not significantly different in the CAC mechanism. RalGAPα2 KO mice had a significantly larger tumor number and size and a significantly higher proportion of tumors invading the submucosa than WT mice. Higher expression levels of matrix metalloproteinase-9 and matrix metalloproteinase-13 were observed in RalGAPα2 KO mice than in WT mice. The expression levels of interleukin 1ß, NLRP3, apoptosis associated speck-like protein containing a CARD, and caspase-1 were apparently increased in the tumors of RalGAPα2 KO mice compared with WT mice. NLRP3 inhibitor reduced the number of invasive tumors. CONCLUSIONS: Ral activation participates in the mechanism of CAC development via NLRP3 inflammasome activation.


Asunto(s)
Neoplasias Asociadas a Colitis/inmunología , Proteínas Activadoras de GTPasa/metabolismo , Inflamasomas/inmunología , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Neoplasias Experimentales/inmunología , Animales , Azoximetano/administración & dosificación , Azoximetano/toxicidad , Neoplasias Asociadas a Colitis/inducido químicamente , Neoplasias Asociadas a Colitis/patología , Colon/efectos de los fármacos , Colon/inmunología , Colon/patología , Regulación hacia Abajo/inmunología , Proteínas Activadoras de GTPasa/genética , Humanos , Inflamasomas/antagonistas & inhibidores , Inflamasomas/metabolismo , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/inmunología , Mucosa Intestinal/patología , Masculino , Ratones , Ratones Noqueados , Proteína con Dominio Pirina 3 de la Familia NLR/antagonistas & inhibidores , Neoplasias Experimentales/inducido químicamente , Neoplasias Experimentales/patología , Proteínas de Unión al GTP ral/metabolismo
4.
Sci Rep ; 9(1): 5184, 2019 03 26.
Artículo en Inglés | MEDLINE | ID: mdl-30914727

RESUMEN

The underlying therapeutic mechanism of renal tubular epithelium repair of diabetic nephropathy (DN) by bone marrow-derived mesenchymal stem cells (BM-MSCs) has not been fully elucidated. Recently, mitochondria (Mt) transfer was reported as a novel action of BM-MSCs to rescue injured cells. We investigated Mt transfer from systemically administered BM-MSCs to renal proximal tubular epithelial cells (PTECs) in streptozotocin (STZ)-induced diabetic animals. BM-MSCs also transferred their Mt to impaired PTECs when co-cultured in vitro, which suppressed apoptosis of impaired PTECs. Additionally, BM-MSC-derived isolated Mt enhanced the expression of mitochondrial superoxide dismutase 2 and Bcl-2 expression and inhibited reactive oxygen species (ROS) production in vitro. Isolated Mt also inhibited nuclear translocation of PGC-1α and restored the expression of megalin and SGLT2 under high glucose condition (HG) in PTECs. Moreover, isolated Mt directly injected under the renal capsule of STZ rats improved the cellular morphology of STZ-PTECs, and the structure of the tubular basement membrane and brush border in vivo. This study is the first to show Mt transfer from systemically administered BM-MSCs to damaged PTECs in vivo, and the first to investigate mechanisms underlying the potential therapeutic effects of Mt transfer from BM-MSCs in DN.


Asunto(s)
Nefropatías Diabéticas/metabolismo , Células Epiteliales/metabolismo , Células Epiteliales/patología , Túbulos Renales Proximales/metabolismo , Células Madre Mesenquimatosas/metabolismo , Mitocondrias/metabolismo , Animales , Apoptosis , Línea Celular , Diabetes Mellitus Experimental/patología , Lectinas/metabolismo , Proteína 2 Relacionada con Receptor de Lipoproteína de Baja Densidad/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Células 3T3 NIH , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Ratas Sprague-Dawley , Especies Reactivas de Oxígeno/metabolismo , Transportador 2 de Sodio-Glucosa/metabolismo , Estreptozocina , Superóxido Dismutasa/metabolismo
5.
PLoS One ; 13(9): e0204252, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30240403

RESUMEN

Increasing evidence suggests that an enriched environment (EE) ameliorates cognitive impairment by promoting repair of brain damage. However, the mechanisms by which this occurs have not been determined. To address this issue, we investigated whether an EE enhanced the capability of endogenous bone marrow-derived mesenchymal stem/stromal cells (BM-MSCs) to prevent hippocampal damage due to diabetes by focusing on miRNA carried in BM-MSC-derived exosomes. In diabetic streptozotocin (STZ) rats housed in an EE (STZ/EE), cognitive impairment was significantly reduced, and both neuronal and astroglial damage in the hippocampus was alleviated compared with STZ rats housed in conventional cages (STZ/CC). BM-MSCs isolated from STZ/CC rats had functional and morphological abnormalities that were not detected in STZ/EE BM-MSCs. The miR-146a levels in exosomes in conditioned medium of cultured BM-MSCs and serum from STZ/CC rats were decreased compared with non-diabetic rats, and the level was restored in STZ/EE rats. Thus, the data suggest that increased levels of miR-146a in sera were derived from endogenous BM-MSCs in STZ/EE rats. To examine the possibility that increased miR-146a in serum may exert anti-inflammatory effects on astrocytes in diabetic rats, astrocytes transfected with miR-146a were stimulated with advanced glycation end products (AGEs) to mimic diabetic conditions. The expression of IRAK1, NF-κB, and tumor necrosis factor-α was significantly higher in AGE-stimulated astrocytes, and these factors were decreased in miR-146a-transfected astrocytes. These results suggested that EEs stimulate up-regulation of exosomal miR-146a secretion by endogenous BM-MSCs, which exerts anti-inflammatory effects on damaged astrocytes and prevents diabetes-induced cognitive impairment.


Asunto(s)
Disfunción Cognitiva/prevención & control , Diabetes Mellitus Experimental/patología , MicroARNs/metabolismo , Animales , Astrocitos/citología , Astrocitos/metabolismo , Células de la Médula Ósea/citología , Región CA1 Hipocampal/metabolismo , Región CA1 Hipocampal/patología , Células Cultivadas , Disfunción Cognitiva/etiología , Diabetes Mellitus Experimental/complicaciones , Exosomas/metabolismo , Productos Finales de Glicación Avanzada/farmacología , Quinasas Asociadas a Receptores de Interleucina-1/metabolismo , Masculino , Aprendizaje por Laberinto , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , MicroARNs/sangre , Estrés Oxidativo , Ratas , Transfección , Factor de Necrosis Tumoral alfa/metabolismo , Regulación hacia Arriba/efectos de los fármacos
6.
Sci Rep ; 8(1): 1161, 2018 01 18.
Artículo en Inglés | MEDLINE | ID: mdl-29348535

RESUMEN

Bone marrow-derived mesenchymal stem cells (BM-MSCs) are the most valuable source of autologous cells for transplantation and tissue regeneration to treat osteoporosis. Although BM-MSCs are the primary cells responsible for maintaining bone metabolism and homeostasis, their regenerative ability may be attenuated in postmenopausal osteoporosis patients. Therefore, we first examined potential abnormalities of BM-MSCs in an oestrogen-deficient rat model constructed by ovariectomy (OVX-MSCs). Cell proliferation, mobilisation, and regulation of osteoclasts were downregulated in OVX-MSCs. Moreover, therapeutic effects of OVX-MSCs were decreased in OVX rats. Accordingly, we developed a new activator for BM-MSCs using human umbilical cord extracts, Wharton's jelly extract supernatant (WJS), which improved cell proliferation, mobilisation and suppressive effects on activated osteoclasts in OVX-MSCs. Bone volume, RANK and TRACP expression of osteoclasts, as well as proinflammatory cytokine expression in bone tissues, were ameliorated by OVX-MSCs activated with WJS (OVX-MSCs-WJ) in OVX rats. Fusion and bone resorption activity of osteoclasts were suppressed in macrophage-induced and primary mouse bone marrow cell-induced osteoclasts via suppression of osteoclast-specific genes, such as Nfatc1, Clcn7, Atp6i and Dc-stamp, by co-culture with OVX-MSCs-WJ in vitro. In this study, we developed a new activator, WJS, which improved the functional abnormalities and therapeutic effects of BM-MSCs on postmenopausal osteoporosis.


Asunto(s)
Densidad Ósea/efectos de los fármacos , Mezclas Complejas/farmacología , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/citología , Osteoclastos/metabolismo , Osteoporosis/terapia , Cordón Umbilical/química , Animales , Biomarcadores/metabolismo , Células de la Médula Ósea/citología , Células de la Médula Ósea/metabolismo , Canales de Cloruro/genética , Canales de Cloruro/metabolismo , Técnicas de Cocultivo , Mezclas Complejas/aislamiento & purificación , Modelos Animales de Enfermedad , Femenino , Expresión Génica , Células Madre Mesenquimatosas/metabolismo , Ratones , Osteoclastos/citología , Osteoporosis/etiología , Osteoporosis/genética , Osteoporosis/patología , Ovariectomía/efectos adversos , Células RAW 264.7 , Ratas , Ratas Wistar , Receptor Activador del Factor Nuclear kappa-B/genética , Receptor Activador del Factor Nuclear kappa-B/metabolismo , Fosfatasa Ácida Tartratorresistente/genética , Fosfatasa Ácida Tartratorresistente/metabolismo , Tomografía Computarizada por Rayos X , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Gelatina de Wharton/química
7.
Sci Rep ; 7(1): 8484, 2017 08 16.
Artículo en Inglés | MEDLINE | ID: mdl-28814814

RESUMEN

Bone marrow-derived mesenchymal stem cells (BM-MSC) has been applied as the most valuable source of autologous cell transplantation for various diseases including diabetic complications. However, hyperglycemia may cause abnormalities in intrinsic BM-MSC which might lose sufficient therapeutic effects in diabetic patients. We demonstrated the functional abnormalities in BM-MSC derived from both type 1 and type 2 diabetes models in vitro, which resulted in loss of therapeutic effects in vivo in diabetic nephropathy (DN). Then, we developed a novel method to improve abnormalities in BM-MSC using human umbilical cord extracts, namely Wharton's jelly extract supernatant (WJs). WJs is a cocktail of growth factors, extracellular matrixes and exosomes, which ameliorates proliferative capacity, motility, mitochondrial degeneration, endoplasmic reticular functions and exosome secretions in both type 1 and type 2 diabetes-derived BM-MSC (DM-MSC). Exosomes contained in WJs were a key factor for this activation, which exerted similar effects to complete WJs. DM-MSC activated by WJs ameliorated renal injury in both type 1 and type 2 DN. In this study, we developed a novel activating method using WJs to significantly increase the therapeutic effect of BM-MSC, which may allow effective autologous cell transplantation.


Asunto(s)
Nefropatías Diabéticas/terapia , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/citología , Animales , Médula Ósea , Células de la Médula Ósea/citología , Células de la Médula Ósea/metabolismo , Diabetes Mellitus Tipo 1/metabolismo , Diabetes Mellitus Tipo 1/terapia , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/terapia , Nefropatías Diabéticas/patología , Modelos Animales de Enfermedad , Exosomas , Humanos , Células Madre Mesenquimatosas/metabolismo , Ratones Endogámicos C57BL , Ratas Endogámicas OLETF , Ratas Sprague-Dawley , Gelatina de Wharton/química
8.
Biochem Biophys Res Commun ; 485(2): 468-475, 2017 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-28192120

RESUMEN

BACKGROUND AND AIMS: Acute graft-versus-host disease (GVHD) is a major complication after allogeneic hematopoietic stem cell transplantation, which often targets gastrointestinal (GI) tract. Osteopontin (OPN) plays an important physiological role in the efficient development of Th1 immune responses and cell survival by inhibiting apoptosis. The role of OPN in acute GI-GVHD is poorly understood. In the present study, we investigated the role of OPN in donor T cells in the pathogenicity of acute GI-GVHD. METHODS: OPN knockout (KO) mice and C57BL/6 (B6) mice were used as donors, and (C57BL/6 × DBA/2) F1 (BDF1) mice were used as allograft recipients. Mice with acute GI-GVHD were divided into three groups: the control group (BDF1→BDF1), B6 group (B6→BDF1), and OPN-KO group (OPN-KO→BDF1). Bone marrow cells and spleen cells from donors were transplanted to lethally irradiated recipients. Clinical GVHD scores were assessed daily. Recipients were euthanized on day 7 after transplantation, and colons and small intestines were collected for various analyses. RESULTS: The clinical GVHD score in the OPN-KO group was significantly increased compared with the B6 and control groups. We observed a difference in the severity of colonic GVHD between the OPN-KO group and B6 group, but not small intestinal-GVHD between these groups. Interferon-γ, Tumor necrosis factor-α, Interleukin-17A, and Interleukin-18 gene expression in the OPN-KO group was differed between the colon and small intestine. Flow cytometric analysis revealed that the fluorescence intensity of splenic and colonic CD8 T cells expressing Fas Ligand was increased in the OPN-KO group compared with the B6 group. CONCLUSION: We demonstrated that the importance of OPN in T cells in the onset of acute GI-GVHD involves regulating apoptosis of the intestinal cell via the Fas-Fas Ligand pathway.


Asunto(s)
Apoptosis/inmunología , Células Epiteliales/inmunología , Enfermedades Gastrointestinales/inmunología , Enfermedad Injerto contra Huésped/inmunología , Osteopontina/inmunología , Enfermedad Aguda , Aloinjertos , Animales , Apoptosis/genética , Trasplante de Médula Ósea/efectos adversos , Trasplante de Médula Ósea/métodos , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Citocinas/genética , Citocinas/inmunología , Citocinas/metabolismo , Proteína Ligando Fas/genética , Proteína Ligando Fas/inmunología , Proteína Ligando Fas/metabolismo , Citometría de Flujo , Enfermedades Gastrointestinales/genética , Enfermedades Gastrointestinales/metabolismo , Expresión Génica/inmunología , Enfermedad Injerto contra Huésped/etiología , Enfermedad Injerto contra Huésped/genética , Mucosa Intestinal/inmunología , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patología , Recuento de Linfocitos , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Ratones Noqueados , Microscopía Fluorescente , Osteopontina/genética , Osteopontina/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Índice de Severidad de la Enfermedad
9.
Sci Rep ; 6: 34842, 2016 10 10.
Artículo en Inglés | MEDLINE | ID: mdl-27721418

RESUMEN

Bone marrow-derived mesenchymal stem cells (MSCs) have contributed to the improvement of diabetic nephropathy (DN); however, the actual mediator of this effect and its role has not been characterized thoroughly. We investigated the effects of MSC therapy on DN, focusing on the paracrine effect of renal trophic factors, including exosomes secreted by MSCs. MSCs and MSC-conditioned medium (MSC-CM) as renal trophic factors were administered in parallel to high-fat diet (HFD)-induced type 2 diabetic mice and streptozotocin (STZ)-induced insulin-deficient diabetic mice. Both therapies showed approximately equivalent curative effects, as each inhibited the exacerbation of albuminuria. They also suppressed the excessive infiltration of BMDCs into the kidney by regulating the expression of the adhesion molecule ICAM-1. Proinflammatory cytokine expression (e.g., TNF-α) and fibrosis in tubular interstitium were inhibited. TGF-ß1 expression was down-regulated and tight junction protein expression (e.g., ZO-1) was maintained, which sequentially suppressed the epithelial-to-mesenchymal transition of tubular epithelial cells (TECs). Exosomes purified from MSC-CM exerted an anti-apoptotic effect and protected tight junction structure in TECs. The increase of glomerular mesangium substrate was inhibited in HFD-diabetic mice. MSC therapy is a promising tool to prevent DN via the paracrine effect of renal trophic factors including exosomes due to its multifactorial action.


Asunto(s)
Medios de Cultivo Condicionados/farmacología , Nefropatías Diabéticas/patología , Nefropatías Diabéticas/terapia , Túbulos Renales/metabolismo , Trasplante de Células Madre Mesenquimatosas/métodos , Albuminuria , Animales , Células de la Médula Ósea/patología , Células Cultivadas , Citocinas/metabolismo , Diabetes Mellitus Experimental/complicaciones , Diabetes Mellitus Experimental/etiología , Diabetes Mellitus Tipo 2/complicaciones , Nefropatías Diabéticas/metabolismo , Dieta Alta en Grasa/efectos adversos , Transición Epitelial-Mesenquimal/efectos de los fármacos , Exosomas/metabolismo , Túbulos Renales/patología , Masculino , Ratones Endogámicos C57BL , Ratones Transgénicos , Ratas Endogámicas Lew
10.
Sci Rep ; 6: 24805, 2016 04 22.
Artículo en Inglés | MEDLINE | ID: mdl-27102354

RESUMEN

The incidence of dementia is higher in diabetic patients, but no effective treatment has been developed. This study showed that rat bone marrow mesenchymal stem cells (BM-MSCs) can improve the cognitive impairments of STZ-diabetic mice by repairing damaged neurons and astrocytes. The Morris water maze test demonstrated that cognitive impairments induced by diabetes were significantly improved by intravenous injection of BM-MSCs. In the CA1 region of the hippocampus, degeneration of neurons and astrocytes, as well as synaptic loss, were prominent in diabetes, and BM-MSC treatment successfully normalized them. Since a limited number of donor BM-MSCs was observed in the brain parenchyma, we hypothesized that humoral factors, especially exosomes released from BM-MSCs, act on damaged neurons and astrocytes. To investigate the effectiveness of exosomes for treatment of diabetes-induced cognitive impairment, exosomes were purified from the culture media and injected intracerebroventricularly into diabetic mice. Recovery of cognitive impairment and histological abnormalities similar to that seen with BM-MSC injection was found following exosome treatment. Use of fluorescence-labeled exosomes demonstrated that injected exosomes were internalized into astrocytes and neurons; these subsequently reversed the dysfunction. The present results indicate that exosomes derived from BM-MSCs might be a promising therapeutic tool for diabetes-induced cognitive impairment.


Asunto(s)
Astrocitos/fisiología , Disfunción Cognitiva/terapia , Complicaciones de la Diabetes/terapia , Exosomas/metabolismo , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/metabolismo , Neuronas/fisiología , Administración Intravenosa , Animales , Región CA1 Hipocampal/patología , Modelos Animales de Enfermedad , Ratones , Ratas , Resultado del Tratamiento
11.
PLoS One ; 10(9): e0137801, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26375822

RESUMEN

BACKGROUND: The common disease-common variant hypothesis is insufficient to explain the complexities of Crohn's disease (CD) genetics; therefore, rare variants are expected to be important in the disease. We explored rare variants associated with susceptibility to CD in Japanese individuals by personal genomic analysis. METHODS: Two-step analyses were performed. The first step was a trio analysis with whole-exome sequence (WES) analysis and the second was a follow-up case-control association study. The WES analysis pipeline comprised Burrows-Wheeler Aligner, Picard, Genome Analysis Toolkit, and SAMTOOLS. Single nucleotide variants (SNVs)/indels were annotated and filtered by using programs implemented in ANNOVAR in combination with identity-by-descent (IBD), subsequently were subjected to the linkage based, and de novo based strategies. Finally, we conducted an association study that included 176 unrelated subjects with CD and 358 healthy control subjects. RESULTS: In family members, 234,067-297,523 SNVs/indels were detected and they were educed to 106-146 by annotation based filtering. Fifty-four CD variants common to both individuals of the affected sib pair were identified. The linkage based strategy detected five candidate variants whereas the de novo based strategy identified no variants. Consequently, five candidates were analyzed in the case-control association study. CD showed a significant association with one variant in exon 4 of IL23R, G149R [rs76418789, P = 3.9E-5, odds ratio (OR) 0.21, 95% confidence interval (CI) 0.09-0.47 for the dominant model (AA + AG versus GG), and P = 7.3E-5, OR 0.21, 95% CI 0.10-0.48 for AG versus GG, and P = 7.2E-5, OR 0.23, 95% CI 0.10-0.50 for the allele model]. CONCLUSIONS: The present study, using personal genomics analysis of a small CD pedigree, is the first to show that the low-frequency non-synonymous variant of IL23R, rs76418789, protects against CD development in Japanese subjects.


Asunto(s)
Enfermedad de Crohn/genética , Predisposición Genética a la Enfermedad , Polimorfismo de Nucleótido Simple/genética , Medicina de Precisión , Receptores de Interleucina/genética , Adulto , Anciano , Estudios de Casos y Controles , Enfermedad de Crohn/patología , Femenino , Genómica , Humanos , Masculino , Persona de Mediana Edad , Linaje , Adulto Joven
12.
J Gastroenterol ; 50(9): 962-74, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25680886

RESUMEN

BACKGROUND: The role of mesenchymal stem/stromal cells (MSCs) in tumorigenesis remains controversial. This study aimed to determine whether heterotypic interactions between MSCs and colon cancer cells can supply contextual signals towards tumor progression. METHODS: Xenografts consisting of co-implanted human colorectal cancer cells with rat MSCs in immunodeficient mice were evaluated by tumor progression, angiogenic profiles, and MSC fate. Furthermore, we investigated how MSCs function as a cancer cell niche by co-culture experiments in vitro. RESULTS: Tumor growth progressed in two ways, either independent of or dependent on MSCs. Such cell line-specific dependency could not be explained by host immune competency. COLO 320 xenograft angiogenesis was MSC-dependent, but less dependent on vascular endothelial growth factor (VEGF), whereas HT-29 angiogenesis was not MSC-dependent, but was VEGF-dependent. MSCs and COLO 320 cells established a functional positive feedback loop that triggered formation of a cancer cell niche, leading to AKT activation. Subsequently, MSCs differentiated into pericytes that enhanced angiogenesis as a perivascular niche. In contrast, the MSC niche conferred an anti-proliferative property to HT-29 cells, through mesenchymal-epithelial transition resulting in p38 activation. CONCLUSIONS: In conclusion, MSCs demonstrate pleiotropic capabilities as a cancer cell or perivascular niche to modulate colorectal cancer cell fate in a cell line-dependent manner in a xenogeneic context.


Asunto(s)
Neoplasias Colorrectales/patología , Células Madre Mesenquimatosas/patología , Neovascularización Patológica/patología , Transducción de Señal/fisiología , Nicho de Células Madre/fisiología , Animales , Apoptosis , Western Blotting , Línea Celular Tumoral , Proliferación Celular , Técnicas de Cocultivo , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Xenoinjertos , Humanos , Hibridación Fluorescente in Situ , Ratones , Análisis por Micromatrices , Procesos Neoplásicos , Ratas , Reacción en Cadena en Tiempo Real de la Polimerasa , Trasplante Heterólogo
13.
J Gastroenterol ; 50(3): 280-6, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25618180

RESUMEN

Inflammatory bowel disease (IBD) could be curable by "immune rest" and correction of the genetic predisposition inherent in allogeneic hematopoietic stem cell transplantation. However, balancing risks against benefits remains challenging. The application of mesenchymal stem cells (MSCs) serving as a site-regulated "drugstore" is a recent concept, which suggests the possibility of an alternative treatment for many intractable diseases such as IBD. Depending on the required function of MSC, such as a cell provider, immune moderator, and/or trophic resource, MSC therapy should be optimized to maximize its therapeutic benefit. Therapeutic effects do not always require full engraftment of MSCs. Therefore, optimization of pleiotropic gut trophic factors produced by MSCs, which favoring not only regulating immune responses but also promoting tissue repair, must directly enhance new drug discoveries for treatment of IBD. Stem cell biology holds great promise for a new era of cell-based therapy, sparking considerable interest among scientists, clinicians, and patients. However, the translational arm of stem cell science remains in a relatively primitive state. Although several clinical studies using MSCs have been initiated, early results suggest several inherent problems. In each study, optimization of MSC therapy appears to be the most urgent problem, and can be resolved only by scientifically unveiling the mechanisms of therapeutic action. In the present review, the authors outline how such information would facilitate the critical steps in the paradigm shift from basic research on stem cell biology to clinical practice of regenerative medicine for conquering IBD in the near future.


Asunto(s)
Enfermedades Inflamatorias del Intestino/terapia , Trasplante de Células Madre/métodos , Trasplante de Células Madre Hematopoyéticas/métodos , Humanos , Enfermedades Inflamatorias del Intestino/inmunología , Trasplante de Células Madre Mesenquimatosas/métodos
14.
Pain ; 155(9): 1762-1772, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24907405

RESUMEN

Chronic neuropathic pain causes abnormal sensitivities such as hyperalgesia and allodynia, and emotional abnormalities such as anxiety and depression. Although spinal cord microglia are involved in abnormal sensitivity to neuropathic pain, no previous studies have examined the mechanism of neuropathic pain-induced anxiety. Here, we examined the involvement of bone marrow (BM)-derived microglia aggregated in the amygdalae of mice with chronic neuropathic pain in the development of anxiety-like behavior. We prepared partial sciatic nerve ligations (PSNL) in mice that received bone marrow transplantation from green fluorescent protein (GFP)-Tg mice after irradiation with head protection, and examined GFP-positive microglia in the central nuclei of the amygdalae (CeA). On day 28 after PSNL, BM-derived microglia aggregated in the CeA concurrent with anxiety-like behavior. BM-derived microglia in the CeA highly expressed interleukin (IL)-1ß and C-C chemokine receptor type 2 (CCR2). In addition, neurons in the CeA highly expressed monocyte chemotactic protein-1 (MCP-1), a ligand for CCR2, in PSNL-treated mice compared to sham-operated mice, suggesting that the MCP-1/CCR2 axis is involved in the recruitment of BM-derived microglia. Oral administration of a CCR2 antagonist decreased the number of BM-derived microglia in the CeA, and successfully reversed the anxiety-like behavior and hypersensitivity to mechanical stimuli in PSNL-treated mice. Microinjections of an IL-1ß receptor antagonist directly into the CeA successfully reversed the anxiety-like behavior in the PSNL-treated mice even though the neuropathic pain persisted. These results suggest that the recruitment of BM-derived microglia to the CeA via the MCP-1/CCR2 axis and neuron-microglia interactions might be important in the pathogenesis of neuropathic pain-induced anxiety.


Asunto(s)
Amígdala del Cerebelo/metabolismo , Ansiedad/etiología , Conducta Animal/fisiología , Médula Ósea/metabolismo , Microglía/metabolismo , Neuralgia/complicaciones , Nervio Ciático/lesiones , Animales , Ansiedad/metabolismo , Quimiocina CCL2/metabolismo , Masculino , Ratones , Ratones Transgénicos , Neuralgia/metabolismo , Neuronas/metabolismo
15.
Stem Cells ; 32(4): 913-25, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24715689

RESUMEN

The role of mesenchymal stem cells (MSCs) in tumorigenesis remains controversial. Therefore, our goal was to determine whether exogenous MSCs possess intrinsic antineoplastic or proneoplastic properties in azoxymethane (AOM)-induced carcinogenesis. Three in vivo models were studied: an AOM/dextran sulfate sodium colitis-associated carcinoma model, an aberrant crypt foci model, and a model to assess the acute apoptotic response of a genotoxic carcinogen (AARGC). We also performed in vitro coculture experiments. As a result, we found that MSCs partially canceled AOM-induced tumor initiation but not tumor promotion. Moreover, MSCs inhibited the AARGC in colonic epithelial cells because of the removal of O(6)-methylguanine (O(6) MeG) adducts through O(6) MeG-DNA methyltransferase activation. Furthermore, MSCs broadly affected the cell-cycle machinery, potentially leading to G1 arrest in vivo. Coculture of IEC-6 rat intestinal cells with MSCs not only arrested the cell cycle at the G1 phase, but also induced apoptosis. The anti-carcinogenetic properties of MSCs in vitro required transforming growth factor (TGF)-ß signaling because such properties were completely abrogated by absorption of TGF-ß under indirect coculture conditions. MSCs inhibited AOM-induced tumor initiation by preventing the initiating cells from sustaining DNA insults and subsequently inducing G1 arrest in the initiated cells that escaped from the AARGC. Furthermore, tumor initiation perturbed by MSCs might potentially dysregulate WNT and TGF-ß-Smad signaling pathways in subsequent tumorigenesis. Obtaining a better understanding of MSC functions in colon carcinogenesis is essential before commencing the broader clinical application of promising MSC-based therapies for cancer-prone patients with inflammatory bowel disease.


Asunto(s)
Azoximetano/toxicidad , Carcinógenos/toxicidad , Neoplasias del Colon/metabolismo , Células Madre Mesenquimatosas/metabolismo , Neoplasias Experimentales/metabolismo , Animales , Células Cultivadas , Técnicas de Cocultivo , Neoplasias del Colon/inducido químicamente , Neoplasias del Colon/patología , Puntos de Control de la Fase G1 del Ciclo Celular/efectos de los fármacos , Humanos , Células Madre Mesenquimatosas/patología , Neoplasias Experimentales/inducido químicamente , Neoplasias Experimentales/patología , Ratas , Ratas Endogámicas Lew , Factor de Crecimiento Transformador beta/metabolismo , Vía de Señalización Wnt/efectos de los fármacos
16.
J Gastroenterol ; 49(8): 1217-30, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23942620

RESUMEN

BACKGROUND: There are substantial differences in inflammatory bowel disease (IBD) genetics depending on the populations examined. We aimed to identify Japanese population-specific or true culprit susceptibility genes through a meta-analysis of past genetic studies of Japanese IBD. METHODS: For this study, we reviewed 2,703 articles. The review process consisted of three screening stages: we initially searched for relevant studies and then relevant single nucleotide polymorphisms (SNPs). Finally, we adjusted them for the meta-analysis. To maximize our chances of analysis, we introduced proxy SNPs during the first stage. To minimize publication bias, no significant SNPs and solitary SNPs without pairs were combined to be reconsidered during the third stage. Additionally, two SNPs were newly genotyped. Finally, we conducted a meta-analysis of 37 published studies in 50 SNPs located at 22 loci corresponding to the total number of 4,853 Crohn's disease (CD), 5,612 ulcerative colitis (UC) patients, and 14,239 healthy controls. RESULTS: We confirmed that the NKX2-3 polymorphism is associated with common susceptibility to IBD and that HLA-DRB1*0450 alleles increase susceptibility to CD but reduce risk for UC while HLA-DRB1*1502 alleles increase susceptibility to UC but reduce CD risk. Moreover, we found individual disease risk loci: TNFSF15 and TNFα to CD and HLA-B*5201, and NFKBIL1 to UC. The genetic risk of HLA was substantially high (odds ratios ranged from 1.54 to 2.69) while that of common susceptibility loci to IBD was modest (odds ratio ranged from 1.13 to 1.24). CONCLUSIONS: Results indicate that Japanese IBD susceptibility loci identified by the meta-analysis are closely associated with the HLA regions.


Asunto(s)
Colitis Ulcerosa/genética , Enfermedad de Crohn/genética , Predisposición Genética a la Enfermedad , Alelos , Pueblo Asiatico/genética , Estudios de Casos y Controles , Sitios Genéticos/genética , Humanos , Japón , Polimorfismo de Nucleótido Simple
17.
J Gastroenterol ; 49(2): 270-82, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24217964

RESUMEN

BACKGROUND: Although mounting evidence implicates mesenchymal stem cells (MSCs) in intestinal tissue repair, controversy remains regarding the engraftment, proliferation, and differentiation for repopulating MSCs in recipient tissues. Therefore, we investigated the paracrine and/or endocrine role of MSCs in experimental colitis. METHODS: We analyzed the therapeutic effects of MSC-conditioned medium (MSC-CM) on dextran sulfate sodium (DSS)- or 2,4,6-trinitrobenzenesulfonic acid (TNBS)-induced colitis. We investigated the effects of MSC-CM on the epithelial cell viability, mobility, cell cycle, and cytokine production in ex vivo lamina propria/mesenteric lymphocytes, a macrophage cell line, and the mixed lymphocyte reaction. An optimal regimen against colitis was explored. The contents of MSC-CM were analyzed using a WNT signaling pathway polymerase chain reaction array, an inflammatory cytokines antibody array, and liquid chromatography-tandem mass spectrometry analysis. RESULTS: Independent of the systemic administration route, MSC-CM concentrates were effective for the inductive phase of TNBS-induced colitis and for the recovery phase of DSS-induced colitis. Hypoxia appeared to be one of the optimal preconditioning factors assessed by cell motility and viability through activating the PI3K-Akt pathway in rat small intestine epithelial cells, IEC-6. Thus, Hypoxia had profound effects on the contents of MSC-CM, which comprised pleiotropic gut trophic factors involved in each wound healing process, including the anti-inflammatory, proliferative, and tissue remodeling phases. CONCLUSIONS: Identification and optimization of potential gut trophic factors in MSC-CM is urgently needed to form the basis for new drug discovery and for optimizing cell-based therapies for inflammatory bowel disease.


Asunto(s)
Colitis/tratamiento farmacológico , Medios de Cultivo Condicionados/farmacología , Citocinas/metabolismo , Células Madre Mesenquimatosas/metabolismo , Animales , Apoptosis , Células CACO-2 , Ciclo Celular/efectos de los fármacos , Hipoxia de la Célula , Movimiento Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Quimiocina CCL2/análisis , Colitis/inducido químicamente , Colitis/patología , Medios de Cultivo Condicionados/química , Citocinas/análisis , Sulfato de Dextran , Células Epiteliales/citología , Células Epiteliales/efectos de los fármacos , Células Epiteliales/fisiología , Humanos , Péptidos y Proteínas de Señalización Intercelular/genética , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/patología , Linfocitos/efectos de los fármacos , Linfocitos/metabolismo , Macrófagos/citología , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Proteínas de la Membrana/genética , Ratones , Análisis de Secuencia por Matrices de Oligonucleótidos , Análisis por Matrices de Proteínas , Proteínas Proto-Oncogénicas/genética , Ratas , Ratas Endogámicas Lew , Ratas Wistar , Ácido Trinitrobencenosulfónico , Regulación hacia Arriba/efectos de los fármacos , Factor A de Crecimiento Endotelial Vascular/análisis , Proteínas Wnt/genética , Vía de Señalización Wnt/genética
18.
Hepatology ; 59(5): 1816-29, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24375439

RESUMEN

UNLABELLED: Although mesenchymal stem cells (MSCs) have been implicated in hepatic injury, the mechanism through which they contribute to diabetic liver disease has not been clarified. In this study, we investigated the effects of MSC therapy on diabetic liver damage with a focus on the role of bone-marrow-derived cells (BMDCs), which infiltrate the liver, and elucidated the mechanism mediating this process. Rat bone-marrow (BM)-derived MSCs were administered to high-fat diet (HFD)-induced type 2 diabetic mice and streptozotocin (STZ)-induced insulin-deficient diabetic mice. MSC-conditioned medium (MSC-CM) was also administered to examine the trophic effects of MSCs on liver damage. Therapeutic effects of MSCs were analyzed by assessing serum liver enzyme levels and histological findings. Kinetic and molecular profiles of BMDCs in the liver were evaluated using BM-chimeric mice. Curative effects of MSC and MSC-CM therapies were similar because both ameliorated the aggravation of aspartate aminotransferase and alanine aminotransferase at 8 weeks of treatment, despite persistent hyperlipidemia and hyperinsulinemia in HFD-diabetic mice and persistent hyperglycemia in STZ-diabetic mice. Furthermore, both therapies suppressed the abnormal infiltration of BMDCs into the liver, reversed excessive expression of proinflammatory cytokines in parenchymal cells, and regulated proliferation and survival signaling in the liver in both HFD- and STZ-diabetic mice. In addition to inducing hepatocyte regeneration in STZ-diabetic mice, both therapies also prevented excessive lipid accumulation and apoptosis of hepatocytes and reversed insulin resistance (IR) in HFD-diabetic mice. CONCLUSION: MSC therapy is a powerful tool for repairing diabetic hepatocyte damage by inhibiting inflammatory reactions induced by BMDCs and IR. These effects are likely the result of humoral factors derived from MSCs.


Asunto(s)
Células de la Médula Ósea/fisiología , Diabetes Mellitus Experimental/terapia , Hígado/patología , Trasplante de Células Madre Mesenquimatosas , Animales , Apoptosis , Comunicación Celular , Diferenciación Celular , Linaje de la Célula , Movimiento Celular , Citocinas/análisis , Diabetes Mellitus Experimental/patología , Dieta Alta en Grasa , Resistencia a la Insulina , Hígado/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratas , Estreptozocina
19.
PLoS One ; 8(11): e81744, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24303068

RESUMEN

BACKGROUND: Microglia of the central nervous system act as sentinels and rapidly react to infection or inflammation. The pathophysiological role of bone marrow-derived microglia is of particular interest because they affect neurodegenerative disorders and neuropathic pain. The hypothesis of the current study is that chronic psychological stress (chronic PS) induces the infiltration of bone marrow-derived microglia into hypothalamus by means of chemokine axes in brain and bone marrow. METHODS AND FINDINGS: Here we show that bone marrow-derived microglia specifically infiltrate the paraventricular nucleus (PVN) of mice that received chronic PS. Bone marrow derived-microglia are CX3CR1(low)CCR2(+)CXCR4(high), as distinct from CX3CR1(high)CCR2(-)CXCR4(low) resident microglia, and express higher levels of interleukin-1ß (IL-1ß) but lower levels of tumor necrosis factor-α (TNF-α). Chronic PS stimulates the expression of monocyte chemotactic protein-1 (MCP-1) in PVN neurons, reduces stromal cell-derived factor-1 (SDF-1) in the bone marrow and increases the frequency of CXCR4(+) monocytes in peripheral circulation. And then a chemokine (C-C motif) receptor 2 (CCR2) or a ß3-adrenoceptor blockade prevents infiltration of bone marrow-derived microglia in the PVN. CONCLUSION: Chronic PS induces the infiltration of bone marrow-derived microglia into PVN, and it is conceivable that the MCP-1/CCR2 axis in PVN and the SDF-1/CXCR4 axis in bone marrow are involved in this mechanism.


Asunto(s)
Células de la Médula Ósea/inmunología , Células de la Médula Ósea/patología , Microglía/inmunología , Microglía/patología , Núcleo Hipotalámico Paraventricular/patología , Estrés Psicológico/inmunología , Estrés Psicológico/patología , Animales , Células de la Médula Ósea/metabolismo , Quimiocina CCL2/metabolismo , Quimiocina CXCL12/metabolismo , Quimiotaxis/inmunología , Hipotálamo/inmunología , Hipotálamo/metabolismo , Hipotálamo/patología , Interleucina-1beta/metabolismo , Masculino , Ratones , Microglía/metabolismo , Neuronas/metabolismo , Núcleo Hipotalámico Paraventricular/inmunología , Núcleo Hipotalámico Paraventricular/metabolismo , ARN Mensajero/genética , Receptores CCR2/metabolismo , Receptores CXCR4/metabolismo , Receptores de Interleucina-1/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Estrés Psicológico/genética , Estrés Psicológico/metabolismo
20.
Am J Physiol Cell Physiol ; 305(7): C693-703, 2013 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-23885062

RESUMEN

Hematopoietic stem cells (HSCs) are maintained, and their division/proliferation and quiescence are regulated in the microenvironments, niches, in the bone marrow. Although diabetes is known to induce abnormalities in HSC mobilization and proliferation through chemokine and chemokine receptors, little is known about the interaction between long-term HSCs (LT-HSCs) and osteopontin-positive (OPN) cells in endosteal niche. To examine this interaction, LT-HSCs and OPN cells were isolated from streptozotocin-induced diabetic and nondiabetic mice. In diabetic mice, we observed a reduction in the number of LT-HSCs and OPN cells and impaired expression of Tie2, ß-catenin, and N-cadherin on LT-HSCs and ß1-integrin, ß-catenin, angiopoietin-1, and CXCL12 on OPN cells. In an in vitro coculture system, LT-HSCs isolated from nondiabetic mice exposed to diabetic OPN cells showed abnormal mRNA expression levels of Tie2 and N-cadherin. Conversely, in LT-HSCs derived from diabetic mice exposed to nondiabetic OPN cells, the decreased mRNA expressions of Tie2, ß-catenin, and N-cadherin were restored to normal levels. The effects of diabetic or nondiabetic OPN cells on LT-HSCs shown in this coculture system were confirmed by the coinjection of LT-HSCs and OPN cells into bone marrow of irradiated nondiabetic mice. Our results provide new insight into the treatment of diabetes-induced LT-HSC abnormalities and suggest that the replacement of OPN cells may represent a novel treatment strategy.


Asunto(s)
Comunicación Celular , Diabetes Mellitus Experimental/metabolismo , Células Madre Hematopoyéticas/metabolismo , Osteoblastos/metabolismo , Osteopontina/metabolismo , Nicho de Células Madre , Animales , Biomarcadores/metabolismo , Trasplante de Médula Ósea , Células Cultivadas , Técnicas de Cocultivo , Diabetes Mellitus Experimental/patología , Regulación de la Expresión Génica , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Osteoblastos/patología , Osteopontina/genética , ARN Mensajero/metabolismo , Transducción de Señal , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA