Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
1.
Biochem Biophys Res Commun ; 696: 149469, 2024 Feb 12.
Artículo en Inglés | MEDLINE | ID: mdl-38194806

RESUMEN

Accumulating data suggest that ribosomal protein S6 kinase 1 (S6K1), an effector in the mammalian target of rapamycin (mTOR) pathway, plays pleiotropic roles in tumor progression. However, to date, while the tumorigenic function of S6K1 in tumor cells has been well elucidated, its role in the tumor stroma remains poorly understood. We recently showed that S6K1 mediates vascular endothelial growth factor A (VEGF-A) production in macrophages, thereby supporting tumor angiogenesis and growth. As macrophage-derived VEGF-A is crucial for both tumor cell intravasation and extravasation across the vascular endothelium, our previous findings suggest that stromal S6K1 signaling is required for tumor metastatic spread. Therefore, we aimed to determine the impact of host S6K1 depletion on tumor metastasis using a murine model of pulmonary metastasis (S6k1-/- mice implanted with B16F10 melanoma). The ablation of S6K1 in the host microenvironment significantly reduced the metastasized B16F10 melanoma cells on the lung surface in both spontaneous and intravenous lung metastasis mouse models without affecting the incidence of metastasis to distant lymph nodes. In addition, stromal S6K1 loss decreased the number of tumor cells circulating in the peripheral blood of mice bearing B16F10 xenografts without affecting the vascular leakage induced by VEGF-A in vivo. These observations demonstrate that S6K1 signaling in host cells other than endothelial cells is required to modulate the host microenvironment to facilitate the metastatic spread of tumors via blood circulation, thus revealing its novel role in the tumor stroma during tumor progression.


Asunto(s)
Neoplasias Pulmonares , Melanoma , Proteínas Quinasas S6 Ribosómicas 90-kDa , Animales , Humanos , Ratones , Células Endoteliales/metabolismo , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Mamíferos/metabolismo , Melanoma/metabolismo , Melanoma/patología , Transducción de Señal , Microambiente Tumoral , Factor A de Crecimiento Endotelial Vascular/metabolismo , Proteínas Quinasas S6 Ribosómicas 90-kDa/metabolismo
2.
Food Chem Toxicol ; 184: 114387, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38123059

RESUMEN

Diisobutyl phthalate (DiBP) is a commonly used plasticizer in manufacturing consumer and industrial products to improve flexibility and durability. Despite of the numerous studies, however, the direct mechanism underlying the male reproductive damage of DiBP is poorly understood. In this study, we investigated the male germ cell toxicity of DiBP using GC-1 spermatogonia (spg) cells. Our results indicated that DiBP exposure causes oxidative stress and apoptosis in GC-1 spg cells. In addition, DiBP-derived autophagy activation and down-regulation of phosphoinositide 3-kinase (PI3K)-AKT and extracellular signal-regulated kinase (ERK) pathways further inhibited GC-1 spg cell proliferation, indicating that DiBP can instigate male germ cell toxicity by targeting several pathways. Importantly, a combined treatment of parthenolide, N-acetylcysteine, and 3-methyladenine significantly reduced DiBP-induced male germ cell toxicity and restored proliferation. Taken together, the results of this study can provide valuable information to the existing literature by enhancing the understanding of single phthalate DiBP-derived male germ cell toxicity and the therapeutic interventions that can mitigate DiBP damage.


Asunto(s)
Acetatos , Dibutil Ftalato , Fenoles , Fosfatidilinositol 3-Quinasas , Humanos , Masculino , Dibutil Ftalato/toxicidad , Células Germinativas
3.
J Vet Sci ; 22(3): e25, 2021 May.
Artículo en Inglés | MEDLINE | ID: mdl-33908202

RESUMEN

BACKGROUND: Malignant lymphoma is the most common hematopoietic malignancy in dogs, and relapse is frequently seen despite aggressive initial treatment. In order for the treatment of these recurrent lymphomas in dogs to be effective, it is important to choose a personalized and sensitive anticancer agent. To provide a reliable tool for drug development and for personalized cancer therapy, it is critical to maintain key characteristics of the original tumor. OBJECTIVES: In this study, we established a model of hybrid tumor/stromal spheroids and investigated the association between canine lymphoma cell line (GL-1) and canine lymph node (LN)-derived stromal cells (SCs). METHODS: A hybrid spheroid model consisting of GL-1 cells and LN-derived SC was created using ultra low attachment plate. The relationship between SCs and tumor cells (TCs) was investigated using a coculture system. RESULTS: TCs cocultured with SCs were found to have significantly upregulated multidrug resistance genes, such as P-qp, MRP1, and BCRP, compared with TC monocultures. Additionally, it was revealed that coculture with SCs reduced doxorubicin-induced apoptosis and G2/M cell cycle arrest of GL-1 cells. CONCLUSIONS: SCs upregulated multidrug resistance genes in TCs and influenced apoptosis and the cell cycle of TCs in the presence of anticancer drugs. This study revealed that understanding the interaction between the tumor microenvironment and TCs is essential in designing experimental approaches to personalized medicine and to predict the effect of drugs.


Asunto(s)
Técnicas de Cocultivo/veterinaria , Enfermedades de los Perros/tratamiento farmacológico , Descubrimiento de Drogas/métodos , Medicina de Precisión/veterinaria , Animales , Línea Celular Tumoral/efectos de los fármacos , Técnicas de Cocultivo/métodos , Modelos Animales de Enfermedad , Perros , Linfoma/tratamiento farmacológico , Medicina de Precisión/métodos , Esferoides Celulares/efectos de los fármacos , Células del Estroma/efectos de los fármacos
4.
Sensors (Basel) ; 20(15)2020 Jul 22.
Artículo en Inglés | MEDLINE | ID: mdl-32707843

RESUMEN

The early detection of tumors improves chances of decreased morbidity and prolonged survival. Serum biomarkers are convenient to use and have several advantages over other approaches, such as accuracy and straightforward protocols. Reliable biomarkers from easily accessible sources are warranted for the development of cost-effective assays for routine screening, particularly in veterinary medicine. Extracellular c-AMP-dependent protein kinase A (ECPKA) is a cytosolic leakage enzyme. The diagnostic accuracy of detecting autoantibodies against ECPKA was found to be higher than that of ECPKA activity from enzymatic assays, which use a complicated method. Here, we investigated the diagnostic significance of measuring serum ECPKA autoantibody levels using an in-house kit (AniScan cancer detection kit; Biattic, Anyang, Korea). We used sera from 550 dogs, including healthy dogs and those with malignant and benign tumors. Serum ECPKA and immunoglobulin G were determined using the AniScan cancer detection kit. ECPKA autoantibody levels were significantly higher (p < 0.01) in malignant tumors than in benign tumors, non-tumor diseases, and healthy controls. On the basis of sensitivity and specificity values, AniScan ECPKA is a rapid and easy-to-use assay that can be applied to screen malignant tumors from benign tumors or other diseases in dogs.


Asunto(s)
Biomarcadores de Tumor , Proteínas Quinasas Dependientes de AMP Cíclico , Enfermedades de los Perros , Neoplasias , Animales , AMP Cíclico , Enfermedades de los Perros/diagnóstico , Perros , Femenino , Masculino , Neoplasias/diagnóstico , República de Corea
5.
Sci Rep ; 10(1): 7585, 2020 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-32350315

RESUMEN

An amendment to this paper has been published and can be accessed via a link at the top of the paper.

6.
Transl Oncol ; 13(4): 100767, 2020 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-32251993

RESUMEN

Two isoforms of the 70-kDa ribosomal protein S6 kinase, S6K1 and S6K2, have been identified and are considered key downstream effectors of the mTOR signaling pathway, which is involved in tumor growth and progression. However, their biological roles in the tumor microenvironment are poorly understood. In this study, utilizing tumor xenograft models in S6k1-/- and S6k2-/- mice, we show that loss of S6K1 but not S6K2 in the tumor stroma suppresses tumor growth, accompanied by attenuated tumor angiogenesis. We found that while S6K1 depletion had no effect on the proangiogenic phenotype of endothelial cells, the growth and angiogenesis of tumor xenografts were significantly reduced in wild-type mice upon reconstitution with S6K1-deficient bone marrow cells. Furthermore, upon S6K1 loss, induction of both mRNA and protein levels of Hif-1α and those of the downstream target, Vegf, was compromised in bone marrow-derived macrophages stimulated with lactate. These findings indicate that S6K1 but not S6K2 contributes to establishing a microenvironment that favors tumor growth through mediating angiogenesis, and suggest that attenuated tumor angiogenesis upon loss of S6K1 in the tumor stroma is, at least in part, attributable to impaired upregulation of Vegf in tumor-associated macrophages.

7.
Sci Rep ; 10(1): 2115, 2020 02 07.
Artículo en Inglés | MEDLINE | ID: mdl-32034203

RESUMEN

The inflammatory bowel diseases (IBD) are characterized by relapsing inflammation and immune activation diseases of the gastrointestinal tract. Extracellular vesicles, which elicit similar biological activity to the stem cell themselves, have been used experimentally to treat dextran sulfate sodium (DSS)-induced colitis in murine models though immunosuppressive potential. In this study, we investigated whether the Extracellular vesicles (EVs) obtained by stimulating inflammatory cytokine on canine adipose mesenchymal stem cells (cASC) improved anti-inflammatory and/or immunosuppressive potential of EVs, and/or their ability to alleviate inflammation in colitis. We also explored the correlation between immune cells and the inflammatory repressive effect of primed EVs. Pro-inflammatory cytokines such as TNF-α and IFN-γ increased immunosuppressive protein such as HGF, TSG-6, PGE2 and TGF-ß in EVs. Moreover, the anti-inflammatory effect of EVs was improved through pretreatment with inflammatory cytokines. Importantly, EVs obtained from primed stem cells effectively induced macrophage polarization toward an anti-inflammatory M2 phenotype and suppressed activated immunity by enhancing regulatory T cells in inflamed colon in mice. Our results provide a new and effective therapy for the EVs obtained from ASC stimulated with TNF-α and IFN-γ against not only IBD, but also immune-mediated disease.

8.
PLoS One ; 15(2): e0220756, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32040478

RESUMEN

Adipose tissue derived mesenchymal stem/stromal cell (ASC)-derived extracellular vesicles (EV) have been reported to be beneficial against dextran sulfate sodium (DSS)-induced colitis in mice. However, the underlying mechanisms have not been fully elucidated. We hypothesize that the tumor necrosis factor-α-stimulated gene/protein 6 (TSG-6) in EVs is a key factor influencing the alleviation of colitis symptoms. DSS-induced colitis mice (C57BL/6, male, Naïve = 6, Sham = 8, PBS = 8 EV = 8, CTL-EV = 8, TSG-6 depleted EV = 8) were intraperitoneally administered EVs (100 ug/mice) on day 1, 3, and 5; colon tissues were collected on day 10 for histopathological, RT-qPCR, western blot and immunofluorescence analyses. In mice injected with EV, inflammation was alleviated. Indeed, EVs regulated the levels of pro- and anti-inflammatory cytokines, such as TNF-α, IL-1ß, IFN-γ, IL-6, and IL-10 in inflamed colons. However, when injected with TSG-6 depleted EV, the degree of inflammatory relief was reduced. Furthermore, TSG-6 in EVs plays a key role in increasing regulatory T cells (Tregs) and polarizing macrophage from M1 to M2 in the colon. In conclusion, this study shows that TSG-6 in EVs is a major factor in the relief of DSS-induced colitis, by increasing the number of Tregs and macrophage polarization from M1 to M2 in the colon.


Asunto(s)
Moléculas de Adhesión Celular/farmacología , Colitis/prevención & control , Vesículas Extracelulares/química , Células Madre Mesenquimatosas/química , Animales , Recuento de Células , Colitis/inducido químicamente , Colitis/terapia , Citocinas/metabolismo , Sulfato de Dextran/efectos adversos , Perros , Vesículas Extracelulares/trasplante , Inflamación/terapia , Macrófagos/citología , Células Madre Mesenquimatosas/ultraestructura , Ratones , Linfocitos T Reguladores/citología
9.
Biol Reprod ; 101(2): 360-367, 2019 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-31187129

RESUMEN

Spermatogonial stem cells (SSCs) are the basis of spermatogenesis in male due to their capability to multiply in numbers by self-renewal and subsequent meiotic processes. However, as SSCs are present in a very small proportion in the testis, in vitro proliferation of undifferentiated SSCs will facilitate the study of germ cell biology. In this study, we investigated the effectiveness of various cell lines as a feeder layer for rat SSCs. Germ cells enriched for SSCs were cultured on feeder layers including SIM mouse embryo-derived thioguanine and ouabain-resistant cells, C166 cells, and mouse and rat testicular endothelial cells (TECs) and their stem cell potential for generating donor-derived colonies and offspring was assessed by transplantation into recipient testes. Rat germ cells cultured on TECs showed increased mRNA and protein levels of undifferentiated spermatogonial markers. Rat SSCs derived from these germ cells underwent spermatogenesis and generated offspring when transplanted into recipients. Collectively, TECs can serve as an effective feeder layer that enhances the proliferative and self-renewal capacity of cultured rat SSCs while preserving their stemness properties.


Asunto(s)
Células Madre Germinales Adultas/fisiología , Células Endoteliales/fisiología , Testículo/citología , Animales , Técnicas de Cultivo de Célula , Proliferación Celular , Trasplante de Células , Células Nutrientes , Masculino , Ratones , Ratones Endogámicos C57BL , Ratas , Ratas Sprague-Dawley
10.
Vet Comp Oncol ; 17(1): 99-106, 2019 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-30411459

RESUMEN

Protein kinase A, a cyclic adenosine monophosphate (AMP)-dependent enzyme, normally exists within mammalian cells; however, in cancer cells, it can leak out and be found in the serum. Extracellular cyclic AMP-dependent protein kinase A (ECPKA) has been determined to increase in the serum of cancer-bearing dogs. However, there have been no reports in the veterinary literature on serum ECPKA autoantibody (ECPKA-Ab) expression in dogs with cancer. The aim of this study was to evaluate ECPKA-Ab and C-reactive protein (CRP) as serum biomarkers for cancer in dogs. ECPKA-Ab and CRP levels were detected by an enzyme-linked immunosorbent assay in serum samples from dogs with malignant tumours (n = 167), benign tumours (n = 42), or non-tumour disease (n = 155) and from healthy control dogs (n = 123). ECPKA-Ab and CRP levels were significantly higher in the dogs with malignant tumours than in those with benign tumours or non-tumour diseases, as well as in the healthy controls (P < 0.001, Kruskal-Wallis test). There was a significant positive correlation between the neoplastic index, which was developed using ECPKA-Ab and CRP levels, and the presence of cancer in dogs (P < 0.001); the area under the receiver-operating characteristic curve was estimated to be >0.85 (P < 0.001). In conclusion, ECPKA-Ab is a potential serum biomarker for a broad spectrum of cancers. Combined measurement of CRP and ECPKA-Ab levels in serum improves the sensitivity and accuracy of a diagnosis of cancer in dogs.


Asunto(s)
Adenosina Monofosfato/metabolismo , Autoanticuerpos/sangre , Proteína C-Reactiva/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/inmunología , Enfermedades de los Perros/diagnóstico , Neoplasias/veterinaria , Animales , Biomarcadores de Tumor/sangre , Proteínas Quinasas Dependientes de AMP Cíclico/clasificación , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Enfermedades de los Perros/sangre , Perros , Femenino , Masculino , Neoplasias/sangre , Neoplasias/diagnóstico
11.
BMC Vet Res ; 14(1): 354, 2018 Nov 20.
Artículo en Inglés | MEDLINE | ID: mdl-30453939

RESUMEN

BACKGROUND: Inflammatory bowel disease (IBD) is an intractable autoimmune disease, relatively common in cats, with chronic vomiting and diarrhea. Previous studies have reported that mesenchymal stem cells (MSCs) alleviate inflammation by modulating immune cells. However, there is a lack of research on cross-talk mechanism between feline adipose tissue-derived mesenchymal stem cells (fAT-MSCs) and immune cells in IBD model. Hence, this study aimed to evaluate the therapeutic effects of fAT-MSC on mice model of colitis and to clarify the therapeutic mechanism of fAT-MSCs. RESULTS: Intraperitoneal infusion of fAT-MSC ameliorated the clinical and histopathologic severity of colitis, including body weight loss, diarrhea, and inflammation in the colon of Dextran sulfate sodium (DSS)-treated mice (C57BL/6). Since regulatory T cells (Tregs) are pivotal in modulating immune responses and maintaining tolerance in colitis, the relation of Tregs with fAT-MSC-secreted factor was investigated in vitro. PGE2 secreted from fAT-MSC was demonstrated to induce elevation of FOXP3 mRNA expression and adjust inflammatory cytokines in Con A-induced feline peripheral blood mononuclear cells (PBMCs). Furthermore, in vivo, FOXP3+ cells of the fAT-MSC group were significantly increased in the inflamed colon, relative to that in the PBS group. CONCLUSION: Our results suggest that PGE2 secreted from fAT-MSC can reduce inflammation by increasing FOXP3+ Tregs in mice model of colitis. Consequently, these results propose the possibility of administration of fAT-MSC to cats with not only IBD but also other immune-mediated inflammatory diseases.


Asunto(s)
Tejido Adiposo/metabolismo , Colitis/tratamiento farmacológico , Dinoprostona/farmacología , Células Madre Mesenquimatosas/metabolismo , Linfocitos T Reguladores/efectos de los fármacos , Animales , Gatos , Colitis/inducido químicamente , Sulfato de Dextran/farmacología , Modelos Animales de Enfermedad , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Reacción en Cadena en Tiempo Real de la Polimerasa
12.
Nat Commun ; 9(1): 4379, 2018 10 22.
Artículo en Inglés | MEDLINE | ID: mdl-30348976

RESUMEN

Maintenance of adult tissues depends on stem cell self-renewal in local niches. Spermatogonial stem cells (SSC) are germline adult stem cells necessary for spermatogenesis and fertility. We show that testicular endothelial cells (TECs) are part of the SSC niche producing glial cell line-derived neurotrophic factor (GDNF) and other factors to support human and mouse SSCs in long-term culture. We demonstrate that FGF-2 binding to FGFR1 on TECs activates the calcineurin pathway to produce GDNF. Comparison of the TEC secretome to lung and liver endothelial cells identified 5 factors sufficient for long-term maintenance of human and mouse SSC colonies in feeder-free cultures. Male cancer survivors after chemotherapy are often infertile since SSCs are highly susceptible to cytotoxic injury. Transplantation of TECs alone restores spermatogenesis in mice after chemotherapy-induced depletion of SSCs. Identifying TECs as a niche population necessary for SSC self-renewal may facilitate fertility preservation for prepubertal boys diagnosed with cancer.


Asunto(s)
Células Endoteliales/citología , Células Endoteliales/efectos de los fármacos , Testículo/citología , Animales , Apoptosis/efectos de los fármacos , Línea Celular , Proliferación Celular/efectos de los fármacos , Preservación de la Fertilidad , Células Germinativas , Factor Neurotrófico Derivado de la Línea Celular Glial/farmacología , Humanos , Masculino , Ratones , Espermatogénesis/efectos de los fármacos , Nicho de Células Madre/efectos de los fármacos
13.
Stem Cell Res Ther ; 9(1): 255, 2018 09 26.
Artículo en Inglés | MEDLINE | ID: mdl-30257717

RESUMEN

BACKGROUND: Through recent studies, the onset of acute pancreatitis in pancreatic acinar cells (PACs) and the regulatory role of PACs in severe acute pancreatitis (SAP) have been revealed. During the early stages of pancreatitis, the endoplasmic reticulum (ER) in PACs undergoes significant changes, including swelling and vacuolization. In response to an increase in the extracellular stress in ER, PACs lose their functions, leading to cell apoptosis and inflammation response. The beneficial effects of human adipose tissue-derived mesenchymal stem cells (hAT-MSCs) on SAP have been well documented in previous studies. However, the underlying mechanism of their action remains controversial. METHODS: In this study, the therapeutic effects of intraperitoneally administered hAT-MSCs in a caerulein (50 µg/kg)- and lipopolysaccharide (LPS) (10 mg/kg)-co-induced SAP mouse model were evaluated. Inflammatory response and ER stress were measured in pancreatic tissue samples, and the beneficial effects were evaluated through quantitative reverse transcription polymerase chain reaction (qRT-PCR), western blot, and immunofluorescence analysis. RESULTS: Inflammatory response and ER stress were ameliorated following hAT-MSC injection, and the beneficial effects were observed in the absence of significant engraftment of hAT-MSCs. hAT-MSCs transfected with siRNA-targeting tumour necrosis factor-α-induced gene/protein 6 (TSG-6) were unable to inhibit ER stress and inflammation. In addition, TSG-6 from hAT-MSCs significantly suppressed ER stress-induced apoptosis and nuclear factor kappa B (NF-κB) activity in SAP model mice. CONCLUSIONS: TSG-6 secreted by hAT-MSCs protects PACs in SAP model mice via the inhibition of ER stress, as well as inflammatory responses. This study has revealed a new area for ER stress-targeted therapy in SAP patients.


Asunto(s)
Moléculas de Adhesión Celular/genética , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/citología , Pancreatitis/terapia , Células Acinares/patología , Enfermedad Aguda/terapia , Tejido Adiposo/citología , Tejido Adiposo/trasplante , Animales , Apoptosis , Modelos Animales de Enfermedad , Estrés del Retículo Endoplásmico/genética , Humanos , Lipopolisacáridos/toxicidad , Ratones , Pancreatitis/inducido químicamente , Pancreatitis/genética , Pancreatitis/fisiopatología
14.
Stem Cell Res Ther ; 9(1): 91, 2018 04 06.
Artículo en Inglés | MEDLINE | ID: mdl-29625582

RESUMEN

BACKGROUND: Inflammatory bowel disease (IBD) is an intractable autoimmune disorder that markedly deteriorates one's quality of life. Mesenchymal stem cells (MSCs) alleviate inflammation by modulating inflammatory cytokines in inflamed tissues, and have been suggested as a promising alternative for IBD treatment in human and veterinary cases. Furthermore, tumor necrosis factor-α-induced gene/protein 6 (TSG-6) is a key factor influencing MSC immunomodulatory properties; however, the precise mechanism of TSG-6 release from canine MSCs in IBD remains unclear. This study aimed to assess the therapeutic effects of canine adipose tissue-derived (cAT)-MSC-produced TSG-6 in an IBD mouse model and to explore the mechanisms underlying the immunomodulatory properties. METHODS: Mice with dextran sulfate sodium-induced colitis were administered cAT-MSCs intraperitoneally; colon tissues were collected on day 10 for histopathological, quantitative real-time polymerase chain reaction, and immunofluorescence analyses. RESULTS: cAT-MSC-secreted TSG-6 ameliorated IBD and regulated colonic expression of pro- and anti-inflammatory cytokines such as tumor necrosis factor-α, interleukin-6, and interleukin-10. To investigate the effect of cAT-MSC-secreted TSG-6 on activated macrophages in vitro, a transwell coculture system was used; TSG-6 released by cAT-MSCs induced a macrophage phenotypic switch from M1 to M2. The cAT-MSC-secreted TSG-6 increased M2 macrophages in the inflamed colon in vivo. CONCLUSIONS: TSG-6 released from cAT-MSCs can alleviate dextran sulfate sodium-induced colitis by inducing a macrophage phenotypic switch to M2 in mice.


Asunto(s)
Moléculas de Adhesión Celular/metabolismo , Diferenciación Celular , Colitis Ulcerosa/terapia , Macrófagos/citología , Trasplante de Células Madre Mesenquimatosas/métodos , Células Madre Mesenquimatosas/metabolismo , Tejido Adiposo/citología , Animales , Moléculas de Adhesión Celular/farmacología , Células Cultivadas , Citocinas/genética , Citocinas/metabolismo , Perros , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL
15.
In Vivo ; 31(6): 1115-1124, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29102933

RESUMEN

BACKGROUND/AIM: The kidney excretes waste materials and regulates important metabolic functions, and renal disorders constitute a significant medical problem and can result in fatalities. In the present study, mesenchymal stem cells derived from canine umbilical cord blood (cUCB-MSCs) were isolated and evaluated for their ability to improve renal function in a canine model of acute kidney injury (AKI). MATERIALS AND METHODS: The canine AKI model was developed by i.v. injection of cisplatin and gentamycin into 14 male beagle dogs. cUCB-MSCs were administered into the renal corticomedullary junction following AKI induction. Survival time, clinical signs, blood analysis and histological parameters were analyzed. RESULTS: The group treated with AKI plus cUCB-MSCs had decreased blood urea nitrogen and creatinine levels, and showed an extended life-span and improved histological manifestations. MSCs were detected around the tubules of these kidneys at the histological level. CONCLUSION: Taken together, our findings suggest that cUCB-MSCs could be an alternative therapeutic agent for canine AKI.


Asunto(s)
Lesión Renal Aguda/terapia , Apoptosis , Riñón/fisiopatología , Trasplante de Células Madre Mesenquimatosas , Lesión Renal Aguda/sangre , Lesión Renal Aguda/genética , Lesión Renal Aguda/fisiopatología , Animales , Nitrógeno de la Urea Sanguínea , Creatinina/sangre , Modelos Animales de Enfermedad , Perros , Humanos , Riñón/lesiones , Células Madre Mesenquimatosas/metabolismo
16.
Cell ; 170(6): 1149-1163.e12, 2017 Sep 07.
Artículo en Inglés | MEDLINE | ID: mdl-28886383

RESUMEN

The diversity of mesenchymal cell types in the lung that influence epithelial homeostasis and regeneration is poorly defined. We used genetic lineage tracing, single-cell RNA sequencing, and organoid culture approaches to show that Lgr5 and Lgr6, well-known markers of stem cells in epithelial tissues, are markers of mesenchymal cells in the adult lung. Lgr6+ cells comprise a subpopulation of smooth muscle cells surrounding airway epithelia and promote airway differentiation of epithelial progenitors via Wnt-Fgf10 cooperation. Genetic ablation of Lgr6+ cells impairs airway injury repair in vivo. Distinct Lgr5+ cells are located in alveolar compartments and are sufficient to promote alveolar differentiation of epithelial progenitors through Wnt activation. Modulating Wnt activity altered differentiation outcomes specified by mesenchymal cells. This identification of region- and lineage-specific crosstalk between epithelium and their neighboring mesenchymal partners provides new understanding of how different cell types are maintained in the adult lung.


Asunto(s)
Pulmón/citología , Mesodermo/citología , Animales , Homeostasis , Pulmón/fisiología , Ratones , Organoides/citología , Alveolos Pulmonares/citología , Receptores Acoplados a Proteínas G/análisis , Análisis de Secuencia de ARN , Análisis de la Célula Individual , Transcripción Genética
17.
Oncotarget ; 7(40): 65429-65440, 2016 10 04.
Artículo en Inglés | MEDLINE | ID: mdl-27589843

RESUMEN

Targeted therapies aimed at tumor vasculature are utilized in combination with chemotherapy to improve drug delivery and efficacy after tumor vascular normalization. Tumor vessels are highly disorganized with disrupted blood flow impeding drug delivery to cancer cells. Although pharmacologic anti-angiogenic therapy can remodel and normalize tumor vessels, there is a limited window of efficacy and these drugs are associated with severe side effects necessitating alternatives for vascular normalization. Recently, moderate aerobic exercise has been shown to induce vascular normalization in mouse models. Here, we provide a mechanistic explanation for the tumor vascular normalization induced by exercise. Shear stress, the mechanical stimuli exerted on endothelial cells by blood flow, modulates vascular integrity. Increasing vascular shear stress through aerobic exercise can alter and remodel blood vessels in normal tissues. Our data in mouse models indicate that activation of calcineurin-NFAT-TSP1 signaling in endothelial cells plays a critical role in exercise-induced shear stress mediated tumor vessel remodeling. We show that moderate aerobic exercise with chemotherapy caused a significantly greater decrease in tumor growth than chemotherapy alone through improved chemotherapy delivery after tumor vascular normalization. Our work suggests that the vascular normalizing effects of aerobic exercise can be an effective chemotherapy adjuvant.


Asunto(s)
Antineoplásicos/uso terapéutico , Doxorrubicina/uso terapéutico , Endotelio Vascular/fisiología , Terapia por Ejercicio , Ejercicio Físico , Neoplasias Experimentales/terapia , Remodelación Vascular , Animales , Calcineurina/metabolismo , Terapia Combinada , Humanos , Mecanotransducción Celular , Melanoma Experimental , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Factores de Transcripción NFATC/metabolismo , Resistencia al Corte , Transducción de Señal , Trombospondina 1/genética , Trombospondina 1/metabolismo
18.
Cancer Res ; 74(8): 2171-81, 2014 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-24590059

RESUMEN

Recent advances in cancer immunotherapy suggest that manipulation of the immune system to enhance the antitumor response may be a highly effective treatment modality. One understudied aspect of immunosurveillance is antiangiogenic surveillance, the regulation of tumor angiogenesis by the immune system, independent of tumor cell lysis. CD4(+) T cells can negatively regulate angiogenesis by secreting antiangiogenic factors such as thrombospondin-1 (TSP-1). In tumor-bearing mice, we show that a Th1-directed viral infection that triggers upregulation of TSP-1 in CD4(+) and CD8(+) T cells can inhibit tumor angiogenesis and suppress tumor growth. Using bone marrow chimeras and adoptive T-cell transfers, we demonstrated that TSP-1 expression in the T-cell compartment was necessary and sufficient to inhibit tumor growth by suppressing tumor angiogenesis after the viral infection. Our results establish that tumorigenesis can be stanched by antiangiogenic surveillance triggered by an acute viral infection, suggesting novel immunologic approaches to achieve antiangiogenic therapy.


Asunto(s)
Inmunoterapia Adoptiva/métodos , Melanoma Experimental/terapia , Linfocitos T/inmunología , Trombospondina 1/inmunología , Animales , Carcinogénesis/inmunología , Técnicas de Cultivo de Célula , Vigilancia Inmunológica , Activación de Linfocitos , Masculino , Melanoma Experimental/irrigación sanguínea , Melanoma Experimental/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones SCID , Ratones Transgénicos , Neovascularización Patológica/inmunología , Neovascularización Patológica/terapia , Trombospondina 1/biosíntesis , Trombospondina 1/genética , Ensayos Antitumor por Modelo de Xenoinjerto
19.
Cell ; 156(3): 440-55, 2014 Jan 30.
Artículo en Inglés | MEDLINE | ID: mdl-24485453

RESUMEN

Lung stem cells are instructed to produce lineage-specific progeny through unknown factors in their microenvironment. We used clonal 3D cocultures of endothelial cells and distal lung stem cells, bronchioalveolar stem cells (BASCs), to probe the instructive mechanisms. Single BASCs had bronchiolar and alveolar differentiation potential in lung endothelial cell cocultures. Gain- and loss-of-function experiments showed that BMP4-Bmpr1a signaling triggers calcineurin/NFATc1-dependent expression of thrombospondin-1 (Tsp1) in lung endothelial cells to drive alveolar lineage-specific BASC differentiation. Tsp1 null mice exhibited defective alveolar injury repair, confirming a crucial role for the BMP4-NFATc1-TSP1 axis in lung epithelial differentiation and regeneration in vivo. Discovery of this pathway points to methods to direct the derivation of specific lung epithelial lineages from multipotent cells. These findings elucidate a pathway that may be a critical target in lung diseases and provide tools to understand the mechanisms of respiratory diseases at the single-cell level.


Asunto(s)
Bronquiolos/citología , Diferenciación Celular , Células Endoteliales/metabolismo , Alveolos Pulmonares/citología , Transducción de Señal , Células Madre/metabolismo , Animales , Proteína Morfogenética Ósea 4/metabolismo , Receptores de Proteínas Morfogenéticas Óseas de Tipo 1/metabolismo , Bronquiolos/metabolismo , Células Cultivadas , Técnicas de Cocultivo , Ratones , Factores de Transcripción NFATC/metabolismo , Alveolos Pulmonares/metabolismo , Células Madre/citología , Trombospondina 1/genética , Trombospondina 1/metabolismo
20.
Cell Biol Int ; 38(1): 106-16, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24115681

RESUMEN

Fibrosis is a common end stage for a variety of liver diseases, including most chronic liver diseases, and results from an imbalance between collagen deposition and degradation. Mesenchymal stem cells (MSCs) have the ability to migrate into fibrotic livers and differentiate into hepatocytes. Hepatocyte growth factor (HGF) has potent anti-apoptotic and mitogenic effects on hepatocytes during liver injury and plays an essential role in the development and regeneration of the liver. In this study, human HGF-overexpressing human umbilical cord blood-derived MSCs (hHGF-HUCB-MSCs) were prepared using the pMEX Expression System, and the upregulation of hHGF expression was confirmed by RT-PCR and ELISA. HGF expressed by hHGF-HUCB-MSCs exerted a stimulatory effect on hepatocyte proliferation in vitro. hHGF-HUCB-MSCs were transplanted to investigate the therapeutic effects of these cells on carbon tetrachloride (CCL4)-induced liver fibrosis in a rat model. After 4 weeks of cell treatment once per week with 2 × 10(6) cells, biochemical analysis of the serum and histopathological analysis of the liver tissue were performed. The results of the biochemical analysis of the serum show that the hHGF-HUCB-MSC-treated group had higher levels of alanine aminotransferase, aspartate aminotransferase and alkaline phosphatase, indicating the improvement of liver function. Histopathology showed that the hHGF-HUCB-MSC-treated group had reduction in the density of collagen fibres. Thus hHGF-HUCB-MSCs can enhance liver regeneration and could be useful for the treatment of patients with liver fibrosis or cirrhosis.


Asunto(s)
Factor de Crecimiento de Hepatocito/metabolismo , Cirrosis Hepática/cirugía , Células Madre Mesenquimatosas/citología , Cordón Umbilical/citología , Alanina Transaminasa/metabolismo , Fosfatasa Alcalina/metabolismo , Animales , Aspartato Aminotransferasas/metabolismo , Tetracloruro de Carbono/toxicidad , Células Cultivadas , Modelos Animales de Enfermedad , Células Hep G2 , Factor de Crecimiento de Hepatocito/genética , Humanos , Hígado/enzimología , Hígado/metabolismo , Hígado/patología , Cirrosis Hepática/inducido químicamente , Regeneración Hepática , Masculino , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/metabolismo , Ratas , Ratas Wistar
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...