Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
BMC Vet Res ; 20(1): 375, 2024 Aug 22.
Artigo em Inglês | MEDLINE | ID: mdl-39174969

RESUMO

BACKGROUND: The aim of this study was to evaluate the adverse effects of allogeneic mesenchymal stem cells (MSCs) transplanted via intravenous infusion in dogs and examine their safety. We performed a retrospective analysis of various clinical assessments, including physical examination, blood tests, and radiographs, and monitored the formation of neoplasms during a 6-month follow-up period in 40 client-owned dogs that received intravenous infusion of adipose tissue-derived MSCs (AT-MSCs) for the treatment of various underlying diseases between 2012 and 2018. RESULTS: No significant adverse effects of MSC therapy were detected by clinical assessment, blood tests, or radiographic examination in the 6-month follow-up period after the first MSC treatment. Additionally no new neoplasms were observed during this period. CONCLUSIONS: To our knowledge, this study is the first to evaluate the safety aspects (≥ 6 months) associated with intravenous allogeneic AT-MSC infusion. These results suggest that allogenic AT-MSC infusion could be a useful and relatively safe therapeutic approach in canines.


Assuntos
Doenças do Cão , Transplante de Células-Tronco Mesenquimais , Animais , Cães , Transplante de Células-Tronco Mesenquimais/veterinária , Transplante de Células-Tronco Mesenquimais/efeitos adversos , Transplante de Células-Tronco Mesenquimais/métodos , Feminino , Masculino , Estudos Retrospectivos , Doenças do Cão/terapia , Células-Tronco Mesenquimais , Transplante Homólogo/veterinária , Injeções Intravenosas/veterinária , Tecido Adiposo/citologia
2.
Vet Med (Praha) ; 67(4): 206-211, 2022 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-39170805

RESUMO

Renal Fanconi syndrome (RFS) affects the proximal tubular resorption in the nephrons. This causes excessive loss of key solutes through the urine. In a canine patient, we successfully managed the renal tubular acidosis and proteinuria caused by RFS via transplantation of canine adipose tissue-derived mesenchymal stem cells (cAT-MSCs). cAT-MSCs were administered ten times at intervals of 2-4 weeks. The post-therapy check-up revealed that the cAT-MSC treatment improved the renal tubular acidosis and proteinuria. Hence, a cAT-MSC transplant may be considered as an adjuvant therapy in veterinary medicine to initiate and maintain relief of RFS-induced acidosis and proteinuria.

3.
J Am Anim Hosp Assoc ; 55(6): 323-327, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31525093

RESUMO

Fanconi syndrome is a renal proximal tubulopathy characterized by excessive urinary loss of glucose, amino acids, several electrolytes, and bicarbonate. Here, we report the case of transient Fanconi syndrome in a dog following administration of firocoxib, cefadroxil, tramadol, and famotidine. A 10 mo old Maltese was presented with lethargy, anorexia, vomiting, and weight loss. Transient Fanconi syndrome without azotemia was associated with firocoxib, cefadroxil, tramadol, and famotidine treatment. The dog received supportive care including IV fluids, gastroprotectants, and oral nutritional supplements. Two months after initial diagnosis and treatment, the dog showed complete resolution of glucosuria and aminoaciduria. The unique features of Fanconi syndrome in this case emphasize the potential renal tubular toxicity of this widely used multiple-drug combination.


Assuntos
4-Butirolactona/análogos & derivados , Cefadroxila/efeitos adversos , Doenças do Cão/induzido quimicamente , Famotidina/efeitos adversos , Síndrome de Fanconi/veterinária , Sulfonas/efeitos adversos , Tramadol/efeitos adversos , 4-Butirolactona/administração & dosagem , 4-Butirolactona/efeitos adversos , Analgésicos Opioides/administração & dosagem , Analgésicos Opioides/efeitos adversos , Animais , Antibacterianos/administração & dosagem , Antibacterianos/efeitos adversos , Antiulcerosos/administração & dosagem , Antiulcerosos/efeitos adversos , Cefadroxila/administração & dosagem , Cães , Famotidina/administração & dosagem , Síndrome de Fanconi/induzido quimicamente , Glucose , Glicosúria , Masculino , Sulfonas/administração & dosagem , Tramadol/administração & dosagem
4.
BMC Vet Res ; 14(1): 354, 2018 Nov 20.
Artigo em Inglês | MEDLINE | ID: mdl-30453939

RESUMO

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.


Assuntos
Tecido Adiposo/metabolismo , Colite/tratamento farmacológico , Dinoprostona/farmacologia , Células-Tronco Mesenquimais/metabolismo , Linfócitos T Reguladores/efeitos dos fármacos , Animais , Gatos , Colite/induzido quimicamente , Sulfato de Dextrana/farmacologia , Modelos Animais de Doenças , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Reação em Cadeia da Polimerase em Tempo Real
5.
J Vet Intern Med ; 38(4): 2158-2164, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39023218

RESUMO

BACKGROUND: Romiplostim, a thrombopoietin analog, is commonly used to treat immune-mediated thrombocytopenia (ITP) in humans, but its use in dogs remains limited. OBJECTIVES: Evaluate the effects and adverse events of romiplostim administration in dogs with thrombocytopenia caused by various underlying diseases. ANIMALS: Forty-two client-owned dogs with naturally occurring thrombocytopenia at 2 referral animal hospitals. METHODS: Retrospective, multi-institutional analysis to evaluate the outcomes of romiplostim treatment in dogs. RESULTS: Among the dogs treated with romiplostim, 27 experienced an increase in platelet count and 26 maintained a platelet count within the reference range. Platelet count improvement was observed in various conditions: primary ITP (90%, n = 18/20), pancytopenia of unknown etiology (42.9%, n = 3/7), chemotherapy-induced thrombocytopenia (50%, n = 3/6), babesiosis (100%, n = 1/1), radiotherapy-induced thrombocytopenia (0%, n = 0/1), and disseminated intravascular coagulopathy (33.3%, n = 2/6). The median time for platelet recovery (>50 000/µL) after romiplostim administration was 4 days, and the median time for platelet count normalization was 7 days. Median hospitalization time for the improvement group (I) was 5 days. The survival-to-discharge rates were 85%, 40%, and 28.6% for dogs with primary ITP, secondary thrombocytopenia, and thrombocytopenia of unknown etiology, respectively. CONCLUSIONS AND CLINICAL IMPORTANCE: Romiplostim is a well-tolerated and promising treatment for primary ITP in dogs, suggesting its potential as a valuable therapeutic option for dogs with thrombocytopenia caused by various underlying conditions. These findings emphasize the need for further research to optimize romiplostim dosing and understand its role in treating secondary thrombocytopenia and pancytopenia of unknown etiology.


Assuntos
Doenças do Cão , Receptores Fc , Proteínas Recombinantes de Fusão , Trombocitopenia , Trombopoetina , Cães , Animais , Trombopoetina/uso terapêutico , Receptores Fc/uso terapêutico , Proteínas Recombinantes de Fusão/uso terapêutico , Proteínas Recombinantes de Fusão/efeitos adversos , Proteínas Recombinantes de Fusão/administração & dosagem , Doenças do Cão/tratamento farmacológico , Estudos Retrospectivos , Feminino , Trombocitopenia/veterinária , Trombocitopenia/tratamento farmacológico , Masculino , Contagem de Plaquetas/veterinária , Resultado do Tratamento , Púrpura Trombocitopênica Idiopática/tratamento farmacológico , Púrpura Trombocitopênica Idiopática/veterinária
6.
Vet Comp Oncol ; 2024 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-39090848

RESUMO

Canine lymphoma, the most prevalent haematopoietic tumour in dogs, presents significant challenges in veterinary oncology. This study investigates the prognostic value of the neutrophil-to-lymphocyte ratio (NLR) in small-sized dogs (≤10 kg) with multicentric lymphoma. In this retrospective study, we examined medical records and haematological data from 35 dogs to assess the association between NLR and two key outcomes: time-to-progression (TTP) and lymphoma-specific survival (LSS) using Cox proportional hazards models. Our findings revealed a significant correlation between elevated NLR and a worse prognosis, as evidenced by TTP (p = 0.005) and LSS (p = 0.001). NLR is linked to increased hazard ratios (HRs) for the time-to-progression rate (TTPR) at 180, 360 and 540 days (p = 0.001, p = 0.003 and p = 0.005, respectively) and the lymphoma-specific survival rate (LSSR) at the same intervals (p = 0.016, p = 0.001 and p = 0.001, respectively). Cutoff value of 3.764 for NLR was established, above which there is a significantly increased risk of early disease progression and decreased survival. Additionally, our analysis indicates that dogs with substage b exhibited earlier progression than those with substage a, evident in overall (p = 0.026) and TTPR at 180 days (p = 0.004), 360 days (p = 0.018), 540 days (p = 0.026) and LSSR at 180 days (p = 0.033). The results underscore the potential of NLR as a prognostic marker in cases of dogs ≤10 kg with multicentric lymphoma, suggesting that higher NLR is associated with a poorer prognosis.

7.
Vet Res Commun ; 2024 Aug 12.
Artigo em Inglês | MEDLINE | ID: mdl-39133399

RESUMO

The prevalence of chronic kidney disease (CKD) in dogs increases with age, and renal fibrosis is an important pathophysiological mechanism in this process. However, only a few drugs that can effectively inhibit fibrosis in the kidneys of dogs are currently available. In this study, we aimed to determine whether pirfenidone, a drug that has shown antifibrotic effects in various clinical studies, also exerts antifibrotic effects on canine renal tubular epithelial cells, Madin-Darby canine kidney cells (MDCK). To this end, we treated MDCK cells with various concentrations of pirfenidone, followed by transforming growth factor-beta1 (TGF-ß1) to stimulate fibrotic conditions. A cell viability assay was performed to determine the effect of pirfenidone on cell survival. Fibrosis-related markers and TGF-ß1 fibrotic pathway-related markers were assessed using qPCR, Western blot analysis and immunocytochemistry. A one-way analysis of variance (ANOVA) was performed, followed by Tukey's post-hoc test for multiple comparisons. Pirfenidone treatment significantly reduced the expression of profibrotic markers such as α-smooth muscle actin, fibronectin, and collagen. Additionally, it upregulated the expression of E-cadherin, an epithelial marker. Furthermore, pirfenidone effectively inhibited the phosphorylation of key factors involved in the TGF-ß1 signaling pathway, including Smad2/3 and ERK1/2. These results demonstrate that pirfenidone suppresses TGF-ß1-induced fibrosis in MDCK cells by attenuating epithelial-mesenchymal transition and the relevant signaling pathways.

8.
J Vet Sci ; 24(6): e76, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-37904638

RESUMO

An 11-year-old neutered male Miniature Poodle with a stage 3 apocrine gland adenocarcinoma was started on chemotherapy with toceranib phosphate after surgery. Beginning on day 10 of toceranib, the dog's foot pads became erythematous and hyperkeratinized. The dog complained of pain, inability to walk, depression, and loss of appetite. The symptoms resolved when toceranib was discontinued and reappeared when toceranib was resumed. Grade 3 palmar-plantar erythrodysesthesia was identified as an adverse event of toceranib based on the VCOG-CTCAE and Naranjo scale. Although very rare in veterinary medicine, clinicians should consider that palmar-plantar erythrodysesthesia can occur after toceranib administration.


Assuntos
Adenocarcinoma , Sacos Anais , Doenças do Cão , Masculino , Cães , Animais , Glândulas Apócrinas , Pirróis/efeitos adversos , Adenocarcinoma/tratamento farmacológico , Adenocarcinoma/veterinária , Adenocarcinoma/induzido quimicamente , Doenças do Cão/tratamento farmacológico
9.
Front Vet Sci ; 10: 1298736, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-38105775

RESUMO

A 13-year-old neutered male Korean short-hair cat presented with anorexia, lethargy, and a severely distended abdomen, suggestive of ascites. Abdominocentesis yielded serosanguineous fluid. A subsequent diagnostic workup, including blood tests, ascitic fluid analysis, imaging studies [radiography, ultrasound, and computed tomography (CT)], and histopathological examination, was performed to identify the underlying cause. Imaging studies revealed characteristics of encapsulating peritoneal sclerosis (EPS) such as peritoneal thickening, fat stranding, and calcification. During laparotomy, fibrous membranes encapsulating the abdominal organs and ascites were observed, and multiple calcified regions were detected on the abdominal wall. Histopathological analysis confirmed the diagnosis of poorly differentiated invasive malignant neoplasms, which were further classified as carcinomatosis based on positive cytokeratin and negative vimentin immunohistochemistry results. To our knowledge, this is the first report of sclerosing peritoneal carcinomatosis with osseous metaplasia in a cat.

10.
Vet Immunol Immunopathol ; 219: 109973, 2020 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-31765882

RESUMO

Preconditioning with hypoxia or hypoxia-mimetic agents has been tried with mesenchymal stem cells (MSCs) to improve the secretion of anti-inflammatory factors. These preconditioning procedures upregulate hypoxia inducible factor (HIF) 1-alpha leading to the transcription of HIF-dependent tissue protective and anti-inflammatory genes. Due to the limited number of studies exploring the activity of deferoxamine (DFO)-a hypoxia-mimetic agent-in MSCs, we aimed to determine whether DFO can enhance the secretion of anti-inflammatory substances in canine adipose tissue-derived (cAT)-MSCs. Furthermore, we investigated whether this activity of DFO could affect macrophage polarization and activate anti-inflammatory reactions. cAT-MSCs preconditioned with DFO exhibited enhanced secretion of anti-inflammatory factors such as prostaglandin E2 and tumor necrosis factor-α-stimulated gene-6. To evaluate the interaction between DFO preconditioned cAT-MSCs and macrophages, RAW 264.7 cells were co-cultured with cAT-MSCs using the Transwell system, and changes in the expression of factors related to macrophage polarization were analyzed using the quantitative real-time PCR and western blot assays. When RAW 264.7 cells were co-cultured with DFO preconditioned cAT-MSCs, the expression of M1 and M2 markers decreased and increased, respectively, compared to co-culturing with non-preconditioned cAT-MSCs. Thus, cAT-MSCs preconditioned with DFO can more effectively direct and reprogram macrophage polarization into the M2 phase, an anti-inflammatory state.


Assuntos
Tecido Adiposo/citologia , Anti-Inflamatórios/farmacologia , Diferenciação Celular/efeitos dos fármacos , Desferroxamina/farmacologia , Macrófagos/citologia , Células-Tronco Mesenquimais/efeitos dos fármacos , Animais , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Técnicas de Cocultura , Cães , Ativação de Macrófagos/efeitos dos fármacos , Camundongos , Células RAW 264.7 , Transdução de Sinais
11.
Vet Comp Oncol ; 18(2): 152-160, 2020 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-31503379

RESUMO

Paclitaxel, a member of the taxane family, exhibits antitumour effects by targeting the microtubules in cancer cells. Recently, oral paclitaxel has been developed to overcome the side effects of intravenous paclitaxel administration in human patients. The objective of this study was to investigate the antitumour effects of oral paclitaxel in vitro and in vivo. Three weeks after inoculation, oral paclitaxel (25 and 50 mg/kg) or saline was administered every week for three consecutive weeks. To explore the underlying mechanism, tumour angiogenesis was examined by immunohistochemistry with an anti-CD31 antibody. Tumour cell apoptosis was detected by Terminal deoxynucleotidyl transferase dUTP Nick-End Labeling assay, and cell cycle arrest was confirmed by western blot analysis. Oral paclitaxel treatment of canine melanoma cells exerted mediated antiproliferative effects and mediated cell cycle arrest in vitro. In animal experiments, after oral paclitaxel administration, the average tumour size decreased to approximately 30% of that in the control. Histologically, oral paclitaxel showed anti-angiogenic effects and induced the apoptosis in tumour tissues. Oral paclitaxel also downregulated the intratumoural expression of cyclin D1 and inhibited cell proliferation. The study findings support potential application of oral paclitaxel as a novel chemotherapeutic strategy to treat canine melanoma. This is the first study to investigate the potential of oral paclitaxel as a therapeutic drug against canine tumours.


Assuntos
Antineoplásicos Fitogênicos/uso terapêutico , Melanoma/tratamento farmacológico , Paclitaxel/uso terapêutico , Administração Oral , Animais , Antineoplásicos Fitogênicos/administração & dosagem , Antineoplásicos Fitogênicos/efeitos adversos , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Cães , Feminino , Camundongos , Camundongos Nus , Neoplasias Experimentais , Paclitaxel/efeitos adversos
12.
PLoS One ; 15(2): e0220756, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32040478

RESUMO

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.


Assuntos
Moléculas de Adesão Celular/farmacologia , Colite/prevenção & controle , Vesículas Extracelulares/química , Células-Tronco Mesenquimais/química , Animais , Contagem de Células , Colite/induzido quimicamente , Colite/terapia , Citocinas/metabolismo , Sulfato de Dextrana/efeitos adversos , Cães , Vesículas Extracelulares/transplante , Inflamação/terapia , Macrófagos/citologia , Células-Tronco Mesenquimais/ultraestrutura , Camundongos , Linfócitos T Reguladores/citologia
13.
J Vet Med Sci ; 81(11): 1663-1670, 2019 Dec 05.
Artigo em Inglês | MEDLINE | ID: mdl-31582601

RESUMO

The paracrine function of mesenchymal stem cells (MSCs) during transplantation has been recently studied due to its poor differentiation ratio. Dimethyloxalylglycine (DMOG) has been used to promote angiogenesis in experimental animal models, however, comparable approaches for canine MSCs are not sufficient. In the present study, we assessed whether DMOG improves angiogenesis in canine adipose tissue-derived mesenchymal stem cells (cAT-MSCs). cAT-MSCs were treated with DMOG and their effect on angiogenesis was investigated by cell proliferation assay, western blotting, and tube formation assay. Dimethyloxalylglycine preconditioning enhanced the expression of vascular endothelial growth factor (VEGF) among pro-angiogenic factors in cAT-MSCs via hypoxia-inducible factor-1α stabilization. Dimethyloxalylglycine primed-cAT-MSC-conditioned media increased angiogenesis in human umbilical vein endothelial cells. These results suggest that DMOG conditioning of cAT-MSCs augmented the secretion of VEGF, which acted as a prominent pro-angiogenic factor during angiogenesis. DMOG-primed cAT-MSCs may have the potential to induce beneficial effects in ischemic diseases in clinical trials.


Assuntos
Cães , Glicina/análogos & derivados , Células-Tronco Mesenquimais/efeitos dos fármacos , Neovascularização Fisiológica/efeitos dos fármacos , Indutores da Angiogênese/metabolismo , Animais , Diferenciação Celular/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Glicina/farmacologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo
14.
Vet Comp Oncol ; 17(1): 99-106, 2019 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-30411459

RESUMO

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.


Assuntos
Monofosfato de Adenosina/metabolismo , Autoanticorpos/sangue , Proteína C-Reativa/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/imunologia , Doenças do Cão/diagnóstico , Neoplasias/veterinária , Animais , Biomarcadores Tumorais/sangue , Proteínas Quinases Dependentes de AMP Cíclico/classificação , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Doenças do Cão/sangue , Cães , Feminino , Masculino , Neoplasias/sangue , Neoplasias/diagnóstico
15.
Res Vet Sci ; 125: 176-184, 2019 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-31247473

RESUMO

Canine inflammatory bowel disease (IBD) is an intractable autoimmune disorder that results in various gastrointestinal and systemic symptoms. Mesenchymal stem cells (MSCs), which release immunomodulatory factors such as tumor necrosis factor-α (TNF-α)-induced gene/protein 6 (TSG-6) and prostaglandin E2 (PGE2), have been suggested as an alternative therapeutic option for IBD treatment in veterinary medicine. Furthermore, although it is known that MSCs pre-treated with pro-inflammatory cytokines show enhanced anti-inflammatory properties via the secretion of soluble factors, the underlying mechanisms of IBD remain unclear. The aim of this study was to demonstrate the therapeutic effects and corresponding mechanisms of canine adipose tissue-derived (cAT)-MSCs stimulated with TNF-α in mouse models of IBD. Mice with dextran sulfate sodium (DSS)- or dinitrobenzene sulfonic acid (DNBS)-induced colitis were injected intraperitoneally with cAT-MSCs pre-treated with TNF-α. Colitis severity was assessed and colon tissues were collected for histopathological, enzyme-linked immunosorbent assay, and flow cytometry analysis. cAT-MSCs stimulated with TNF-α secreted higher concentrations of immunomodulatory factors such as TSG-6 and PGE2, which play a key role in inducing phenotypic alterations in macrophages. Consequently, TNF-α-pre-treated cAT-MSCs further regulated colonic inflammatory cytokines such as interleukin (IL)-1ß, IL-6, and IL-10, and ameliorated DSS- or DNBS-induced colitis in mice. Additionally, we demonstrated that M1 macrophages (F4/80+/iNOS+ cells) were decreased in colon tissues from mice treated with TNF-α-pre-treated cAT-MSCs, whereas M2 macrophages (F4/80+/CD206+ cells) were increased. These results may suggest a new cell-based therapeutic option for treating IBD.


Assuntos
Colite/induzido quimicamente , Cães , Doenças Inflamatórias Intestinais/terapia , Macrófagos/fisiologia , Células-Tronco Mesenquimais/fisiologia , Fator de Necrose Tumoral alfa/farmacologia , Animais , Colite/terapia , Citocinas/genética , Citocinas/metabolismo , Sulfato de Dextrana , Dinoprostona/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Camundongos , Fator de Necrose Tumoral alfa/administração & dosagem
16.
Stem Cell Res Ther ; 9(1): 255, 2018 09 26.
Artigo em Inglês | MEDLINE | ID: mdl-30257717

RESUMO

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.


Assuntos
Moléculas de Adesão Celular/genética , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/citologia , Pancreatite/terapia , Células Acinares/patologia , Doença Aguda/terapia , Tecido Adiposo/citologia , Tecido Adiposo/transplante , Animais , Apoptose , Modelos Animais de Doenças , Estresse do Retículo Endoplasmático/genética , Humanos , Lipopolissacarídeos/toxicidade , Camundongos , Pancreatite/induzido quimicamente , Pancreatite/genética , Pancreatite/fisiopatologia
17.
J Vet Med Sci ; 80(1): 152-155, 2018 Feb 02.
Artigo em Inglês | MEDLINE | ID: mdl-29142147

RESUMO

Two dogs presented to the emergency service after accidental ingestion of afloqualone tablets, a muscle relaxant used for back pain in humans. Toxic effects of the drug in these dogs included vomiting, respiratory depression, seizures, ataxia, bradycardia, and hematuria. Treatment consisted of fluid diuresis, furosemide, and propofol. Flumazenil, a gamma-amino butyric acid antagonist, was administered intravenously; however, it was not effective in stopping the seizures in these dogs. Both dogs recovered with supportive treatment. To the authors' knowledge, this is the first documented report of afloqualone intoxication in dogs.


Assuntos
Doenças do Cão/induzido quimicamente , Quinazolinas/intoxicação , Animais , Ataxia/induzido quimicamente , Ataxia/veterinária , Bradicardia/induzido quimicamente , Bradicardia/veterinária , Cães , Flumazenil/uso terapêutico , Masculino , Quinazolinas/antagonistas & inibidores , Insuficiência Respiratória/induzido quimicamente , Insuficiência Respiratória/veterinária , Convulsões/induzido quimicamente , Convulsões/veterinária , Vômito/induzido quimicamente , Vômito/veterinária
18.
Res Vet Sci ; 119: 19-26, 2018 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-29783120

RESUMO

Mesenchymal stem cells (MSCs) have been used in studies on treatment of various diseases, and their application to immune-mediated diseases has garnered interest. Various methods for enhancing the immunomodulation effect of human MSCs have been used; however, similar approaches for canine MSCs are relatively unexplored. Accordingly, we evaluated immunomodulatory effects and mechanisms in canine MSCs treated with TNF-α and IFN-γ. Lipopolysaccharide (LPS)-stimulated RAW 264.7 cells were incubated with the conditioned media (CM) from canine MSCs for 48 h. Expression of RNA was assessed by quantitative reverse transcription PCR (qRT-PCR), and protein levels were assessed by western blot. Expression of inducible nitric oxide synthase (iNOS), IL-6 and IL-1ß was significantly (one-way ANOVA) decreased in LPS-stimulated RAW 264.7 cells incubated with CM from canine MSCs compared to that in LPS-stimulated RAW 264.7 cells alone. Furthermore, anti-inflammatory effects of TNF-α- and IFN-γ-primed canine MSCs were significantly increased compared with those of naïve canine MSCs. Expression of cyclooxygenase 2 (COX-2) and prostaglandin E2 (PGE2) were likewise significantly increased in primed canine MSCs. The level of iNOS protein in LPS-stimulated RAW 264.7 cells incubated with CM from the primed canine MSCs was decreased, but it increased when the cells were treated with NS-398(PGE2 inhibitor). In conclusion, compared with naïve canine MSCs, cells primed with TNF-α and IFN-γ cause a greater reduction in release of anti-inflammatory cytokines from LPS-stimulated RAW 264.7 cells; the mechanism is upregulation of the COX-2/PGE2 pathway.


Assuntos
Ciclo-Oxigenase 2/imunologia , Dinoprostona/imunologia , Interferon gama/farmacologia , Células-Tronco Mesenquimais/metabolismo , Fator de Necrose Tumoral alfa/farmacologia , Animais , Anti-Inflamatórios , Citocinas , Cães , Humanos , Lipopolissacarídeos , Macrófagos , Óxido Nítrico
19.
Stem Cell Res Ther ; 9(1): 91, 2018 04 06.
Artigo em Inglês | MEDLINE | ID: mdl-29625582

RESUMO

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.


Assuntos
Moléculas de Adesão Celular/metabolismo , Diferenciação Celular , Colite Ulcerativa/terapia , Macrófagos/citologia , Transplante de Células-Tronco Mesenquimais/métodos , Células-Tronco Mesenquimais/metabolismo , Tecido Adiposo/citologia , Animais , Moléculas de Adesão Celular/farmacologia , Células Cultivadas , Citocinas/genética , Citocinas/metabolismo , Cães , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL
20.
In Vivo ; 31(3): 349-356, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28438862

RESUMO

BACKGROUND/AIM: The transcription factors Oct4 and Sox2 enhance the proliferation and pluripotency of human adipose tissue-derived mesenchymal stem cells (hAT-MSCs); however, the anti-inflammatory effects of Oct4- and Sox2-overexpressing hAT-MSCs (Oct4/Sox2-hAT-MSCs) are unclear. Here, we evaluated the anti-inflammatory effects of Oct4/Sox2-hAT-MSCs in vitro and in vivo. MATERIALS AND METHODS: Supernatants from green-fluorescent protein (GFP)- and Oct4/Sox2-hAT-MSCs were used to treat lipopolysaccharide (LPS)-stimulated RAW264.7 cells and inflammatory cytokine expression was determined. In LPS-induced mice, GFP- and Oct4/Sox2-hAT-MSCs were injected intraperitoneally and survival rates, as well as sickness scores of mice, were monitored. RESULTS: Decreased expression of pro-inflammatory cytokines was observed in Oct4/Sox2-hAT-MSC supernatant-exposed RAW264.7 cells compared to that in GFP-hAT-MSC supernatant-exposed RAW264.7 cells. The sickness score was reduced to 34.9% and the survival rate was increased by 11.1% in Oct4/Sox2-hAT-MSC-injected mice compared to that in GFP-hAT-MSC-injected mice. CONCLUSION: Our findings provide important insights into the development of therapies utilizing Oct4/Sox2-hAT-MSCs in inflammatory diseases.


Assuntos
Tecido Adiposo/metabolismo , Anti-Inflamatórios/metabolismo , Células-Tronco Mesenquimais/metabolismo , Fator 3 de Transcrição de Octâmero/metabolismo , Fatores de Transcrição SOXB1/metabolismo , Animais , Células Cultivadas , Citocinas/metabolismo , Proteínas de Fluorescência Verde/metabolismo , Humanos , Inflamação/metabolismo , Camundongos , Células RAW 264.7 , Taxa de Sobrevida
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa