Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 159
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Stem Cells ; 2024 May 25.
Artículo en Inglés | MEDLINE | ID: mdl-38795363

RESUMEN

Companion animals in veterinary medicine develop multiple naturally occurring diseases analogous to human conditions. We previously reported a comprehensive review on the feasibility, safety, and biologic activity of using novel stem cell therapies to treat a variety of inflammatory conditions in dogs and cats (2008-2015) [1]. The purpose of this review is to provide an updated summary of current studies in companion animal disease models that have evaluated stem cell therapeutics that are relevant to human disease. Here we have reviewed the literature from 2015 to 2023 for publications on stem cell therapies that have been evaluated in companion animals, including dogs, cats, and horses. The review excluded case reports or studies performed in experimentally induced models of disease, studies involving cancer, or studies in purpose-bred laboratory species such as rodents. We identified 45 manuscripts meeting these criteria, an increase from 19 that were described in the previous review [1]. The majority of studies were performed in dogs (n=28), with additional studies in horses (n=9) and cats (n=8). Disease models included those related to musculoskeletal disease (osteoarthritis, tendon/ligament injury), neurologic disease (canine cognitive dysfunction, intervertebral disc disease, spinal cord injury) gingival/dental disease (gingivostomatitis), dermatologic disease (atopic dermatitis), chronic multi-drug resistant infections, ophthalmic disease (keratoconjunctivitis sicca, eosinophilic keratitis, immune mediated keratitis), cardiopulmonary disease (asthma, degenerative valve disease, dilated cardiomyopathy), gastrointestinal disease (inflammatory bowel disease, chronic enteropathy) and renal disease (chronic kidney disease). The majority of studies reported beneficial responses to stem cell treatment, with the exception of those related to more chronic processes such as spinal cord injury and chronic kidney disease. However, it should also be noted that 22 studies were open-label, baseline-controlled trials and only 12 studies were randomized and controlled, making overall study interpretation difficult. As noted in the previous review, improved regulatory oversight and consistency in manufacturing of stem cell therapies is needed. Enhanced understanding of the temporal course of disease processes using advanced -omics approaches may further inform mechanisms of action and help define appropriate timing of interventions. Future directions of stem cell-based therapies could include use of stem-cell derived extracellular vesicles, or cell conditioning approaches to direct cells to specific pathways that are tailored to individual disease processes and stages of illness.

2.
Cancer Immunol Immunother ; 73(5): 77, 2024 Mar 30.
Artículo en Inglés | MEDLINE | ID: mdl-38554158

RESUMEN

The use of large animal spontaneous models of solid cancers, such as dogs with osteosarcoma (OS), can help develop new cancer immunotherapy approaches, including chimeric antigen receptor (CAR) T cells. The goal of the present study was to generate canine CAR T cells targeting the B7-H3 (CD276) co-stimulatory molecule overexpressed by several solid cancers, including OS in both humans and dogs, and to assess their ability to recognize B7-H3 expressed by canine OS cell lines or by canine tumors in xenograft models. A second objective was to determine whether a novel dual CAR that expressed a chemokine receptor together with the B7-H3 CAR improved the activity of the canine CAR T cells. Therefore, in the studies reported here we examined B7-H3 expression by canine OS tumors, evaluated target engagement by canine B7-H3 CAR T cells in vitro, and compared the relative effectiveness of B7-H3 CAR T cells versus B7-H3-CXCR2 dual CAR T cells in canine xenograft models. We found that most canine OS tumors expressed B7-H3; whereas, levels were undetectable on normal dog tissues. Both B7-H3 CAR T cells demonstrated activation and OS-specific target killing in vitro, but there was significantly greater cytokine production by B7-H3-CXCR2 CAR T cells. In canine OS xenograft models, little anti-tumor activity was generated by B7-H3 CAR T cells; whereas, B7-H3-CXCR2 CAR T cells significantly inhibited tumor growth, inducing complete tumor elimination in most treated mice. These findings indicated therefore that addition of a chemokine receptor could significantly improve the anti-tumor activity of canine B7-H3 CAR T cells, and that evaluation of this new dual CAR construct in dogs with primary or metastatic OS is warranted since such studies could provide a critical and realistic validation of the chemokine receptor concept.


Asunto(s)
Neoplasias Óseas , Osteosarcoma , Humanos , Perros , Animales , Ratones , Antígenos B7/metabolismo , Osteosarcoma/terapia , Neoplasias Óseas/patología , Linfocitos T , Receptores de Quimiocina , Línea Celular Tumoral
3.
Cancer Immunol Immunother ; 72(5): 1185-1198, 2023 May.
Artículo en Inglés | MEDLINE | ID: mdl-36367558

RESUMEN

Ocular surface squamous neoplasia (OSSN) is the major cause of corneal cancer in man and horses worldwide, and the prevalence of OSSN is increasing due to greater UVB exposure globally. Currently, there are no approved treatments for OSSN in either species, and most patients are managed with surgical excision or off-label treatment with locally injected interferon alpha, or topically applied cytotoxic drugs such as mitomycin C. A more broadly effective and readily applied immunotherapy could exert a significant impact on management of OSSN worldwide. We therefore evaluated the effectiveness of a liposomal TLR complex (LTC) immunotherapy, which previously demonstrated strong antiviral activity in multiple animal models following mucosal application, for ocular antitumor activity in a horse spontaneous OSSN model. In vitro studies demonstrated strong activation of interferon responses in horse leukocytes by LTC and suppression of OSSN cell growth and migration. In a trial of 8 horses (9 eyes), treatment with topical or perilesional LTC resulted in an overall tumor response rate of 67%, including durable regression of large OSSN tumors. Repeated treatment with LTC ocular immunotherapy was also very well tolerated clinically. We conclude therefore that ocular immunotherapy with LTC warrants further investigation as a novel approach to management of OSSN in humans.


Asunto(s)
Antineoplásicos , Carcinoma de Células Escamosas , Neoplasias de la Conjuntiva , Neoplasias del Ojo , Humanos , Caballos , Animales , Interferón alfa-2/uso terapéutico , Carcinoma de Células Escamosas/patología , Antineoplásicos/uso terapéutico , Neoplasias de la Conjuntiva/tratamiento farmacológico , Neoplasias de la Conjuntiva/patología , Neoplasias de la Conjuntiva/cirugía , Interferón-alfa , Neoplasias del Ojo/terapia , Inmunoterapia , Estudios Retrospectivos
4.
Vet Clin North Am Equine Pract ; 39(3): 565-578, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37442729

RESUMEN

Increasing antimicrobial resistance in veterinary practice has driven the investigation of novel therapeutic strategies including regenerative and biologic therapies to treat bacterial infection. Integration of biological approaches such as platelet lysate and mesenchymal stromal cell (MSC) therapy may represent adjunctive treatment strategies for bacterial infections that minimize systemic side effects and local tissue toxicity associated with traditional antibiotics and that are not subject to antibiotic resistance. In this review, we will discuss mechanisms by which biological therapies exert antimicrobial effects, as well as potential applications and challenges in clinical implementation in equine practice.


Asunto(s)
Enfermedades de los Caballos , Células Madre Mesenquimatosas , Caballos , Animales , Enfermedades de los Caballos/terapia , Plaquetas , Antibacterianos
5.
Int J Mol Sci ; 23(2)2022 Jan 13.
Artículo en Inglés | MEDLINE | ID: mdl-35055015

RESUMEN

Stereotactic body radiotherapy (SBRT) is known to induce important immunologic changes within the tumor microenvironment (TME). However, little is known regarding the early immune responses within the TME in the first few weeks following SBRT. Therefore, we used the canine spontaneous tumor model to investigate TME responses to SBRT, and how local injection of immune modulatory antibodies to OX40 and TLR 3/9 agonists might modify those responses. Pet dogs with spontaneous cancers (melanoma, carcinoma, sarcoma, n = 6 per group) were randomized to treatment with either SBRT or SBRT combined with local immunotherapy. Serial tumor biopsies and serum samples were analyzed for immunologic responses. SBRT alone resulted at two weeks after treatment in increased tumor densities of CD3+ T cells, FoxP3+ Tregs, and CD204+ macrophages, and increased expression of genes associated with immunosuppression. The addition of OX40/TLR3/9 immunotherapy to SBRT resulted in local depletion of Tregs and tumor macrophages and reduced Treg-associated gene expression (FoxP3), suppressed macrophage-associated gene expression (IL-8), and suppressed exhausted T cell-associated gene expression (CTLA4). Increased concentrations of IL-7, IL-15, and IL-18 were observed in serum of animals treated with SBRT and immunotherapy, compared to animals treated with SBRT. A paradoxical decrease in the density of effector CD3+ T cells was observed in tumor tissues that received combined SBRT and immunotherapy as compared to animals treated with SBRT only. In summary, these results obtained in a spontaneous large animal cancer model indicate that addition of OX40/TLR immunotherapy to SBRT modifies important immunological effects both locally and systemically.


Asunto(s)
Antineoplásicos Inmunológicos/farmacología , Enfermedades de los Perros/terapia , Neoplasias/veterinaria , Radiocirugia/métodos , Receptores OX40/antagonistas & inhibidores , Receptores Toll-Like/antagonistas & inhibidores , Animales , Terapia Combinada , Citocinas , Enfermedades de los Perros/diagnóstico , Enfermedades de los Perros/etiología , Perros , Femenino , Expresión Génica , Linfocitos Infiltrantes de Tumor/inmunología , Linfocitos Infiltrantes de Tumor/metabolismo , Linfocitos Infiltrantes de Tumor/patología , Masculino , Neovascularización Patológica/metabolismo , Radioterapia Guiada por Imagen , Tomografía Computarizada por Rayos X , Resultado del Tratamiento , Microambiente Tumoral/inmunología
6.
J Immunol ; 202(10): 3087-3102, 2019 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-30971441

RESUMEN

Inflammatory monocytes have been shown to play key roles in cancer metastasis through promotion of tumor cell extravasation, growth, and angiogenesis. Monocyte recruitment to metastases is mediated primarily via the CCL2-CCR2 chemotactic axis. Thus, disruption of this axis represents an attractive therapeutic target for the treatment of metastatic disease. Losartan, a type I angiotensin II receptor (AT1R) antagonist, has been previously shown to have immunomodulatory actions involving monocyte and macrophage activity. However, the exact mechanisms accounting for these effects have not been fully elucidated. Therefore, we investigated the effects of losartan and its primary metabolite on CCL2-mediated monocyte recruitment and CCR2 receptor function using mouse tumor models and in vitro human monocyte cultures. We show, in this study, that losartan and its metabolite potently inhibit monocyte recruitment through the noncompetitive inhibition of CCL2-induced ERK1/2 activation, independent of AT1R activity. Studies in experimental metastasis models demonstrated that losartan treatment significantly reduced the metastatic burden in mice, an effect associated with a significant decrease in CD11b+/Ly6C+-recruited monocytes in the lungs. Collectively, these results indicate that losartan can exert antimetastatic activity by inhibiting CCR2 signaling and suppressing monocyte recruitment and therefore suggest that losartan (and potentially other AT1R blocker drugs) could be repurposed for use in cancer immunotherapy.


Asunto(s)
Bloqueadores del Receptor Tipo 1 de Angiotensina II/farmacología , Losartán/farmacología , Neoplasias Pulmonares , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Monocitos/inmunología , Proteínas de Neoplasias/inmunología , Neoplasias Experimentales , Receptor de Angiotensina Tipo 1/inmunología , Receptores CCR2/inmunología , Animales , Línea Celular Tumoral , Femenino , Humanos , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/inmunología , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/secundario , Sistema de Señalización de MAP Quinasas/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos ICR , Ratones Noqueados , Monocitos/patología , Metástasis de la Neoplasia , Neoplasias Experimentales/tratamiento farmacológico , Neoplasias Experimentales/inmunología , Neoplasias Experimentales/patología
7.
Vet Surg ; 50(4): 858-871, 2021 May.
Artículo en Inglés | MEDLINE | ID: mdl-33797775

RESUMEN

OBJECTIVE: To evaluate effects of Toll-like and nucleotide-binding oligomerization domain (NOD)-like receptor (TLR, NLR) ligand stimulation of equine mesenchymal stromal cells (MSCs) on antibacterial and immunomodulatory properties in vitro. STUDY DESIGN: Controlled laboratory study. SAMPLE POPULATION: Equine bone-marrow-derived MSCs (three horses). METHODS: MSCs were stimulated with TLR (polyinosinic:polycytidylic acid [pIC] and lipopolysaccharide [LPS]) and NLR agonists (γ-d-Glu-mDAP [IE-DAP]) for 2 h, and plated at 1 × 105 cells/well 24 h. MSC-conditioned media (MSC-CM) were collected and assessed for antimicrobial peptide cathelicidin/LL-37 production, bactericidal action against multidrug-resistant planktonic and biofilm Staphylococcus aureus and neutrophil phagocytosis. Bacterial growth was measured by plating bacteria and counting viable colonies, reading culture absorbance, and live-dead staining with confocal microscopy imaging. Following initial comparison of activating stimuli, TLR3-agonist pIC protocols (cell density during activation and plating, culture time, %serum) were further optimized for bactericidal activity and secretion of interleukin-8 (IL-8), monocyte-chemoattractant-protein (MCP-1), and cathelicidin/LL37. RESULTS: MSCs stimulation with pIC (p = .004) and IE-DAP (p = .03) promoted increased bactericidal activity, evidenced by reduced viable planktonic colony counts. PIC stimulation (2 × 106 cells/ml, 2 h, 10 µg/ml) further suppressed biofilm formation (p = .001), enhanced neutrophil bacterial phagocytosis (p = .009), increased MCP-1 secretion (p < .0001), and enhanced cathelicidin/LL-37 production, which was apparent when serum concentration in media was reduced to 1% (p = .01) and 2.5% (p = .05). CONCLUSION: TLR-3 pIC MSCs activation was most effective to enhance antibacterial and cytokine responses, which were affected by serum reduction. CLINICAL SIGNIFICANCE: In vitro TLR-3 activation of equine MSCs tested here may be a strategy to improve antibacterial properties of MSCs to treat antibiotic-resistant infections.


Asunto(s)
Péptidos Catiónicos Antimicrobianos/biosíntesis , Caballos/inmunología , Inmunomodulación/genética , Células Madre Mesenquimatosas/inmunología , Fagocitosis/efectos de los fármacos , Staphylococcus aureus/fisiología , Receptores Toll-Like/metabolismo , Animales , Biopelículas , Citocinas/biosíntesis , Farmacorresistencia Bacteriana Múltiple/inmunología , Neutrófilos/efectos de los fármacos , Catelicidinas
8.
Vet Surg ; 50(3): 650-658, 2021 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-33606293

RESUMEN

OBJECTIVE: To evaluate relative cytotoxicity of antibiotics to normal canine joint tissues in vitro. STUDY DESIGN: Experimental in vitro study. SAMPLE POPULATION: Chondrocytes and synoviocytes (three dogs); cartilage explants (three dogs); six dogs total. METHODS: Chondrocytes and synoviocytes from normal femoropatellar joints of three dogs were plated on 24-well plates (50 000 cells/cm2 , triplicate, 48 hours) and exposed to antibiotics (ampicillin sulbactam, vancomycin, cefazolin, ceftazidime, amikacin, enrofloxacin; 0.39-25 mg/mL, 24 hours). Viability was assessed by using trypan blue dye exclusion. Antibiotic concentrations at which 50% cell death occurred (half-maximal inhibitory concentration) were determined to rank antibiotics for relative cytotoxicity. Occurrence of caspase-3 expression after antibiotic exposure was assessed as an indication of apoptosis induction. Cartilage explants from three different dogs were minced and exposed to antibiotics (amikacin, ceftazidime, cefazolin, enrofloxacin; 5 mg/mL, 72 hours). Live/dead staining was performed, and fluorescence was visualized by using confocal microscopy. Percentage of live vs dead cells was quantitated. RESULTS: Viability of chondrocytes and synoviocytes decreased with increasing antibiotic concentrations. Half-maximal inhibitory concentrations were determined for synoviocytes (vancomycin 13.77, ampicillin sulbactam 3.07, amikacin 2.26, ceftazidime 1.62, cefazolin 1.48, enrofloxacin 1.25 mg/mL) and chondrocytes (vancomycin 8.65, ampicillin sulbactam 8.63, ceftazidime 3.16, amikacin 2.74, cefazolin 1.67, enrofloxacin 0.78 mg/mL). Caspase-3 expression was upregulated, providing evidence that apoptotic pathways were active in cell death. CONCLUSION: Half-maximal inhibitory concentration data provided evidence of lower toxicity of vancomycin and ampicillin sulbactam to joint tissues in vitro. CLINICAL SIGNIFICANCE: These results provide evidence to justify future in vitro work with osteoarthritic joint tissues and in vivo clinical trials to evaluate safety and efficacy of intra-articular antibiotics to treat dogs with septic arthritis.


Asunto(s)
Antibacterianos/toxicidad , Cartílago/efectos de los fármacos , Supervivencia Celular , Condrocitos/efectos de los fármacos , Perros , Sinoviocitos/efectos de los fármacos , Animales , Cadáver , Cartílago/trasplante , Supervivencia Celular/efectos de los fármacos , Femenino , Masculino
9.
J Immunol ; 200(4): 1261-1269, 2018 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-29352000

RESUMEN

Mesenchymal stem cells (MSC) exert immune modulatory properties and previous studies demonstrated suppressive effects of MSC treatment in animal models of allergic airway inflammation. However, the underlying mechanisms have not been fully elucidated. We studied the role of MSC in immune activation and subsequent recruitment of monocytes in suppressing airway hyperresponsiveness and airway inflammation using a mouse model of allergic airway inflammation. MSC administration prior to or after allergen challenge inhibited the development of airway inflammation in allergen-sensitized mice. This was accompanied by an influx of CCR2-positive monocytes, which were localized around injected MSC in the lungs. Notably, IL-10-producing monocytes and/or macrophages were also increased in the lungs. Systemic administration of liposomal clodronate or a CCR2 antagonist significantly prevented the suppressive effects of MSC. Activation of MSC by IFN-γ leading to the upregulation of CCL2 expression was essential for the suppressive effects, as administration of wild-type MSC into IFN-γ-deficient recipients, or IFN-γ receptor-deficient or CCL2-deficient MSC into wild-type mice failed to suppress airway inflammation. These results suggest that MSC activation by IFN-γ, followed by increased expression of CCL2 and recruitment of monocytes to the lungs, is essential for suppression by MSC in allergen-induced airway hyperresponsiveness and airway inflammation.


Asunto(s)
Células Madre Mesenquimatosas/inmunología , Monocitos/inmunología , Receptores CCR2/inmunología , Hipersensibilidad Respiratoria/inmunología , Animales , Movimiento Celular/inmunología , Femenino , Inflamación/inmunología , Inflamación/metabolismo , Células Madre Mesenquimatosas/metabolismo , Ratones , Ratones Endogámicos C57BL , Receptores CCR2/biosíntesis , Hipersensibilidad Respiratoria/metabolismo
10.
Vet Surg ; 49(5): 914-922, 2020 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-32310309

RESUMEN

OBJECTIVE: To determine the value of initial aerobic bacterial cultures of acute open traumatic wounds to predict bacterial species in wounds that become infected. STUDY DESIGN: Prospective clinical trial. ANIMALS: Sixty-four dogs with naturally occurring acute cutaneous traumatic wounds (2017-2018). METHODS: Initial swabs were taken from each wound prior to and after lavage and debridement for quantitative and qualitative aerobic bacterial culture. Cultures were repeated on wounds that displayed any clinical sign of infection within 14 days of presentation. RESULTS: Fewer bacteria were cultured from postlavage than from prelavage swabs in 43 of 50 (86%) acute wounds. All primary clinicians prescribed ß-lactam antibiotics to the dogs at initial presentation. All bacteria cultured from postlavage/debridement cultures at initial presentation were susceptible to the prophylactic antimicrobial prescribed. Postoperative infection was subsequently diagnosed in 14 of 64 (22%) dogs; 13 of these dogs had positive culture results. No correlation was detected between the results of initial wound cultures and the subsequent development of wound infection. Bacterial species present in the initial wound swab did not correlate with those subsequently cultured from infected tissues. CONCLUSION: Results of pretreatment wound cultures from open traumatic wounds in dogs were not predictive of bacterial species subsequently recovered from infected wounds. The bacterial burden present in pretreatment wounds was not predictive of whether wounds would ultimately become infected after surgical management. CLINICAL SIGNIFICANCE: Routine bacterial culturing of acute wounds is not likely to help predict subsequent wound infection, nor is it likely to accurately guide early selection of antimicrobials to treat wounds that become infected.


Asunto(s)
Antibacterianos/uso terapéutico , Bacterias/efectos de los fármacos , Infecciones Bacterianas/veterinaria , Perros/lesiones , Infección de Heridas/tratamiento farmacológico , Animales , Infecciones Bacterianas/tratamiento farmacológico , Femenino , Masculino , Estudios Prospectivos
11.
BMC Vet Res ; 15(1): 330, 2019 Sep 13.
Artículo en Inglés | MEDLINE | ID: mdl-31519215

RESUMEN

BACKGROUND: Non-specific immunotherapeutics have been evaluated previously in dogs, primarily for cancer treatment. However, there remains a need for a more broadly targeted, general purpose immunotherapeutic capable of activating innate immune defenses for non-specific protection or early treatment of viral and bacterial infections. To address need, our group has developed a liposomal immune stimulant (liposome-TLR complexes, LTC) containing TLR 3 and 9 agonists specifically designed to activate mucosal immune defenses in sites such as nasal cavity and oropharynx, following topical delivery. In this study, we evaluated the local immune stimulatory properties of LTC in vitro and in healthy purpose-bred dogs, including activation of cellular recruitment and cytokine production. The ability of LTC treatment to elicit effective antiviral immunity was assessed in dogs following a canine herpesvirus outbreak, and the impact of LTC treatment on the local microbiome of the oropharynx was also investigated. RESULTS: These studies revealed that LTC potently activated innate immune responses in vitro and triggered significant recruitment of inflammatory monocytes and T cells into the nasal cavity and oropharynx of healthy dogs. Administration of LTC to dogs shortly after an outbreak of canine herpesvirus infection resulted in significant reduction in clinical signs of infection. Interestingly, administration of LTC to healthy dogs did not disrupt the microbiome in the oropharynx, suggesting resiliency of the microflora to transient immune activation. CONCLUSIONS: Taken together, these results indicate that LTC administration mucosally to dogs can trigger local innate immune activation and activation of antiviral immunity, without significantly disrupting the composition of the local microbiome. Thus, the LTC immune stimulant has potential for use as a non-specific immunotherapy for prevention or early treatment of viral and bacterial infections in dogs.


Asunto(s)
Perros/inmunología , Inmunidad Innata/efectos de los fármacos , Liposomas/administración & dosificación , Membrana Mucosa/efectos de los fármacos , Administración a través de la Mucosa , Animales , Enfermedades de los Perros/inmunología , Enfermedades de los Perros/virología , Infecciones por Herpesviridae/veterinaria , Herpesvirus Cánido 1 , Membrana Mucosa/inmunología , Ácidos Nucleicos/inmunología , Orofaringe/microbiología
12.
Vet Dermatol ; 29(6): 482-e162, 2018 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-30141276

RESUMEN

BACKGROUND: Juvenile onset generalized demodicosis (JOGD) is thought to occur due to immunological abnormalities and is over-represented in pit bull terrier-type dogs. ANIMALS: Twelve pit bull terrier-type dogs with JOGD and 12 age-matched healthy pit bull terrier-type dogs. OBJECTIVE: To investigate immunological differences between age-matched healthy and JOGD pit bull terrier-type dogs by flow cytometry, multiplex, molecular and serological assays. METHODS AND MATERIALS: Flow cytometry quantified B cells expressing MHCII or surface-bound IgG, CD4+ T cells expressing MHCII, CD8 T cells expressing MHCII or CD11a, neutrophil and monocyte markers. Surface expression was quantified by calculating the geometric mean fluorescence index. The Wilcoxon rank sum test was used to compare median results for IL-2, IL-4, IL-6, IL-7, IL-8, IL-10, IL-13, IL-18, FOXP3, monocyte chemoattractant protein-1, GM-CSF, KC, IgE, IgA, IgG, IgM, C-reactive protein, lymphocyte, neutrophil and monocyte in the groups. IFN-gamma, IP-10, IL-15, IL-31 and TNF-alpha also were measured; however, insufficient dogs (<5) had values that were in range of the assay to allow for statistical evaluation. Significance was defined as P < 0.05. RESULTS: Serum concentrations of IL-2, IL-18 and MCP-1 were significantly higher (P = 0.01, P = 0.01, P = 0.04) in the JOGD group. Also, IgA median value was significantly higher (P = 0.002) in pit bull terrier-type dogs with JOGD. Flow cytometry revealed that T-cell, neutrophil and monocyte markers were not different between groups. CONCLUSIONS: Results suggest an appropriate compensatory immune response by pit bull terrier-type dogs in the JOGD group and do not support the explanation of global immune deficiency in these dogs.


Asunto(s)
Enfermedades de los Perros/parasitología , Infestaciones por Ácaros/veterinaria , Factores de Edad , Animales , Linfocitos B/inmunología , Linfocitos T CD4-Positivos/inmunología , Estudios de Casos y Controles , Quimiocina CCL2/sangre , Enfermedades de los Perros/inmunología , Perros , Femenino , Citometría de Flujo/veterinaria , Interleucinas/sangre , Masculino , Infestaciones por Ácaros/inmunología , Infestaciones por Ácaros/parasitología , Ácaros/inmunología
13.
Cancer Immunol Immunother ; 66(4): 523-535, 2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-28184968

RESUMEN

PD-L1 is an immune checkpoint protein that has emerged as a major signaling molecule involved with tumor escape from T cell immune responses. Studies have shown that intra-tumoral expression of PD-L1 can inhibit antitumor immune responses. However, it has recently been shown that expression of PD-L1 on myeloid cells from the tumor is a stronger indicator of prognosis than tumor cell PD-L1 expression. Therefore, it is important to understand the factors that govern the regulation of PD-L1 expression on tumor-infiltrating myeloid cells. We found that immature bone marrow monocytes in tumor-bearing mice had low levels of PD-L1 expression, while higher levels of expression were observed on monocytes in circulation. In contrast, macrophages found in tumor tissues expressed much higher levels of PD-L1 than circulating monocytes, implying upregulation by the tumor microenvironment. We demonstrated that tumor-conditioned media strongly induced increased PD-L1 expression by bone marrow-derived monocytes and TNF-α to be a cytokine that causes an upregulation of PD-L1 expression by the monocytes. Furthermore, we found production of TNF-α by the monocytes themselves to be a TLR2-dependent response to versican secreted by tumor cells. Thus, PD-L1 expression by tumor macrophages appears to be regulated in a different manner than by tumor cells themselves.


Asunto(s)
Antígeno B7-H1/metabolismo , Macrófagos/inmunología , Melanoma/inmunología , Monocitos/inmunología , Factor de Necrosis Tumoral alfa/inmunología , Animales , Antígeno B7-H1/genética , Regulación de la Expresión Génica , Humanos , Melanoma Experimental , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Neoplasias Experimentales , Receptor Toll-Like 2/metabolismo , Escape del Tumor , Microambiente Tumoral , Versicanos/metabolismo
14.
Stem Cells ; 34(7): 1709-29, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27066769

RESUMEN

Studies to evaluate the therapeutic potential of stem cells in humans would benefit from more realistic animal models. In veterinary medicine, companion animals naturally develop many diseases that resemble human conditions, therefore, representing a novel source of preclinical models. To understand how companion animal disease models are being studied for this purpose, we reviewed the literature between 2008 and 2015 for reports on stem cell therapies in dogs and cats, excluding laboratory animals, induced disease models, cancer, and case reports. Disease models included osteoarthritis, intervertebral disc degeneration, dilated cardiomyopathy, inflammatory bowel diseases, Crohn's fistulas, meningoencephalomyelitis (multiple sclerosis-like), keratoconjunctivitis sicca (Sjogren's syndrome-like), atopic dermatitis, and chronic (end-stage) kidney disease. Stem cells evaluated in these studies included mesenchymal stem-stromal cells (MSC, 17/19 trials), olfactory ensheathing cells (OEC, 1 trial), or neural lineage cells derived from bone marrow MSC (1 trial), and 16/19 studies were performed in dogs. The MSC studies (13/17) used adipose tissue-derived MSC from either allogeneic (8/13) or autologous (5/13) sources. The majority of studies were open label, uncontrolled studies. Endpoints and protocols were feasible, and the stem cell therapies were reportedly safe and elicited beneficial patient responses in all but two of the trials. In conclusion, companion animals with naturally occurring diseases analogous to human conditions can be recruited into clinical trials and provide realistic insight into feasibility, safety, and biologic activity of novel stem cell therapies. However, improvements in the rigor of manufacturing, study design, and regulatory compliance will be needed to better utilize these models. Stem Cells 2016;34:1709-1729.


Asunto(s)
Ensayos Clínicos como Asunto , Mascotas , Trasplante de Células Madre , Células Madre/citología , Animales , Modelos Animales de Enfermedad , Humanos
15.
Antimicrob Agents Chemother ; 58(10): 5954-63, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25070108

RESUMEN

Burkholderia pseudomallei, a facultative intracellular pathogen, causes severe infections and is inherently refractory to many antibiotics. Previous studies from our group have shown that interferon gamma (IFN-γ) interacts synergistically with the antibiotic ceftazidime to kill bacteria in infected macrophages. The present study aimed to identify the underlying mechanism of that interaction. We first showed that blocking reactive oxygen species (ROS) pathways reversed IFN-γ- and ceftazidime-mediated killing, which led to our hypothesis that IFN-γ-induced ROS interacted with ceftazidime to synergistically kill Burkholderia bacteria. Consistent with this hypothesis, we also observed that buthionine sulfoximine (BSO), another inducer of ROS, could substitute for IFN-γ to similarly potentiate the effect of ceftazidime on intracellular killing. Next, we observed that IFN-γ induced ROS-mediated killing of intracellular but not extracellular bacteria. On the other hand, ceftazidime effectively reduced extracellular bacteria but was not capable of intracellular killing when applied at 10 µg/ml. We investigated the exact role of IFN-γ-induced ROS responses on intracellular bacteria and notably observed a lack of actin polymerization associated with Burkholderia bacteria in IFN-γ-treated macrophages, which led to our finding that IFN-γ-induced ROS blocks vacuolar escape. Based on these results, we propose a model in which synergistically reduced bacterial burden is achieved primarily through separate and compartmentalized killing: intracellular killing by IFN-γ-induced ROS responses and extracellular killing by ceftazidime. Our findings suggest a means of enhancing antibiotic activity against Burkholderia bacteria through combination with drugs that induce ROS pathways or otherwise target intracellular spread and/or replication of bacteria.


Asunto(s)
Burkholderia pseudomallei/efectos de los fármacos , Ceftazidima/farmacología , Interferón gamma/farmacología , Especies Reactivas de Oxígeno/metabolismo , Animales , Ceftazidima/química , Línea Celular , Células Cultivadas , Femenino , Citometría de Flujo , Macrófagos , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Endogámicos ICR , Microscopía Fluorescente , Especies Reactivas de Oxígeno/química
16.
PLoS Pathog ; 8(3): e1002587, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22438809

RESUMEN

Pulmonary Francisella tularensis and Burkholderia pseudomallei infections are highly lethal in untreated patients, and current antibiotic regimens are not always effective. Activating the innate immune system provides an alternative means of treating infection and can also complement antibiotic therapies. Several natural agonists were screened for their ability to enhance host resistance to infection, and polysaccharides derived from the Acai berry (Acai PS) were found to have potent abilities as an immunotherapeutic to treat F. tularensis and B. pseudomallei infections. In vitro, Acai PS impaired replication of Francisella in primary human macrophages co-cultured with autologous NK cells via augmentation of NK cell IFN-γ. Furthermore, Acai PS administered nasally before or after infection protected mice against type A F. tularensis aerosol challenge with survival rates up to 80%, and protection was still observed, albeit reduced, when mice were treated two days post-infection. Nasal Acai PS administration augmented intracellular expression of IFN-γ by NK cells in the lungs of F. tularensis-infected mice, and neutralization of IFN-γ ablated the protective effect of Acai PS. Likewise, nasal Acai PS treatment conferred protection against pulmonary infection with B. pseudomallei strain 1026b. Acai PS dramatically reduced the replication of B. pseudomallei in the lung and blocked bacterial dissemination to the spleen and liver. Nasal administration of Acai PS enhanced IFN-γ responses by NK and γδ T cells in the lungs, while neutralization of IFN-γ totally abrogated the protective effect of Acai PS against pulmonary B. pseudomallei infection. Collectively, these results demonstrate Acai PS is a potent innate immune agonist that can resolve F. tularensis and B. pseudomallei infections, suggesting this innate immune agonist has broad-spectrum activity against virulent intracellular pathogens.


Asunto(s)
Arecaceae/química , Inmunidad Innata/efectos de los fármacos , Melioidosis/prevención & control , Neumonía/tratamiento farmacológico , Polisacáridos/farmacología , Tularemia/prevención & control , Administración Intranasal , Animales , Burkholderia pseudomallei/efectos de los fármacos , Burkholderia pseudomallei/inmunología , Modelos Animales de Enfermedad , Femenino , Francisella tularensis/efectos de los fármacos , Francisella tularensis/inmunología , Interferón gamma/metabolismo , Células Asesinas Naturales/efectos de los fármacos , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/metabolismo , Longevidad/efectos de los fármacos , Pulmón/efectos de los fármacos , Pulmón/inmunología , Pulmón/metabolismo , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Macrófagos/metabolismo , Melioidosis/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Extractos Vegetales/administración & dosificación , Extractos Vegetales/farmacología , Neumonía/inmunología , Neumonía/microbiología , Polisacáridos/administración & dosificación , Polisacáridos/aislamiento & purificación , Tularemia/inmunología
17.
J Immunol ; 189(12): 5612-21, 2012 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-23136203

RESUMEN

Vaccine adjuvant-induced inflammation augments vaccine immunity in part by recruiting APCs to vaccine draining lymph nodes (LNs). However, the role of one APC subtype, inflammatory monocytes, in regulating vaccine immunity in healthy animals has not been fully examined in detail. Therefore, vaccine-mediated monocyte recruitment and subsequent immune responses were investigated using murine vaccination models and in vitro assays. Recruitment of inflammatory monocytes to vaccine draining LNs was rapid and mediated primarily by local production of MCP-1, as revealed by studies in MCP-1(-/-) mice. Interrupting monocyte recruitment to LNs by either transient monocyte depletion or monocyte migration blockade led to marked amplification of both cellular and humoral immune responses to vaccination. These results were most consistent with the idea that rapidly mobilized inflammatory monocytes were actually suppressing vaccine responses. The suppressive nature of vaccine-elicited monocytes was confirmed using in vitro cocultures of murine monocytes and T cells. Furthermore, it was determined that inflammatory monocytes suppressed T cell responses by sequestering cysteine, as cysteine supplementation in vitro and in vivo appreciably augmented vaccine responses. These findings indicated, therefore, that vaccination-elicited inflammation, although necessary for effective immunity, also generated potent counter-regulatory immune responses that were mediated primarily by inflammatory monocytes. Therefore, interrupting monocyte-mediated vaccine counterregulatory responses may serve as an effective new strategy for broadly amplifying vaccine immunity.


Asunto(s)
Vacunas contra el Cáncer/antagonistas & inhibidores , Vacunas contra el Cáncer/inmunología , Tolerancia Inmunológica/inmunología , Monocitos/inmunología , Monocitos/patología , Vacunas de ADN/antagonistas & inhibidores , Vacunas de ADN/inmunología , Animales , Vacunas contra el Cáncer/administración & dosificación , Cationes , Línea Celular Tumoral , Inhibición de Migración Celular/genética , Inhibición de Migración Celular/inmunología , Cisteína/administración & dosificación , Tolerancia Inmunológica/genética , Inflamación/genética , Inflamación/inmunología , Inflamación/patología , Liposomas , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Endogámicos ICR , Ratones Noqueados , Monocitos/metabolismo , Receptores CCR2/antagonistas & inhibidores , Receptores CCR2/deficiencia , Receptores CCR2/genética , Vacunas de ADN/administración & dosificación
18.
J Am Vet Med Assoc ; 262(S1): S121-S130, 2024 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-38437789

RESUMEN

Neurological diseases and injuries in veterinary patients (horses, dogs, and cats) are complex, and effective treatment options are limited. Neuronal loss, damage to nerve conduction pathways, and inflammation and scarring associated with spinal cord injury pose major challenges in managing many neurological diseases. Furthermore, most of these neuropathologies lack definitive pharmacological treatments, driving interest and research into novel interventions. Our objective is to provide a narrative review of the current literature surrounding cellular therapies including neuronal and glial stem cells, neurotrophic factors, mesenchymal stem or stromal cells, and cells derived from induced pluripotent stem cells for the treatment of diverse neurological pathologies. Cellular therapies have the potential for cellular replacement, immune modulation, and paracrine signaling and the flexibility of being used alone or alongside surgical intervention. Mesenchymal stem or stromal cells are arguably the most researched cellular therapy and have been administered intrathecally, IV, intra-arterially, intranasally, and intraspinally with few adverse reactions. Limited clinical and experimental studies have suggested efficacy in diseases including acute spinal cord injury and intervertebral disc disease. Little is currently known about the safety and efficacy of neural stem cells, precursor cell administration, and induced pluripotent stem cell-derived treatments. Further research is necessary to determine the efficacy and long-term safety of cellular therapies. Future aims should include larger controlled clinical trials in companion animals for common neurologic conditions including acute spinal cord injury, intervertebral disc disease, peripheral nerve injury, degenerative neuropathies, and age-associated cognitive decline.


Asunto(s)
Enfermedades de los Perros , Enfermedades de los Caballos , Enfermedades del Sistema Nervioso , Animales , Perros , Enfermedades de los Perros/terapia , Caballos , Enfermedades del Sistema Nervioso/veterinaria , Enfermedades del Sistema Nervioso/terapia , Enfermedades de los Caballos/terapia , Tratamiento Basado en Trasplante de Células y Tejidos/veterinaria
19.
Front Immunol ; 15: 1397590, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38933260

RESUMEN

Chronic inflammatory enteropathy (CIE) is a common condition in dogs causing recurrent or persistent gastrointestinal clinical signs. Pathogenesis is thought to involve intestinal mucosal inflammatory infiltrates, but histopathological evaluation of intestinal biopsies from dogs with CIE fails to guide treatment, inform prognosis, or correlate with clinical remission. We employed single-cell RNA sequencing to catalog and compare the diversity of cells present in duodenal mucosal endoscopic biopsies from 3 healthy dogs and 4 dogs with CIE. Through characterization of 35,668 cells, we identified 31 transcriptomically distinct cell populations, including T cells, epithelial cells, and myeloid cells. Both healthy and CIE samples contributed to each cell population. T cells were broadly subdivided into GZMAhigh (putatively annotated as tissue resident) and IL7Rhigh (putatively annotated as non-resident) T cell categories, with evidence of a skewed proportion favoring an increase in the relative proportion of IL7Rhigh T cells in CIE dogs. Among the myeloid cells, neutrophils from CIE samples exhibited inflammatory (SOD2 and IL1A) gene expression signatures. Numerous differentially expressed genes were identified in epithelial cells, with gene set enrichment analysis suggesting enterocytes from CIE dogs may be undergoing stress responses and have altered metabolic properties. Overall, this work reveals the previously unappreciated cellular heterogeneity in canine duodenal mucosa and provides new insights into molecular mechanisms which may contribute to intestinal dysfunction in CIE. The cell type gene signatures developed through this study may also be used to better understand the subtleties of canine intestinal physiology in health and disease.


Asunto(s)
Enfermedades de los Perros , Duodeno , Perfilación de la Expresión Génica , Análisis de la Célula Individual , Transcriptoma , Animales , Perros , Duodeno/patología , Duodeno/inmunología , Duodeno/metabolismo , Enfermedades de los Perros/genética , Enfermedades de los Perros/inmunología , Enfermedades de los Perros/patología , Mucosa Intestinal/inmunología , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patología , Enfermedad Crónica , Masculino , Femenino , Linfocitos T/inmunología , Linfocitos T/metabolismo
20.
Vet Sci ; 11(4)2024 Apr 07.
Artículo en Inglés | MEDLINE | ID: mdl-38668434

RESUMEN

Ocular surface squamous neoplasia (OSSN) represents the most common conjunctival tumor in horses and frequently results in vision loss and surgical removal of the affected globe. Multiple etiologic factors have been identified as contributing to OSSN progression, including solar radiation exposure, genetic mutations, and a lack of periocular pigmentation. Response to conventional treatments has been highly variable, though our recent work indicates that these tumors are highly responsive to local immunotherapy. In the present study, we extended our investigation of OSSN in horses to better understand how the ocular transcriptome responds to the presence of the tumor and how the ocular surface microbiome may also be altered by the presence of cancer. Therefore, we collected swabs from the ventral conjunctival fornix from 22 eyes in this study (11 with cytologically or histologically confirmed OSSN and 11 healthy eyes from the same horses) and performed RNA sequencing and 16S microbial sequencing using the same samples. Microbial 16s DNA sequencing and bulk RNA sequencing were both conducted using an Illumina-based platform. In eyes with OSSN, we observed significantly upregulated expression of genes and pathways associated with inflammation, particularly interferon. Microbial diversity was significantly reduced in conjunctival swabs from horses with OSSN. We also performed interactome analysis and found that three bacterial taxa (Actinobacillus, Helcococcus and Parvimona) had significant correlations with more than 100 upregulated genes in samples from animals with OSSN. These findings highlight the inflammatory nature of OSSN in horses and provide important new insights into how the host ocular surface interacts with certain microbial populations. These findings suggest new strategies for the management of OSSN in horses, which may entail immunotherapy in combination with ocular surface probiotics or prebiotics to help normalize ocular cell and microbe interactions.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA