Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 66
Filtrar
1.
Front Vet Sci ; 11: 1382652, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38803805

RESUMO

Low back pain poses a significant societal burden, with progressive intervertebral disc degeneration (IDD) emerging as a pivotal contributor to chronic pain. Improved animal models of progressive IDD are needed to comprehensively investigate new diagnostic and therapeutic approaches to managing IDD. Recent studies underscore the immune system's involvement in IDD, particularly with regards to the role of immune privileged tissues such as the nucleus pulposus (NP) becoming an immune targeting following initial disc injury. We therefore hypothesized that generating an active immune response against NP antigens with an NP vaccine could significantly accelerate and refine an IDD animal model triggered by mechanical puncture of the disc. To address this question, rabbits were immunized against NP antigens following disc puncture, and the impact on development of progressive IDD was assessed radiographically, functionally, and histologically compared between vaccinated and non-vaccinated animals over a 12-week period. Immune responses to NP antigens were assessed by ELISA and Western blot. We found that the vaccine elicited strong immune responses against NP antigens, including a dominant ~37 kD antigen. Histologic evaluation revealed increases IDD in animals that received the NP vaccine plus disc puncture, compared to disc puncture and vaccine only animals. Imaging evaluation evidenced a decrease in disc height index and higher scores of disc degeneration in animals after disc punctures and in those animals that received the NP vaccine in addition to disc puncture. These findings therefore indicate that it is possible to elicit immune responses against NP antigens in adult animals, and that these immune responses may contribute to accelerated development of IDD in a novel immune-induced and accelerated IDD model.

2.
Stem Cells ; 2024 May 25.
Artigo em Inglês | MEDLINE | ID: mdl-38795363

RESUMO

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.

3.
Vet Clin North Am Equine Pract ; 39(3): 565-578, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37442729

RESUMO

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.


Assuntos
Doenças dos Cavalos , Células-Tronco Mesenquimais , Cavalos , Animais , Doenças dos Cavalos/terapia , Plaquetas , Antibacterianos
4.
J Orthop Res ; 41(4): 902-912, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-36030381

RESUMO

Osteoarthritis (OA) is a leading cause of morbidity among aging populations, yet symptom and/or disease-modification remains elusive. Adipose-derived mesenchymal stromal cells (adMSCs) have demonstrated immunomodulatory and anti-inflammatory properties that may alleviate clinical signs and interrupt disease onset and progression. Indeed, multiple manuscripts have evaluated intra-articular administration of adMSCs as a therapeutic; however, comparatively few evaluations of systemic delivery methods have been published. Therefore, the aim of this study was to evaluate the short-term impact of intravenous (IV) delivery of allogeneic adMSCs in an established model of spontaneous OA, the Hartley guinea pig. Animals with moderate OA received once weekly injections of 2 × 106 adMSCs or vehicle control for 4 weeks in peripheral veins; harvest occurred 2 weeks after the final injection. Systemic administration of adMSCs resulted in no adverse effects and was efficacious in reducing clinical signs of OA (as assessed by computer-aided gait analysis) compared to control injected animals. Further, there were significant decreases in key inflammatory mediators (including monocyte chemoattractant protein-1, tumor necrosis factor, and prostaglandin E2 ) both systemically (liver, kidney, and serum) and locally in the knee (joint tissues and synovial fluid) in animals treated with IV adMSCs relative to controls (as per enzyme-linked immunosorbent assay and/or immunohistochemistry, dictated by tissue sample). Thus, systemic administration of adMSCs by IV injection significantly improved gait parameters and reduced both systemic and intra-articular inflammatory mediators in animals with OA. These findings demonstrate the potential utility of alternative delivery approaches for cellular therapy of OA, particularly for patients with multiple affected joints.


Assuntos
Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais , Osteoartrite do Joelho , Osteoartrite , Animais , Cobaias , Injeções Intravenosas , Osteoartrite/patologia , Articulação do Joelho/patologia , Inflamação , Injeções Intra-Articulares , Osteoartrite do Joelho/patologia , Transplante de Células-Tronco Mesenquimais/métodos
5.
Front Vet Sci ; 10: 1293199, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-38162475

RESUMO

Introduction: Equine recurrent uveitis (ERU), an immune mediated disease characterized by repeated episodes of intra-ocular inflammation, affects 25% of horses in the USA and is the most common cause of glaucoma, cataracts, and blindness. Mesenchymal stromal cells (MSCs) have immunomodulatory properties, which are upregulated by preconditioning with toll-like receptor agonists. The objective was to evaluate safety and migration of TLR-3 agonist polyinosinic, polycytidylic acid (pIC)-activated MSCs injected subconjunctivally in healthy horses prior to clinical application in horses with ERU. We hypothesized that activated allogeneic MSCs injected subconjunctivally would not induce ocular or systemic inflammation and would remain in the conjunctiva for >14 days. Methods: Bulbar subconjunctiva of two horses was injected with 10 × 106 pIC-activated (10 µg/mL, 2 h) GFP-labeled MSCs from one donor three times at two-week intervals. Vehicle (saline) control was injected in the contralateral conjunctiva. Horses received physical and ophthalmic exams [slit lamp biomicroscopy, rebound tonometry, fundic examination, and semiquantitative preclinical ocular toxicology scoring (SPOTS)] every 1-3 days. Systemic inflammation was assessed via CBC, fibrinogen, and serum amyloid A (SAA). Horses were euthanized 14 days following final injection. Full necropsy and histopathology were performed to examine ocular tissues and 36 systemic organs for MSC presence via IVIS Spectrum. Anti-GFP immunohistochemistry was performed on ocular tissues. Results: No change in physical examinations was noted. Bloodwork revealed fibrinogen 100-300 mg/dL (ref 100-400) and SAA 0-25 µg/mL (ref 0-20). Ocular effects of the subjconjucntival injection were similar between MSC and control eyes on SPOTS grading system, with conjunctival hypermia, chemosis and ocular discharge noted bilaterally, which improved without intervention within 14 days. All other ocular parameters were unaffected throughout the study. Necropsy and histopathology revealed no evidence of systemic inflammation. Ocular histopathology was similar between MSC and control eyes. Fluorescent imaging analysis did not locate MSCs. Immunohistochemistry did not identify intact MSCs in the conjunctiva, but GFP-labeled cellular components were present in conjunctival phagocytic cells. Discussion: Allogeneic pIC-activated conjunctival MSC injections were well tolerated. GFP-labeled tracking identified MSC components phagocytosed by immune cells subconjunctivally. This preliminary safety and tracking information is critical towards advancing immune conditioned cellular therapies to clinical trials in horses.

6.
Vet Sci ; 9(11)2022 Nov 04.
Artigo em Inglês | MEDLINE | ID: mdl-36356087

RESUMO

Antimicrobial resistance and biofilm formation both present challenges to treatment of bacterial infections with conventional antibiotic therapy and serve as the impetus for development of improved therapeutic approaches. Mesenchymal stromal cell (MSC) therapy exerts an antimicrobial effect as demonstrated in multiple acute bacterial infection models. This effect can be enhanced by pre-conditioning the MSC with Toll or Nod-like receptor stimulation, termed activated cellular therapy (ACT). The purpose of this review is to summarize the current literature on mechanisms of antimicrobial activity of MSC with emphasis on enhanced effects through receptor agonism, and data supporting use of ACT in treatment of bacterial infections in veterinary species including dogs, cats, and horses with implications for further treatment applications. This review will advance the field's understanding of the use of activated antimicrobial cellular therapy to treat infection, including mechanisms of action and potential therapeutic applications.

7.
Clin Cancer Res ; 28(4): 662-676, 2022 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-34580111

RESUMO

PURPOSE: There is increasing recognition that progress in immuno-oncology could be accelerated by evaluating immune-based therapies in dogs with spontaneous cancers. Osteosarcoma (OS) is one tumor for which limited clinical benefit has been observed with the use of immune checkpoint inhibitors. We previously reported the angiotensin receptor blocker losartan suppressed metastasis in preclinical mouse models through blockade of CCL2-CCR2 monocyte recruitment. Here we leverage dogs with spontaneous OS to determine losartan's safety and pharmacokinetics associated with monocyte pharmacodynamic endpoints, and assess its antitumor activity, in combination with the kinase inhibitor toceranib. PATIENTS AND METHODS: CCL2 expression, monocyte infiltration, and monocyte recruitment by human and canine OS tumors and cell lines were assessed by gene expression, ELISA, and transwell migration assays. Safety and efficacy of losartan-toceranib therapy were evaluated in 28 dogs with lung metastatic OS. Losartan PK and monocyte PD responses were assessed in three dose cohorts of dogs by chemotaxis, plasma CCL2, and multiplex cytokine assays, and RNA-seq of losartan-treated human peripheral blood mononuclear cells. RESULTS: Human and canine OS cells secrete CCL2 and elicit monocyte migration, which is inhibited by losartan. Losartan PK/PD studies in dogs revealed that a 10-fold-higher dose than typical antihypertensive dosing was required for blockade of monocyte migration. Treatment with high-dose losartan and toceranib was well-tolerated and induced a clinical benefit rate of 50% in dogs with lung metastases. CONCLUSIONS: Losartan inhibits the CCL2-CCR2 axis, and in combination with toceranib, exerts significant biological activity in dogs with metastatic osteosarcoma, supporting evaluation of this drug combination in patients with pediatric osteosarcoma. See related commentary by Weiss et al., p. 571.


Assuntos
Neoplasias Ósseas , Doenças do Cão , Osteossarcoma , Animais , Neoplasias Ósseas/tratamento farmacológico , Neoplasias Ósseas/veterinária , Doenças do Cão/tratamento farmacológico , Cães , Humanos , Leucócitos Mononucleares , Losartan/farmacologia , Losartan/uso terapêutico , Camundongos , Monócitos , Osteossarcoma/tratamento farmacológico , Osteossarcoma/veterinária
8.
Vet Surg ; 50(3): 650-658, 2021 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-33606293

RESUMO

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.


Assuntos
Antibacterianos/toxicidade , Cartilagem/efeitos dos fármacos , Sobrevivência Celular , Condrócitos/efeitos dos fármacos , Cães , Sinoviócitos/efeitos dos fármacos , Animais , Cadáver , Cartilagem/transplante , Sobrevivência Celular/efeitos dos fármacos , Feminino , Masculino
9.
J Vet Emerg Crit Care (San Antonio) ; 30(5): 525-533, 2020 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-32705762

RESUMO

OBJECTIVE: To evaluate the clinical efficacy of a single infusion of hyperimmune plasma (HIP) in dogs with canine parvovirus (CPV). DESIGN: Prospective, randomized, placebo-controlled clinical trial. SETTING: University teaching hospital. ANIMALS: Client-owned dogs with naturally occurring CPV. INTERVENTIONS: Dogs presenting for CPV treatment (n = 31) underwent cardiovascular resuscitation and were randomized to receive a single dose of either HIP (10 mL/kg IV) or placebo (0.9% sodium chloride [10 mL/kg IV]) during the first 6 hours of hospitalization. All dogs were treated with a standardized treatment protocol (IV fluid therapy [120 mL/kg/d isotonic crystalloids], cefoxitin [30 mg/kg IV q 8 h], maropitant [1 mg/kg IV q 24 h], and buprenorphine [0.01-0.02 mg/kg IV q 8 h]) until hospital discharge. MEASUREMENTS AND MAIN RESULTS: Dogs treated with HIP (n = 16) demonstrated a lower shock index at 24 hours (median = 0.77, range: 0.5-1.5) than those treated with placebo (n = 15, median = 1.34, range: 0.5-1.7; P = 0.02). Plasma lactate concentration was lower at 24 hours in HIP-treated dogs (median = 1.3 mmol/L, range: 0.9-3.4 mmol/L) than in placebo-treated dogs (median = 2.1 mmol/L, range: 1.1-3.4 mmol/L; P = 0.01). There was no difference in duration of hospitalization when comparing HIP-treated dogs (median = 3.2 days, range: 0.83-10 days) to placebo-treated dogs (median = 2.83 days, range: 1-8.38 days; P = 0.35). Survival was 16 of 16 (100%) for the HIP group and 14 of 15 (93.3%) for the placebo group (P = 0.32). CONCLUSIONS: HIP at 10 mL/kg IV administered to dogs with CPV within the first 6 hours of hospitalization improves markers of shock during the initial 24 hours of hospitalization. No effects were observed on duration of hospitalization or mortality; however, this study was underpowered to evaluate these effects. HIP was well tolerated in this population of critically ill dogs.


Assuntos
Doenças do Cão/terapia , Enterite/veterinária , Imunização Passiva/veterinária , Infecções por Parvoviridae/veterinária , Parvovirus Canino , Animais , Cães , Enterite/terapia , Feminino , Hidratação/veterinária , Masculino , Infecções por Parvoviridae/terapia , Estudos Prospectivos
10.
Vet Surg ; 49(5): 914-922, 2020 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-32310309

RESUMO

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.


Assuntos
Antibacterianos/uso terapêutico , Bactérias/efeitos dos fármacos , Infecções Bacterianas/veterinária , Cães/lesões , Infecção dos Ferimentos/tratamento farmacológico , Animais , Infecções Bacterianas/tratamento farmacológico , Feminino , Masculino , Estudos Prospectivos
11.
Equine Vet J ; 52(5): 654-663, 2020 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-31971273

RESUMO

Mesenchymal stem cells (MSCs) are widely used for treatment of musculoskeletal diseases in horses, but there is ongoing debate regarding the relative safety and efficacy of allogeneic MSCs, compared with autologous equine MSCs. This review summarises the currently available published data regarding the therapeutic use of autologous and allogeneic MSCs in horses. Arguments that have been advanced against the use of allogeneic MSCs include higher risk of immunological reactions and shorter cell survival times following injection. Arguments favouring the use of allogeneic MSCs include the ability to bank cells and reduce the time to treatment, to collect MSCs from younger donor animals and the ability to manipulate banked cells prior to administration. In vitro studies and a limited set of experimental in vivo studies have indicated that adverse immunological reactions may occur when allogeneic MSCs are administered to horses. However, newer studies lack evidence of inflammatory reactions or adverse clinical responses when allogeneic MSCs are administered and compared with autologous MSCs. Thus, while the relative merits of allogeneic vs autologous MSCs for treatment of musculoskeletal injuries in horses have not been fully established, accumulating evidence from studies in horses suggests that allogeneic MSCs maybe a safe alternative to autologous MSCs. Large, properly designed, randomised trials in addition to careful immunological evaluation of short-term and long-term, local and systemic immune responses are needed to more fully resolve the issue.


Assuntos
Transplante de Células-Tronco Hematopoéticas/veterinária , Doenças dos Cavalos , Transplante de Células-Tronco Mesenquimais/veterinária , Células-Tronco Mesenquimais , Doenças Musculoesqueléticas/veterinária , Animais , Cavalos , Transplante Homólogo/veterinária
12.
Equine Vet J ; 52(4): 601-612, 2020 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-31821594

RESUMO

BACKGROUND: Allogeneic and autologous bone marrow-derived mesenchymal stem cells (BMDMSCs) have been administered in equine joints for their anti-inflammatory effects. However, allogeneic BMDMSC offer multiple clinical and practical advantages. Therefore, it is important to determine the relative effectiveness of allogeneic vs autologous BMDMSCs. OBJECTIVES: The objective of the study was to compare the inflamed joint response to autologous vs allogeneic BMDMSCs injections, and to determine if either treatment generated an anti-inflammatory effect. STUDY DESIGN: Randomised controlled study. METHOD: Bone marrow was harvested from eight horses. Autologous BMDMSCs and pooled allogeneic BMDMSCs were culture expanded, cryopreserved and thawed immediately prior to administration. Ten million autologous BMDMSCs were administered with 75 ng rIL-1ß into one tarsocrural joint and the contralateral tarsocrural joint received allogeneic BMDMSC plus 75 ng rIL-1ß. Repeat injections were performed with the same treatment administered into the same joint. Four additional horses received 75 ng rIL-1ß alone in a single tarsocrural joint. Clinical parameters (lameness, joint circumference and joint effusion) and synovial fluid parameters, including nucleated cell count (NCC), differential cell count, total protein (TP), prostaglandin E2 (PGE2 ) and C-reactive protein (CRP), were measured at baseline, 6, 12, 24, 72, 168 and 336 hours post-injection. RESULTS: No difference was detected between autologous and allogeneic treatment groups with respect to subjective lameness, joint effusion, joint circumference, NCC, TP, differential cell count, CRP or PGE2 . Neither autologous nor allogeneic treatments resulted in an improvement in clinical or cytological parameters over that elicited by rIL-1ß alone. MAIN LIMITATIONS: A single dose of rIL-1ß was evaluated and resulted in a severe synovitis which may have been too severe to observe a BMDMSC-mediated effect. CONCLUSIONS: This study revealed that allogeneic and autologous BMDMSCs resulted in an equivalent clinical and cytological response. Allogeneic and autologous BMDMSCs were equally ineffective in reducing the inflammatory response from acute rIL-1ß-induced joint inflammation in horses.


Assuntos
Transplante de Células-Tronco Hematopoéticas/veterinária , Doenças dos Cavalos , Transplante de Células-Tronco Mesenquimais/veterinária , Células-Tronco Mesenquimais , Sinovite/veterinária , Animais , Medula Óssea , Cavalos , Inflamação/veterinária , Injeções Intra-Articulares/veterinária , Interleucina-1beta , Líquido Sinovial
13.
J Immunol ; 202(10): 3087-3102, 2019 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-30971441

RESUMO

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.


Assuntos
Bloqueadores do Receptor Tipo 1 de Angiotensina II/farmacologia , Losartan/farmacologia , Neoplasias Pulmonares , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Monócitos/imunologia , Proteínas de Neoplasias/imunologia , Neoplasias Experimentais , Receptor Tipo 1 de Angiotensina/imunologia , Receptores CCR2/imunologia , Animais , Linhagem Celular Tumoral , Feminino , Humanos , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/imunologia , Neoplasias Pulmonares/patologia , Neoplasias Pulmonares/secundário , Sistema de Sinalização das MAP Quinases/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos ICR , Camundongos Knockout , Monócitos/patologia , Metástase Neoplásica , Neoplasias Experimentais/tratamento farmacológico , Neoplasias Experimentais/imunologia , Neoplasias Experimentais/patologia
14.
Front Vet Sci ; 5: 208, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30234134

RESUMO

Background: The effects of recombinant interleukin-1ß (rIL-1ß) have been described for the middle carpal joint (MCJ). However, we are unaware of any studies that have described the cytological response of the tibiotarsal joint (TTJ) to rIL-1ß or compared the clinical and cytological responses of the MCJ to the TTJ following the administration of intra-articular rIL-1ß. Such information is critical for researchers planning to use rIL-1ß to create acute synovitis models in horses. Objectives: To compare the clinical and cytological responses of the MCJ to the TTJ following administration of intra-articular rIL-1ß. Methods: Twelve horses were used for the study. Eight horses received 75 ng of rIL-1ß into the MCJ and four horses received 75 ng of rIL-1ß into the TTJ. Clinical and cytological outcome parameters including lameness, joint circumference, joint effusion score, total nucleated cell count, cellular differentials, C-reactive protein, and prostaglandin-E2 concentrations were determined at baseline and multiple post-treatment time points over a 336 h period (2 weeks). Results: Recombinant IL-1ß administered into the TTJ resulted in a significantly greater respiratory rate at 24 h and heart rate at 12 h when compared to rIL-1ß administered into the MCJ. In addition, the TTJ had a significantly greater increase in joint circumference at 24 post-injection hour (PIH) and subjective effusion grade at 24 PIH and 336 PIH. The MCJ had significantly higher total protein concentration at 6 PIH, and a significantly higher NCC at 24 and 72 PIH when compared to the TTJ. Conversely, the TTJ had significantly higher neutrophilic infiltration than the MCJ at 6 PIH and 168 PIH. Conclusions: This study establishes that the same intra-articular dose of rIL-1 ß elicits significantly different clinical and cytological responses in the MCJ compared to the TTJ in the equine model of intra-articular synovitis. In addition, clinical and cytological evidence of synovitis may persist up to or >1 week following intra-articular administration of rIL-1 ß.

15.
Vet Dermatol ; 29(6): 482-e162, 2018 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-30141276

RESUMO

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.


Assuntos
Doenças do Cão/parasitologia , Infestações por Ácaros/veterinária , Fatores Etários , Animais , Linfócitos B/imunologia , Linfócitos T CD4-Positivos/imunologia , Estudos de Casos e Controles , Quimiocina CCL2/sangue , Doenças do Cão/imunologia , Cães , Feminino , Citometria de Fluxo/veterinária , Interleucinas/sangue , Masculino , Infestações por Ácaros/imunologia , Infestações por Ácaros/parasitologia , Ácaros/imunologia
16.
Cancer Immunol Res ; 6(10): 1260-1273, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-30012633

RESUMO

Tumor-associated macrophages (TAMs) express programmed cell death ligand 1 (PD-L1) and contribute to the immune-suppressive tumor microenvironment. Although the role of the PD-L1 and PD-1 interaction to regulate T-cell suppression is established, less is known about PD-L1 signaling in macrophages and how these signals may affect the function of TAMs. We used in vitro and in vivo models to investigate PD-L1 signaling in macrophages and the effects of PD-L1 antibody treatment on TAM responses. Treatment of mouse and human macrophages with PD-L1 antibodies increased spontaneous macrophage proliferation, survival, and activation (costimulatory molecule expression, cytokine production). Similar changes were observed in macrophages incubated with soluble CD80 and soluble PD-1, and in PD-L1-/- macrophages. Macrophage treatment with PD-L1 antibodies upregulated mTOR pathway activity, and RNAseq analysis revealed upregulation of multiple macrophage inflammatory pathways. In vivo, treatment with PD-L1 antibody resulted in increased tumor infiltration with activated macrophages. In tumor-bearing RAG-/- mice, upregulated costimulatory molecule expression by TAMs and reduced tumor growth were observed. Combined PD-1/ PD-L1 antibody treatment of animals with established B16 melanomas cured half of the treated mice, whereas treatment with single antibodies had little therapeutic effect. These findings indicate that PD-L1 delivers a constitutive negative signal to macrophages, resulting in an immune-suppressive cell phenotype. Treatment with PD-L1 antibodies reverses this phenotype and triggers macrophage-mediated antitumor activity, suggesting a distinct effect of PD-L1, but not PD-1, antibody treatment. Cancer Immunol Res; 6(10); 1260-73. ©2018 AACR.


Assuntos
Antígeno B7-H1/imunologia , Macrófagos/imunologia , Melanoma Experimental/imunologia , Animais , Anticorpos/farmacologia , Anticorpos/uso terapêutico , Antígeno B7-H1/antagonistas & inibidores , Proliferação de Células , Células Cultivadas , Humanos , Ativação de Macrófagos , Macrófagos/efeitos dos fármacos , Melanoma Experimental/tratamento farmacológico , Melanoma Experimental/patologia , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Receptor de Morte Celular Programada 1/antagonistas & inibidores , Receptor de Morte Celular Programada 1/imunologia , Transdução de Sinais
17.
Vaccine ; 35(48 Pt B): 6611-6619, 2017 12 04.
Artigo em Inglês | MEDLINE | ID: mdl-29079105

RESUMO

Leishmaniasis is an arthropod vectored disease causing considerable human morbidity and mortality. Vaccination remains the most realistic and practical means to interrupt the growing number and diversity of sand fly vectors and reservoirs of Leishmania. Since transmission of Leishmania is achieved exclusively by sand fly vectors via immune-modulating salivary substances, conventional vaccination requiring an unmodified host immune response for success are potentially destined to fail unless immunomodulatory factors are somehow neutralized. Using cationic liposome DNA complexes (CLDC) as an adjuvant system along with Lu. longipalpis sand fly salivary component maxadilan (MAX) as antigen (Ag), we show that mice are protected from the MAX-induced exacerbation of infection with Leishmania major (Lm). The CLDC adjuvant and alum were comparable in terms of lesion induration and decreased parasite burden, however the alum adjuvant imposed more inflammation at the injection site. BALB/c, C3H and C57BL/6 mice vaccinated with MAX-CLDC containing either the full-length MAX or peptides spanning the N- and C-terminal regions of MAX are protected against footpad challenges with Lm co-injected with MAX. When compared to unvaccinated controls, all strains of mice immunized with CLDC containing either peptides encompassing the first 20 N-terminal AA or those spanning the last 15 AA of the C-terminal domain of MAX demonstrated decreased parasite burden after 9 or 18 weeks post challenge with Lm + MAX. MAX-CLDC immunized mice showed increased IFNγ-secreting and decreased IL-4-secreting CD4+ cells in footpad-draining lymph nodes. Antisera from C-terminal peptide (P11) MAX-CLDC-vaccinated animals was capable of recognizing FL-MAX and its C-terminal domain and also blocked MAX-mediated reprogramming of bone marrow-derived dendritic cells (BM-DC) in vitro. This peptide vaccine targeting sand fly MAX, improves host immunity against MAX-mediated immunomodulation.


Assuntos
Proteínas de Insetos/química , Proteínas de Insetos/imunologia , Leishmaniose Cutânea/prevenção & controle , Peptídeos/imunologia , Saliva/química , Adjuvantes Imunológicos/administração & dosagem , Compostos de Alúmen/administração & dosagem , Animais , Linfócitos T CD4-Positivos/imunologia , Cátions , DNA/química , Modelos Animais de Doenças , Imunização , Proteínas de Insetos/administração & dosagem , Leishmania major/imunologia , Leishmaniose Cutânea/imunologia , Lipossomos/administração & dosagem , Lipossomos/química , Lipossomos/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Peptídeos/administração & dosagem , Psychodidae/química , Psychodidae/imunologia
18.
Stem Cells Dev ; 26(7): 503-511, 2017 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-27958776

RESUMO

The use of allogeneic bone marrow-derived mesenchymal stem cells (BMDMSCs) may provide an effective alternative to autologous BMDMSCs for treatment of equine musculoskeletal injuries. However, concerns have been raised regarding the potential safety and effectiveness of allogeneic BMDMSCs. We conducted studies to assess the immunological properties of equine allogeneic BMDMSCs compared with those of autologous BMDMSCs. For assessment of inherent immunogenicity, the relative ability of allogeneic and autologous BMDMSCs to stimulate spontaneous proliferation of equine lymphocytes was compared. The immunosuppressive activity of the two cell types was evaluated by adding autologous or allogeneic BMDMSCs to activated lymphocytes and assessing suppression of lymphocyte proliferation and IFNγ production. Fifty-six allogeneic and 12 autologous combinations were evaluated. Studies were also done to elucidate mechanisms by which equine mesenchymal stem cells (MSCs) suppress lymphocyte function. Potential mechanisms evaluated included production of prostaglandin E2 (PGE2), nitric oxide, transforming growth factor-beta, and indoleamine 2,3-dioxygenase. We found that autologous and allogeneic BMDMSCs both induced mild but equivalent levels of spontaneous lymphocyte activation in vitro. In in vitro assays assessing the ability of BMDMSCs to suppress activated lymphocytes, both allogeneic and autologous BMDMSCs suppressed T cell proliferation and IFNγ production to an equal degree. The primary mechanism of equine BMDMSC suppression of T cells was mediated by PGE2. We concluded that allogeneic and autologous BMDMSCs are equivalent in terms of their immunomodulatory properties, and stimulated peripheral blood mononuclear cells appear to trigger the immunosuppressive properties of MSCs. Therefore, both cell types appear to have equal potency in modulating inflammatory processes related to acute or chronic musculoskeletal injuries in the horse.


Assuntos
Autoantígenos/imunologia , Células da Medula Óssea/imunologia , Proliferação de Células/fisiologia , Leucócitos Mononucleares/imunologia , Linfocinas/imunologia , Células-Tronco Mesenquimais/imunologia , Células da Medula Óssea/citologia , Células Cultivadas , Humanos , Ativação Linfocitária/fisiologia , Células-Tronco Mesenquimais/citologia , Linfócitos T/imunologia
19.
Stem Cells Dev ; 26(4): 249-262, 2017 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-27842458

RESUMO

Mesenchymal stem cells (MSC) represent a readily accessible source of cells with potent immune modulatory activity. MSC can suppress ongoing inflammatory responses by suppressing T cell function, while fewer studies have examined the impact of MSC on dendritic cell (DC) function. The dog spontaneous disease model represents an important animal model with which to evaluate the safety and effectiveness of cellular therapy with MSC. This study evaluated the effects of canine MSC on the activation and maturation of canine monocyte-derived DC, as well as mechanisms underlying these effects. Adipose-derived canine MSC were cocultured with canine DC, and the MSC effects on DC maturation and activation were assessed by flow cytometry, cytokine ELISA, and confocal microscopy. We found that canine MSC significantly suppressed lipopolysaccharide (LPS)-stimulated upregulation of DC activation markers such as major histocompatibility class II (MHCII), CD86, and CD40. Furthermore, pretreatment of MSC with interferon gamma (IFNγ) augmented this suppressive activity. IFNγ-activated MSC also significantly reduced LPS-elicited DC secretion of tumor necrosis factor alpha without reducing secretion of interleukin-10. The suppressive effect of IFNγ-treated MSC on LPS-induced DC activation was mediated by soluble factors secreted by both MSC and DC. Pathways of DC functional suppression included programmed death ligand-1 expression and secretion of nitrous oxide, prostaglandin E2, and adenosine by activated MSC. Coculture of DC with IFNγ-treated MSC maintained DC in an immature state and prolonged DC antigen uptake during LPS maturation stimulus. Taken together, canine MSC are capable of potently suppressing DC function in a potentially inflammatory microenvironment through several separate immunological pathways and confirm the potential for immune therapy with MSC in canine immune-mediated disease models.


Assuntos
Diferenciação Celular , Citocinas/farmacologia , Células Dendríticas/citologia , Células Dendríticas/metabolismo , Mediadores da Inflamação/farmacologia , Células-Tronco Mesenquimais/metabolismo , Transdução de Sinais/efeitos dos fármacos , Animais , Antígenos/metabolismo , Antígenos CD/metabolismo , Diferenciação Celular/efeitos dos fármacos , Técnicas de Cocultura , Células Dendríticas/efeitos dos fármacos , Cães , Endossomos/efeitos dos fármacos , Endossomos/metabolismo , Fibroblastos/citologia , Fibroblastos/efeitos dos fármacos , Antígenos de Histocompatibilidade Classe II/metabolismo , Imunomodulação/efeitos dos fármacos , Terapia de Imunossupressão , Interferon gama/farmacologia , Lipopolissacarídeos/farmacologia , Células-Tronco Mesenquimais/efeitos dos fármacos , Monócitos/citologia , Fenótipo , Pele/citologia , Solubilidade , Fator de Necrose Tumoral alfa/metabolismo
20.
Stem Cells ; 34(7): 1709-29, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27066769

RESUMO

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.


Assuntos
Ensaios Clínicos como Assunto , Animais de Estimação , Transplante de Células-Tronco , Células-Tronco/citologia , Animais , Modelos Animais de Doenças , Humanos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...