Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Animals (Basel) ; 11(1)2021 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-33467761

RESUMEN

Despite the possibilities of routine clinical measures and assays on readily accessible bio-samples, it is not always essential in animals to investigate the dynamics of disease longitudinally. In this regard, minimally invasive imaging methods provide powerful tools in preclinical research. They can contribute to the ethical principle of gathering as much relevant information per animal as possible. Besides, with an obvious parallel to clinical diagnostic practice, such imaging platforms are potent and valuable instruments leading to a more refined use of animals from a welfare perspective. Non-human primates comprise highly relevant species for preclinical research to enhance our understanding of disease mechanisms and/or the development of improved prophylactic or therapeutic regimen for various human diseases. In this paper, we describe parameters that critically affect the quality of integrated positron emission tomography and computed tomography (PET-CT) in non-human primates. Lessons learned are exemplified by results from imaging experimental infectious respiratory disease in macaques; specifically tuberculosis, influenza, and SARS-CoV-2 infection. We focus on the thorax and use of 18F-fluorodeoxyglucose as a PET tracer. Recommendations are provided to guide various stages of PET-CT-supported research in non-human primates, from animal selection, scan preparation, and operation, to processing and analysis of imaging data.

2.
Cell Rep Med ; 2(1): 100187, 2021 01 19.
Artículo en Inglés | MEDLINE | ID: mdl-33521701

RESUMEN

To fight tuberculosis, better vaccination strategies are needed. Live attenuated Mycobacterium tuberculosis-derived vaccine, MTBVAC, is a promising candidate in the pipeline, proven to be safe and immunogenic in humans so far. Independent studies have shown that pulmonary mucosal delivery of Bacillus Calmette-Guérin (BCG), the only tuberculosis (TB) vaccine available today, confers superior protection over standard intradermal immunization. Here we demonstrate that mucosal MTBVAC is well tolerated, eliciting polyfunctional T helper type 17 cells, interleukin-10, and immunoglobulins in the airway and yielding a broader antigenic profile than BCG in rhesus macaques. Beyond our previous work, we show that local immunoglobulins, induced by MTBVAC and BCG, bind to M. tuberculosis and enhance pathogen uptake. Furthermore, after pulmonary vaccination, but not M. tuberculosis infection, local T cells expressed high levels of mucosal homing and tissue residency markers. Our data show that pulmonary MTBVAC administration has the potential to enhance its efficacy and justifies further exploration of mucosal vaccination strategies in preclinical efficacy studies.


Asunto(s)
Vacuna BCG/administración & dosificación , Inmunidad Mucosa , Mycobacterium tuberculosis/inmunología , Mucosa Respiratoria/inmunología , Vacunas contra la Tuberculosis/administración & dosificación , Tuberculosis Pulmonar/prevención & control , Administración Intranasal , Animales , Reprogramación Celular/genética , Reprogramación Celular/inmunología , Femenino , Regulación de la Expresión Génica , Inyecciones Intradérmicas , Interleucina-10/genética , Interleucina-10/inmunología , Interleucina-17/genética , Interleucina-17/inmunología , Pulmón/efectos de los fármacos , Pulmón/inmunología , Pulmón/microbiología , Macaca mulatta , Masculino , Monocitos/efectos de los fármacos , Monocitos/inmunología , Monocitos/microbiología , Mycobacterium tuberculosis/patogenicidad , Mucosa Respiratoria/microbiología , Células TH1/inmunología , Células TH1/microbiología , Células Th17/inmunología , Células Th17/microbiología , Tuberculosis Pulmonar/genética , Tuberculosis Pulmonar/inmunología , Tuberculosis Pulmonar/microbiología , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/inmunología
3.
Cell Rep Med ; 2(1): 100185, 2021 01 19.
Artículo en Inglés | MEDLINE | ID: mdl-33521699

RESUMEN

BCG vaccination can strengthen protection against pathogens through the induction of epigenetic and metabolic reprogramming of innate immune cells, a process called trained immunity. We and others recently demonstrated that mucosal or intravenous BCG better protects rhesus macaques from Mycobacterium tuberculosis infection and TB disease than standard intradermal vaccination, correlating with local adaptive immune signatures. In line with prior mouse data, here, we show in rhesus macaques that intravenous BCG enhances innate cytokine production associated with changes in H3K27 acetylation typical of trained immunity. Alternative delivery of BCG does not alter the cytokine production of unfractionated bronchial lavage cells. However, mucosal but not intradermal vaccination, either with BCG or the M. tuberculosis-derived candidate MTBVAC, enhances innate cytokine production by blood- and bone marrow-derived monocytes associated with metabolic rewiring, typical of trained immunity. These results provide support to strategies for improving TB vaccination and, more broadly, modulating innate immunity via mucosal surfaces.


Asunto(s)
Vacuna BCG/administración & dosificación , Inmunidad Mucosa , Mycobacterium tuberculosis/inmunología , Mucosa Respiratoria/inmunología , Vacunas contra la Tuberculosis/administración & dosificación , Tuberculosis Pulmonar/prevención & control , Acetilación , Administración Intranasal , Animales , Médula Ósea/efectos de los fármacos , Médula Ósea/inmunología , Médula Ósea/microbiología , Reprogramación Celular/genética , Reprogramación Celular/inmunología , Femenino , Regulación de la Expresión Génica , Histonas/genética , Histonas/inmunología , Inyecciones Intravenosas , Interleucina-1beta/genética , Interleucina-1beta/inmunología , Interleucina-6/genética , Interleucina-6/inmunología , Pulmón/efectos de los fármacos , Pulmón/inmunología , Pulmón/microbiología , Macaca mulatta , Masculino , Monocitos/efectos de los fármacos , Monocitos/inmunología , Monocitos/microbiología , Mycobacterium tuberculosis/patogenicidad , Mucosa Respiratoria/microbiología , Tuberculosis Pulmonar/genética , Tuberculosis Pulmonar/inmunología , Tuberculosis Pulmonar/microbiología , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/inmunología
4.
NPJ Vaccines ; 5(1): 39, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32435513

RESUMEN

Tuberculosis (TB) still is the principal cause of death from infectious disease and improved vaccination strategies are required to reduce the disease burden and break TB transmission. Here, we investigated different routes of administration of vectored subunit vaccines based on chimpanzee-derived adenovirus serotype-3 (ChAd3) for homologous prime-boosting and modified vaccinia virus Ankara (MVA) for heterologous boosting with both vaccine vectors expressing the same antigens from Mycobacterium tuberculosis (Ag85B, ESAT6, Rv2626, Rv1733, RpfD). Prime-boost strategies were evaluated for immunogenicity and protective efficacy in highly susceptible rhesus macaques. A fully parenteral administration regimen was compared to exclusive respiratory mucosal administration, while parenteral ChAd3-5Ag prime-boosting and mucosal MVA-5Ag boosting were applied as a push-and-pull strategy from the periphery to the lung. Immune analyses corroborated compartmentalized responses induced by parenteral versus mucosal vaccination. Despite eliciting TB-specific immune responses, none of the investigational regimes conferred a protective effect by standard readouts of TB compared to non-vaccinated controls, while lack of protection by BCG underpinned the stringency of this non-human primate test modality. Yet, TB manifestation after full parenteral vaccination was significantly less compared to exclusive mucosal vaccination.

5.
Front Immunol ; 10: 2479, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31736945

RESUMEN

While tuberculosis continues to afflict mankind, the immunological mechanisms underlying TB disease development are still incompletely understood. Advanced preclinical models for TB research include both rhesus and cynomolgus macaques (Macaca mulatta and Macaca fascicularis, respectively), with rhesus typically being more susceptible to acute progressive TB disease than cynomolgus macaques. To determine which immune mechanisms are responsible for this dissimilar disease development, we profiled a broad range of innate and adaptive responses, both local and peripheral, following experimental pulmonary Mycobacterium tuberculosis (Mtb) infection of both species. While T-cell and antibody responses appeared indistinguishable, we identified anti-inflammatory skewing of peripheral monocytes in rhesus and a more prominent local pro-inflammatory cytokine release profile in cynomolgus macaques associated with divergent TB disease outcome. Importantly, these differences were detectable both before and early after infection. This work shows that inflammatory and innate immune status prior to and at early stages after infection, critically affects outcome of TB infection.


Asunto(s)
Macaca fascicularis/inmunología , Macaca mulatta/inmunología , Mycobacterium tuberculosis , Tuberculosis Pulmonar/inmunología , Animales , Citocinas/inmunología , Inmunidad Innata , Pulmón/inmunología , Pulmón/microbiología , Pulmón/patología , Masculino , Tuberculosis Pulmonar/microbiología , Tuberculosis Pulmonar/patología
6.
Nat Med ; 25(2): 255-262, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30664782

RESUMEN

Tuberculosis (TB) remains the deadliest infectious disease1, and the widely used Bacillus Calmette-Guérin (BCG) vaccine fails to curb the epidemic. An improved vaccination strategy could provide a cost-effective intervention to break the transmission cycle and prevent antimicrobial resistance2,3. Limited knowledge of the host responses critically involved in protective immunity hampers the development of improved TB vaccination regimens. Therefore, assessment of new strategies in preclinical models to select the best candidate vaccines before clinical vaccine testing remains indispensable. We have previously established in rhesus macaques (Macaca mulatta) that pulmonary mucosal BCG delivery reduces TB disease where standard intradermal injection fails4,5. Here, we show that pulmonary BCG prevents infection by using a repeated limiting-dose Mycobacterium tuberculosis challenge model and identify polyfunctional T-helper type 17 (TH17) cells, interleukin-10 and immunoglobulin A as correlates of local protective immunity. These findings warrant further research into mucosal immunization strategies and their translation to clinical application to more effectively prevent the spread of TB.


Asunto(s)
Vacuna BCG/inmunología , Tuberculosis/inmunología , Tuberculosis/prevención & control , Animales , Carga Bacteriana , Relación Dosis-Respuesta Inmunológica , Inmunidad Humoral , Interferón gamma/metabolismo , Pulmón/inmunología , Pulmón/microbiología , Pulmón/patología , Macaca mulatta , Masculino , Membrana Mucosa/inmunología , Vacunación
7.
Drug Discov Today ; 12(7-8): 327-35, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17395093

RESUMEN

The costs for the development of new drugs have increased dramatically over the past 30 years. One of the main reasons for this increase is the low success rate of new drugs being approved for patient use, which is, in part, a consequence of the common use of rodent models for preclinical validation of efficacy. Especially in the development of biologicals, which are now successfully used in the treatment of rheumatoid arthritis, the selection of the right animal model is pivotal. Non-human primates could help to bridge the evolutionary gap between rodent models and human patients.


Asunto(s)
Artritis Experimental/tratamiento farmacológico , Artritis Reumatoide/tratamiento farmacológico , Evaluación Preclínica de Medicamentos/métodos , Animales , Artritis Experimental/metabolismo , Artritis Experimental/fisiopatología , Artritis Reumatoide/metabolismo , Artritis Reumatoide/fisiopatología , Biomarcadores/análisis , Diseño de Fármacos , Humanos , Macaca mulatta , Ratones
8.
Methods Mol Biol ; 1559: 411-417, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28063060

RESUMEN

Animal models of rheumatoid arthritis are important in the elucidation of etiopathogenic mechanisms of the disease and for the development of promising new therapies. Species specificity of new biological compounds and their mode of action preclude safety and efficacy testing in rodent models of disease. Nonhuman primates (NHP) can fill this niche and provide the only relevant model. Over the last two decades models of collagen-induced arthritis (CIA) were developed in the rhesus monkey and the common marmoset. However, NHP are higher-order animals and complex sentient beings. So especially in models where pain is an intricate part of the disease, analgesia needs to be addressed because of ethical considerations. In our model, a morphine-based pain relief was used that does not interfere with the normal development of disease allowing us to evaluate important mechanistic aspects of the arthritis.


Asunto(s)
Analgésicos Opioides , Artritis Experimental/complicaciones , Buprenorfina , Manejo del Dolor/métodos , Dolor/prevención & control , Animales , Artritis Experimental/patología , Artritis Experimental/fisiopatología , Callithrix , Esquema de Medicación , Macaca mulatta , Dolor/complicaciones , Dolor/fisiopatología , Dimensión del Dolor/métodos , Índice de Severidad de la Enfermedad
9.
Arthritis Res Ther ; 19(1): 246, 2017 Nov 02.
Artículo en Inglés | MEDLINE | ID: mdl-29096669

RESUMEN

BACKGROUND: Rheumatoid factor (RF), anti-citrullinated protein antibodies (ACPA) and anti-carbamylated protein (anti-CarP) antibodies are rheumatoid arthritis (RA)-associated autoantibodies. Besides their presence in human serum, anti-CarP antibodies have also been described in rodent models of arthritis, while ACPA are not consistently detectable. Data on these RA-associated autoantibodies in primates are not available. Therefore, we investigated the presence of RF, anti-CarP antibodies and ACPA in rhesus monkeys before and after collagen-induced arthritis immunizations. METHODS: In previous studies, arthritis was induced in groups of rhesus monkeys by immunisation with collagen following pre-treatment with placebo, abatacept or Roactemra. Previously collected serum was used to measure, autoantibodies by ELISA, detecting anti-CarP antibodies, RF-IgM and antibodies against CCP2, citrullinated myelin basic protein and citrullinated fibrinogen. RESULTS: Out of the three autoantibodies, only anti-CarP antibodies were detectable in resus monkeys with arthritis. RF-IgM and ACPA were undetectable and below the detection limit of the ELISA. The level of anti-CarP antibodies increases over time and, similar to in humans and mice, these autoantibodies were already detectable before clinical disease onset. Furthermore, preventive treatment with abatacept (CTLA4/IgG1-Fc fusion protein) inhibited the development of anti-CarP antibodies after immunization, while this was less evident for preventive Roactemra (anti-IL6-receptor) treatment. Moreover, disease progression was only reduced following abatacept treatment. CONCLUSION: Rhesus monkeys develop anti-CarP antibodies upon induction of collagen-induced arthritis, while we were unable to detect RF or ACPA. Also, the development of anti-CarP antibodies could be inhibited by preventive abatacept treatment.


Asunto(s)
Artritis Experimental/inmunología , Autoanticuerpos/inmunología , Macaca mulatta/inmunología , Proteínas/inmunología , Abatacept/farmacología , Animales , Antirreumáticos/farmacología , Artritis Experimental/prevención & control , Carbamatos/metabolismo , Ensayo de Inmunoadsorción Enzimática , Femenino , Masculino , Proteínas/metabolismo , Factores de Tiempo
10.
Cancer Res ; 62(21): 6187-93, 2002 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-12414646

RESUMEN

Overexpression of p53 is found in approximately 50% of human cancers, making it an attractive target antigen for immunotherapy of cancer. Research in this area has thus far primarily focused on p53-specific CTLs. Although these CTLs were shown to be highly effective against p53-overexpressing tumors in vivo, immunological tolerance seems to strongly restrict the spectrum of the p53-specific CTL repertoire in p53(+/+) subjects. In view of the emerging role of CD4(+) Th (Th) cells in the antitumor response, we investigated the specificity and antitumor efficacy of the p53-specific Th response in mice. Our data show that high affinity Th cells against the naturally processed epitope p53(108-122) can be elicited in both p53(-/-) and p53(+/+) mice, indicating that the p53-specific T-cell response is not affected by tolerance at the Th level. Furthermore, p53(108-122)-specific Th cells were effective in enabling p53-specific CTLs to control the growth of p53-overexpressing tumors in vivo. Therefore, exploitation of the p53-specific Th response appears to be a highly useful aspect of immunotherapeutic strategies against cancers.


Asunto(s)
Linfocitos T Colaboradores-Inductores/inmunología , Proteína p53 Supresora de Tumor/inmunología , Secuencia de Aminoácidos , Animales , Células Dendríticas/inmunología , Epítopos de Linfocito T/inmunología , Femenino , Inmunoterapia Adoptiva , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Desnudos , Datos de Secuencia Molecular , Neoplasias Experimentales/inmunología , Neoplasias Experimentales/terapia , Fragmentos de Péptidos/inmunología , Linfocitos T Citotóxicos/inmunología , Transfección , Proteína p53 Supresora de Tumor/genética
11.
Hum Gene Ther Clin Dev ; 26(2): 103-12, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-26086763

RESUMEN

Preclinical studies to assess biodistribution, safety, and initial efficacy of ART-I02, an adeno-associated type 5 (rAAV5) vector expressing human interferon ß (hIFN-ß), were performed in a total of 24 rhesus monkeys with collagen-induced arthritis. All monkeys were naïve or showed limited neutralizing antibody (Nab) titers to AAV5 at the start of the study. Animals were injected with a single intra-articular dose of ART-I02 or placebo, consisting of 3.2×10(13) vg (Dose A=maximum feasible dose), 4.58×10(12) vg (Dose B), or placebo in the first affected finger joint, the ipsilateral knee, and ankle joint at the same time point. Animals were monitored for clinical parameters and well-being with a maximum of 4 weeks, with the option that the severity of arthritis could necessitate an earlier time point of sacrifice. No adverse events were noted after injection of ART-I02. No abnormalities were observed after histological evaluation of all organs. At both dose levels, immunohistochemical staining indicated expression of hIFN-ß. In animals injected with Dose A, we observed stabilization or a reduction in swelling in the finger joint in which vector was administered. The highest copy numbers of vector DNA were detected in synovial tissue of the injected joint and the draining lymph node of the injected knee. High titers of Nab to rAAV5 were observed at the end of the study. Five monkeys developed an rAAV5-specific T-cell response. Two monkeys developed Nab to hIFN-ß. In conclusion, intra-articular injection of ART-I02 was well-tolerated and did not induce adverse events. After administration of Dose A of ART-I02, we observed a beneficial effect on joint swelling, substantiated by decreased histological inflammation and bone erosion scores. A GMP vector for clinical application has been manufactured and is currently being tested in GLP rodent studies, with the aim to move forward to a clinical trial.


Asunto(s)
Artritis Experimental/terapia , Dependovirus/genética , Interferón beta/genética , Animales , Articulación del Tobillo/patología , Artritis Experimental/metabolismo , Artritis Experimental/patología , Femenino , Articulaciones de los Dedos/patología , Terapia Genética , Vectores Genéticos , Humanos , Inyecciones Intraarticulares , Interferón beta/metabolismo , Articulación de la Rodilla/patología , Macaca mulatta , Masculino , Distribución Tisular , Resultado del Tratamiento
12.
Transpl Immunol ; 11(2): 215-22, 2003.
Artículo en Inglés | MEDLINE | ID: mdl-12799206

RESUMEN

The development of an in vitro assay predicting the chances of graft survival after treatment with immunoregulatory agents is a major topic in transplantation. Antibodies (Abs) interfering in the costimulatory pathway are promising candidates for the induction of tolerance. To evaluate these antibodies for clinical use studies non-human primates are the only feasible option due to species specificity of the antibodies. Peripheral blood mononuclear cells, isolated from a large panel of rhesus monkeys, were used in a unidirectional mixed lymphocyte reaction to evaluate the ability of antibodies blocking the costimulatory pathway, to affect both primary and secondary proliferative and cytolytic allospecific immune responses in vitro. These blocking antibodies were also used in protocols prolonging allograft survival in a life-supporting kidney allotransplant model in rhesus macaques. The ultimate aim is to establish a correlation between parameters obtained in vitro and the success of transplantation in vivo. The combination of anti-CD80 and anti-CD86 resulted in a complete abrogation of the primary alloresponse as measured in a proliferation assay. Adding anti-CD40 significantly reduced this inhibitory effect although the in vivo effects of this antibody have been shown to be beneficial. The secondary response was most prominently inhibited by the combination of anti-CD80/86. Paradoxically, anti-CD40 alone markedly inhibited the secondary proliferative response, but did not add to the inhibitory effect of the combination of anti-CD80/86. The cytolytic response was inhibited maximally only when CsA was added to the combination of anti-CD80/86. Treatment with monoclonal antibodies alone without immunosuppressive drugs was sufficient to maintain graft survival during the time of treatment in most animals. However, rejection was initiated as soon as the treatment ceased and no tolerance, resulting in long-term graft and patient survival, was established. The complete inhibition of primary alloresponses and the partial inhibition of secondary proliferative alloresponses correlate with prolonged graft survival during treatment, but have no predictive value for the success of tolerance induction for kidney allografts in rhesus monkeys.


Asunto(s)
Anticuerpos Bloqueadores/uso terapéutico , Rechazo de Injerto/prevención & control , Supervivencia de Injerto/efectos de los fármacos , Trasplante de Riñón/inmunología , Linfocitos/efectos de los fármacos , Tolerancia al Trasplante , Animales , Anticuerpos Monoclonales/uso terapéutico , Antígenos CD/inmunología , Antígeno B7-1/inmunología , Antígeno B7-2 , Antígenos CD40/inmunología , Células Cultivadas , Supervivencia de Injerto/inmunología , Prueba de Cultivo Mixto de Linfocitos , Linfocitos/inmunología , Glicoproteínas de Membrana/inmunología
13.
Arthritis Res Ther ; 15(6): R207, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24299175

RESUMEN

INTRODUCTION: Targeting the CD20 antigen has been a successful therapeutic intervention in the treatment of rheumatoid arthritis (RA). However, in some patients with an inadequate response to anti-CD20 therapy, a persistence of CD20- plasmablasts is noted. The strong expression of CD319 on CD20- plasmablast and plasma cell populations in RA synovium led to the investigation of the potential of CD319 as a therapeutic target. METHODS: PDL241, a novel humanized IgG1 monoclonal antibody (mAb) to CD319, was generated and examined for its ability to inhibit immunoglobulin production from plasmablasts and plasma cells generated from peripheral blood mononuclear cells (PBMC) in the presence and absence of RA synovial fibroblasts (RA-SF). The in vivo activity of PDL241 was determined in a human PBMC transfer into NOD scid IL-2 gamma chain knockout (NSG) mouse model. Finally, the ability of PDL241 to ameliorate experimental arthritis was evaluated in a collagen-induced arthritis (CIA) model in rhesus monkeys. RESULTS: PDL241 bound to plasmablasts and plasma cells but not naïve B cells. Consistent with the binding profile, PDL241 inhibited the production of IgM from in vitro PBMC cultures by the depletion of CD319+ plasmablasts and plasma cells but not B cells. The activity of PDL241 was dependent on an intact Fc portion of the IgG1 and mediated predominantly by natural killer cells. Inhibition of IgM production was also observed in the human PBMC transfer to NSG mouse model. Treatment of rhesus monkeys in a CIA model with PDL241 led to a significant inhibition of anti-collagen IgG and IgM antibodies. A beneficial effect on joint related parameters, including bone remodeling, histopathology, and joint swelling was also observed. CONCLUSIONS: The activity of PDL241 in both in vitro and in vivo models highlights the potential of CD319 as a therapeutic target in RA.


Asunto(s)
Anticuerpos Monoclonales Humanizados/inmunología , Formación de Anticuerpos/efectos de los fármacos , Artritis Reumatoide/inmunología , Células Plasmáticas/inmunología , Receptores Inmunológicos/inmunología , Animales , Citometría de Flujo , Xenoinjertos , Humanos , Inmunoglobulinas/biosíntesis , Inmunohistoquímica , Macaca mulatta , Ratones , Ratones Endogámicos NOD , Ratones Noqueados , Ratones SCID , Familia de Moléculas Señalizadoras de la Activación Linfocitaria , Membrana Sinovial/inmunología , Membrana Sinovial/metabolismo
14.
Arthritis Res Ther ; 12(5): R200, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20977720

RESUMEN

INTRODUCTION: There is an ever-increasing need for animal models to evaluate efficacy and safety of new therapeutics in the field of rheumatoid arthritis (RA). Particularly for the early preclinical evaluation of human-specific biologicals targeting the progressive phase of the disease, there is a need for relevant animal models. In response to this requirement we set out to develop a model of collagen-induced arthritis (CIA) in a small-sized nonhuman primate species (300 to 400 g at adult age); that is, the common marmoset (Callithrix jacchus). METHODS: Twenty-two animals divided into three experiments were immunized with collagen type II (CII) of either bovine or chicken origin with different immunization strategies. The animals were analyzed for clinical manifestation of arthritis, hematology and clinical chemistry, immunological responses against CII and histopathological features of the arthritis. RESULTS: Clinically manifest arthritis was observed in almost 100% (21 out of 22) of the animals. Fifty percent of the animals developed semi-acute CIA while the other 50% displayed a more chronic disease. Both cellular (CD3/CD4 and CD3/CD8) and humoral responses (IgM and IgG) against CII were involved in the development of the disease. Besides mild histopathological changes in bone and cartilage, severe inflammation in extraarticular tissues like periosteum and subcutaneous tissues was observed. CONCLUSIONS: This new model in marmosets more closely resembles chronic RA with respect to the chronic disease course and pathomorphological presentation than the more acute monophasic and destructive CIA model in macaques. This model can therefore fill a niche in preclinical testing of new human specific therapeutics.


Asunto(s)
Artritis Experimental/inmunología , Artritis Experimental/patología , Artritis Reumatoide/patología , Animales , Artritis Experimental/metabolismo , Artritis Reumatoide/inmunología , Artritis Reumatoide/metabolismo , Biomarcadores/análisis , Callithrix , Separación Celular , Enfermedad Crónica , Femenino , Citometría de Flujo , Inmunoensayo , Masculino
15.
Arthritis Res Ther ; 7(4): 145-54, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-15987497

RESUMEN

Models of rheumatoid arthritis (RA) in laboratory animals are important tools for research into pathogenic mechanisms and the development of effective, safe therapies. Rodent models (rats and mice) have provided important information about the pathogenic mechanisms. However, the evolutionary distance between rodents and humans hampers the translation of scientific principles into effective therapies. The impact of the genetic distance between the species is especially seen with treatments based on biological molecules, which are usually species-specific. The outbred nature and the closer anatomical, genetic, microbiological, physiological, and immunological similarity of nonhuman primates to humans may help to bridge the wide gap between inbred rodent strain models and the heterogeneous RA patient population. Here we review clinical, immunological and pathological aspects of the rhesus monkey model of collagen-induced arthritis, which has emerged as a reproducible model of human RA in nonhuman primates.


Asunto(s)
Artritis Experimental/inmunología , Artritis Reumatoide/inmunología , Modelos Animales de Enfermedad , Animales , Artritis Experimental/patología , Artritis Reumatoide/patología , Humanos , Macaca mulatta , Primates
16.
Arthritis Rheum ; 52(2): 627-36, 2005 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-15693002

RESUMEN

OBJECTIVE: Collagen-induced arthritis (CIA) in the rhesus monkey is a nonhuman primate model of rheumatoid arthritis (RA). The close phylogenetic relationship between humans and the rhesus monkey makes this model useful for the preclinical safety and efficacy testing of new therapies that are inactive in animals more distinctly related to humans. In this study, we tested the therapeutic potential of a novel, small molecular weight antagonist of CCR5, SCH-X, in this model. METHODS: CIA was induced in 10 rhesus monkeys. The animals were allocated to receive SCH-X or saline as the control (n = 5 in each group). Treatment was initiated on the day of CIA induction and continued for 45 days. Monkeys were monitored before and 63 days after CIA induction for macroscopic signs of clinical arthritis, such as soft-tissue swelling and body weight. Furthermore, markers of inflammation and joint degradation were monitored to follow the disease course. RESULTS: Only 2 of 5 animals in the SCH-X-treated group displayed prominent soft-tissue swelling, compared with all 5 saline-treated monkeys. In addition to the suppression of joint inflammation, treatment with SCH-X resulted in a reduction in joint destruction, as demonstrated by lower rates of urinary excretion of collagen crosslinks, with confirmation by histology. Whereas in all saline-treated monkeys, marked erosion of joint cartilage was observed, this was absent in 4 of the 5 SCH-X-treated monkeys. CONCLUSION: The systemic effects of treatment with SCH-X were a suppressed acute-phase reaction (reduction in C-reactive protein level) in the 3 treated monkeys with CIA that remained asymptomatic, and an altered antibody response toward type II collagen. The results suggest that the CCR5 antagonist SCH-X might have a strong clinical potential for treatment during periods of active inflammation, as seen in RA.


Asunto(s)
Artritis Experimental/prevención & control , Antagonistas de los Receptores CCR5 , Animales , Artritis Experimental/inducido químicamente , Artritis Experimental/patología , Cartílago Articular/patología , Colágeno Tipo II , Estudios de Seguimiento , Macaca mulatta , Masculino
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA