Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
1.
Anal Bioanal Chem ; 415(3): 391-404, 2023 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-36346456

RESUMO

The SARS-CoV-2 pandemic has shown the importance of rapid and comprehensive diagnostic tools. While there are numerous rapid antigen tests available, rapid serological assays for the detection of neutralizing antibodies are and will be needed to determine not only the amount of antibodies formed after infection or vaccination but also their neutralizing potential, preventing the cell entry of SARS-CoV-2. Current active-virus neutralization assays require biosafety level 3 facilities, while virus-free surrogate assays are more versatile in applications, but still take typically several hours until results are available. To overcome these disadvantages, we developed a competitive chemiluminescence immunoassay that enables the detection of neutralizing SARS-CoV-2 antibodies within 7 min. The neutralizing antibodies bind to the viral receptor binding domain (RBD) and inhibit the binding to the human angiotensin-converting enzyme 2 (ACE2) receptor. This competitive binding inhibition test was characterized with a set of 80 samples, which could all be classified correctly. The assay results favorably compare to those obtained with a more time-intensive ELISA-based neutralization test and a commercial surrogate neutralization assay. Our test could further be used to detect individuals with a high total IgG antibody titer, but only a low neutralizing titer, as well as for monitoring neutralizing antibodies after vaccinations. This effective performance in SARS-CoV-2 seromonitoring delineates the potential for the test to be adapted to other diseases in the future.


Assuntos
COVID-19 , SARS-CoV-2 , Humanos , Anticorpos Neutralizantes , Anticorpos Antivirais , COVID-19/diagnóstico , Ensaio de Imunoadsorção Enzimática , Imunoensaio , Luminescência , Automação Laboratorial
2.
Cells ; 11(8)2022 04 07.
Artigo em Inglês | MEDLINE | ID: mdl-35455934

RESUMO

Dysregulation of platelet function is causally connected to thrombus formation and cardiovascular diseases. Therefore, assessing platelet reactivity is crucial. However, current platelet function tests come with pitfalls, limiting clinical use. Plasma miRNA signatures have been suggested as novel biomarkers for predicting/diagnosing cardiovascular diseases and monitoring antiplatelet therapy. Here, we provide results from a comprehensive study on the feasibility of using circulatory platelet miRNAs as surrogate markers of platelet activation. We performed small RNA-Seq on different blood cell types to confirm known and identify novel platelet-enriched miRNAs and validated a panel of 16 miRNAs using RT-qPCR. To identify the main carrier of these blood-based platelet miRNAs, we enriched and analyzed distinct microvesicle populations. Platelets were stimulated with GPVI and P2Y12 agonists in vitro to monitor the release of the selected miRNAs following activation. Finally, the miRNA panel was also measured in plasma from mice undergoing the Folts intervention (recurrent thrombus formation in the carotid artery). Applying an unbiased bioinformatics-supported workflow to our NGS data, we were able to confirm a panel of previously established miRNA biomarker candidates and identify three new candidates (i.e., miR-199a-3p, miR-151a-5p, and miR-148b-3p). Basal levels of platelet-derived miRNAs in plasma were mainly complexed with proteins, not extracellular vesicles. We show that changes in miRNA levels due to platelet activation are detectable using RT-qPCR. In addition, we highlight limitations of studying the in vitro release of miRNAs from platelets. In vivo thrombosis resulted in significant elevations of platelet-derived miRNA levels in mice. In conclusion, we provide in-depth evidence that activated platelets release miRNAs, resulting in measurable changes in circulatory miRNA levels, rendering them promising biomarker candidates.


Assuntos
Doenças Cardiovasculares , MicroRNAs , Animais , Biomarcadores/metabolismo , Plaquetas/metabolismo , Doenças Cardiovasculares/metabolismo , Camundongos , MicroRNAs/metabolismo , Ativação Plaquetária
3.
PLoS One ; 17(4): e0266701, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35468147

RESUMO

OBJECTIVE: We investigated blood samples from fully SARS-CoV2-vaccinated subjects and from previously positive tested patients up to one year after infection with SARS-CoV2, and compared short- and long-term T cell and antibody responses, with a special focus on the recently emerged delta variant (B.1.617.2). METHODS AND RESULTS: In 23 vaccinated subjects, we documented high anti-SARS-CoV2 spike protein receptor binding domain (RBD) antibody titers. Average virus neutralization by antibodies, assessed as inhibition of ACE2 binding to RBD, was 2.2-fold reduced for delta mutant vs. wild type (wt) RBD. The mean specific antibody titers were lower one year after natural infection than after vaccination; ACE2 binding to delta mutant vs. wt RBD was 1.65-fold reduced. In an additional group, omicron RBD binding was reduced compared to delta. Specific CD4+ T cell responses were measured after stimulation with peptides pools from wt, alpha, beta, gamma, or delta variant SARS-CoV2 spike proteins by flow cytometric intracellular cytokine staining. There was no significant difference in cytokine production of IFN-γ, TNF-α, or IL-2 between vaccinated subjects. T cell responses to wt or mutant SARS-CoV2 spike were significantly weaker after natural occurring infections compared to those in vaccinated individuals. CONCLUSION: Antibody neutralisation of the delta mutant was reduced compared to wt, as assessed in a novel inhibition assay with a finger prick blood drop. Strong CD4 T cell responses were present against wt and mutant SARS-CoV2 variants, including the delta (B.1.617.2) strain, in fully vaccinated individuals, whereas they were partly weaker 1 year after natural infection. Hence, immune responses after vaccination are stronger compared to those after naturally occurring infection, pointing out the need of the vaccine to overcome the pandemic.


Assuntos
COVID-19 , Vacinas Virais , Enzima de Conversão de Angiotensina 2 , COVID-19/prevenção & controle , Citocinas , Humanos , RNA Viral , SARS-CoV-2 , Glicoproteína da Espícula de Coronavírus/genética , Linfócitos T , Vacinação , Proteínas do Envelope Viral
4.
PLoS One ; 16(8): e0255363, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34347814

RESUMO

The standard histological processing procedure, which produces excellent staining of sections for most tissues, fails to yield satisfactory results in adult mouse orbits or eyeballs. Here, we show that a protocol using tissue block staining and domestic adhesive tapes resulted in qualified integral serial cryo-sections of whole orbits or eyeballs, and the fine structures were well preserved. The histological processing protocol comprises paraformaldehyde fixation, ethylenediaminetetraacetic acid decalcification, tissue block staining with hematoxylin and eosin, embedding, adhesive tape aided sectioning, and water-soluble mounting. This protocol was proved to be the best in comparison with seven other related existing histological traditional or non-traditional processing methods, according to the staining slice quality. We observed a hundred percent success rate in sectioning, collection, and mounting with this method. The reproducibility tested on qualified section success rates and slice quality scores confirmed that the technique is reliable. The feasibility of the method to detect target molecules in orbits was verified by successful trial tests on block immunostaining and adhesive tape-aided sectioning. Application of this protocol in joints, brains, and so on,-the challenging integral sectioning tissues, also generated high-quality histological staining sections.


Assuntos
Olho/anatomia & histologia , Órbita/anatomia & histologia , Preservação de Tecido/instrumentação , Animais , Criopreservação , Estudos de Viabilidade , Feminino , Camundongos , Microtomia , Coloração e Rotulagem , Fita Cirúrgica , Inclusão do Tecido , Fixação de Tecidos , Preservação de Tecido/métodos
5.
Sci Rep ; 11(1): 17345, 2021 08 30.
Artigo em Inglês | MEDLINE | ID: mdl-34462464

RESUMO

Rheumatoid arthritis (RA) is a systemic inflammatory autoimmune disease which affects primarily the joints. Peptides of several proteins have shown an effect in some experimental animal models of RA. We investigated arthritis development in male DBA/1 mice which were injected with bovine collagen II (bCII) and human fibrinogen (hFib) on days 0 and 21, leading to stable and reproducible disease induction in 100% of immunized mice (FIA-CIA). In a second study, two bCII-derived peptides were given three times in the course of 6 weeks after FIA-CIA induction to test for impact on arthritis. Mice were scored weekly for arthritis and anti-citrullinated peptide antibodies (ACPAs) were determined in the sera taken on days 0, 14, 35, 56 and 84. Histology of the hind paws was performed at the end of the experiment. Intravenous administration of peptide 90578, a novel fructosylated peptide derived from the immunodominant T cell epitope of bCII, at a dosage of 1 mg/kg resulted in significant beneficial effects on clinical outcome parameters and on the arthritis histology scores which was sustained over 12 weeks. Survival tended to be improved in peptide 90578-treated mice. Intravenous administration of pure soluble peptide 90578 without adjuvants is a promising approach to treat RA, with treatment starting at a time when ACPAs are already present. The results complement existing data on peptide "vaccination" of healthy animals, or on treatment using recombinant peptide expressing virus or complex biological compounds.


Assuntos
Artrite Reumatoide/imunologia , Artrite/imunologia , Artrite/metabolismo , Epitopos de Linfócito T/química , Frutose/química , Peptídeos/química , Animais , Antígenos de Diferenciação de Linfócitos B , Autoimunidade , Bovinos , Citrulina/química , Colágeno Tipo II/química , Antígenos de Histocompatibilidade Classe II , Inflamação , Masculino , Camundongos , Camundongos Endogâmicos DBA , Peptídeos Cíclicos
6.
J Autoimmun ; 122: 102666, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34144327

RESUMO

BACKGROUND: BALB/c mice which received long-term immunizations of adenovirus (Ad) expressing thyrotropin receptor A-subunits (TSHR) developed stable Graves' disease (GD). TSHR-derived cyclic peptide 19 (P19) was identified as effective therapy in this model. METHODS: In Ad-TSHR mice, we investigated shorter disease intervals up to 4 months for histological alterations of the orbits, fine tuning of anti-TSHR antibodies (Ab) and free thyroxine (fT4) hormone levels by using novel detection methods in an independent laboratory. Therapy (0.3 mg/kg P19 or vehicle) was given intravenously after the fourth Ad-TSHR immunization (week 11) and continued until week 19. RESULTS: Thyrotropin binding inhibitory immunoglobulins (TBII, bridge immunoassay), blocking (TBAb) and stimulating (TSAb) TSHR-Ab (both cell-based bioassays) and serum levels of fT4 were significantly elevated at week 11 in Ad-TSHR-immunized mice versus none in control mice. For the first time, TSAb, TBAb, and thyroperoxidase-Ab were detected in 17 of 19, 12/19 and 6/19 Ad-TSHR immunized mice, respectively at week 21. Also, for the first time, this study showed that P19 treatment markedly reduced serum TBII (p < 0.0001), serum fT4 (p = 0.02), and acidic mucins and collagen content in the orbital tissue of Ad-TSHR-immunized mice. CONCLUSION: P19 significantly improved thyroid function, confirming previous results in an independent second laboratory. A relevant shift of anti-TSHR antibody subpopulations in response to P19 therapy may help explain its immunological effects. Moreover, P19 exerted a beneficial effect on mucine and collagen content of orbital tissue. Hence, P19 offers a potential novel therapeutic approach for GD and associated orbitopathy.


Assuntos
Doença de Graves/tratamento farmacológico , Oftalmopatia de Graves/tratamento farmacológico , Peptídeos Cíclicos/farmacologia , Animais , Colágeno/análise , Modelos Animais de Doenças , Feminino , Doença de Graves/sangue , Doença de Graves/imunologia , Doença de Graves/fisiopatologia , Oftalmopatia de Graves/imunologia , Oftalmopatia de Graves/patologia , Humanos , Imunoglobulinas Estimuladoras da Glândula Tireoide/sangue , Imunoglobulinas Estimuladoras da Glândula Tireoide/imunologia , Camundongos , Mucinas/análise , Órbita/efeitos dos fármacos , Órbita/patologia , Peptídeos Cíclicos/genética , Peptídeos Cíclicos/uso terapêutico , Receptores da Tireotropina/administração & dosagem , Receptores da Tireotropina/genética , Receptores da Tireotropina/imunologia , Glândula Tireoide/efeitos dos fármacos , Glândula Tireoide/imunologia , Glândula Tireoide/fisiopatologia
7.
Cells ; 9(8)2020 08 18.
Artigo em Inglês | MEDLINE | ID: mdl-32824700

RESUMO

Blood-derived microRNA signatures have emerged as powerful biomarkers for predicting and diagnosing cardiovascular disease, cancer, and metabolic disorders. Platelets and platelet-derived microvesicles are a major source of microRNAs. We have previously shown that the inappropriate anticoagulation and storage of blood samples causes substantial platelet activation that is associated with the release of platelet-stored molecules into the plasma. However, it is currently unclear if circulating microRNA levels are affected by artificial platelet activation due to suboptimal plasma preparation. To address this issue, we used a standardized RT-qPCR test for 12 microRNAs (thrombomiR®, TAmiRNA GmbH, Vienna, Austria) that have been associated with cardiovascular and thrombotic diseases and were detected in platelets and/other hematopoietic cells. Blood was prevented from coagulating with citrate-theophylline-adenosine-dipyridamole (CTAD), sodium citrate, or ethylenediaminetetraacetic acid (EDTA) and stored for different time periods either at room temperature or at 4 °C prior to plasma preparation and the subsequent quantification of microRNAs. We found that five microRNAs (miR-191-5p, miR-320a, miR-21-5p, miR-23a-3p, and miR-451a) were significantly increased in the EDTA plasma. Moreover, we observed a time-dependent increase in plasma microRNAs that was most pronounced in the EDTA blood stored at room temperature for 24 h. Furthermore, significant correlations between microRNA levels and plasma concentrations of platelet-stored molecules pointed towards in vitro platelet activation. Therefore, we strongly recommend to (i) use CTAD as an anticoagulant, (ii) process blood samples as quickly as possible, and (iii) store blood samples at 4 °C whenever immediate plasma preparation is not feasible to generate reliable data on blood-derived microRNA signatures.


Assuntos
Anticoagulantes/farmacologia , Coagulação Sanguínea/efeitos dos fármacos , Coleta de Amostras Sanguíneas/métodos , Dipiridamol/farmacologia , Ácido Edético/farmacologia , MicroRNAs/sangue , MicroRNAs/genética , Citrato de Sódio/farmacologia , Adulto , Biomarcadores/sangue , Doadores de Sangue , Plaquetas/metabolismo , Feminino , Humanos , Masculino , Ativação Plaquetária/efeitos dos fármacos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Temperatura
8.
J Endocrinol ; 246(2): 135-147, 2020 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-32573180

RESUMO

Graves' disease is an autoimmune disorder, which is characterized by stimulatory antibodies targeting the human thyrotropin receptor (TSHR), resulting in hyperthyroidism and multiple organ damage. We systematically investigated monomeric and dimeric fusion proteins of the A subunit of TSHR for efficacy to bind to the monoclonal patient antibody M22, to interact with Graves' patient serum samples, and to impact on anti-TSHR antibody titers, hyperthyroidism, tachycardia and other in vivo read-outs in a long-term mouse model of Graves' disease induced by immunization with a recombinant adenovirus encoding TSHR A. Binding assays and functional measurements of TSHR-dependent cAMP formation showed binding of monomeric TSHR-His and dimeric TSHR-Fc to the anti-TSHR antibody M22 at low-effective concentrations (EC50 of 5.7 nmol/L and 8.6 nmol/L) and inhibition of the effects of this antibody at high efficiencies (IC50 values of 16-20 nmol/L). Both proteins also block the effects of polyclonal anti-TSHR antibodies occurring in Graves' patient sera with somewhat lower average efficiencies (mean IC50 values of 29 nmol/L and 68 nmol/L). However, in vivo characterization of epicutaneous patch administrations of TSHR-Fc at doses of 0.3 and 0.6 mg/kg body weight in a murine Graves' disease model did not result in any improvement of disease parameters. In conclusion, high affinity binding of TSHR-Fc to pathological anti-TSHR antibodies was not matched by efficacy to improve Graves' disease parameter in a long-term mouse model.


Assuntos
Doença de Graves/metabolismo , Receptores da Tireotropina/metabolismo , Animais , Autoimunidade/genética , Autoimunidade/fisiologia , AMP Cíclico/genética , AMP Cíclico/metabolismo , Eletrocardiografia , Ensaio de Imunoadsorção Enzimática , Feminino , Doença de Graves/genética , Frequência Cardíaca/genética , Frequência Cardíaca/fisiologia , Humanos , Masculino , Camundongos , Peptídeos/genética , Peptídeos/metabolismo , Receptores da Tireotropina/genética , Temperatura
9.
Eur Thyroid J ; 9(Suppl 1): 51-58, 2020 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-33511085

RESUMO

INTRODUCTION: A novel long-term murine model for Graves' disease (GD) using repeated, long-term immunizations with recombinant adenovirus expressing the extracellular A-subunit of the human thyrotropin receptor (Ad-TSHR) was applied to evaluate the functional anti-TSHR-antibody (TSHR-Ab) profile. METHODS: BALB/c mice received 7 immunizations with either 1010 plaque-forming units of Ad-TSHR or control Ad-GFP. Naïve (nonimmuized native) mice were also studied. Three 3-weekly immunizations were followed by 4-weekly boosts until the 7th immunization. Blocking (TBAb) and stimulating (TSAb) TSHR-Ab were measured with bioassays. Assay cut-offs for TBAb/TSAb were at 34% inhibition and a specimen-to-reference ratio (SRR) of 140%. RESULTS: Nineteen (8 Ad-TSHR-, 4 Ad-GFP-immunized, and 7 native) mice were investigated. All native mice were negative for TSHR-binding inhibitory immunoglobulins (TBII) prior to immunization. Native and Ad-GFP mice were negative in weeks 17 and 27 for TBII and TBAb/TSAb. In native mice, the free thyroxine (fT4) levels (median [25th percentile; 75th percentile]) were in the upper normal range (1.2 ng/mL [1.1; 1.6]) prior to immunization, at weeks 17 (2.2 ng/mL [2.1; 2.4]) and 27 (1.4 ng/mL [1.1; 1.7]), respectively. In contrast, in Ad-TSHR-immunized mice, fT4 values were markedly increased at weeks 17 (4.4 ng/mL [3.9; 6]) and 27 (4.5 ng/mL [4.2; 6]) compared to those in Ad-GFP mice (2 ng/mL [1.8; 2.1] and 1.4 ng/mL [1.1; 1.6]), respectively (p = 0.0008, p = 0.001). In contrast, at week 17, in Ad-TSHR mice, the mean TBII, TBAb, and TSAb levels were 40 IU/L (40; 40); 62% inhibition (38; 69), and 116% SRR (97; 185), respectively; at week 27, they were 40 IU/L (39; 40); 65% inhibition (34; 80) and 95% SRR (63; 187), respectively. Three serum samples from Ad-TSHR mice (38%) demonstrated dual TBAb/TSAb positivity. CONCLUSIONS: TBAb/TSAb were highly prevalent in Ad-TSHR-immunized mice, thus confirming the successful establishment of a novel, long-term murine model for GD. All TBAb- and TSAb-positive Ad-TSHR-immunized mice were TBII-positive. Thus, the binding immunoassay did not differentiate between TSHR-Ab functionality.

10.
Thyroid ; 29(2): 258-267, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30618332

RESUMO

BACKGROUND: Cyclic peptides derived from some cylindrical loops of the leucine-rich repeat domain (LRD) of the thyrotropin receptor (TSHR) have been shown to treat disease manifestations in a mouse model of Graves' disease during a long-term protocol of four-weekly immunizations with adenovirus coding for the TSHR A-subunit (Ad-TSHR289). METHODS: In a follow-up study, two additional cyclic peptides were tested, which were shortened in order to obtain additional information on the minimally involved epitopes and to enable easier production conditions. In addition, a linear peptide was tested, which mimics parts of three loops of the native TSHR LRD structure, and is potentially able to block the discontinuous epitopes of anti-TSHR antibodies. RESULTS: The novel peptides markedly reduced thyroid size, serum thyroxine levels, retro-orbital fibrosis, and tachycardia in Ad-TSHR289-immunized mice. In immunologically naïve mice, administration of the peptides did not induce any immune response. CONCLUSIONS: In summary, novel cyclic peptides mitigate many clinical findings in a mouse model of established Graves' disease and orbitopathy, and may therefore provide an additional therapeutic option compared to existing drugs or interventions.


Assuntos
Oftalmopatia de Graves/imunologia , Órbita/fisiopatologia , Peptídeos Cíclicos/uso terapêutico , Adenoviridae , Animais , Modelos Animais de Doenças , Epitopos/química , Feminino , Fibrose , Oftalmopatia de Graves/terapia , Células HEK293 , Humanos , Imunoglobulinas Estimuladoras da Glândula Tireoide , Camundongos , Camundongos Endogâmicos BALB C , Receptores da Tireotropina/genética , Taquicardia/genética , Tiroxina/sangue
11.
PLoS One ; 13(8): e0201160, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30125285

RESUMO

RATIONALE: Despite advances in pharmacotherapy, heart failure still incurs significant morbidity and mortality. Stimulating antibodies directed against the secondextracellular loop of the human ß1-adrenergic receptor (anti-ß1EC2) cause myocyte damage and heart failure in rats. This receptor domain is 100% homologous between rats and humans. OBJECTIVE: ß1EC2-mimicking cyclopeptides (25-meric) markedly improved the development and/or course of anti-ß1EC2-mediated cardiomyopathy. Further developments should be investigated. METHODS AND RESULTS: The shortened 18-meric cyclic peptide COR-1, in which one of the two disulphide bonds was removed to enable reproducible GMP production, can also be used to treat cardiomyopathic rats. Echocardiography, catheterization and histopathology of the rat hearts revealed that monthly intravenous administrations of COR-1 almost fully reversed the cardiomyopathic phenotype within 6 months at doses of 1 to 4 mg/kg body weight. Administration of COR-1 resulted in markedly reduced anti-ß1EC2-expressing memory B lymphocytes in the spleen despite continued antigenic boosts, but did not significantly decrease overall peripheral anti-ß1EC2 titers. COR-1 did not induce any anti-ß1EC2 or other immune response in naïve rats (corresponding to findings in healthy human volunteers). It did not cause any toxic side effects in GLP studies in dogs, rats or mice, and the "no observed adverse effect level" (NOAEL) exceeded the therapeutic doses by 100-fold. CONCLUSION: The second generation immunomodulating epitope-mimicking cyclopeptide COR-1 (also termed JNJ-5442840) offers promise to treat immune-mediated cardiac diseases.


Assuntos
Insuficiência Cardíaca/tratamento farmacológico , Peptídeos Cíclicos/uso terapêutico , Receptores Adrenérgicos beta 1/imunologia , Sequência de Aminoácidos , Animais , Anticorpos Monoclonais/administração & dosagem , Especificidade de Anticorpos , Modelos Animais de Doenças , Feminino , Cobaias , Insuficiência Cardíaca/etiologia , Insuficiência Cardíaca/fisiopatologia , Humanos , Masculino , Mimetismo Molecular/imunologia , Miocárdio/metabolismo , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/uso terapêutico , Peptídeos Cíclicos/química , Peptídeos Cíclicos/toxicidade , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos , Ratos Endogâmicos Lew , Ratos Wistar , Receptores Adrenérgicos beta 1/química , Receptores Adrenérgicos beta 1/genética
12.
Front Biosci (Landmark Ed) ; 23(11): 2044-2052, 2018 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-29772544

RESUMO

Graves´ disease is an autoimmune disorder, which is characterized by stimulatory antibodies targeting the human thyrotropin receptor (TSHR), resulting in hyperthyroidism and multiple organ damage. The disease can be modelled in mice using adenoviral immunizations with the extracellular A subunit of the TSHR, which induces a long-term stable disease state. TSHR binding cAMP-stimulatory antibodies, thyroid enlargement, elevated serum thyroxin levels, tachycardia, cardiac hypertrophy and orbitopathy are observed in these Ad-TSHR-immunized mice. T cell epitope-derived linear peptides have been identified using immunized HLA-DR3 transgenic mice, which may induce tolerance towards TSHR. A combination of such peptides are being investigated in a first clinical phase I trial in patients with Graves´ disease. Alternatively, intravenous administration of cyclic peptides derived from the interaction site of the TSHR A domain with stimulatory anti-TSHR antibodies can re-establish tolerance towards the antigen in immunized mice, improving symptoms of Graves´ disease within 3 - 4 months after starting these therapies. In immunologically naïve mice, administration of the cyclic peptides did not induce any immune response.


Assuntos
Antígenos/imunologia , Doença de Graves/imunologia , Oftalmopatia de Graves/imunologia , Tolerância Imunológica/imunologia , Receptores da Tireotropina/imunologia , Animais , Modelos Animais de Doenças , Doença de Graves/metabolismo , Doença de Graves/terapia , Oftalmopatia de Graves/metabolismo , Oftalmopatia de Graves/terapia , Humanos , Imunização/métodos , Camundongos Endogâmicos BALB C , Camundongos Transgênicos , Peptídeos Cíclicos/administração & dosagem , Peptídeos Cíclicos/imunologia , Receptores da Tireotropina/antagonistas & inibidores , Receptores da Tireotropina/metabolismo
13.
Endocrinology ; 158(7): 2376-2390, 2017 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-28368444

RESUMO

A model for human Graves disease in mice was used to compare several treatment approaches. The mice received regular adenovirus (Ad) thyroid-stimulating hormone receptor (TSHR) A subunit immunizations (injections every 4 weeks). The generation of anti-TSHR antibodies, enlarged thyroid sizes (goiter), elevated serum thyroxine levels, retro-orbital fibrosis, and cardiac involvement (tachycardia and hypertrophy) were consistently observed over 9 months. Treatment of established disease in these mice using cyclic peptides that mimic one of the cylindrical loops of the TSHR leucine-rich repeat domain improved or cured all investigated parameters after six consecutive monthly injections. The first significant beneficial effects were observed 3 to 4 months after starting these therapies. In immunologically naïve mice, administration of any of the cyclic peptides did not induce any immune response. In contrast, monthly injections of the full antigenic TSHR A domain as fusion protein with immunoglobulin G crystallizable fragment induced clinical signs of allergy in Ad-TSHR-immunized mice and anti-TSHR antibodies in naïve control mice. In conclusion, cyclic peptides resolved many clinical findings in a mouse model of established Graves disease and orbitopathy. In contrast to blocking TSHR by allosteric modulation, the approach does not incur a direct receptor antagonism, which might offer a favorable side effect profile.


Assuntos
Doença de Graves/tratamento farmacológico , Oftalmopatia de Graves/tratamento farmacológico , Peptídeos Cíclicos/uso terapêutico , Receptores da Tireotropina/química , Animais , Células CHO , Cricetinae , Cricetulus , Modelos Animais de Doenças , Feminino , Doença de Graves/sangue , Doença de Graves/complicações , Doença de Graves/patologia , Oftalmopatia de Graves/sangue , Oftalmopatia de Graves/patologia , Células HEK293 , Humanos , Imunoglobulinas Estimuladoras da Glândula Tireoide/sangue , Camundongos , Camundongos Endogâmicos BALB C , Peptídeos Cíclicos/química
14.
Clin Rev Allergy Immunol ; 52(2): 182-193, 2017 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-27368808

RESUMO

Various approaches have been used to model human Graves' disease in mice, including transfected fibroblasts, and plasmid or adenoviral immunisations with the extracellular A subunit of the human thyrotropin receptor (TSHR). Some of these models were only observed for a short time period or were self-limiting. A long-term model for human Graves' disease was established in mice using continuing immunisations (4-weekly injections) with recombinant adenovirus expressing TSHR. Generation of TSHR binding cAMP-stimulatory antibodies, thyroid enlargement and alterations, elevated serum thyroxin levels, tachycardia and cardiac hypertrophy were maintained for at least 9 months in all Ad-TSHR-immunised mice. Here, we show that these mice suffer from orbitopathy, which was detected by serial orbital sectioning and histomorphometry. Attempts to treat established Graves' disease in preclinical mouse model studies have included small molecule allosteric antagonists and specific antagonist antibodies which were isolated from hypothyroid patients. In addition, novel peptides have been conceived which mimic the cylindrical loops of the TSHR leucine-rich repeat domain, in order to re-establish tolerance toward the antigen. Here, we show preliminary results that one set of these peptides improves or even cures all signs and symptoms of Graves' disease in mice after six consecutive monthly injections. First beneficial effects were observed 3-4 months after starting these therapies. In immunologically naïve mice, administration of the peptides did not induce any immune response.


Assuntos
Modelos Animais de Doenças , Doença de Graves/imunologia , Tolerância Imunológica/imunologia , Receptores da Tireotropina/imunologia , Animais , Doença de Graves/patologia , Imunização , Camundongos , Doenças Orbitárias/etiologia , Doenças Orbitárias/imunologia , Doenças Orbitárias/patologia
15.
Heart Int ; 11(1): e10-e16, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27924212

RESUMO

BACKGROUND: Thrombolytic therapy with recombinant tissue plasminogen activator (rtPA) remains the only approved medication for acute ischemic stroke, but incurs significant bleeding risks. Therefore, approaches to combine lower doses of thrombolytic therapy with other effective drugs aim at improving efficacy and reducing bleeding rates. We examined the safety and therapeutic effects of various dosings of rtPA, either alone or combined with glycoprotein VI-Fc fusion protein (GPVI-Fc, Revacept) on experimental stroke in mice. METHODS AND RESULTS: The effect of filament-induced intracerebral thrombus formation and embolization was investigated after a one-hour occlusion of the middle cerebral artery. In accordance with previous studies, treatment with 10 mg/kg rtPA significantly improved functional outcome, cerebral infarct size and edema, but also resulted in markedly increased intracranial bleeding volumes. In contrast, low doses of rtPA (0.1 or 0.35 mg/kg body weight) did not change outcome parameters. However, addition of 1 mg/kg Revacept to 0.35 mg/kg rtPA led to improved reperfusion compared to rtPA alone. Moreover, these combined treatments resulted in improved grip strength, compared to the respective dose of rtPA alone. Infarct-surrounding edema improved after combined treatments, but not after respective single rtPA dosings. Intracranial bleeding volumes were below controls after all low-dose rtPA therapies, given either alone or combined with Revacept. CONCLUSIONS: In contrast to using the equally effective full dose of rtPA, intracranial bleeding was not increased by low-dose rtPA combined with Revacept. Therefore, addition of Revacept to low-dose rtPA does not incur safety risks, but improves efficacy of treatment.

16.
Endocrinology ; 156(4): 1577-89, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25562617

RESUMO

A transient model for human Graves' disease was successfully established in mice using up to 3 immunizations with recombinant adenovirus expressing the extracellular A-subunit of the human TSH receptor (TSHR) (Ad-TSHR). We studied extension of adenovirally induced TSHR A-subunit immunization in mice by using a novel protocol of long-term 3- and 4-weekly injections. Generation of TSHR binding stimulatory antibodies (capacity to stimulate cAMP activity in TSHR-expressing test cells), goiter, and histological thyroid alterations were maintained for at least 9 months in all Ad-TSHR-immunized mice. In response to injection of 10(10) plaque-forming units of Ad-TSHR, also elevated mean serum T4 levels were observed throughout the study. Moreover, cardiac organ involvement (tachycardia and hypertrophy) were consistently observed in these mice. Higher doses of Ad-TSHR (10(11) plaque-forming units) did not produce consistent elevation of T4 and were not associated with a clear increase in heart rate vs controls, probably because these high doses provoked an immune response-induced tachycardia on their own. In summary, a long-term model of Graves' disease induced by a relatively simple protocol of continuing monthly immunizations should allow to investigate long-term disease mechanisms and may possibly obviate the need for more complicated disease models. Moreover, the clinical outcome predictor of tachycardia and cardiac involvement was reliably detected in the model.


Assuntos
Cardiomegalia/etiologia , Doença de Graves/etiologia , Receptores da Tireotropina/imunologia , Taquicardia/etiologia , Adenoviridae , Animais , Autoanticorpos , Cardiomegalia/imunologia , Modelos Animais de Doenças , Feminino , Doença de Graves/imunologia , Imunização , Camundongos , Taquicardia/imunologia , Tiroxina/sangue
17.
PLoS One ; 8(7): e66960, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23935828

RESUMO

OBJECTIVES: We examined the effect of Revacept, an Fc fusion protein which is specifically linked to the extracellular domain of glycoprotein VI (GPVI), on thrombus formation after vessel wall injury and on experimental stroke in mice. BACKGROUND: Several antiplatelet drugs for the treatment of myocardial infarction or ischemic stroke with potent anti-ischemic effects have been developed, but all incur a significant risk of bleeding. METHODS: Platelet adhesion and thrombus formation after endothelial injury was monitored in the carotid artery by intra-vital fluorescence microscopy. The morphological and clinical consequences of stroke were investigated in a mouse model with a one hour-occlusion of the middle cerebral artery. RESULTS: Thrombus formation was significantly decreased after endothelial injury by 1 mg/kg Revacept i.v., compared to Fc only. 1 mg/kg Revacept i.v. applied in mice with ischemic stroke immediately before reperfusion significantly improved functional outcome, cerebral infarct size and edema compared to Fc only. Also treatment with 10 mg/kg rtPA was effective, and functional outcome was similar in both treatment groups. The combination of Revacept with rtPA leads to increased reperfusion compared to treatment with either agent alone. In contrast to rtPA, however, there were no signs of increased intracranial bleeding with Revacept. Both rtPA and Revacept improved survival after stroke compared to placebo treatment. Revacept and vWF bind to collagen and Revacept competitively prevented the binding of vWF to collagen. CONCLUSIONS: Revacept reduces arterial thrombus formation, reduces cerebral infarct size and edema after ischemic stroke, improves functional and prognostic outcome without intracranial bleeding. Revacept not only prevents GPVI-mediated, but probably also vWF-mediated platelet adhesion and aggregate formation. Therefore Revacept might be a potent and safe tool to treat ischemic complications of stroke.


Assuntos
Infarto Cerebral/tratamento farmacológico , Infarto Cerebral/fisiopatologia , Glicoproteínas/uso terapêutico , Fragmentos Fc das Imunoglobulinas/uso terapêutico , Proteínas Recombinantes de Fusão/uso terapêutico , Recuperação de Função Fisiológica , Acidente Vascular Cerebral/tratamento farmacológico , Acidente Vascular Cerebral/fisiopatologia , Animais , Ligação Competitiva/efeitos dos fármacos , Plaquetas/efeitos dos fármacos , Plaquetas/patologia , Comunicação Celular/efeitos dos fármacos , Infarto Cerebral/patologia , Colágeno/metabolismo , Modelos Animais de Doenças , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/patologia , Glicoproteínas/farmacologia , Humanos , Fragmentos Fc das Imunoglobulinas/farmacologia , Infarto da Artéria Cerebral Média/complicações , Infarto da Artéria Cerebral Média/tratamento farmacológico , Infarto da Artéria Cerebral Média/patologia , Infarto da Artéria Cerebral Média/fisiopatologia , Inflamação/patologia , Camundongos , Ligação Proteica/efeitos dos fármacos , Proteínas Recombinantes de Fusão/farmacologia , Recuperação de Função Fisiológica/efeitos dos fármacos , Traumatismo por Reperfusão/complicações , Traumatismo por Reperfusão/patologia , Traumatismo por Reperfusão/fisiopatologia , Acidente Vascular Cerebral/complicações , Acidente Vascular Cerebral/patologia , Trombose/complicações , Trombose/patologia , Trombose/fisiopatologia , Resultado do Tratamento , Trombose Venosa/complicações , Trombose Venosa/tratamento farmacológico , Trombose Venosa/patologia , Trombose Venosa/fisiopatologia , Fator de von Willebrand/metabolismo
18.
PLoS One ; 8(8): e71193, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23951109

RESUMO

AIMS: Glycoprotein VI (GPVI) is a key platelet receptor which mediates plaque-induced platelet activation and consecutive atherothrombosis, but GPVI is also involved in platelet-mediated atheroprogression. Therefore, interference in GPVI-mediated platelet activation has the potential to combine short-term and long-term beneficial effects, specificity and safety especially regarding bleeding complications. METHODS AND RESULTS: We investigated the effects of the soluble dimeric GPVI receptor fusion protein, Revacept, an antagonist of collagen-mediated platelet activation, in an animal model of atherosclerosis: twenty week old rabbits, which had been fed on a cholesterol-rich diet for 8 weeks, received Revacept (8 mg/kg) or control twice weekly for 4 weeks. Pharmacokinetics indicated a slight accumulation of the drug in the serum after repeated dosing of Revacept for 3 weeks. A significant improvement of endothelial dysfunction after 0.06 and 0.6 µg/min acetylcholine and a significant decrease of vessel wall thickening were found after Revacept treatment. Accordingly, aortic vessel weight was reduced, and plaque sizes, macrophage and T-cell invasion tended to be reduced in histological evaluations. Bleeding time was determined after tail clipping in mice. Revacept alone or in combination with widely used anti-platelet drugs revealed a high safety margin with no prolongation of bleeding times. CONCLUSION: Repeated doses of Revacept led to a significant improvement of endothelial dysfunction and vascular morphology in atherosclerotic rabbits. Furthermore, no influence of Revacept on bleeding time alone or in combinations with various anti-platelet drugs was found in mice. Thus, the inhibition of collagen-mediated platelet interaction with the atherosclerotic endothelium by Revacept exerts beneficial effects on morphology and vascular function in vivo and seems to have a wide therapeutic window without influencing the bleeding time.


Assuntos
Aterosclerose/complicações , Aterosclerose/fisiopatologia , Glicoproteínas/farmacologia , Fragmentos Fc das Imunoglobulinas/farmacologia , Trombose/etiologia , Acetilcolina/farmacologia , Animais , Aterosclerose/sangue , Tempo de Sangramento , Peso Corporal , Colesterol/sangue , Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/patologia , Glicoproteínas/administração & dosagem , Fragmentos Fc das Imunoglobulinas/administração & dosagem , Linfócitos/patologia , Macrófagos/patologia , Masculino , Camundongos , Placa Aterosclerótica/patologia , Coelhos
19.
Heart Int ; 7(2): e12, 2012 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-23185679

RESUMO

The ABCG1 protein is centrally involved in reverse cholesterol transport from the vessel wall. Investigation of the effects of ABCG1 overexpression or knockdown in vivo has produced controversial results and strongly depended on the gene intervention model in which it was studied. Therefore, we investigated the effect of local overexpression of human ABCG1 in a novel model of vessel wall-directed adenoviral gene transfer in atherosclerotic rabbits. We conducted local, vascular-specific gene transfer by adenoviral delivery of human ABCG1 (Ad-ABCG1-GFP) in cholesterol-fed atherosclerotic rabbits in vivo. Endothelial overexpression of ABCG1 markedly reduced atheroprogression (plaque size) and almost blunted vascular inflammation, as shown by markedly reduced macrophage and smooth muscle cell invasion into the vascular wall. Also endothelial function, as determined by vascular ultrasound in vivo, was improved in rabbits after gene transfer with Ad-ABCG1-GFP. Therefore, both earlier and later stages of atherosclerosis were improved in this model of somatic gene transfer into the vessel wall. In contrast to results in transgenic mice, over-expression of ABCG1 by somatic gene transfer to the atherosclerotic vessel wall results in a significant improvement of plaque morphology and composition, and of vascular function in vivo.

20.
Eur J Heart Fail ; 14(11): 1230-9, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22968742

RESUMO

AIMS: A novel concept for the treatment of heart failure is the neutralization of antibodies against the ß(1)-adrenergic receptor (anti-ß(1)AR-ab). In a rat model of autoimmune cardiomyopathy, the cyclic peptide COR-1 (given i.v. once monthly) neutralized anti-ß(1)AR-abs and prevented anti-ß(1)AR-ab-induced myocardial damage, and completely reverted cardiac dysfunction over 3-6 months. METHODS AND RESULTS: A clinical phase I trial was designed as a single-blinded, placebo-controlled study. Fifty human volunteers received COR-1 or matching placebo as a single i.v. administration with ascending doses (10-240 mg). Primary endpoints were safety and tolerability, while the pharmacokinetic profile of COR-1 was assessed as a secondary endpoint. All five investigated dose groups were well tolerated; no drug-related side effects occurred. Pharmacokinetics revealed a favourable profile with an almost complete plasma clearance within 60 min after administration. Pharmacodynamic investigation showed dose-dependent efficacy with almost complete scavenging of pathological anti-ß(1)AR-abs ex vivo at the two highest doses. No anti-COR-1 autoantibodies occurred. No other effects on the immune system (such as an increase of crucial cytokines) were observed up to 43 days after drug administration, nor upon incubation of anti-ß(1)AR-ab-positive patient blood samples with COR-1 ex vivo. CONCLUSIONS: COR-1 was shown to be safe after i.v. administration in vivo; no relevant side effects occurred. Efficacy was estimated from ex vivo investigation of the potency to neutralize specific anti-ß(1)-AR-abs. TRIAL REGISTRATION: NCT 01043146, Eudra CT 2008-007745-31.


Assuntos
Antagonistas Adrenérgicos/uso terapêutico , Insuficiência Cardíaca/imunologia , Sistema Imunitário/efeitos dos fármacos , Peptídeos Cíclicos/uso terapêutico , Receptores Adrenérgicos/efeitos dos fármacos , Antagonistas Adrenérgicos/administração & dosagem , Adulto , Análise de Variância , Animais , Área Sob a Curva , Autoanticorpos/imunologia , Citocinas , Feminino , Insuficiência Cardíaca/tratamento farmacológico , Insuficiência Cardíaca/patologia , Hemodinâmica , Humanos , Sistema Imunitário/imunologia , Masculino , Peptídeos Cíclicos/administração & dosagem , Ratos , Método Simples-Cego
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA