Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 52
Filtrar
1.
J Asthma ; 59(2): 243-254, 2022 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-33211619

RESUMO

OBJECTIVE: The aim of this study was to determine if there is an association between the salivary protein profile and disease control in asthma. METHODS: Thirty asthmatic patients (17 adults and 13 children) participated in this study. Saliva samples were collected from healthy subjects, controlled and uncontrolled asthmatics. Individual samples from each group were combined to form a pooled sample, from which proteomic analysis was performed using gel-based quantitative proteomics. RESULTS: Fourteen out of thirty asthmatics were classified to be controlled asthma. Most of asthmatics received inhaled corticosteroids as the controller medications. SDS-PAGE showed predominant bands at high molecular weight in asthmatic saliva compared to that of the controls. Shotgun proteomic analyses indicated that 193 salivary proteins were expressed in both controlled and uncontrolled asthmatics. They were predicted to associate with proteins involved in pathogenesis of asthma including IL-5, IL-6, MCP-1, VEGF, and periostin and asthma medicines (Cromolyn, Nedocromil, and Theophylline). Nucleoside diphosphate kinase (NME1-NME2) only expressed in controlled asthmatics whereas polycystic kidney and hepatic disease 1 (PKHD1)/fibrocystin, zinc finger protein 263 (ZNF263), uncharacterized LOC101060047 (ENSG00000268865), desmoglein 2 (DSG2) and S100 calcium binding protein A2 (S100A2) were only found in uncontrolled asthma. Therefore, the six proteins were associated with disease control in children and adults with asthma. CONCLUSION: Our findings suggest that NME1-NME2, PKHD1, ZNF 263, uncharacterized LOC101060047, DSG 2 and S100 A2 in saliva are associated with disease control in asthma.


Assuntos
Asma , Proteômica , Corticosteroides/uso terapêutico , Adulto , Asma/tratamento farmacológico , Asma/metabolismo , Biomarcadores , Criança , Humanos , Proteínas e Peptídeos Salivares/uso terapêutico
2.
Angew Chem Int Ed Engl ; 60(10): 5348-5356, 2021 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-33345438

RESUMO

Blood feeding arthropods, such as leeches, ticks, flies and mosquitoes, provide a privileged source of peptidic anticoagulant molecules. These primarily operate through inhibition of the central coagulation protease thrombin by binding to the active site and either exosite I or exosite II. Herein, we describe the rational design of a novel class of trivalent thrombin inhibitors that simultaneously block both exosites as well as the active site. These engineered hybrids were synthesized using tandem diselenide-selenoester ligation (DSL) and native chemical ligation (NCL) reactions in one-pot. The most potent trivalent inhibitors possessed femtomolar inhibition constants against α-thrombin and were selective over related coagulation proteases. A lead hybrid inhibitor possessed potent anticoagulant activity, blockade of both thrombin generation and platelet aggregation in vitro and efficacy in a murine thrombosis model at 1 mg kg-1 . The rational engineering approach described here lays the foundation for the development of potent and selective inhibitors for a range of other enzymatic targets that possess multiple sites for the disruption of protein-protein interactions, in addition to an active site.


Assuntos
Anticoagulantes/uso terapêutico , Inibidores da Agregação Plaquetária/uso terapêutico , Proteínas e Peptídeos Salivares/uso terapêutico , Trombose/tratamento farmacológico , Amblyomma/química , Animais , Anopheles/química , Anticoagulantes/síntese química , Anticoagulantes/metabolismo , Domínio Catalítico , Humanos , Masculino , Camundongos Endogâmicos C57BL , Inibidores da Agregação Plaquetária/síntese química , Inibidores da Agregação Plaquetária/metabolismo , Ligação Proteica , Engenharia de Proteínas , Proteínas e Peptídeos Salivares/síntese química , Proteínas e Peptídeos Salivares/metabolismo , Trombina/química , Trombina/metabolismo , Moscas Tsé-Tsé/química
3.
Sci Rep ; 10(1): 6388, 2020 04 14.
Artigo em Inglês | MEDLINE | ID: mdl-32286411

RESUMO

We have investigated Amblyomin-X-treated horse melanomas to better understand its mode of action through transcriptome analysis and the in vivo model. Amblyomin-X is a Kunitz-type homologous protein that selectively leads to the death of tumor cells via ER stress and apoptosis, currently under investigation as a new drug candidate for cancer treatment. Melanomas are immunogenic tumors, and a better understanding of the immune responses is warranted. Equine melanomas are spontaneous and not so aggressive as human melanomas are, as this study shows that the in vivo treatment of encapsulated horse melanoma tumors led to a significant reduction in the tumor size or even the complete disappearance of the tumor mass through intratumoral injections of Amblyomin-X. Transcriptome analysis identified ER- and mitochondria-stress, modulation of the innate immune system, apoptosis, and possibly immunogenic cell death activation. Interactome analysis showed that Amblyomin-X potentially interacts with key elements found in transcriptomics. Taken together, Amblyomin-X modulated the tumor immune microenvironment in different ways, at least contributing to induce tumor cell death.


Assuntos
Antineoplásicos/uso terapêutico , Proteínas de Artrópodes/uso terapêutico , Doenças dos Cavalos/tratamento farmacológico , Melanoma/veterinária , Proteínas e Peptídeos Salivares/uso terapêutico , Animais , Morte Celular/efeitos dos fármacos , Descoberta de Drogas , Cavalos , Masculino , Melanoma/tratamento farmacológico , Microambiente Tumoral/efeitos dos fármacos
4.
Arch Oral Biol ; 99: 31-42, 2019 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-30599395

RESUMO

Saliva contains a large number of proteins that play various crucial roles to maintain the oral health and tooth integrity. This oral fluid is proposed to be one of the most important host factors, serving as a special medium for monitoring aspects of microorganisms, diet and host susceptibility involved in the caries process. Extensive salivary proteomic and peptidomic studies have resulted in considerable advances in the field of biomarkers discovery for dental caries. These salivary biomarkers may be exploited for the prediction, diagnosis, prognosis and treatment of dental caries, many of which could also provide the potential templates for bioactive peptides used for the biomimetic management of dental caries, rather than repairing caries lesions with artificial materials. A comprehensive understanding of the biological function of salivary proteins as well as their derived biomimetic peptides with promising potential against dental caries has been long awaited. This review overviewed a collection of current literature and addressed the majority of different functions of salivary proteins and peptides with their potential as functional biomarkers for caries risk assessment and clinical prospects for the anti-caries application.


Assuntos
Cárie Dentária/diagnóstico , Cárie Dentária/terapia , Diagnóstico Precoce , Peptidomiméticos/metabolismo , Peptidomiméticos/uso terapêutico , Proteínas e Peptídeos Salivares/fisiologia , Proteínas e Peptídeos Salivares/uso terapêutico , Anti-Infecciosos/farmacologia , Biomarcadores/metabolismo , Cariostáticos , Cárie Dentária/metabolismo , Suscetibilidade à Cárie Dentária/fisiologia , Humanos , Saúde Bucal , Proteômica , Medição de Risco , Saliva/química , Proteínas e Peptídeos Salivares/classificação , Remineralização Dentária
5.
Oncotarget ; 7(38): 62255-62266, 2016 09 20.
Artigo em Inglês | MEDLINE | ID: mdl-27566592

RESUMO

Renal cell carcinoma (RCC), also called kidney cancer or renal adenocarcinoma, is highly resistant to current treatments. It has been previously reported that a Kunitz-type inhibitor domain-containing protein, isolated from the salivary glands of the Amblyomma cajennense tick, triggers apoptosis in murine renal adenocarcinoma cells (Renca) by inhibiting the proteasome and endoplasmic reticulum stress. Of note, Amblyomin-X is the corresponding recombinant protein identified in the cDNA library from A. cajennense salivary glands. Herein, using orthotopic kidney tumors in mice, we demonstrate that Amblyomin-X is able to drastically reduce the incidence of lung metastases by inducing cell cycle arrest and apoptosis. The in vitro assays show that Amblyomin-X is capable of reducing the proliferation rate of Renca cells, promoting cell cycle arrest, and down-regulating the expression of crucial proteins (cyclin D1, Ki67 and Pgp) involved in the aggressiveness and resistance of RCC. Regarding non-tumor cells (NIH3T3), Amblyomin-X produced minor effects in the cyclin D1 levels. Interestingly, observing the image assays, the fluorescence-labelled Amblyomin-X was indeed detected in the tumor stroma whereas in healthy animals it was rapidly metabolized and excreted. Taken the findings together, Amblyomin-X can be considered as a potential anti-RCC drug candidate.


Assuntos
Apoptose/efeitos dos fármacos , Carcinoma de Células Renais/tratamento farmacológico , Neoplasias Renais/tratamento farmacológico , Inibidores de Proteassoma/farmacologia , Proteínas e Peptídeos Salivares/farmacologia , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/metabolismo , Animais , Proteínas de Artrópodes , Carcinoma de Células Renais/patologia , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Ciclina D1/metabolismo , Regulação para Baixo , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Humanos , Antígeno Ki-67/metabolismo , Rim/patologia , Neoplasias Renais/patologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Células NIH 3T3 , Complexo de Endopeptidases do Proteassoma/efeitos dos fármacos , Inibidores de Proteassoma/uso terapêutico , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Proteínas Recombinantes/farmacologia , Proteínas Recombinantes/uso terapêutico , Proteínas e Peptídeos Salivares/uso terapêutico , Testes de Toxicidade , Ensaios Antitumorais Modelo de Xenoenxerto
6.
Clin Rheumatol ; 35(10): 2585-9, 2016 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-27448151

RESUMO

The objective of this study is to assess the occurrence of and risk factors for serious infections in rheumatoid arthritis (RA)-associated interstitial lung disease (ILD). All patients with RA-ILD (ACR 1987 criteria for RA) seen at a single center from 1998 to 2014 were identified and manually screened for study inclusion. Follow-up data were abstracted until death or December 31, 2015. Serious infection was defined as requiring antimicrobial therapy and hospitalization. Risk of infection was analyzed by person-year (py) methods using time-dependent covariates started when the medication was first used and stopped 30 days after the medication was discontinued. Of the 181 included patients, 87 (48 %) were female. The mean age at ILD diagnosis was 67.4 (±9.9) years, and median follow-up time was 3.1 (range: 0.01 to 14.8) years. Higher infection rates were observed during the first year after ILD diagnosis (14.1 per 100 py) than subsequently (5.7 per 100 py; p = 0.001). Pneumonia was the most common (3.9 per 100 py). Overall infection risk was higher in organizing pneumonia (OP) (27.1 per 100 py) than usual interstitial pneumonia (7.7 per 100 py) or non-specific interstitial pneumonia (5.5 per 100 py) (p < 0.001). The highest infection rate observed was with a daily prednisone use >10 mg per day (15.4 per 100 py). Patients with RA-ILD are at risk of serious infection. Prednisone use >10 mg per day was associated with higher rates of infection. Immunosuppressive drug use governed by concern for risk of infection in patients with ILD resulting in channeling bias cannot be excluded.


Assuntos
Antirreumáticos/efeitos adversos , Artrite Reumatoide/complicações , Doenças Pulmonares Intersticiais/complicações , Pneumonia/etiologia , Prednisona/efeitos adversos , Proteínas e Peptídeos Salivares/efeitos adversos , Idoso , Antirreumáticos/uso terapêutico , Artrite Reumatoide/tratamento farmacológico , Feminino , Humanos , Incidência , Masculino , Pessoa de Meia-Idade , Pneumonia/epidemiologia , Prednisona/uso terapêutico , Risco , Proteínas e Peptídeos Salivares/uso terapêutico
7.
Arch Med Res ; 46(6): 502-6, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26143971

RESUMO

Opioid peptides are potent analgesics with therapeutic potential in the treatment of acute and chronic pain. Their efficacy is limited by peptidases (enkephalinases). Opiorphin pentapeptide (QRFSR) is the first characterized human endogenous inhibitor of enkephalinases. The peptide is able to increase the binding and affinity of endogenous opiates to mu opioid receptors; thus, the mechanism of opiorphin may provide a new therapeutic approach in pain management. The analgesic effect of opiorphin was proven in several earlier published in vitro and in vivo studies. Our aim was to test the transfer of opiorphin through a blood-brain barrier model for the first time. The flux of opiorphin was tested on a blood-brain barrier culture model consisting of rat brain endothelial, glial and pericyte cells. Brain endothelial cells in this triple co-culture model form tight monolayers characterized by transendothelial electrical resistance measurement. Relative quantity of the peptide was estimated by mass spectrometry. The transfer of opiorphin through the blood-brain barrier model was estimated to be ∼3%, whereas the permeability coefficient was 0.53 ± 1.36 × 10(-6) cm/s (n = 4). We also observed rapid conversion of N-terminal glutamine into pyroglutamic acid during the transfer experiments. Our results indicate that opiorphin crosses cultured brain endothelial cells in the absence of serum factors in a significant amount. This is in agreement with previous in vivo data showing potentiation of enkephalin-mediated antinociception. We suggest that opiorphin may have a potential as a centrally acting novel drug to treat pain.


Assuntos
Analgésicos/uso terapêutico , Barreira Hematoencefálica/química , Encéfalo/metabolismo , Células Endoteliais/metabolismo , Oligopeptídeos/uso terapêutico , Peptídeos Opioides/química , Proteínas e Peptídeos Salivares/uso terapêutico , Animais , Encéfalo/citologia , Humanos , Ratos , Ratos Wistar
8.
Biomed Res Int ; 2015: 813632, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25834826

RESUMO

OBJECTIVE: To kill urothelial cancer cells while preserving healthy cells, this study used photothermal therapy (PTT). PTT techniques target urothelial cancer cells using gold nanoparticles (GNPs) and a green light laser. MATERIALS AND METHODS: The GNPs were conjugated with anti-Mucin 7 antibodies, which acted as a probe for targeting tumor cells. Conjugated GNPs were exposed to a green light laser (532 nm) with sufficient thermal energy to kill the transitional cell carcinomas (TCCs). RESULTS: According to our results, nanoparticles conjugated with Mucin 7 antibodies damaged all types of cancer cells (MBT2, T24, 9202, and 8301) at relatively low energy levels (i.e., 500 laser shots at 10 W/cm(2) in power, 1.6 Hz in frequency, and 300 ms in duration). Nonconjugated nanoparticles required 30 W/cm(2) or more to achieve the same effect. Cell damage was directly related to irradiation time and applied laser energy. CONCLUSIONS: The minimally invasive PTT procedure combined with Mucin 7 targeted GNPs is able to kill cancer cells and preserve healthy cells. The success of this treatment technique can likely be attributed to the lower amount of energy required to kill targeted cancer cells compared with that required to kill nontargeted cancer cells. Our in vitro pilot study yielded promising results; however, additional animal studies are required to confirm these findings.


Assuntos
Anticorpos Anti-Idiotípicos/uso terapêutico , Nanopartículas Metálicas/uso terapêutico , Mucinas/uso terapêutico , Neoplasias/terapia , Proteínas e Peptídeos Salivares/uso terapêutico , Animais , Anticorpos Anti-Idiotípicos/química , Linhagem Celular Tumoral , Ouro/administração & dosagem , Ouro/química , Humanos , Nanopartículas Metálicas/química , Mucinas/química , Mucinas/imunologia , Neoplasias/imunologia , Neoplasias/patologia , Fototerapia , Proteínas e Peptídeos Salivares/química , Proteínas e Peptídeos Salivares/imunologia , Urotélio/imunologia , Urotélio/patologia
9.
Peptides ; 55: 47-51, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24486428

RESUMO

Human opiorphin is a recently identified endogenous pentapeptide, encoded by ProL1 multigenes family that contributes to cardiovascular modulation. The aim of this study was to evaluate the effect of opiorphin through intravenous injection (i.v.) on mean arterial pressure (MAP) regulation. To investigate the bioactivity of opiorphin, a rat cannulation model was developed for MAP measurement and blood sampling. In our present study, opiorphin (200-700 nmol/kg) increased MAP in dose-related and time-dependent manner in conscious rats, which associated highly with the elevation of angiotensin II (AngII) levels in serum. Furthermore, the MAP elevation induced by opiorphin was completely blocked by AngII receptor antagonist valsartan and partially attenuated by angiotensin-converting enzyme inhibitor captopril. Finally, we tested the effect of opiorphin in hypoxia condition, which exhibited that opiorphin reversed hypoxia induced hypotension in conscious rats. Taken together, these results indicated that opiorphin may play an important role in the modulation of blood pressure through AngII dependent pathway, which may help future development of potent clinical therapeutics for emergency treatment.


Assuntos
Oligopeptídeos/fisiologia , Sistema Renina-Angiotensina , Proteínas e Peptídeos Salivares/fisiologia , Angiotensina II/sangue , Animais , Anti-Hipertensivos/farmacologia , Pressão Sanguínea , Captopril/farmacologia , Hipotensão/tratamento farmacológico , Hipotensão/etiologia , Hipóxia/complicações , Hipóxia/fisiopatologia , Masculino , Oligopeptídeos/farmacologia , Oligopeptídeos/uso terapêutico , Ratos Wistar , Proteínas e Peptídeos Salivares/farmacologia , Proteínas e Peptídeos Salivares/uso terapêutico , Tetrazóis/farmacologia , Valina/análogos & derivados , Valina/farmacologia , Valsartana
10.
Biomed Pharmacother ; 67(3): 192-6, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23433900

RESUMO

Cancer is linked to hypercoagulability, and many studies have shown that anticoagulant drugs affect tumor progression. In this study was demonstrated that the Amblyomin-X (which is a recombinant protein that exerts similarity to the Kunitz-type inhibitors and shows pro-apoptotic effects in different tumor cell lines) and heparin (a classic anticoagulant) have similar effects on cancer progression and on normalization of the hypercoagulable state. However, Amblyomin-X showed a distinct mechanism in triggering its effects in vitro, because it exerted a cytotoxic effect in cancer cells by inducing apoptosis and promoting cell cycle arrest.


Assuntos
Apoptose/fisiologia , Progressão da Doença , Inibidores do Fator Xa , Melanoma Experimental/terapia , Proteínas e Peptídeos Salivares/uso terapêutico , Trombofilia/terapia , Animais , Proteínas de Artrópodes , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Masculino , Melanoma Experimental/metabolismo , Melanoma Experimental/patologia , Camundongos , Camundongos Endogâmicos C57BL , Proteínas e Peptídeos Salivares/fisiologia , Trombofilia/metabolismo , Trombofilia/patologia , Ensaios Antitumorais Modelo de Xenoenxerto/métodos
12.
Thromb Res ; 130(3): e163-70, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22683021

RESUMO

Melanoma is a highly metastatic cancer and there is strong evidence that the clotting initiator protein, tissue factor (TF), contributes to its aggressive pattern. TF inhibitors may attenuate primary tumor growth and metastasis. In this study, we evaluated the effect of ixolaris, a TF inhibitor, on a murine model of melanoma B16F10 cells. Enzymatic assays performed with B16F10 and human U87-MG tumor cells as the TF source showed that ixolaris inhibits the generation of FX in either murine, human or hybrid FVIIa/TF complexes. The effect of ixolaris on the metastatic potential was further estimated by intravenous injection of B16F10 cells in C57BL/6 mice. Ixolaris (250 µg/kg) dramatically decreased the number of pulmonary tumor nodules (4 ± 1 compared to 47 ± 10 in the control group). Furthermore, a significant decrease in tumor weights was observed in primary tumor growth assays in animals treated with ixolaris (250 µg/kg) from days 3 to 18 after a subcutaneous inoculation of melanoma cells. Remarkably, immunohistochemical analyses showed that inhibition of melanoma growth by ixolaris is accompanied by a significant downregulation of both vascular endothelial growth factor (VEGF) expression and microvascular density in the tumor mass. Our data demonstrate that ixolaris targets B16F10 cell-derived TF, resulting in the reduction of both the primary tumor growth and the metastatic potential of melanoma, as well as the inhibition of tumor angiogenesis. Therefore TF may be a potential target for the treatment of this aggressive malignancy.


Assuntos
Melanoma/tratamento farmacológico , Melanoma/secundário , Proteínas e Peptídeos Salivares/uso terapêutico , Tromboplastina/antagonistas & inibidores , Animais , Crescimento Celular , Linhagem Celular Tumoral , Proliferação de Células , Humanos , Melanoma/patologia , Camundongos , Camundongos Endogâmicos C57BL , Resultado do Tratamento
13.
PLoS One ; 7(4): e35627, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22558182

RESUMO

CLINICAL RELEVANCE: Late complications can occur with use of current antimetabolites to prevent scarring following glaucoma filtration surgery (GFS). Safer, more targeted, anti-fibrosis agents are sought. OBJECTIVES: The protein saratin has been shown to exhibit anti-fibrotic and anti-thrombotic properties in response to injury, but had not been used for glaucoma surgery. The goal of this study was to compare the efficacy of saratin with that of the widely accepted mitomycin-C (MMC) in prolonging bleb survival following GFS in the rabbit model. Two saratin delivery routes were compared; a single intraoperative topical application versus a combination of intraoperative topical application with two additional postoperative injections. METHODS: Twenty-four New Zealand White rabbits underwent GFS and received either intraoperative topical saratin, intraoperative topical saratin plus two injections on post-operative days 4 and 8, balanced saline solution (BSS), or MMC. The bleb tissues and their elevation durations were compared based on clinical and histological findings. RESULTS: Rabbits receiving topical+injections of saratin had a mean bleb survival of 33.6±8.5 days, significantly higher than the negative BSS controls, which averaged 17.4±6.0 days (p = 0.018). No improvement over BSS was seen for rabbits receiving topical saratin only (15.5±4.8 days, p = 0.749). Rabbits receiving saratin did not develop bleb avascularity and thinning associated with MMC treatment and there were no apparent clinical signs of toxicity. CONCLUSIONS: Treatment with a single intraoperative topical application plus two additional postoperative injections significantly prolonged bleb elevation comparable to MMC, but without toxicity; however, topical application alone was ineffective.


Assuntos
Cicatriz/prevenção & controle , Fibrose/prevenção & controle , Cirurgia Filtrante/efeitos adversos , Glaucoma/cirurgia , Proteínas e Peptídeos Salivares/administração & dosagem , Trombose/prevenção & controle , Administração Tópica , Animais , Plaquetas/efeitos dos fármacos , Cicatriz/etiologia , Modelos Animais de Doenças , Olho/efeitos dos fármacos , Olho/patologia , Fibrose/etiologia , Glaucoma/patologia , Humanos , Injeções Intraoculares , Mitomicina/administração & dosagem , Mitomicina/uso terapêutico , Coelhos , Proteínas e Peptídeos Salivares/uso terapêutico , Trombose/etiologia
14.
Thromb Haemost ; 107(1): 111-23, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22159626

RESUMO

Salivary glands from haematophagous animals express a notable diversity of negative modulators of platelet function. Triplatin is an inhibitor of collagen-induced platelet aggregation which has been described as an antagonist of glycoprotein VI (GPVI). Because triplatin displays sequence homology to members of the lipocalin family of proteins, we investigated whether triplatin mechanism of action could be explained by interaction with pro-haemostatic prostaglandins. Our results demonstrate that triplatin inhibits platelet aggregation induced by low doses of collagen, thromboxane A2 (TXA(2)) mimetic (U46619), and arachidonic acid (AA). On the other hand, it does not inhibit platelet aggregation by convulxin, PMA, or low-dose ADP. Isothermal titration calorimetry (ITC) revealed that triplatin binds AA, cTXA(2), TXB(2), U46619 or prostaglandin (PG)H(2) mimetic (U51605). Consistent with its ligand specificity, triplatin induces relaxation of rat aorta contracted with U46619. Triplatin also interacts with PGF(2α) and PGJ(2), but not with leukotrienes, AA or biogenic amines. Surface plasmon resonance experiments failed to demonstrate interaction of triplatin with GPVI; it also did to inhibit platelet adhesion to fibrillar or soluble collagen. Because triplatin displays sequence similarity to apolipoprotein D (ApoD) - a lipocalin associated with high-density lipoprotein, ApoD was tested as a putative TXA(2)-binding molecule. ITC failed to demonstrate binding of ApoD to all prostanoids described above, or to AA. Furthermore, ApoD was devoid of inhibitory properties towards platelets activation by AA, collagen, or U46619. In conclusion, triplatin mechanism of action has been elucidated without ambiguity as a novel TXA(2)- and PGF(2α)- binding protein. It conceivably blocks platelet aggregation and vasoconstriction, thus contributing to successful blood feeding at the vector-host interface.


Assuntos
Inibidores da Agregação Plaquetária/farmacologia , Glicoproteínas da Membrana de Plaquetas/química , Glândulas Salivares/metabolismo , Proteínas e Peptídeos Salivares/uso terapêutico , Tromboxano A2/metabolismo , Ácido 15-Hidroxi-11 alfa,9 alfa-(epoximetano)prosta-5,13-dienoico/farmacologia , Aminas/química , Animais , Ácido Araquidônico/metabolismo , Cavalos , Humanos , Ligantes , Modelos Biológicos , Agregação Plaquetária/efeitos dos fármacos , Prostaglandinas H/farmacologia , Ligação Proteica , Serpentes
15.
J Immunol ; 187(9): 4913-9, 2011 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-21964028

RESUMO

Experimental evidence suggests that C inhibition and more particularly combined inhibition of C and the TLR coreceptor CD14 may be of therapeutic benefit in sepsis and other inflammatory conditions. A barrier to the testing and further development of many inhibitors is that their activity is species specific. Pig is a relevant species for experimental models of human disease, and this study undertakes a comprehensive comparison of the inhibitory efficacy of the C5 inhibitor Ornithodoros moubata C inhibitor (OmCI) in human and porcine whole blood ex vivo models of Escherichia coli-induced sepsis. The effect of OmCI on complement activity in pigs undergoing E. coli sepsis was also examined. Porcine and human serum, and whole blood anticoagulated with lepirudin, was incubated with E. coli and the effect of OmCI investigated. The ex vivo results were virtually identical in pig and human. OmCI completely ablated the activity of all three C pathways at 0.64 µM. E. coli-induced C activation and expression of CD11b (wCD11R3 in the pig), was abolished ex vivo at 0.32 µM OmCI. Combining anti-CD14 and OmCI reduced the formation of IL-8 and TNF-α more potently than the single inhibitors. OmCI also efficiently bound E. coli-induced leukotriene B(4) in pig and human plasma. In support of our ex vivo findings, in vivo the activity of all C pathways was inhibited at 0.6 mg OmCI/kg pig. In conclusion, OmCI efficiently inhibited pig and human C activation, has accompanying anti-inflammatory effects and is a promising candidate inhibitor for further in vivo studies of sepsis.


Assuntos
Complemento C5a/antagonistas & inibidores , Proteínas Inativadoras do Complemento/fisiologia , Ornithodoros/imunologia , Animais , Complemento C5a/metabolismo , Proteínas Inativadoras do Complemento/uso terapêutico , Via Alternativa do Complemento/imunologia , Via Clássica do Complemento/imunologia , Modelos Animais de Doenças , Infecções por Escherichia coli/imunologia , Infecções por Escherichia coli/prevenção & controle , Feminino , Humanos , Masculino , Projetos Piloto , Proteínas e Peptídeos Salivares/fisiologia , Proteínas e Peptídeos Salivares/uso terapêutico , Sepse/imunologia , Sepse/prevenção & controle , Suínos
16.
J Clin Periodontol ; 38 Suppl 11: 126-41, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21323710

RESUMO

AIMS: The goal of this review is to identify the antimicrobial proteins in the oral fluids, saliva and gingival crevicular fluid and identify functional families and candidates for antibacterial treatment. RESULTS: Periodontal biofilms initiate a cascade of inflammatory and immune processes that lead to the destruction of gingival tissues and ultimately alveolar bone loss and tooth loss. Treatment of periodontal disease with conventional antibiotics does not appear to be effective in the absence of mechanical debridement. An alternative treatment may be found in antimicrobial peptides and proteins, which can be bactericidal and anti-inflammatory and block the inflammatory effects of bacterial toxins. The peptides have co-evolved with oral bacteria, which have not developed significant peptide resistance. Over 45 antibacterial proteins are found in human saliva and gingival crevicular fluid. The proteins and peptides belong to several different functional families and offer broad protection from invading microbes. Several antimicrobial peptides and proteins (AMPs) serve as templates for the development of therapeutic peptides and peptide mimetics, although to date none have demonstrated efficacy in human trials. CONCLUSIONS: Existing and newly identified AMPs may be developed for therapeutic use in periodontal disease or can serve as templates for peptide and peptide mimetics with improved therapeutic indices.


Assuntos
Anti-Infecciosos/análise , Peptídeos Catiônicos Antimicrobianos/análise , Doenças Periodontais/metabolismo , Anti-Infecciosos/uso terapêutico , Anti-Inflamatórios/análise , Anti-Inflamatórios/uso terapêutico , Peptídeos Catiônicos Antimicrobianos/uso terapêutico , Biofilmes , Líquido do Sulco Gengival/química , Humanos , Doenças Periodontais/tratamento farmacológico , Doenças Periodontais/microbiologia , Saliva/química , Proteínas e Peptídeos Salivares/análise , Proteínas e Peptídeos Salivares/uso terapêutico
17.
Toxicon ; 56(7): 1145-54, 2010 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-20570593

RESUMO

The aim of this study was to evaluate the anti-tumor activity of Amblyomin-X, a serine protease Kunitz-type inhibitor. Amblyomin-X induced tumor mass regression and decreased number of metastatic events in a B16F10 murine melanoma model. Alterations on expression of several genes related to cell cycle were observed when two tumor cell lines were treated with Amblyomin-X. PSMB2, which encodes a proteasome subunit, was differentially expressed, in agreement to inhibition of proteasomal activity in both cell lines. In conclusion, our results indicate that Amblyomin-X selectively acts on tumor cells by inducing apoptotic cell death, possibly by targeting the ubiquitin-proteasome system.


Assuntos
Antineoplásicos/uso terapêutico , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Ixodidae/química , Melanoma Experimental/tratamento farmacológico , Complexo de Endopeptidases do Proteassoma/metabolismo , Proteínas e Peptídeos Salivares/uso terapêutico , Inibidores de Serina Proteinase/uso terapêutico , Ubiquitina/metabolismo , Animais , Antineoplásicos/química , Antineoplásicos/isolamento & purificação , Apoptose/efeitos dos fármacos , Proteínas de Artrópodes , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Modelos Animais de Doenças , Ixodidae/genética , Melanoma Experimental/patologia , Camundongos , Camundongos Endogâmicos C57BL , Metástase Neoplásica , Complexo de Endopeptidases do Proteassoma/efeitos dos fármacos , Glândulas Salivares/química , Proteínas e Peptídeos Salivares/química , Proteínas e Peptídeos Salivares/isolamento & purificação , Inibidores de Serina Proteinase/química , Inibidores de Serina Proteinase/isolamento & purificação
18.
J Thromb Haemost ; 7(11): 1855-64, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19624457

RESUMO

BACKGROUND: The expression levels of the clotting initiator protein Tissue Factor (TF) correlate with vessel density and the histological malignancy grade of glioma patients. Increased procoagulant tonus in high grade tumors (glioblastomas) also indicates a potential role for TF in progression of this disease, and suggests that anticoagulants could be used as adjuvants for its treatment. OBJECTIVES: We hypothesized that blocking of TF activity with the tick anticoagulant Ixolaris might interfere with glioblastoma progression. METHODS AND RESULTS: TF was identified in U87-MG cells by flow-cytometric and functional assays (extrinsic tenase). In addition, flow-cytometric analysis demonstrated the exposure of phosphatidylserine in the surface of U87-MG cells, which supported the assembly of intrinsic tenase (FIXa/FVIIIa/FX) and prothrombinase (FVa/FXa/prothrombin) complexes, accounting for the production of FXa and thrombin, respectively. Ixolaris effectively blocked the in vitro TF-dependent procoagulant activity of the U87-MG human glioblastoma cell line and attenuated multimolecular coagulation complexes assembly. Notably, Ixolaris inhibited the in vivo tumorigenic potential of U87-MG cells in nude mice, without observable bleeding. This inhibitory effect of Ixolaris on tumor growth was associated with downregulation of VEGF and reduced tumor vascularization. CONCLUSION: Our results suggest that Ixolaris might be a promising agent for anti-tumor therapy in humans.


Assuntos
Proliferação de Células/efeitos dos fármacos , Glioblastoma/tratamento farmacológico , Neovascularização Patológica/tratamento farmacológico , Proteínas e Peptídeos Salivares/farmacologia , Tromboplastina/antagonistas & inibidores , Animais , Linhagem Celular Tumoral , Progressão da Doença , Regulação para Baixo/efeitos dos fármacos , Glioblastoma/patologia , Humanos , Camundongos , Camundongos Nus , Proteínas e Peptídeos Salivares/uso terapêutico , Fator A de Crescimento do Endotélio Vascular/genética
19.
Ann Neurol ; 65(1): 67-75, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19194881

RESUMO

OBJECTIVE: Complement mediated injury of the neuromuscular junction is considered a primary disease mechanism in human myasthenia gravis and animal models of experimentally acquired myasthenia gravis (EAMG). We utilized active and passive models of EAMG to investigate the efficacy of a novel C5 complement inhibitor rEV576, recombinantly produced protein derived from tick saliva, in moderating disease severity. METHODS: Standardized disease severity assessment, serum complement hemolytic activity, serum cytotoxicity, acetylcholine receptor (AChR) antibody concentration, IgG subclassification, and C9 deposition at the neuromuscular junction were used to assess the effect of complement inhibition on EAMG induced by administration of AChR antibody or immunization with purified AChR. RESULTS: Administration of rEV576 in passive transfer EAMG limited disease severity as evidenced by 100% survival rate and a low disease severity score. In active EAMG, rats with severe and mild EAMG were protected from worsening of disease and had limited weight loss. Serum complement activity (CH(50)) in severe and mild EAMG was reduced to undetectable levels during treatment, and C9 deposition at the neuromuscular junction was reduced. Treatment with rEV576 resulted in reduction of toxicity of serum from severe and mild EAMG rats. Levels of total AChR IgG, and IgG(2a) antibodies were similar, but unexpectedly, the concentration of complement fixing IgG(1) antibodies was lower in a group of rEV576-treated animals, suggesting an effect of rEV576 on cellular immunity. INTERPRETATION: Inhibition of complement significantly reduced weakness in two models of EAMG. C5 inhibition could prove to be of significant therapeutic value in human myasthenia gravis.


Assuntos
Complemento C5/antagonistas & inibidores , Proteínas Inativadoras do Complemento/uso terapêutico , Miastenia Gravis Autoimune Experimental/prevenção & controle , Animais , Anticorpos/efeitos adversos , Proteínas de Artrópodes , Linhagem Celular Tumoral , Complemento C9/metabolismo , Proteínas Inativadoras do Complemento/imunologia , Proteínas Inativadoras do Complemento/metabolismo , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Feminino , Imunoglobulina G/sangue , Imunoglobulina G/classificação , Proteínas de Insetos/imunologia , Proteínas de Insetos/uso terapêutico , Força Muscular/efeitos dos fármacos , Miastenia Gravis Autoimune Experimental/etiologia , Miastenia Gravis Autoimune Experimental/imunologia , Miastenia Gravis Autoimune Experimental/patologia , Junção Neuromuscular/efeitos dos fármacos , Junção Neuromuscular/metabolismo , Ratos , Ratos Endogâmicos Lew , Receptores Colinérgicos/imunologia , Rabdomiossarcoma/metabolismo , Rabdomiossarcoma/patologia , Proteínas e Peptídeos Salivares/sangue , Proteínas e Peptídeos Salivares/imunologia , Proteínas e Peptídeos Salivares/uso terapêutico , Índice de Gravidade de Doença , Fatores de Tempo , Redução de Peso/efeitos dos fármacos
20.
J Dent Res ; 87(10): 915-27, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18809744

RESUMO

Peptides with broad-spectrum antimicrobial activity are found in the mucosal surfaces at many sites in the body, including the airway, the oral cavity, and the digestive tract. Based on their in vitro antimicrobial and other immunomodulatory activities, these host defense peptides have been proposed to play an important role in the innate defense against pathogenic microbial colonization. The genes that encode these peptides are up-regulated by pathogens, further supporting their role in innate immune defense. However, the differences in the local microbial environments between the generally sterile airway and the highly colonized oral cavity suggest a more complex role for these peptides in innate immunity. For example, beta-defensin genes are induced in the airway by all bacteria and Toll-like receptor (TLR) agonists primarily through an NF-kappaB-mediated pathway. In contrast, the same genes are induced in the gingival epithelium by only a subset of bacteria and TLR ligands, via different pathways. Furthermore, the environments into which the peptides are secreted--specifically saliva, gingival crevicular fluid, and airway surface fluid--differ greatly and can effect their respective activities in host defense. In this review, we examine the differences and similarities between host defense peptides in the oral cavity and the airway, to gain a better understanding of their contributions to immunity.


Assuntos
Peptídeos Catiônicos Antimicrobianos/imunologia , Mucosa Bucal/química , Mucosa Bucal/imunologia , Mucosa Respiratória/química , Mucosa Respiratória/imunologia , Animais , Peptídeos Catiônicos Antimicrobianos/química , Peptídeos Catiônicos Antimicrobianos/genética , Peptídeos Catiônicos Antimicrobianos/uso terapêutico , Infecções Bacterianas/imunologia , Células Epiteliais/química , Células Epiteliais/imunologia , Expressão Gênica , Humanos , Imunidade Inata , Mutação , NF-kappa B/metabolismo , Proteínas e Peptídeos Salivares/química , Proteínas e Peptídeos Salivares/genética , Proteínas e Peptídeos Salivares/imunologia , Proteínas e Peptídeos Salivares/uso terapêutico , Receptores Toll-Like/metabolismo , Viroses/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...