Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
1.
Arch Razi Inst ; 78(3): 963-972, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-38028841

RESUMEN

This study aimed to detect the levels of apurinic/apyrimidinic endonuclease 1 (APE1) gene expression and C-type lectin domain family 4 member M (CLEC4M) and their association with cisplatin chemotherapy in lung cancer patients. Overall, 105 individuals who attended the Al-Amal National Hospital for Cancer Management, Baghdad, Iraq, were enrolled in the study and divided into three equal groups. The groups included the patients newly diagnosed with lung cancer, cancer patients who received cisplatin, and the healthy control group. All study groups were subjected to the sampling of the venous blood for molecular analysis by real-time polymerase chain reaction (RT-PCR) to detect the APE1 gene and enzyme-linked immunosorbent assay (ELISA) for serological testing to measure the concentration of CLEC4M protein. Significantly, the values of both cancer groups were higher than those reported in the control group. The relative index revealed a significant difference in the mean fold change level of APE1 in the newly diagnosed group (3 fold) and cisplatin therapy patients group (2 fold), compared to the control group (P=0.005). No significant differences were detected between the two cancer groups in terms of fold change mean of expression, demographic characteristics, and cancer histological type. Regarding human CLEC4M protein level, cases receiving cisplatin (139.2±25.9) and newly diagnosed patients (331.0±38.1) had a highly significant difference with the control group (100.3±47.5, P<0.001). There was no significant difference between the concentration level of CLEC4M and all parameters in demographic characteristics and cancer histological type. This was the first study to demonstrate that higher expression levels of new APE1, CLEC4M, and glutathione, especially after chemotherapy, are beneficial as diagnostic and prognostic markers for resistance to platinum chemotherapy in Iraqi lung cancer patients.


Asunto(s)
Neoplasias Pulmonares , Humanos , Neoplasias Pulmonares/genética , Cisplatino/efectos adversos , Endonucleasas/uso terapéutico , Ensayo de Inmunoadsorción Enzimática , Irak , ADN-(Sitio Apurínico o Apirimidínico) Liasa/genética , ADN-(Sitio Apurínico o Apirimidínico) Liasa/metabolismo , Receptores de Superficie Celular/uso terapéutico , Moléculas de Adhesión Celular/uso terapéutico , Lectinas Tipo C/genética , Lectinas Tipo C/uso terapéutico
2.
Malays J Pathol ; 45(1): 65-76, 2023 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-37119247

RESUMEN

INTRODUCTION: Acute myeloid leukaemia (AML) is a heterogeneous malignant disease with a high degree of treatment failure using chemotherapy. Leukaemia stem cells (LSCs) are CD34+CD38- early progenitors associated with poor prognosis in AML. A unique LSC phenotype that excludes rare normal haematopoietic stem cells (HSC) is still elusive. This study aimed to determine expression of selected potential LSC markers in normal and leukaemic myeloid cells and correlate prognosis in AML patients. MATERIALS AND METHODS: Flow cytometry and RT-qPCR measured expressions of ALDH, IL3RA/CD123, CLEC12A/CLL-1/CD371, HOXA3 and ENPP4. Normal cord blood (n=3) and blood monocytes (n=5) represented HSC and mature cells, respectively. Myeloid leukaemia cell lines (THP-1, KG-1a, K562 and HL-60) represented progenitor cells at various stages of maturation. AML samples included chemo-resistant (n=8), early relapse (n=2) and late relapse (n=18). RESULTS: Combining protein/gene expressions, CD34+CD38- was a feature of immature cells seen in cord blood, KG-1a, and K562 but not more mature cells (blood monocytes and HL-60). Normal cells expressed CD371 while mature cells (blood monocytes and HL-60) lacked CD123. ENPP4 was not expressed on normal cells while HOXA3 was expressed only on cord blood and THP-1. In AML, CD123, HOXA3, ENPP4 (but not CD371) were significantly increased in the CD34+CD38- fraction of chemo-resistant patients while ALDH was associated with chemo-resistance. CONCLUSION: CD34+CD38- presented an immature phenotype and with ALDH were associated with poor prognosis. CD123, HOXA3 and ENPP4 further enriched the LSC population. ENPP4 has not been reported and has the advantage of not being expressed on HSC and normal monocytes.


Asunto(s)
Subunidad alfa del Receptor de Interleucina-3 , Leucemia Mieloide Aguda , Humanos , Subunidad alfa del Receptor de Interleucina-3/metabolismo , Subunidad alfa del Receptor de Interleucina-3/uso terapéutico , Leucemia Mieloide Aguda/genética , Células Madre Hematopoyéticas/metabolismo , Células Madre Hematopoyéticas/patología , Antígenos CD34/metabolismo , Antígenos CD34/uso terapéutico , Recurrencia , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Receptores Mitogénicos/metabolismo , Receptores Mitogénicos/uso terapéutico , Lectinas Tipo C/metabolismo , Lectinas Tipo C/uso terapéutico , Proteínas de Homeodominio/metabolismo , Proteínas de Homeodominio/uso terapéutico
3.
Adv Drug Deliv Rev ; 196: 114776, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-36934519

RESUMEN

Combatting the ever-increasing threat from invasive fungal pathogens faces numerous fundamental challenges, including constant human exposure to large reservoirs of species in the environment, the increasing population of immunocompromised or immunosuppressed individuals, the unsatisfactory efficacy of current antifungal drugs and their associated toxicity, and the scientific and economic barriers limiting a new antifungal pipeline. DectiSomes represent a new drug delivery platform that enhances antifungal efficacy for diverse fungal pathogens and reduces host toxicity for current and future antifungals. DectiSomes employ pathogen receptor proteins - C-type lectins - to target drug-loaded liposomes to conserved fungal cognate ligands and away from host cells. DectiSomes represent one leap forward for urgently needed effective pan-antifungal therapy. Herein, we discuss the problems of battling fungal diseases and the state of DectiSome development.


Asunto(s)
Antifúngicos , Micosis , Humanos , Antifúngicos/farmacología , Antifúngicos/uso terapéutico , Liposomas , Lectinas Tipo C/uso terapéutico , Micosis/tratamiento farmacológico
4.
Gut ; 72(3): 443-450, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36008101

RESUMEN

OBJECTIVE: Despite being in sustained and stable remission, patients with Crohn's disease (CD) stopping anti-tumour necrosis factor α (TNFα) show a high rate of relapse (~50% within 2 years). Characterising non-invasively the biological profiles of those patients is needed to better guide the decision of anti-TNFα withdrawal. DESIGN: Ninety-two immune-related proteins were measured by proximity extension assay in serum of patients with CD (n=102) in sustained steroid-free remission and stopping anti-TNFα (infliximab). As previously shown, a stratification based on time to clinical relapse was used to characterise the distinct biological profiles of relapsers (short-term relapsers: <6 months vs mid/long-term relapsers: >6 months). Associations between protein levels and time to clinical relapse were determined by univariable Cox model. RESULTS: The risk (HR) of mid/long-term clinical relapse was specifically associated with a high serum level of proteins mainly expressed in lymphocytes (LAG3, SH2B3, SIT1; HR: 2.2-4.5; p<0.05), a low serum level of anti-inflammatory effectors (IL-10, HSD11B1; HR: 0.2-0.3; p<0.05) and cellular junction proteins (CDSN, CNTNAP2, CXADR, ITGA11; HR: 0.4; p<0.05). The risk of short-term clinical relapse was specifically associated with a high serum level of pro-inflammatory effectors (IL-6, IL12RB1; HR: 3.5-3.6; p<0.05) and a low or high serum level of proteins mainly expressed in antigen presenting cells (CLEC4A, CLEC4C, CLEC7A, LAMP3; HR: 0.4-4.1; p<0.05). CONCLUSION: We identified distinct blood protein profiles associated with the risk of short-term and mid/long-term clinical relapse in patients with CD stopping infliximab. These findings constitute an advance for the development of non-invasive biomarkers guiding the decision of anti-TNFα withdrawal.


Asunto(s)
Enfermedad de Crohn , Humanos , Infliximab/uso terapéutico , Enfermedad de Crohn/tratamiento farmacológico , Recurrencia Local de Neoplasia , Factor de Necrosis Tumoral alfa , Antiinflamatorios/uso terapéutico , Biomarcadores , Recurrencia , Inducción de Remisión , Glicoproteínas de Membrana , Receptores Inmunológicos , Lectinas Tipo C/uso terapéutico , Péptidos y Proteínas de Señalización Intercelular
5.
Hum Vaccin Immunother ; 18(1): 1873056, 2022 12 31.
Artículo en Inglés | MEDLINE | ID: mdl-33625943

RESUMEN

Dendritic cell (DC) vaccines are a safe and effective means of inducing tumor immune responses, however, a better understanding of DC biology is required in order to realize their full potential. Recent advances in DC biology have identified a crucial role for cDC1 in tumor immune responses, making this DC subset an attractive vaccine target. Human cDC1 exclusively express the C-type-lectin-like receptor, CLEC9A (DNGR-1) that plays an important role in cross-presentation, the process by which effective CD8+ T cell responses are generated. CLEC9A antibodies deliver antigen specifically to cDC1 for the induction of humoral, CD4+ and CD8+ T cell responses and are therefore promising candidates to develop as vaccines for infectious diseases and cancer. The development of human CLEC9A antibodies now facilitates their application as vaccines for cancer immunotherapy. Here we discuss the recent advances in CLEC9A targeting antibodies as vaccines for cancer and their translation to the clinic.


Asunto(s)
Vacunas contra el Cáncer , Inmunogenicidad Vacunal , Lectinas Tipo C , Neoplasias , Receptores Mitogénicos , Linfocitos T CD8-positivos , Reactividad Cruzada , Células Dendríticas , Humanos , Lectinas Tipo C/uso terapéutico , Neoplasias/patología , Neoplasias/terapia , Receptores Mitogénicos/uso terapéutico
6.
Sci Rep ; 11(1): 11663, 2021 06 03.
Artículo en Inglés | MEDLINE | ID: mdl-34083615

RESUMEN

The interaction of platelet GPIbα with von Willebrand factor (VWF) is essential to initiate platelet adhesion and thrombosis, particularly under high shear stress conditions. However, no drug targeting GPIbα has been developed for clinical practice. Here we characterized anfibatide, a GPIbα antagonist purified from snake (Deinagkistrodon acutus) venom, and evaluated its interaction with GPIbα by surface plasmon resonance and in silico modeling. We demonstrated that anfibatide interferds with both VWF and thrombin binding, inhibited ristocetin/botrocetin- and low-dose thrombin-induced human platelet aggregation, and decreased thrombus volume and stability in blood flowing over collagen. In a single-center, randomized, and open-label phase I clinical trial, anfibatide was administered intravenously to 94 healthy volunteers either as a single dose bolus, or a bolus followed by a constant rate infusion of anfibatide for 24 h. Anfibatide inhibited VWF-mediated platelet aggregation without significantly altering bleeding time or coagulation. The inhibitory effects disappeared within 8 h after drug withdrawal. No thrombocytopenia or anti-anfibatide antibodies were detected, and no serious adverse events or allergic reactions were observed during the studies. Therefore, anfibatide was well-tolerated among healthy subjects. Interestingly, anfibatide exhibited pharmacologic effects in vivo at concentrations thousand-fold lower than in vitro, a phenomenon which deserves further investigation.Trial registration: Clinicaltrials.gov NCT01588132.


Asunto(s)
Plaquetas/efectos de los fármacos , Plaquetas/metabolismo , Venenos de Crotálidos/uso terapéutico , Fibrinolíticos/uso terapéutico , Lectinas Tipo C/uso terapéutico , Complejo GPIb-IX de Glicoproteína Plaquetaria/antagonistas & inhibidores , Venenos de Serpiente/uso terapéutico , Animales , Coagulación Sanguínea/efectos de los fármacos , Venenos de Crotálidos/química , Venenos de Crotálidos/aislamiento & purificación , Venenos de Crotálidos/farmacocinética , Crotalinae , Fibrinolíticos/química , Fibrinolíticos/aislamiento & purificación , Fibrinolíticos/farmacocinética , Voluntarios Sanos , Humanos , Lectinas Tipo C/química , Lectinas Tipo C/aislamiento & purificación , Modelos Moleculares , Adhesividad Plaquetaria/efectos de los fármacos , Agregación Plaquetaria/efectos de los fármacos , Recuento de Plaquetas , Complejo GPIb-IX de Glicoproteína Plaquetaria/química , Unión Proteica , Conformación Proteica , Ristocetina/farmacología , Venenos de Serpiente/química , Venenos de Serpiente/aislamiento & purificación , Venenos de Serpiente/farmacocinética , Relación Estructura-Actividad , Trombina/farmacología , Trombosis/prevención & control , Factor de von Willebrand/química , Factor de von Willebrand/metabolismo
7.
mBio ; 12(1)2021 02 23.
Artículo en Inglés | MEDLINE | ID: mdl-33622715

RESUMEN

Invasive fungal diseases cause millions of deaths each year. There are currently approximately 300,000 acute cases of aspergillosis, most of which result from a pulmonary infection of immunocompromised patients by the common soil organism and opportunistic pathogen Aspergillus fumigatus Patients are treated with antifungal drugs, such as amphotericin B (AmB). However, AmB has serious limitations due to human organ toxicity. AmB is slightly less toxic if loaded in liposomes, such as AmBisome or AmB-loaded liposomes (AmB-LLs). Even with antifungal therapy, recurrent infections are common, and 1-year fatality rates may exceed 50%. We have previously shown that coating AmB-LLs with the extracellular oligomannan-binding domain of the C-type lectin receptor Dectin-2 (DEC2-AmB-LLs) effectively targets DEC2-AmB-LLs to cell walls, exopolysaccharide matrices, and biofilms of fungal pathogens in vitroIn vitro, DEC2-AmB-LLs reduce the effective dose of AmB for 95% inhibition and killing of A. fumigatus 10-fold compared to that of untargeted AmB-LLs. Herein we tested the antifungal activity of DEC2-AmB-LLs relative to that of untargeted AmB-LLs in immunosuppressed mice with pulmonary aspergillosis. Remarkably, DEC2-AmB-LLs bound 30-fold more efficiently to A. fumigatus at sites of infection in the lungs. Furthermore, Dectin-2-targeted liposomes delivering AmB at a dose of 0.2 mg/kg of body weight significantly reduced the fungal burden in lungs compared to results with untargeted AmB-LLs at 0.2 mg/kg and micellar voriconazole at 20 mg/kg and prolonged mouse survival. By dramatically increasing the efficacy of antifungal drugs at low doses, targeted liposomes have the potential to create a new clinical paradigm to treat diverse fungal diseases.IMPORTANCE Invasive aspergillosis (IA) generally results from a pulmonary infection of immunocompromised patients by the common soil organism and opportunistic pathogen Aspergillus fumigatus The susceptible population has expanded rapidly due to the increased number of cancer patients with immunocompromising chemotherapy and transplant patients taking immunosuppressants. Patients are treated with antifungals, such as liposomal amphotericin B, with per-patient costs exceeding $50,000 in the United States. However, AmB has serious side effects due to host toxicity, which limits its usage and contributes to the lack of fungal clearance in patients at safe doses. Fifty percent of IA patients die within a year. Herein, we employed liposomal amphotericin B coated with the innate immune receptor Dectin-2 to direct antifungals specifically to the fungal pathogen. Using two mouse models of pulmonary aspergillosis, we demonstrate that Dectin-2-targeted delivery of amphotericin B to A. fumigatus resulted in remarkably higher efficacy than that of the untargeted antifungal formulations.


Asunto(s)
Antifúngicos/uso terapéutico , Aspergillus fumigatus/efectos de los fármacos , Pared Celular/metabolismo , Lectinas Tipo C/metabolismo , Lectinas Tipo C/uso terapéutico , Liposomas/química , Aspergilosis Pulmonar/tratamiento farmacológico , Anfotericina B/metabolismo , Anfotericina B/uso terapéutico , Animales , Pared Celular/efectos de los fármacos , Femenino , Lectinas Tipo C/genética , Liposomas/uso terapéutico , Ratones , Neutropenia
8.
Brain Res Bull ; 170: 49-57, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33556561

RESUMEN

RATIONALE: Hematoma expansion (HE) aggravates brain injury after intracerebral hemorrhage (ICH) and hypertension is a key contributor to HE. Plasma kallikrein (PK) is involved in hemorrhagic transformation in ischemic stroke mice. This study was conducted to explore the role of PK in HE in hypertensive ICH. METHODS: Hypertension was achieved by continuous infusion of angiotensin II (Ang II) with an osmotic pump in C57BL/6 mice. ICH was achieved by stereotactic intrastriatal injection of blood. PK-specific antibody and platelet glycoprotein VI (GPVI) agonists were administered to intervene in hematoma expansion. The hematoma volume was indicated by the erythrocyte components hemoglobin and carbonic anhydrase-1 in the ipsilateral brain hemisphere. RESULTS: Ang II-induced hypertensive mice showed enhanced hematoma expansion and worsened neurologic deficits after ICH modeling. Moreover, intrastriatal injection of blood from Ang II-treated mice into normal mice increased the area of secondary hemorrhage more than blood from untreated mice. Mechanistically, elevated PK was found in Ang II-infused mice whereas, inhibition of PK and administration of the GPVI agonist convulxin decreased hematoma expansion and improved neurologic deficits after ICH. CONCLUSIONS: These findings suggest that PK inhibition and GPVI agonist treatment might serve as potential methods to intervene in HE after ICH.


Asunto(s)
Hemorragia Cerebral/tratamiento farmacológico , Hipertensión/tratamiento farmacológico , Lectinas Tipo C/uso terapéutico , Calicreína Plasmática/antagonistas & inhibidores , Angiotensina II , Animales , Presión Sanguínea/fisiología , Hemorragia Cerebral/complicaciones , Hemorragia Cerebral/patología , Venenos de Crotálidos/farmacología , Venenos de Crotálidos/uso terapéutico , Modelos Animales de Enfermedad , Hipertensión/inducido químicamente , Hipertensión/complicaciones , Hipertensión/patología , Masculino , Ratones , Resultado del Tratamiento
9.
J Breath Res ; 15(1): 016012, 2020 11 20.
Artículo en Inglés | MEDLINE | ID: mdl-33108776

RESUMEN

In children with asthma, the responsiveness of inhaled corticosteroids (ICS) is depended on asthma endotype and phenotype. This study aimed to describe the clinical and biological characteristics, and its correlation with polymorphism of rs28364072 in FCER2 of asthmatic children. This work aimed to study the correlation between fractional concentration of exhaled nitric oxide (FENO) level and rs28364072 polymorphism of FCER2 gene with ICS responsiveness and disease control in children with asthma. This study was a prospective and descriptive study. All clinical characteristics, FENO, blood eosinophil counts (BEC), skin prick test (SPT), total IgE, asthma control test, and FCER2 gene polymorphism were performed for each patient. One hundred and seven asthmatic children who were over 5 years old (9.2 ± 2.6), were included. Patients with FENO > 20 ppb had higher percentage of positive SPT, total IgE level, and BEC (89.2 vs 80.0%, 851.1 vs 656.9 UI ml-1, and 785 ± 576 G L-1 vs 425 ± 364 G L-1; respectively). Among them, there were 54.2% of homozygous TT, 36.4% of heterozygous TC, and 9.4% of homozygous CC of rs28364072 polymorphism in FCER2. The percentage of patients with controlled asthma was increasing after 1 month and 3 months (47.1% and 58.8%; respectively). During the study, the ICS was decreasing as indicated by asthma control (348 ± 118 mcg at 1st month vs 329 ± 119 mcg at 3rd month; p < 0.05). CC genotype had the lowest level of increasing FEV1 compared to that in genotype TC and TT (8.4% vs 8.7% and 27.1%; p > 0.05 and p < 0.05; respectively). The percentage of polymorphism in rs28364072 of FCER2 was significant higher in patients with controlled asthma compared to uncontrolled asthma. Asthmatic children with high FENO and rs28364072 polymorphism in FCER2 gene are good responders to ICS; however, asthmatic children with homozygous variant CC of rs28364072 are poorly responsive to ICS.


Asunto(s)
Corticoesteroides/administración & dosificación , Asma/tratamiento farmacológico , Asma/genética , Pruebas Respiratorias , Espiración , Lectinas Tipo C/genética , Óxido Nítrico/metabolismo , Polimorfismo de Nucleótido Simple/genética , Receptores de IgE/genética , Administración por Inhalación , Adolescente , Corticoesteroides/uso terapéutico , Asma/sangre , Asma/fisiopatología , Niño , Preescolar , Femenino , Humanos , Inmunoglobulina E/sangre , Lectinas Tipo C/uso terapéutico , Masculino , Estudios Prospectivos , Receptores de IgE/uso terapéutico
10.
Expert Rev Hematol ; 13(11): 1153-1164, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32876503

RESUMEN

Introduction: Acquired thrombotic thrombocytopenic purpura (aTTP) is a thrombotic microangiopathy caused by inhibitory autoantibodies against ADAMTS13 protein. Until recently, the combination of plasma exchange (PEX) and immunosuppression has been the standard front-line treatment in this disorder. However, aTTP-related mortality, refractoriness, and relapse are still a matter of concern. Areas covered: The better understanding of the pathophysiological mechanisms of aTTP has allowed substantial improvements in the diagnosis and treatment of this disease. Recently, the novel anti-VWF nanobody caplacizumab has been approved for acute episodes of aTTP. Caplacizumab is capable to block the adhesion of platelets to VWF, therefore inhibiting microthrombi formation in the ADAMTS13-deficient circulation. In this review, the characteristics of caplacizumab together with the available data of its efficacy and safety in the clinical setting will be analyzed. Besides, the current scenario of aTTP treatment will be provided, including the role of other innovative drugs. Expert opinion: With no doubt, caplacizumab is going to change the way we treat aTTP. In combination with standard treatment, caplacizumab can help to significantly reduce aTTP-related mortality and morbidity and could spare potential long-term consequences by minimizing the risk of exacerbation.


Asunto(s)
Fibrinolíticos/uso terapéutico , Terapia Molecular Dirigida , Púrpura Trombocitopénica Trombótica/tratamiento farmacológico , Anticuerpos de Dominio Único/uso terapéutico , Factor de von Willebrand/antagonistas & inhibidores , Proteína ADAMTS13/deficiencia , Proteína ADAMTS13/inmunología , Proteína ADAMTS13/uso terapéutico , Acetilcisteína/uso terapéutico , Aptámeros de Nucleótidos/uso terapéutico , Autoantígenos/inmunología , Ensayos Clínicos como Asunto , Terapia Combinada , Venenos de Crotálidos/uso terapéutico , Aprobación de Drogas , Quimioterapia Combinada , Drogas en Investigación/uso terapéutico , Humanos , Factores Inmunológicos/uso terapéutico , Inmunosupresores/uso terapéutico , Lectinas Tipo C/uso terapéutico , Estudios Multicéntricos como Asunto , Intercambio Plasmático , Adhesividad Plaquetaria/efectos de los fármacos , Dominios Proteicos/inmunología , Púrpura Trombocitopénica Trombótica/fisiopatología , Púrpura Trombocitopénica Trombótica/terapia , Proteínas Recombinantes/uso terapéutico , Anticuerpos de Dominio Único/inmunología , Anticuerpos de Dominio Único/farmacología , Resultado del Tratamiento , Factor de von Willebrand/inmunología
11.
J Mol Neurosci ; 70(1): 71-83, 2020 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-31478134

RESUMEN

The disruption of the blood-brain barrier (BBB) and the consequent brain edema are major contributors to the pathogenesis of cerebral ischemia/reperfusion injury. RhoA is generally thought to play a crucial role in the process of BBB disruption and participate in the signaling pathways emanating from TLR4. However, it remains unverified the regulatory role of TLR4 in the RhoA/ROCK pathway in cerebral I/R injury and its effects on the BBB as well. The present study probes into the protective effect of ANF on the BBB after cerebral I/R injury and the possible mechanisms. Focal cerebral ischemia was induced by 120 min of transient middle cerebral artery occlusion (MCAO). ANF (1, 2, 4 µg/kg) was achieved by intravenous injection after 120 min of MCAO followed by 1, 24, 48, and 72 h reperfusion. Evans blue extravasation, brain water content, RhoA activity, and the expressions of TLR4, ROCK1/2, p-MLC2, MMP-2/9, ZO-1, occludin, and claudin-5 protein in rat brain were evaluated 72 h after reperfusion. ANF could significantly reduce the Evans blue extravasation and water content in the ipsilateral hemisphere and obviously increase the occludin, claudin-5, and ZO-1 expression after cerebral I/R injury. Furthermore, cerebral I/R injury induced apparently increased expression of TLR4, RhoA-GTP, ROCK1/2, p-MLC2, and MMMP-2/9, which, however, could be remarkably alleviated by ANF intervention. Taken together, the TLR4/RhoA/ROCK signaling pathway is implicated in BBB breakdown after cerebral I/R injury, and ANF preserves BBB integrity, probably via inhibiting the TLR4/RhoA/ROCK signaling pathway.


Asunto(s)
Barrera Hematoencefálica/efectos de los fármacos , Venenos de Crotálidos/uso terapéutico , Infarto de la Arteria Cerebral Media/tratamiento farmacológico , Lectinas Tipo C/uso terapéutico , Fármacos Neuroprotectores/uso terapéutico , Daño por Reperfusión/tratamiento farmacológico , Receptor Toll-Like 4/metabolismo , Animales , Barrera Hematoencefálica/metabolismo , Miosinas Cardíacas/metabolismo , Venenos de Crotálidos/administración & dosificación , Venenos de Crotálidos/farmacología , Lectinas Tipo C/administración & dosificación , Masculino , Metaloproteinasas de la Matriz/metabolismo , Cadenas Ligeras de Miosina/metabolismo , Fármacos Neuroprotectores/efectos adversos , Fármacos Neuroprotectores/farmacología , Ratas , Ratas Sprague-Dawley , Transducción de Señal , Proteínas de Unión al GTP rho/metabolismo , Quinasas Asociadas a rho/metabolismo
12.
Hematology Am Soc Hematol Educ Program ; 2018(1): 539-547, 2018 11 30.
Artículo en Inglés | MEDLINE | ID: mdl-30504355

RESUMEN

The advent of plasma exchange has dramatically changed the prognosis of acute thrombotic thrombocytopenic purpura (TTP). Recent insights into TTP pathogenesis have led to the development of novel therapies targeting pathogenic anti-ADAMTS13 antibody production, von Willebrand factor (VWF)-platelet interactions, and ADAMTS13 replacement. Retrospective and prospective studies have established the efficacy of rituximab as an adjunct to plasma exchange for patients with acute TTP, either upfront or for refractory disease. Relapse prevention is a major concern for survivors of acute TTP, and emerging data support the prophylactic use of rituximab in patients with persistent or recurrent ADAMTS13 deficiency in clinical remission. Capalcizumab, a nanobody directed against domain A1 of VWF that prevents the formation of VWF-platelet aggregates, recently completed phase 2 (TITAN) and 3 (HERCULES) trials with encouraging results. Compared with placebo, caplacizumab shortened the time to platelet recovery and may protect against microthrombotic tissue injury in the acute phase of TTP, though it does not modify the underlying immune response. Other promising therapies including plasma cell inhibitors (bortezomib), recombinant ADAMTS13, N-acetyl cysteine, and inhibitors of the VWF-glycoprotein Ib/IX interaction (anfibatide) are in development, and several of these agents are in prospective clinical studies to evaluate their efficacy and role in TTP. In the coming years, we are optimistic that novel therapies and international collaborative efforts will usher in even more effective, evidence-based approaches to address refractory acute TTP and relapse prevention.


Asunto(s)
Bortezomib/uso terapéutico , Venenos de Crotálidos/uso terapéutico , Lectinas Tipo C/uso terapéutico , Púrpura Trombocitopénica Trombótica/tratamiento farmacológico , Rituximab/uso terapéutico , Anticuerpos de Dominio Único/uso terapéutico , Proteína ADAMTS13/antagonistas & inhibidores , Proteína ADAMTS13/sangre , Enfermedad Aguda , Autoanticuerpos/sangre , Ensayos Clínicos Fase II como Asunto , Ensayos Clínicos Fase III como Asunto , Supervivencia sin Enfermedad , Humanos , Intercambio Plasmático , Recuento de Plaquetas , Púrpura Trombocitopénica Trombótica/sangre , Púrpura Trombocitopénica Trombótica/mortalidad , Púrpura Trombocitopénica Trombótica/patología , Recurrencia , Tasa de Supervivencia , Factor de von Willebrand/antagonistas & inhibidores , Factor de von Willebrand/metabolismo
13.
Brain Behav Immun ; 69: 255-263, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29195783

RESUMEN

Blood-brain barrier (BBB) disruption, thrombus formation and immune-mediated inflammation are important steps in the pathophysiology of cerebral ischemia-reperfusion injury but are still inaccessible to therapeutic interventions. Recent studies have provided increasing evidence that blocking of platelet glycoprotein (GP) receptor Ib might represent a novel target in treating acute ischemic stroke. This research was conducted to explore the therapeutic efficacy and potential mechanisms of GPIbα inhibitor (anfibatide) in a model of brain ischemia-reperfusion injury in mice. Male mice underwent 90 min of right middle cerebral artery occlusion (MCAO) followed by 24 h of reperfusion. Anfibatide (1, 2, 4 ug/kg) or tirofiban were administered intravenously 1 h after reperfusion. The results showed that anfibatide could significantly reduce infarct volumes, increase the number of intact neuronal cells and improve neurobehavioral function. Moreover, anfibatide could reduce post ischemic BBB damage by attenuating increased paracellular permeability in the ischemia hemisphere significantly. Stroke-induced increases in activity and protein expression of macrophage-1 antigen (MAC-1) and P-selectin were also reduced by anfibatide intervention. Finally, anfibatide exerted antithrombotic effects upon stroke by decreased the number of microthrombi formation. This is the first demonstration of anfibatide's efficacy in protecting the BBB integrity and decreasing neutrophil inflammation response mediated by MAC-1 besides microthrombus formation inhibition in the brain during reperfusion. Anfibatide, as a promising anti-thrombo-inflammation agent, could be beneficial for the treatment of ischemic stroke.


Asunto(s)
Barrera Hematoencefálica/efectos de los fármacos , Venenos de Crotálidos/uso terapéutico , Fibrinolíticos/uso terapéutico , Lectinas Tipo C/uso terapéutico , Complejo GPIb-IX de Glicoproteína Plaquetaria/antagonistas & inhibidores , Daño por Reperfusión/tratamiento farmacológico , Tirofibán/uso terapéutico , Animales , Barrera Hematoencefálica/metabolismo , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Venenos de Crotálidos/farmacología , Fibrinolíticos/farmacología , Infarto de la Arteria Cerebral Media/metabolismo , Masculino , Ratones , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Daño por Reperfusión/metabolismo , Tirofibán/farmacología
14.
Proc Natl Acad Sci U S A ; 114(10): 2675-2680, 2017 03 07.
Artículo en Inglés | MEDLINE | ID: mdl-28223515

RESUMEN

The advances in subunit vaccines development have intensified the search for potent adjuvants, particularly adjuvants inducing cell-mediated immune responses. Identification of the C-type lectin Mincle as one of the receptors underlying the remarkable immunogenicity of the mycobacterial cell wall, via recognition of trehalose-6,6'-dimycolate (TDM), has opened avenues for the rational design of such molecules. Using a combination of chemical synthesis, biological evaluation, molecular dynamics simulations, and protein mutagenesis, we gained insight into the molecular bases of glycolipid recognition by Mincle. Unexpectedly, the fine structure of the fatty acids was found to play a key role in the binding of a glycolipid to the carbohydrate recognition domain of the lectin. Glucose and mannose esterified at O-6 by a synthetic α-ramified 32-carbon fatty acid showed agonist activity similar to that of TDM, despite their much simpler structure. Moreover, they were seen to stimulate proinflammatory cytokine production in primary human and murine cells in a Mincle-dependent fashion. Finally, they were found to induce strong Th1 and Th17 immune responses in vivo in immunization experiments in mice and conferred protection in a murine model of Mycobacterium tuberculosis infection. Here we describe the rational development of new molecules with powerful adjuvant properties.


Asunto(s)
Adyuvantes Inmunológicos/química , Lectinas Tipo C/inmunología , Receptores Inmunológicos/inmunología , Tuberculosis/prevención & control , Vacunas de Subunidad/inmunología , Inmunidad Adaptativa/efectos de los fármacos , Adyuvantes Inmunológicos/uso terapéutico , Animales , Pared Celular/efectos de los fármacos , Pared Celular/inmunología , Factores Cordón/química , Factores Cordón/inmunología , Humanos , Lectinas Tipo C/química , Lectinas Tipo C/uso terapéutico , Ratones , Simulación de Dinámica Molecular , Mutagénesis/efectos de los fármacos , Mycobacterium/inmunología , Mycobacterium/patogenicidad , Receptores Inmunológicos/química , Tuberculosis/inmunología , Tuberculosis/microbiología , Vacunas de Subunidad/uso terapéutico
15.
J Hematol Oncol ; 10(1): 28, 2017 01 21.
Artículo en Inglés | MEDLINE | ID: mdl-28109307

RESUMEN

BACKGROUND: Tumor metastasis is an essential cause of the poor prognosis of colon cancer. DC-SIGNR is a C-type lectin that is frequently found on human liver sinusoidal endothelial cells. LSECtin, which is a homologue of DC-SIGNR, has been demonstrated to participate in colon cancer liver metastasis. Due to the similarities in the expression pattern and structure of the two proteins, we speculated that DC-SIGNR could also be involved in this process. METHODS: Colon cancer cells were treated with the DC-SIGNR protein or control IgG, after which cell migration, invasion, and morphology were assayed. Xenograft mouse models were used to determine the role of DC-SIGNR in colon cancer liver metastasis in vivo. In addition, a human gene expression array was used to detect differential gene expression in colon cancer cells stimulated with the DC-SIGNR protein. The serum level of DC-SIGNR was examined in colon cancer patients by ELISA, and the significance of DC-SIGNR was determined. RESULTS: In our research, we investigated whether DC-SIGNR promotes colon cancer cell adhesion, migration, and invasion. Knocking down mouse DC-SIGNR decreased the liver metastatic potency of colon cancer cells and increased survival time. Expressing human DC-SIGNR enhanced colon cancer liver metastasis. Furthermore, DC-SIGNR conferred metastatic capability on cancer cells by upregulating various metallothionein isoforms. To validate the above results, we also found that the serum DC-SIGNR level was statistically higher in colon cancer patients with liver metastasis compared with those without metastasis. CONCLUSIONS: These results imply that DC-SIGNR may promote colon carcinoma hepatic metastasis and could serve as a promising therapeutic target for anticancer treatment.


Asunto(s)
Moléculas de Adhesión Celular/farmacología , Neoplasias del Colon/patología , Lectinas Tipo C/fisiología , Neoplasias Hepáticas/secundario , Animales , Adhesión Celular , Moléculas de Adhesión Celular/uso terapéutico , Movimiento Celular , Modelos Animales de Enfermedad , Xenoinjertos , Humanos , Lectinas Tipo C/uso terapéutico , Ratones , Invasividad Neoplásica , Metástasis de la Neoplasia , Receptores de Superficie Celular/uso terapéutico , Células Tumorales Cultivadas
16.
FASEB J ; 31(3): 1107-1119, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-27974593

RESUMEN

Angiogenesis is a cause of visual impairment and blindness in the wet form of age-related macular degeneration and in ischemic retinopathies. Current therapies include use of anti-VEGF agents to reduce choroidal neovascularization (CNV) and edema. These treatments are effective in most cases, but spontaneous or acquired resistance to anti-VEGF and possible adverse effects of long-term VEGF inhibition in the retina and choroid highlight a need for additional alternative therapies. Integrins αvß3 and αvß5, which regulate endothelial cell proliferation and stabilization, have been implicated in ocular angiogenesis. Lebecetin (LCT) is a 30-kDa heterodimeric C-type lectin that is isolated from Macrovipera lebetina venom and interacts with α5ß1- and αv-containing integrins. We previously showed that LCT inhibits human brain microvascular endothelial cell adhesion, migration, proliferation, and tubulogenesis. To evaluate the inhibitory effect of LCT on ocular angiogenesis, we cultured aortic and choroidal explants in the presence of LCT and analyzed the effect of LCT on CNV in the mouse CNV model and on retinal neovascularization in the oxygen-induced retinopathy model. Our data demonstrate that a single injection of LCT efficiently reduced CNV and retinal neovascularization in these models.-Montassar, F., Darche, M., Blaizot, A., Augustin, S., Conart, J.-B., Millet, A., Elayeb, M., Sahel, J.-A., Réaux-Le Goazigo, A., Sennlaub, F., Marrakchi, N., Messadi, E., Guillonneau, X. Lebecetin, a C-type lectin, inhibits choroidal and retinal neovascularization.


Asunto(s)
Coroides/efectos de los fármacos , Lectinas Tipo C/uso terapéutico , Degeneración Macular/tratamiento farmacológico , Neovascularización Patológica/tratamiento farmacológico , Venenos de Víboras/uso terapéutico , Animales , Aorta/citología , Aorta/efectos de los fármacos , Endotelio Vascular/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratas , Ratas Endogámicas Lew , Venenos de Víboras/farmacología
17.
PLoS One ; 11(3): e0150733, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26983031

RESUMEN

OBJECTIVE: No efficient medical treatment is available for severe acute hepatitis (SAH) except N-acetylcysteine for acetaminophen-induced acute liver failure. The human C-type lectin Reg3α, referred to as ALF-5755, improved survival in an animal model of acute liver failure and was well tolerated in a phase 1 trial in humans. We performed a phase 2a trial of ALF5755 in non-acetaminophen induced SAH. DESIGN: double-blind, randomized, placebo-controlled study. The primary end-point was the improvement in the coagulation protein synthesis assessed by the change of Prothrombin (PR) during the 72 hours following treatment initiation calculated as PRH0 minus PRH72 divided by 72 (PR slope H0H72). Intention to treat (ITT) and per-protocol (PP) analysis of the entire group and the Hepatitis B virus (HBV)/AIH (auto-immune hepatitis) sub-group were done separately. RESULTS: 57 patients were included. Twenty-eight received ALF-5755, 29 the placebo. Etiologies were: Hepatitis A (n = 10), HBV (n = 13), AIH (n = 9), drug-induced (n = 8), other (n = 17). On the whole group, nor the PR slope H0H72 (0.18±0.31 vs 0.25±0.32), nor the transplant-free survival rate at day 21 (75 vs 86%) differed between groups. Conversely, in the HBV-AIH subgroup, in which ALF was more severe, PR slope H0-H72 was higher in the ALF-5755 arm, the difference being significant in PP analysis (0.048±0.066 vs -0.040±0.099, p = 0.04); the median length of hospitalization was lower in the ALF-5755 group (8 vs 14 days, p = 0.02). CONCLUSION: ALF-5755 was not efficient in a ITT analysis performed on the whole sample; however it led to a significant, although moderate, clinical benefit in a PP analysis of the sub-group of patients with HBV or AIH related SAH. As HBV is the major cause of SAH in Asia and Africa and AIH a growing cause, this study emphasizes the need to pursuit the evaluation of this novel medical treatment of SAH. TRIAL REGISTRATION: ClinicalTrials.gov NCT01318525.


Asunto(s)
Antígenos de Neoplasias/uso terapéutico , Antioxidantes/uso terapéutico , Biomarcadores de Tumor/uso terapéutico , Matriz Extracelular/efectos de los fármacos , Lectinas Tipo C/uso terapéutico , Hepatopatías/tratamiento farmacológico , Proteínas Recombinantes/uso terapéutico , Enfermedad Aguda , Adulto , Antígenos de Neoplasias/efectos adversos , Antígenos de Neoplasias/farmacología , Antioxidantes/farmacocinética , Antioxidantes/farmacología , Área Bajo la Curva , Biomarcadores de Tumor/efectos adversos , Biomarcadores de Tumor/farmacocinética , Biomarcadores de Tumor/farmacología , Método Doble Ciego , Femenino , Humanos , Masculino , Persona de Mediana Edad , Proteínas Asociadas a Pancreatitis , Placebos , Pronóstico , Proteínas Recombinantes/efectos adversos , Proteínas Recombinantes/farmacocinética , Proteínas Recombinantes/farmacología
18.
Int Immunol ; 28(7): 329-38, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27006304

RESUMEN

Accumulated evidence obtained from various clinical trials and animal studies suggested that cancer vaccines need better adjuvants than those that are currently licensed, which include the most commonly used alum and incomplete Freund's adjuvant, because of either a lack of potent anti-tumor immunity or the induction of undesired immunity. Several clinical trials using immunostimulatory adjuvants, particularly agonistic as well as non-agonistic ligands for TLRs, C-type lectin receptors, retinoic acid-inducible gene I-like receptors and stimulator of interferon genes, have revealed their therapeutic potential not only as vaccine adjuvants but also as anti-tumor agents. Recently, combinations of such immunostimulatory or immunomodulatory adjuvants have shown superior efficacy over their singular use, suggesting that seeking optimal combinations of the currently available or well-characterized adjuvants may provide a better chance for the development of novel adjuvants for cancer immunotherapy.


Asunto(s)
Adyuvantes Inmunológicos/uso terapéutico , Compuestos de Alumbre/uso terapéutico , Vacunas contra el Cáncer/inmunología , Inmunoterapia/métodos , Lectinas Tipo C/uso terapéutico , Neoplasias/terapia , Oligodesoxirribonucleótidos/uso terapéutico , Animales , Ensayos Clínicos como Asunto , Humanos , Inmunomodulación , Neoplasias/inmunología , Vacunación
19.
J Biol Chem ; 290(37): 22474-84, 2015 Sep 11.
Artículo en Inglés | MEDLINE | ID: mdl-26198633

RESUMEN

The capacity of natural killer (NK) cells to mediate Fc receptor-dependent effector functions, such as antibody-dependent cellular cytotoxicity (ADCC), largely contributes to their clinical application. Given that activation-induced C-type lectin (AICL), an identified ligand for the NK-activating receptor NKp80, is frequently highly expressed on leukemia cells, the lack of therapeutic AICL-specific antibodies limits clinical application. Here we explore a strategy to reinforce NK anti-leukemia reactivity by combining targeting AICL-expressing leukemia cells with the induction of NK cell ADCC using NKp80-Fc fusion proteins. The NKp80-Fc fusion protein we generated bound specifically to leukemia cells in an AICL-specific manner. Cell binding assays between NK and leukemia cells showed that NKp80-Fc significantly increased NK target cell conjugation. In functional analyses, treatment with NKp80-Fc clearly induced the ADCC effect of NK cells. NKp80-Fc not only promoted NK-mediated leukemia cell apoptosis in the early stage of cell conjugation but also enhanced NK cell degranulation and cytotoxicity activity in the late stage. The bifunctional NKp80-Fc could redirect NK cells toward leukemia cells and triggered NK cell killing in vitro. Moreover, NKp80-Fc enhanced the lysis of NK cells against tumors in leukemia xenograft non-obese diabetic/severe combined immunodeficiency mice. Taken together, our results demonstrate that NKp80-Fc potently amplifies NK cell anti-leukemia effects in vitro and in vivo through induction of the NK cell ADCC effect. This method could potentially be useful for molecular targeted therapy, and the fusion proteins may be a promising drug for immunotherapy of leukemia.


Asunto(s)
Fragmentos Fc de Inmunoglobulinas , Células Asesinas Naturales/inmunología , Lectinas Tipo C , Leucemia/tratamiento farmacológico , Leucemia/inmunología , Receptores de Células Asesinas Naturales , Proteínas Recombinantes de Fusión , Animales , Citotoxicidad Celular Dependiente de Anticuerpos/efectos de los fármacos , Apoptosis/efectos de los fármacos , Apoptosis/genética , Apoptosis/inmunología , Línea Celular Tumoral , Xenoinjertos , Humanos , Fragmentos Fc de Inmunoglobulinas/genética , Fragmentos Fc de Inmunoglobulinas/inmunología , Fragmentos Fc de Inmunoglobulinas/farmacología , Lectinas Tipo C/genética , Lectinas Tipo C/inmunología , Lectinas Tipo C/uso terapéutico , Leucemia/genética , Leucemia/patología , Ratones , Ratones Endogámicos NOD , Ratones SCID , Trasplante de Neoplasias , Receptores de Células Asesinas Naturales/genética , Receptores de Células Asesinas Naturales/inmunología , Receptores de Células Asesinas Naturales/uso terapéutico , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/inmunología , Proteínas Recombinantes de Fusión/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto
20.
J Thromb Haemost ; 12(4): 540-9, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24479713

RESUMEN

BACKGROUND AND OBJECTIVES: Podoplanin, a transmembrane sialoglycoprotein, is expressed by lymphatic endothelial cells and many tumor cells, and is involved in tumor cell-induced platelet aggregation and tumor metastasis. A recent study found that C-type lectin-like receptor 2 (CLEC-2) is a physiologic receptor for podoplanin. Previous studies showed that aggretin, a snake venom-derived protein, activates platelets by targeting platelet CLEC-2. We hypothesized that the C-terminal fragment of aggretin may bind to platelet CLEC-2 and displace podoplanin, in turn exerting antitumor metastatic effects. METHODS AND RESULTS: Aggretin α-chain C-terminus (residues 106-136; AACT) prolonged the lag phase of platelet aggregation induced by aggretin in human washed platelets, indicating that AACT may target the binding site of CLEC-2. HepG2 cells, which are podoplanin-expressing hepatoma cells, induced platelet aggregation with a lag phase. Pretreatment with AACT inhibited platelet aggregation and prolonged the lag phase induced by HepG2 cells. This inhibitory effect was also found with another hepatocarcinoma cell line, HuH-7. AACT inhibited the interaction between HuH-7 cells and platelets, and a specific binding assay demonstrated that CLEC-2 was the binding site for AACT on platelets. In addition, the invasive ability of HepG2 cells was abolished by AACT in a chick embryo chorioallantoic membrane model. Furthermore, formation of lung metastases after intravenous administration of HuH-7 cells was significantly reduced when mice were treated with AACT. CONCLUSIONS: AACT interacts with CLEC-2 of platelets, leading to interference with platelet aggregation and the subsequent metastatic potential of tumor cells. These results suggest that aggretin AACT is a potential candidate for the treatment of tumor metastasis through CLEC-2 blockade.


Asunto(s)
Lectinas Tipo C/química , Lectinas Tipo C/uso terapéutico , Péptidos/química , Agregación Plaquetaria/efectos de los fármacos , Venenos de Víboras/química , Venenos de Víboras/uso terapéutico , Animales , Antineoplásicos/química , Sitios de Unión , Carcinoma Hepatocelular/metabolismo , Línea Celular Tumoral , Pollos , Células Endoteliales/citología , Células Hep G2 , Humanos , Neoplasias Hepáticas/metabolismo , Neoplasias Pulmonares/tratamiento farmacológico , Células MCF-7 , Glicoproteínas de Membrana/metabolismo , Ratones , Metástasis de la Neoplasia , Trasplante de Neoplasias , Estructura Terciaria de Proteína
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...