Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 57
Filtrar
1.
Microbiome ; 12(1): 4, 2024 Jan 03.
Artículo en Inglés | MEDLINE | ID: mdl-38172943

RESUMEN

BACKGROUND: The overgrowth of Desulfovibrio, an inflammation promoting flagellated bacteria, has been found in ulcerative colitis (UC) patients. However, the molecular mechanism in promoting colitis remains unestablished. METHODS: The relative abundance Desulfovibrio vulgaris (D. vulgaris) in stool samples of UC patients was detected. Mice were treated with dextran sulfate sodium to induce colitis with or without administration of D. vulgaris or D. vulgaris flagellin (DVF), and the severity of colitis and the leucine-rich repeat containing 19 (LRRC19) signaling were assessed. The interaction between DVF and LRRC19 was identified by surface plasmon resonance and intestinal organoid culture. Lrrc19-/- and Tlr5-/- mice were used to investigate the indispensable role of LRRC19. Finally, the blockade of DVF-LRRC19 interaction was selected through virtual screening and the efficacy in colitis was assessed. RESULTS: D. vulgaris was enriched in fecal samples of UC patients and was correlated with the disease severity. D. vulgaris or DVF treatment significantly exacerbated colitis in germ-free mice and conventional mice. Mechanistically, DVF could interact with LRRC19 (rather than TLR5) in colitis mice and organoids, and then induce the production of pro-inflammatory cytokines. Lrrc19 knockdown blunted the severity of colitis. Furthermore, typhaneoside, a blockade of binding interfaces, blocked DVF-LRRC19 interaction and dramatically ameliorated DVF-induced colitis. CONCLUSIONS: D. vulgaris could promote colitis through DVF-LRRC19 interaction. Targeting DVF-LRRC19 interaction might be a new therapeutic strategy for UC therapy. Video Abstract.


Asunto(s)
Colitis Ulcerosa , Colitis , Desulfovibrio vulgaris , Humanos , Ratones , Animales , Receptor Toll-Like 5/metabolismo , Receptor Toll-Like 5/uso terapéutico , Desulfovibrio vulgaris/metabolismo , Colitis/inducido químicamente , Colitis/metabolismo , Colitis Ulcerosa/microbiología , Inflamación/metabolismo , Sulfato de Dextran/efectos adversos , Modelos Animales de Enfermedad , Ratones Endogámicos C57BL , Colon/metabolismo , Receptores de Superficie Celular/metabolismo , Receptores de Superficie Celular/uso terapéutico
2.
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
3.
J Biol Chem ; 299(9): 104927, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37330175

RESUMEN

Methicillin-resistant Staphylococcus aureus, or MRSA, is one of the major causative agents of hospital-acquired infections worldwide. Novel antimicrobial strategies efficient against antibiotic-resistant strains are necessary and not only against S. aureus. Among those, strategies that aim at blocking or dismantling proteins involved in the acquisition of essential nutrients, helping the bacteria to colonize the host, are intensively studied. A major route for S. aureus to acquire iron from the host organism is the Isd (iron surface determinant) system. In particular, the hemoglobin receptors IsdH and IsdB located on the surface of the bacterium are necessary to acquire the heme moiety containing iron, making them a plausible antibacterial target. Herein, we obtained an antibody of camelid origin that blocked heme acquisition. We determined that the antibody recognized the heme-binding pocket of both IsdH and IsdB with nanomolar order affinity through its second and third complementary-determining regions. The mechanism explaining the inhibition of acquisition of heme in vitro could be described as a competitive process in which the complementary-determining region 3 from the antibody blocked the acquisition of heme by the bacterial receptor. Moreover, this antibody markedly reduced the growth of three different pathogenic strains of MRSA. Collectively, our results highlight a mechanism for inhibiting nutrient uptake as an antibacterial strategy against MRSA.


Asunto(s)
Anticuerpos Antibacterianos , Staphylococcus aureus Resistente a Meticilina , Receptores de Superficie Celular , Anticuerpos de Dominio Único , Humanos , Antibacterianos/farmacología , Hemo/metabolismo , Staphylococcus aureus Resistente a Meticilina/efectos de los fármacos , Receptores de Superficie Celular/metabolismo , Receptores de Superficie Celular/uso terapéutico , Anticuerpos de Dominio Único/biosíntesis , Anticuerpos de Dominio Único/química , Anticuerpos de Dominio Único/metabolismo , Anticuerpos de Dominio Único/farmacología , Infecciones Estafilocócicas/tratamiento farmacológico , Antígenos Bacterianos/inmunología , Anticuerpos Antibacterianos/genética , Anticuerpos Antibacterianos/inmunología , Camélidos del Nuevo Mundo , Animales , Unión Proteica/efectos de los fármacos , Modelos Moleculares , Simulación de Dinámica Molecular
4.
Pharmacology ; 107(3-4): 216-226, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35073541

RESUMEN

INTRODUCTION: The interindividual variability of the antiplatelet effect of clopidogrel is determined by multiple clinical and genetic factors. A lot of genotype-oriented studies have concentrated on the impact of CYP2C19 gene polymorphisms on platelet aggregation in patients receiving clopidogrel. However, the influence of this polymorphism may be only 12-20%, so other genetic markers should also be investigated. The aim of this work was to study the impact of carriage of CES1, PON1, ABCG2, CYP4F2, CYP3A4, IGTB3, P2Y12, PEAR1, and B4GALT2 polymorphisms on antiplatelet effect of clopidogrel and clinical outcomes in patients with acute coronary syndrome (ACS) and atrial fibrillation (AF). METHODS: 103 patients who underwent ACS with or without percutaneous coronary intervention and concomitant nonvalvular AF were included in an open multicenter prospective study to assess efficacy and safety of combined antithrombotic therapy. The study assessed the frequency of different primary clinical outcomes (incidence of major bleeding, hospital mortality, cardiovascular mortality, stroke and transient ischemic attacks (TIAs), renal mortality) and secondary outcomes (resistance to therapy - high residual platelet reactivity, excessive platelet suppression). Residual platelet reactivity was examined using the VerifyNow system (Accumetrics, Latham, NY, USA). RESULTS: None of the studied genetic markers had no statistically significant effect on the antiaggregant response to clopidogrel in patients with ACS and AF. However, CYP4F2 C(Val433Met) T, PEAR1 rs41273215 C>T were statistically significantly associated with an increased frequency of bleeding on antithrombotic therapy. B4GALT2 rs1061781 was statistically significantly associated with increased frequency of strokes and TIA. CONCLUSION: In our study, we determined that carriers of CYP4F2 gene polymorphisms C(Val433Met)T, PEAR1 rs41273215 C>T (CT+TT) were associated with lower safety of antithrombotic therapy in patients with ACS and AF. And, the B4GALT2 rs1061781 gene polymorphism was associated with a greater risk of insufficient efficacy of the therapy. The data obtained in our study may improve the understanding of the effect of less studied genetic markers on the efficacy and safety of antithrombotic therapy in patients with ACS and AF.


Asunto(s)
Síndrome Coronario Agudo , Fibrilación Atrial , Intervención Coronaria Percutánea , Síndrome Coronario Agudo/tratamiento farmacológico , Síndrome Coronario Agudo/genética , Arildialquilfosfatasa/genética , Arildialquilfosfatasa/uso terapéutico , Fibrilación Atrial/inducido químicamente , Fibrilación Atrial/tratamiento farmacológico , Fibrilación Atrial/genética , Clopidogrel/efectos adversos , Citocromo P-450 CYP2C19/genética , Humanos , Intervención Coronaria Percutánea/efectos adversos , Inhibidores de Agregación Plaquetaria/efectos adversos , Estudios Prospectivos , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/uso terapéutico , Ticlopidina/efectos adversos , Resultado del Tratamiento
5.
Mol Med Rep ; 24(2)2021 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-34165155

RESUMEN

Non­small cell lung cancer (NSCLC) is one of the most common malignancies with high rates of mortality. Although great progress has been made with the development of novel immunotherapies and targeted therapeutic strategies, the 5­year total survival rate of lung cancer has remained unchanged over the past few decades. Therefore, more effective therapeutics are urgently needed. Tumor endothelial marker 8 (TEM8) is an integrin­like cell surface transmembrane protein that has been demonstrated to be upregulated in numerous cancer types and previously showed promise for targeted cancer therapy. However, the role of TEM8 in NSCLC remains poorly understood. The present study aimed to investigate the effects of silencing TEM8 on expression and regulation of extracellular signal­regulated kinase (ERK)1/2 signaling pathways in NSCLC. In the present study, a lentiviral vector that encoded a short hairpin RNA targeting TEM8 was designed and transfected into Xuanwei Lung Cancer (XWLC)­05 lung cancer cells to silence TEM8 expression. Male BALB/c­nu/nu mice were then given subcutaneous injections in the right dorsal flank with XWLC­05 cells. Microvessel density was measured using an anti­CD34 antibody. The mRNA and protein levels of ERK1/2 and Bcl­2 in XWLC­05 cells or xenograft tumor tissues were detected by reverse transcription­quantitative polymerase chain reaction and western blotting. TEM8 knockdown was found to significantly inhibit tumor growth and conferred an anti­angiogenic ability in vivo. Furthermore, TEM8 knockdown suppressed the expression of Bcl­2 mediated by ERK1/2 activity in XWLC­05 cells or tissues from mice with NSCLC. To conclude, these results suggest that the targeted silencing of TEM8 may serve as an effective method of treating NSCLC.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Neoplasias Pulmonares/metabolismo , Proteínas de Microfilamentos/genética , Proteínas de Microfilamentos/metabolismo , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/metabolismo , Transducción de Señal , Animales , Carcinoma de Pulmón de Células no Pequeñas/patología , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica , Técnicas de Silenciamiento del Gen , Silenciador del Gen , Pulmón/patología , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/terapia , Ratones , Ratones Endogámicos BALB C , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Receptores de Superficie Celular/uso terapéutico , Ensayos Antitumor por Modelo de Xenoinjerto
6.
Eur Rev Med Pharmacol Sci ; 25(8): 3325-3337, 2021 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-33928620

RESUMEN

Since the reports in Wuhan (China), in December 2019, of the first cluster of cases of pneumonia caused by the new Coronavirus called 2019-nCoV or SARS-CoV-2, there has been a pandemic spread of the infection. By now, we have no specific therapy to counteract this emergency. The latest epidemiological data suggest that children are just as likely as adults to get infected by the virus. Most of them show mild clinical pictures or are completely asymptomatic, but there is an increased risk for severe disease in infancy (<12 months of age) and in children with underlying medical conditions. In this article, research achievements on the treatment of pediatric SARS-CoV-2 infection are examined.


Asunto(s)
Antivirales/uso terapéutico , COVID-19/terapia , Factores Inmunológicos/uso terapéutico , Adenosina Monofosfato/análogos & derivados , Adenosina Monofosfato/uso terapéutico , Adolescente , Factores de Edad , Alanina/análogos & derivados , Alanina/uso terapéutico , Anticuerpos Monoclonales Humanizados/uso terapéutico , Anticoagulantes/uso terapéutico , Portador Sano , Niño , Preescolar , Cloroquina/uso terapéutico , Terapia de Reemplazo Renal Continuo , Combinación de Medicamentos , Oxigenación por Membrana Extracorpórea , Glucocorticoides/uso terapéutico , Humanos , Hidroxicloroquina/uso terapéutico , Inmunización Pasiva , Inmunoglobulinas Intravenosas/uso terapéutico , Lactante , Recién Nacido , Interferón-alfa/uso terapéutico , Lopinavir/uso terapéutico , Oseltamivir/uso terapéutico , Receptores de Superficie Celular/uso terapéutico , Respiración Artificial , Ribavirina/uso terapéutico , Ritonavir/uso terapéutico , SARS-CoV-2 , Índice de Severidad de la Enfermedad , Trombosis/prevención & control , Tratamiento Farmacológico de COVID-19 , Sueroterapia para COVID-19
7.
Support Care Cancer ; 29(5): 2415-2421, 2021 May.
Artículo en Inglés | MEDLINE | ID: mdl-32918133

RESUMEN

PURPOSE: Chemotherapy-induced gastrointestinal toxicity is a common adverse event during chemotherapeutic treatment. No uniformly applicable strategies exist to predict, prevent, or treat gastrointestinal toxicity. Thus, a goal of mucositis research is to identify targets for therapeutic interventions and individualized risk prediction. Fibrinogen C domain containing 1 (FIBCD1) is a transmembrane protein expressed in human intestinal epithelial cells with functions in the innate immune system. Previous observations have shown that FIBCD1 ameliorates dextran sulfate sodium (DSS)-induced intestinal inflammation in vivo. We evaluated the effect of FIBCD1 in a murine model of chemotherapy-induced gastrointestinal toxicity and inflammation. METHODS: Transgenic (Tg) mice overexpressing FIBCD1 in the intestinal epithelium (Fibcd1Tg) and wild-type (WT) littermates (C57BL/6N) were randomized to receive an intraperitoneal injection of doxorubicin 20 mg/kg or saline and were terminated 2 or 7 days after the injection. Gastrointestinal toxicity was evaluated by weight change, intestinal length, villus height/crypt depth, and histological mucositis score. Expression of inflammatory markers (IL-6, IL-1ß, and Tnfα) was measured by quantitative real-time PCR in intestinal tissue samples. RESULTS: Following doxorubicin treatment, WT mice exhibited an increased weight loss compared with Tg littermates (p < 0.001). No differences between genotypes were seen in mucositis score, intestinal length, villus height/crypt depth, or IL-6, IL-1ß, and Tnfα expression. CONCLUSION: Our findings suggest that FIBCD1 could ameliorate chemotherapy-induced gastrointestinal toxicity by reducing weight loss; however, the mechanism of this possible protective effect remains to be defined warranting additional investigations.


Asunto(s)
Antineoplásicos/uso terapéutico , Mucositis/inducido químicamente , Mucositis/tratamiento farmacológico , Receptores de Superficie Celular/uso terapéutico , Pérdida de Peso/efectos de los fármacos , Animales , Modelos Animales de Enfermedad , Genotipo , Inyecciones Intraperitoneales , Masculino , Ratones , Ratones Endogámicos C57BL
8.
Cell Signal ; 66: 109487, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31778739

RESUMEN

Extracellular matrix (ECM) macromolecules together with a multitude of different molecules residing in the extracellular space play a vital role in the regulation of cellular phenotype and behavior. This is achieved via constant reciprocal interactions between the molecules of the ECM and the cells. The ECM-cell interactions are mediated via cell surface receptors either directly or indirectly with co-operative molecules. The ECM is also under perpetual remodeling process influencing cell-signaling pathways on its part. The fragmentation of ECM macromolecules provides even further complexity for the intricate environment of the cells. However, as long as the interactions between the ECM and the cells are in balance, the health of the body is retained. Alternatively, any dysregulation in these interactions can lead to pathological processes and finally to various diseases. Thus, therapeutic applications that are based on retaining normal ECM-cell interactions are highly rationale. Moreover, in the light of the current knowledge, also concurrent multi-targeting of the complex ECM-cell interactions is required for potent pharmacotherapies to be developed in the future.


Asunto(s)
Proteínas de la Matriz Extracelular , Matriz Extracelular/metabolismo , Receptores de Superficie Celular , Animales , Proteínas de la Matriz Extracelular/metabolismo , Proteínas de la Matriz Extracelular/uso terapéutico , Humanos , Receptores de Superficie Celular/metabolismo , Receptores de Superficie Celular/uso terapéutico , Transducción de Señal
9.
Oncogene ; 38(8): 1183-1199, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30258108

RESUMEN

Glioma is a devastating cancer with a dismal prognosis and there is an urgent need to discover novel glioma-specific antigens for glioma therapy. Previous studies have identified CD163-positive tumour cells in certain solid tumours, but CD163 expression in glioma remains unknown. In this study, via an analysis of public datasets, we demonstrated that CD163 overexpression in glioma specimens correlated with an unfavourable patient prognosis. CD163 expression was increased in glioma cells, especially primary glioma cells. The loss of CD163 expression inhibited both cell cycle progression and the proliferation of glioblastoma multiforme (GBM) cell lines and primary glioma cells. CD163 interacted directly with casein kinase 2 (CK2) and CD163 silencing reduced AKT/GSK3ß/ß-catenin/cyclin D1 pathway activity via CK2. Moreover, CD163 was upregulated in CD133-positive glioma stem cells (GSCs), and CD163 downregulation decreased the expression of GSC markers, including CD133, ALDH1A1, NANOG and OCT4. The knockdown of CD163 impaired GSC stemness by inhibiting the CK2/AKT/GSK3ß/ß-catenin pathway. Finally, a CD163 antibody successfully induced complement-dependent cytotoxicity against glioma cells. Our findings indicate that CD163 contributes to gliomagenesis via CK2 and provides preclinical evidence that CD163 and the CD163 pathway might serve as a therapeutic target for glioma.


Asunto(s)
Antígenos CD/genética , Antígenos de Diferenciación Mielomonocítica/genética , Quinasa de la Caseína II/genética , Glioblastoma/genética , Terapia Molecular Dirigida , Receptores de Superficie Celular/genética , Animales , Antígenos CD/uso terapéutico , Antígenos de Diferenciación Mielomonocítica/uso terapéutico , Apoptosis/genética , Línea Celular Tumoral , Proliferación Celular/genética , Regulación Neoplásica de la Expresión Génica , Técnicas de Silenciamiento del Gen , Glioblastoma/patología , Glucógeno Sintasa Quinasa 3 beta/genética , Humanos , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Pronóstico , Receptores de Superficie Celular/uso terapéutico , Transducción de Señal , beta Catenina/genética
10.
Am J Physiol Renal Physiol ; 315(5): F1416-F1421, 2018 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-30019932

RESUMEN

The antidiuretic hormone vasopressin (VP) is produced by the hypothalamus and is stored and secreted from the posterior pituitary. VP acts via VP type 2 receptors (V2Rs) on the basolateral membrane of principal cells of the collecting duct (CD) to regulate fluid permeability. The VP-evoked endocrine pathway is essential in determining urine concentrating capability. For example, a defect in any component of the VP signaling pathway can result in polyuria, polydipsia, and hypotonic urine, collectively termed diabetes insipidus (DI). A lack of VP production precipitates central diabetes insipidus (CDI), which can be managed effectively by VP supplementation. A majority of cases of nephrogenic diabetes insipidus (NDI) result from V2R mutations that impair receptor sensitivity. No specific therapy is currently available for management of NDI. Evidence is evolving that (pro)renin receptor (PRR), a newly identified member of the renin-angiotensin system, is capable of regulating VP production and action. As such, PRR should be considered strongly as a therapeutic target for treating CDI and NDI. The current review will summarize recent advances in understanding the physiology of renal and central PRR as it relates to the two types of DI.


Asunto(s)
Fármacos Antidiuréticos/uso terapéutico , Diabetes Insípida/tratamiento farmacológico , Diuresis/efectos de los fármacos , Riñón/efectos de los fármacos , Receptores de Superficie Celular/uso terapéutico , Sistema Renina-Angiotensina/efectos de los fármacos , Animales , Diabetes Insípida/enzimología , Diabetes Insípida/fisiopatología , Predisposición Genética a la Enfermedad , Humanos , Riñón/enzimología , Riñón/patología , Mutación , Fenotipo , Receptores de Superficie Celular/metabolismo , Receptores de Vasopresinas/genética , Vasopresinas/metabolismo , Receptor de Prorenina
11.
Physiol Rep ; 5(21)2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-29138356

RESUMEN

Earlier we reported that the recombinant soluble (pro) renin receptor sPRR-His upregulates renal aquoporin-2 (AQP2) expression, and attenuates polyuria associated with nephrogenic diabetes insipidus (NDI) induced by vasopressin type 2 receptor (V2R) antagonism. Patients that receive lithium therapy develop polyuria associated NDI that might be secondary to downregulation of renal AQP2. We hypothesized that sPRR-His attenuates indices of NDI associated with lithium treatment. Eight-week-old male C57/BL6 mice consumed chow supplemented with LiCl (40 mmol/kg diets) for 14 days. For the last 7 days mice received either sPRR-His [30 µg/(kg day), i.v.; sPRR] or vehicle (Veh) via minipump. Control (Con) mice consumed standard chow for 14 days. Compared to Con mice, 14-d LiCl treatment elevated water intake and urine volume, and decreased urine osmolality, regardless of sPRR-His or Veh administration. These data indicate that sPRR-His treatment does not attenuate indices of NDI evoked by lithium. Unexpectedly, epididymal fat mass was lower, adipocyte UCP1 mRNA and protein expression were higher, and multilocular lipid morphology was enhanced, in LiCl-fed mice treated with sPRR-His versus vehicle. The beiging of white adipose tissue is a novel metabolic benefit of manipulating the sPRR in the context of lithium-induced NDI.


Asunto(s)
Tejido Adiposo Blanco/efectos de los fármacos , Antimaníacos/toxicidad , Diabetes Insípida Nefrogénica/inducido químicamente , Cloruro de Litio/toxicidad , Receptores de Superficie Celular/uso terapéutico , Animales , Acuaporina 2/biosíntesis , Diabetes Insípida Nefrogénica/metabolismo , Diabetes Insípida Nefrogénica/fisiopatología , Diabetes Insípida Nefrogénica/prevención & control , Evaluación Preclínica de Medicamentos/métodos , Regulación de la Expresión Génica/efectos de los fármacos , Masculino , Ratones Endogámicos C57BL , ARN Mensajero/genética , Proteínas Recombinantes/farmacología , Solubilidad , Proteína Desacopladora 1/biosíntesis , Proteína Desacopladora 1/genética , Micción/efectos de los fármacos , Receptor de Prorenina
12.
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
13.
J Neuroinflammation ; 13: 43, 2016 Feb 18.
Artículo en Inglés | MEDLINE | ID: mdl-26891688

RESUMEN

BACKGROUND: Interaction of CD200 with its receptor CD200R has an immunoregulatory role and attenuates various types of neuroinflammatory diseases. METHODS: Immunofluorescence staining, western blot analysis, and RT-PCR were used to investigate the modulatory effects of CD200 fusion protein (CD200Fc) on activation of microglia and astrocytes as well as synthesis of pro- (TNF, IL-1ß, IL-6) and anti-inflammatory (IL-4, IL-10) cytokines in the L4-L5 spinal cord segments in relation to behavioral signs of neuropathic pain after unilateral sterile chronic constriction injury (sCCI) of the sciatic nerve. Withdrawal thresholds for mechanical hypersensitivity and latencies for thermal hypersensitivity were measured in hind paws 1 day before operation; 1, 3, and 7 days after sCCI operation; and then 5 and 24 h after intrathecal application of artificial cerebrospinal fluid or CD200Fc. RESULTS: Seven days from sCCI operation and 5 h from intrathecal application, CD200Fc reduced mechanical and thermal hypersensitivity when compared with control animals. Simultaneously, CD200Fc attenuated activation of glial cells and decreased proinflammatory and increased anti-inflammatory cytokine messenger RNA (mRNA) levels. Administration of CD200Fc also diminished elevation of CD200 and CD200R proteins as a concomitant reaction of the modulatory system to increased neuroinflammatory reactions after nerve injury. The anti-inflammatory effect of CD200Fc dropped at 24 h after intrathecal application. CONCLUSIONS: Intrathecal administration of the CD200R1 agonist CD200Fc induces very rapid suppression of neuroinflammatory reactions associated with glial activation and neuropathic pain development. This may constitute a promising and novel therapeutic approach for the treatment of neuropathic pain.


Asunto(s)
Antígenos de Superficie/uso terapéutico , Citocinas/metabolismo , Hiperalgesia/etiología , Inflamación/etiología , Receptores de Superficie Celular/uso terapéutico , Ciática/complicaciones , Animales , Antígenos CD/metabolismo , Antígenos de Superficie/farmacología , Citocinas/genética , Modelos Animales de Enfermedad , Regulación de la Expresión Génica/efectos de los fármacos , Proteína Ácida Fibrilar de la Glía/metabolismo , Hiperalgesia/tratamiento farmacológico , Inflamación/tratamiento farmacológico , Inflamación/metabolismo , Inyecciones Espinales , Masculino , Neuroglía/efectos de los fármacos , Neuroglía/metabolismo , Receptores de Orexina , Umbral del Dolor/efectos de los fármacos , Estimulación Física/efectos adversos , Ratas , Ratas Wistar , Receptores de Superficie Celular/antagonistas & inhibidores , Ciática/tratamiento farmacológico , Médula Espinal/efectos de los fármacos , Médula Espinal/metabolismo , Factores de Tiempo
14.
J Am Soc Nephrol ; 27(8): 2331-45, 2016 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26701976

RESUMEN

AKI confers increased risk of progression to CKD. αKlotho is a cytoprotective protein, the expression of which is reduced in AKI, but the relationship of αKlotho expression level to AKI progression to CKD has not been studied. We altered systemic αKlotho levels by genetic manipulation, phosphate loading, or aging and examined the effect on long-term outcome after AKI in two models: bilateral ischemia-reperfusion injury and unilateral nephrectomy plus contralateral ischemia-reperfusion injury. Despite apparent initial complete recovery of renal function, both types of AKI eventually progressed to CKD, with decreased creatinine clearance, hyperphosphatemia, and renal fibrosis. Compared with wild-type mice, heterozygous αKlotho-hypomorphic mice (αKlotho haploinsufficiency) progressed to CKD much faster, whereas αKlotho-overexpressing mice had better preserved renal function after AKI. High phosphate diet exacerbated αKlotho deficiency after AKI, dramatically increased renal fibrosis, and accelerated CKD progression. Recombinant αKlotho administration after AKI accelerated renal recovery and reduced renal fibrosis. Compared with wild-type conditions, αKlotho deficiency and overexpression are associated with lower and higher autophagic flux in the kidney, respectively. Upregulation of autophagy protected kidney cells in culture from oxidative stress and reduced collagen 1 accumulation. We propose that αKlotho upregulates autophagy, attenuates ischemic injury, mitigates renal fibrosis, and retards AKI progression to CKD.


Asunto(s)
Lesión Renal Aguda/complicaciones , Autofagia/fisiología , Progresión de la Enfermedad , Receptores de Superficie Celular/fisiología , Insuficiencia Renal Crónica/etiología , Animales , Autofagia/efectos de los fármacos , Glucuronidasa , Proteínas Klotho , Ratones , Receptores de Superficie Celular/uso terapéutico , Insuficiencia Renal Crónica/prevención & control
15.
Nat Neurosci ; 18(3): 331-8, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25710834

RESUMEN

Current treatment options for epilepsy are inadequate, as too many patients suffer from uncontrolled seizures and from negative side effects of treatment. In addition to these clinical challenges, our scientific understanding of epilepsy is incomplete. Optogenetic and designer receptor technologies provide unprecedented and much needed specificity, allowing for spatial, temporal and cell type-selective modulation of neuronal circuits. Using such tools, it is now possible to begin to address some of the fundamental unanswered questions in epilepsy, to dissect epileptic neuronal circuits and to develop new intervention strategies. Such specificity of intervention also has the potential for direct therapeutic benefits, allowing healthy tissue and network functions to continue unaffected. In this Perspective, we discuss promising uses of these technologies for the study of seizures and epilepsy, as well as potential use of these strategies for clinical therapies.


Asunto(s)
Anticonvulsivantes/uso terapéutico , Epilepsia/patología , Epilepsia/terapia , Red Nerviosa/efectos de los fármacos , Optogenética , Animales , Epigénesis Genética , Epilepsia/genética , Humanos , Red Nerviosa/fisiología , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/fisiología , Receptores de Superficie Celular/uso terapéutico
16.
Curr Opin Biotechnol ; 24(6): 1072-7, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23587963

RESUMEN

Protein-based biologics, which leverage the inherent affinity and specificity of protein-protein interactions, offer an effective strategy for targeting and modulating disease pathways. Despite the broad diversity of the proteome, monoclonal antibodies have been the major focus of such drug discovery efforts. While antibodies have shown great clinical value, the breadth and complexity of human disease highlight the need for alternatives that expand the therapeutic repertoire beyond this single class of proteins. The elucidation of molecular mechanisms underlying human disease has provided new opportunities for protein-based drugs to address challenging clinical problems. Natural ligands and receptors, which inherently modulate complex biological processes, have emerged as promising candidates for protein-based drug discovery efforts. Protein engineering strategies, guided by biological principles, are allowing ligands and receptors to be developed as next-generation therapeutics with improved safety and efficacy.


Asunto(s)
Descubrimiento de Drogas/métodos , Ingeniería de Proteínas/métodos , Proteínas/uso terapéutico , Anticuerpos Monoclonales/uso terapéutico , Descubrimiento de Drogas/tendencias , Humanos , Ligandos , Ingeniería de Proteínas/tendencias , Proteínas/efectos adversos , Proteínas/metabolismo , Receptor Cross-Talk/efectos de los fármacos , Receptores de Superficie Celular/antagonistas & inhibidores , Receptores de Superficie Celular/metabolismo , Receptores de Superficie Celular/uso terapéutico
17.
Minerva Med ; 103(6): 513-24, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23229370

RESUMEN

There is an ongoing search for biomarkers that can facilitate the diagnosis of subclinical or clinically manifest cardiovascular disease. One of the emerging biomarkers currently under investigation is ST2, which is the receptor of Interleukin-33 (IL-33). ST2 is a member of the Interleukin-1 receptor family and exists in a transmembrane (ST2L) and a soluble form (sST2) due to alternative splicing. Several groups have reported sST2 elevations in serum of cardiovascular disease patients. There is consisting evidence that sST2 is independently predictive for mortality in patients with heart failure or myocardial infarction. In addition to its potential as a biomarker for adverse cardiovascular events, ST2 is considered to play a causal role in chronic cardiovascular diseases such as atherosclerosis and heart failure. Signaling of IL-33 via ST2 has been shown to be cardioprotective in mouse models of myocardial infarction, heart transplantation and cardiac hypertrophy and fibrosis. Furthermore, treatment with IL-33 reduced the development of plaques in atherosclerotic mice. In this paper we will review the currently available literature on sST2 as a biomarker for adverse cardiovascular events. In addition, we will elaborate on the potential mechanistic role of the IL-33/ST2 pathway in chronic inflammatory cardiovascular diseases.


Asunto(s)
Enfermedades Cardiovasculares , Interleucinas/fisiología , Receptores de Superficie Celular/fisiología , Animales , Biomarcadores/metabolismo , Enfermedades Cardiovasculares/tratamiento farmacológico , Enfermedades Cardiovasculares/metabolismo , Enfermedades Cardiovasculares/mortalidad , Humanos , Inmunosupresores/efectos adversos , Inmunosupresores/uso terapéutico , Proteína 1 Similar al Receptor de Interleucina-1 , Interleucina-33 , Interleucinas/uso terapéutico , Ratones , Receptores de Superficie Celular/uso terapéutico
18.
Exp Neurol ; 237(1): 55-69, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22728374

RESUMEN

Several pharmacological approaches to promote neural repair and recovery after CNS injury have been identified. Blockade of either astrocyte-derived chondroitin sulfate proteoglycans (CSPGs) or oligodendrocyte-derived NogoReceptor (NgR1) ligands reduces extrinsic inhibition of axonal growth, though combined blockade of these distinct pathways has not been tested. The intrinsic growth potential of adult mammalian neurons can be promoted by several pathways, including pre-conditioning injury for dorsal root ganglion (DRG) neurons and macrophage activation for retinal ganglion cells (RGCs). Singly, pharmacological interventions have restricted efficacy without foreign cells, mechanical scaffolds or viral gene therapy. Here, we examined combinations of pharmacological approaches and assessed the degree of axonal regeneration. After mouse optic nerve crush injury, NgR1-/- neurons regenerate RGC axons as extensively as do zymosan-injected, macrophage-activated WT mice. Synergistic enhancement of regeneration is achieved by combining these interventions in zymosan-injected NgR1-/- mice. In rats with a spinal dorsal column crush injury, a preconditioning peripheral sciatic nerve axotomy, or NgR1(310)ecto-Fc decoy protein treatment or ChondroitinaseABC (ChABC) treatment independently support similar degrees of regeneration by ascending primary afferent fibers into the vicinity of the injury site. Treatment with two of these three interventions does not significantly enhance the degree of axonal regeneration. In contrast, triple therapy combining NgR1 decoy, ChABC and preconditioning, allows axons to regenerate millimeters past the spinal cord injury site. The benefit of a pre-conditioning injury is most robust, but a peripheral nerve injury coincident with, or 3 days after, spinal cord injury also synergizes with NgR1 decoy and ChABC. Thus, maximal axonal regeneration and neural repair are achieved by combining independently effective pharmacological approaches.


Asunto(s)
Axones/fisiología , Inhibición Neural/fisiología , Neuroglía/fisiología , Traumatismos de la Médula Espinal/fisiopatología , Traumatismos de la Médula Espinal/rehabilitación , Animales , Axones/patología , Condroitina ABC Liasa/administración & dosificación , Condroitina ABC Liasa/uso terapéutico , Femenino , Proteínas Ligadas a GPI/antagonistas & inhibidores , Proteínas Ligadas a GPI/deficiencia , Proteínas Ligadas a GPI/uso terapéutico , Activación de Macrófagos/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas de la Mielina/antagonistas & inhibidores , Proteínas de la Mielina/deficiencia , Proteínas de la Mielina/uso terapéutico , Inhibición Neural/genética , Neuroglía/patología , Receptor Nogo 1 , Traumatismos del Nervio Óptico/genética , Traumatismos del Nervio Óptico/patología , Traumatismos del Nervio Óptico/fisiopatología , Ratas , Ratas Sprague-Dawley , Receptores de Superficie Celular/antagonistas & inhibidores , Receptores de Superficie Celular/deficiencia , Receptores de Superficie Celular/uso terapéutico , Traumatismos de la Médula Espinal/genética , Zimosan/administración & dosificación
19.
Med Res Rev ; 32(2): 349-87, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20577974

RESUMEN

Twenty-seven years after the discovery of HIV as the cause of AIDS more than 25 drugs directed against four different viral targets (i.e. reverse transcriptase, protease, integrase, envelope gp41) and one cellular target (i.e. CCR5 co-receptor) are available for treatment. However, the search for an efficient vaccine is still ongoing. One of the main problems is the presence of a continuously evolving dense carbohydrate shield, consisting of N-linked glycans that surrounds the virion and protects it against efficient recognition and persistent neutralization by the immune system. However, several lectins from the innate immune system specifically bind to these glycans in an attempt to process the virus antigens to provoke an immune response. Across a wide variety of different species in nature lectins can be found that can interact with the glycosylated envelope of HIV-1 and can block the infection of susceptible cells by the virus. In this review, we will give an overview of the lectins from non-mammalian origin that are endowed with antiviral properties and discuss the complex interactions between lectins of the innate immune system and HIV-1. Also, attention will be given to different carbohydrate-related modalities that can be exploited for antiviral chemotherapy.


Asunto(s)
Antivirales/uso terapéutico , Proteínas Bacterianas/uso terapéutico , Proteínas Portadoras/uso terapéutico , Infecciones por VIH/tratamiento farmacológico , Infecciones por VIH/inmunología , VIH-1/inmunología , VIH-1/metabolismo , Humanos , Inmunidad Innata , Lectinas/uso terapéutico , Receptores de Superficie Celular/uso terapéutico , Proteínas del Envoltorio Viral/efectos de los fármacos
20.
Am J Med Sci ; 342(6): 494-502, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22108171

RESUMEN

This study was designed to determine the effects of in vivo anticubilin antisense RNA on the uptake of albumin in tubules and on the tubulointerstitial injury in adriamycin-induced proteinuric rats. Adriamycin-treated rats were subjected to intrarenal delivery of adenoviral vectors encoding empty plasmid, cubilin sense RNA expression vector pAd-CUB or anticubilin antisense RNA expression vector pAd-ACUB on day 3. On days 14 and 28, half of the rats in each group were randomly selected to be killed, and blood samples, kidney tissues and 24-hour urine were collected. The diseased rats treated with pAdEasy-ACUB showed a 60% decrease in serum creatinine and glomerular filtration rate. Interestingly, the anticubilin antisense treatment led to a marked increase in albuminuria. Antisense treatment attenuated the histologic changes on both day 14 and day 28. The antisense treatment induced more than 60% recovery of adriamycin-induced injury, accompanied with 85% knockdown in the expression of cubilin protein and markedly decreased albumin deposition. Adriamycin induced an increase in the expression of monocyte chemoattractant protein-1, transforming growth factor-ß and regulated on activation in normal T-cell expressed and secreted and the number of infiltrating cells, which was reversed by the antisense treatment. Anticubilin antisense RNA delivered by an adenoviral vector ameliorates albuminuria-induced glomerulosclerosis and tubulointerstitial damage in adriamycin nephrotic rats, indicating that cubilin could be a potential therapeutic target in proteinuric nephropathy.


Asunto(s)
Albuminuria , Doxorrubicina/efectos adversos , Enfermedades Renales/inducido químicamente , ARN sin Sentido/uso terapéutico , Adenoviridae , Albuminuria/tratamiento farmacológico , Animales , Western Blotting , Quimiocina CCL2/metabolismo , Creatinina/sangre , Creatinina/orina , Vectores Genéticos , Tasa de Filtración Glomerular , Enfermedades Renales/patología , Enfermedades Renales/fisiopatología , Masculino , Modelos Animales , Proteinuria/tratamiento farmacológico , Distribución Aleatoria , Ratas , Ratas Sprague-Dawley , Receptores de Superficie Celular/uso terapéutico , Albúmina Sérica/metabolismo , Factor de Crecimiento Transformador beta/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...