Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 62
Filtrar
1.
J Biomed Sci ; 31(1): 54, 2024 May 24.
Artículo en Inglés | MEDLINE | ID: mdl-38790021

RESUMEN

BACKGROUND: Alcohol-related liver disease (ALD) is a major health concern worldwide, but effective therapeutics for ALD are still lacking. Tumor necrosis factor-inducible gene 6 protein (TSG-6), a cytokine released from mesenchymal stem cells, was shown to reduce liver fibrosis and promote successful liver repair in mice with chronically damaged livers. However, the effect of TSG-6 and the mechanism underlying its activity in ALD remain poorly understood. METHODS: To investigate its function in ALD mice with fibrosis, male mice chronically fed an ethanol (EtOH)-containing diet for 9 weeks were treated with TSG-6 (EtOH + TSG-6) or PBS (EtOH + Veh) for an additional 3 weeks. RESULTS: Severe hepatic injury in EtOH-treated mice was markedly decreased in TSG-6-treated mice fed EtOH. The EtOH + TSG-6 group had less fibrosis than the EtOH + Veh group. Activation of cluster of differentiation 44 (CD44) was reported to promote HSC activation. CD44 and nuclear CD44 intracellular domain (ICD), a CD44 activator which were upregulated in activated HSCs and ALD mice were significantly downregulated in TSG-6-exposed mice fed EtOH. TSG-6 interacted directly with the catalytic site of MMP14, a proteolytic enzyme that cleaves CD44, inhibited CD44 cleavage to CD44ICD, and reduced HSC activation and liver fibrosis in ALD mice. In addition, a novel peptide designed to include a region that binds to the catalytic site of MMP14 suppressed CD44 activation and attenuated alcohol-induced liver injury, including fibrosis, in mice. CONCLUSIONS: These results demonstrate that TSG-6 attenuates alcohol-induced liver damage and fibrosis by blocking CD44 cleavage to CD44ICD and suggest that TSG-6 and TSG-6-mimicking peptide could be used as therapeutics for ALD with fibrosis.


Asunto(s)
Moléculas de Adhesión Celular , Receptores de Hialuranos , Cirrosis Hepática , Hepatopatías Alcohólicas , Animales , Masculino , Ratones , Moléculas de Adhesión Celular/administración & dosificación , Etanol , Receptores de Hialuranos/metabolismo , Receptores de Hialuranos/genética , Cirrosis Hepática/metabolismo , Cirrosis Hepática/tratamiento farmacológico , Cirrosis Hepática/inducido químicamente , Hepatopatías Alcohólicas/metabolismo , Hepatopatías Alcohólicas/tratamiento farmacológico , Ratones Endogámicos C57BL , Péptidos/farmacología , Péptidos/metabolismo
2.
J Stroke Cerebrovasc Dis ; 29(12): 104986, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-32992175

RESUMEN

BACKGROUND: Early brain injury (EBI) refers to acute brain injury during the first 72 h after subarachnoid hemorrhage (SAH), which is one of the major causes of poor prognosis after SAH. Here, we investigated the effect and the related mechanism of TSG-6 on EBI after SAH. MATERIALS AND METHODS: The Sprague-Dawley rat model of SAH was developed by the endovascular perforation method. TSG-6 (5µg) was administered by an intraventricular injection within 1.5 h after SAH. The effects of TSG-6 on EBI were assessed by neurological score, brain water content (BWC) and TUNEL staining. Immunofluorescence staining was used to assay NF-κB/p-NF-κB expression in microglia. Protein expression levels of heme oxygenase-1 (HO-1), NADPH oxidase 2 (Nox2), Bcl-2, Bax, and cleaved-caspase-3 were measured to investigate the potential mechanism. The enzyme activity of superoxide dismutase (SOD) and glutathione peroxidase (GSH-Px) and the level of reactive oxygen species (ROS) were analyzed using commercially available kits. RESULTS: The results showed that TSG-6 treatment alleviated the neurobehavioral dysfunction and reduced BWC and the number of TUNEL-positive neurons in EBI after SAH. TSG-6 decreased the ROS level and enhanced the enzyme activity of SOD and GSH-Px after SAH. Furthermore TSG-6 inhibited the NF-κB activation, increased the protein expression levels of HO-1 and Bcl-2 and decreased the expression levels of Nox2, Bax, and cleaved-caspase-3. The administration of TSG-6 siRNA abolished the protective effects of TSG-6 on EBI after SAH. CONCLUSION: We found that TSG-6 attenuated oxidative stress and apoptosis in EBI after SAH partly by inhibiting NF-κB and activating HO-1 pathway in brain tissue.


Asunto(s)
Antioxidantes/administración & dosificación , Encéfalo/efectos de los fármacos , Moléculas de Adhesión Celular/administración & dosificación , Hemo Oxigenasa (Desciclizante)/metabolismo , NADPH Oxidasa 2/metabolismo , Fármacos Neuroprotectores/administración & dosificación , Estrés Oxidativo/efectos de los fármacos , Hemorragia Subaracnoidea/tratamiento farmacológico , Animales , Apoptosis/efectos de los fármacos , Encéfalo/enzimología , Encéfalo/patología , Moléculas de Adhesión Celular/genética , Moléculas de Adhesión Celular/metabolismo , Modelos Animales de Enfermedad , Inyecciones Intraventriculares , FN-kappa B/metabolismo , Ratas Sprague-Dawley , Transducción de Señal , Hemorragia Subaracnoidea/enzimología , Hemorragia Subaracnoidea/genética , Hemorragia Subaracnoidea/patología , Factores de Tiempo
3.
Biochem Biophys Res Commun ; 496(4): 1376-1381, 2018 02 19.
Artículo en Inglés | MEDLINE | ID: mdl-29421653

RESUMEN

In active thyroid eye disease (TED), the extraocular muscles feature excessive hyaluronan (HA) accumulation and increased fibroblast proliferation. To investigate the effects of HA on proliferation, we cultured perimysial fibroblasts from extraocular muscles of active TED patients, and adopted IGF-1 and PH20 as modulators for HA concentration and HA polymer size. Based on the results, IGF-1 increased HA concentration, promoted high molecular weight HA (HMW-HA) proportion and stimulated fibroblast proliferation. Hyaluronidase PH20 decreased HA concentration, but caused HMW-HA accumulation and exaggerating proliferation as well. Combined treatment with both reagents resulted in retention of low molecular weight HA (LMW-HA), and suppressed fibroblast proliferation. Pearson correlation demonstrated no significance between HA concentration and proliferation. Mitogenic investigation unveiled the stimulatory effects of HMW-HA via membrane depolarization and inhibitive effects of LMW-HA via membrane hyperpolarization. Our findings offer insights into the essential role of HA molecular weight during TED pathogenesis.


Asunto(s)
Moléculas de Adhesión Celular/administración & dosificación , Fibroblastos/patología , Oftalmopatía de Graves/tratamiento farmacológico , Oftalmopatía de Graves/metabolismo , Ácido Hialurónico/metabolismo , Hialuronoglucosaminidasa/administración & dosificación , Factor I del Crecimiento Similar a la Insulina/administración & dosificación , Músculos Oculomotores/patología , Adulto , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Femenino , Fibroblastos/efectos de los fármacos , Oftalmopatía de Graves/patología , Humanos , Ácido Hialurónico/química , Masculino , Persona de Mediana Edad , Músculos Oculomotores/efectos de los fármacos , Tamaño de la Partícula , Relación Estructura-Actividad
4.
Reprod Fertil Dev ; 28(4): 416-27, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25209425

RESUMEN

The key goals of immunocontraception research are to obtain full contraceptive effects using vaccines administered to both males and females. Current research concerning human anti-sperm contraceptive vaccines is focused on delineating infertility-related epitopes to avoid autoimmune disease. We constructed phage-display peptide libraries to select epitope peptides derived from human posterior head 20 (hPH20) and homo sapiens sperm acrosome associated 1 (hSPACA1) using sera collected from infertile women harbouring anti-sperm antibodies. Following five rounds of selection, positive colonies were reconfirmed for reactivity with the immunoinfertile sera. We biopanned and analysed the chemical properties of four epitope peptides, named P82, Sa6, Sa37 and Sa76. Synthetic peptides were made and coupled to either bovine serum albumin (BSA) or ovalbumin. We used the BSA-conjugated peptides to immunise BALB/c mice and examined the effects on fertility in female and male mice. The synthetic peptides generated a sperm-specific antibody response in female and male mice that caused a contraceptive state. The immunocontraceptive effect was reversible and, with the disappearance of peptide-specific antibodies, there was complete restoration of fertility. Vaccinations using P82, Sa6 and Sa76 peptides resulted in no apparent side effects. Thus, it is efficient and practical to identify epitope peptide candidates by phage display. These peptides may find clinical application in the specific diagnosis and treatment of male and female infertility and contraceptive vaccine development.


Asunto(s)
Anticuerpos/inmunología , Moléculas de Adhesión Celular/administración & dosificación , Anticoncepción Inmunológica/métodos , Fertilidad/efectos de los fármacos , Hialuronoglucosaminidasa/administración & dosificación , Epítopos Inmunodominantes , Isoantígenos/administración & dosificación , Fragmentos de Péptidos/administración & dosificación , Proteínas de Plasma Seminal/administración & dosificación , Espermatozoides/inmunología , Vacunas de Subunidad/administración & dosificación , Adulto , Animales , Moléculas de Adhesión Celular/inmunología , Técnicas de Visualización de Superficie Celular , Mapeo Epitopo , Femenino , Humanos , Hialuronoglucosaminidasa/inmunología , Inmunización , Infertilidad Femenina/inmunología , Infertilidad Femenina/fisiopatología , Isoantígenos/inmunología , Masculino , Mesocricetus , Ratones Endogámicos BALB C , Fragmentos de Péptidos/inmunología , Biblioteca de Péptidos , Proteínas de Plasma Seminal/inmunología , Vacunas de Subunidad/inmunología , Adulto Joven
5.
Toxicol Mech Methods ; 26(1): 32-5, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26313688

RESUMEN

CONTEXT: Paraquat exposure commonly occurs in the developing countries and the mortality rate is high. However, there is currently no consensus on the efficacy of treatment for paraquat exposure. OBJECTIVE: The study was aimed to explore the effects of tumor necrosis factor-α (TNF-α) induced protein 6 (TSG-6) on acute lung injury (ALI) following paraquat exposure in rats. MATERIALS AND METHODS: Male Sprague-Dawley (SD) rats were randomly divided into the sham group (n = 8), the paraquat group (n = 8), and the paraquat TSG-6-treated group (n = 8). Rats were administered with 50 mg/kg of paraquat intraperitoneally. At 1 h after exposure, rats were treated with 30 µg of recombinant human TSG-6 (rhTSG-6) intraperitoneally. After 6 h of exposure, ALI scores were evaluated by histology and the expression of pro-inflammatory cytokines in lung was assayed using real-time RT-PCR. RESULTS: ALI scores were significantly lower in the paraquat TSG-6-treated group, compared with the paraquat group (p < 0.05). The expression of interleukin (IL)-1ß, IL-6, and TNF-α mRNA was significantly lower in the paraquat TSG-6-treated group, compared with the paraquat group (p < 0.01, respectively). DISCUSSION AND CONCLUSION: Administration of rhTSG-6 attenuates ALI following paraquat exposure by suppressing inflammatory response.


Asunto(s)
Lesión Pulmonar Aguda/inducido químicamente , Moléculas de Adhesión Celular/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Paraquat/toxicidad , Proteínas Recombinantes/farmacología , Lesión Pulmonar Aguda/tratamiento farmacológico , Animales , Moléculas de Adhesión Celular/administración & dosificación , Citocinas/genética , Citocinas/metabolismo , Pulmón/metabolismo , Masculino , Ratas , Ratas Sprague-Dawley , Proteínas Recombinantes/administración & dosificación
6.
Gut ; 62(1): 112-20, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22466618

RESUMEN

OBJECTIVE: Pancreatic ductal adenocarcinoma (PDA) is characterised by stromal desmoplasia and vascular dysfunction, which critically impair drug delivery. This study examines the role of an abundant extracellular matrix component, the megadalton glycosaminoglycan hyaluronan (HA), as a novel therapeutic target in PDA. METHODS: Using a genetically engineered mouse model of PDA, the authors enzymatically depleted HA by a clinically formulated PEGylated human recombinant PH20 hyaluronidase (PEGPH20) and examined tumour perfusion, vascular permeability and drug delivery. The preclinical utility of PEGPH20 in combination with gemcitabine was assessed by short-term and survival studies. RESULTS: PEGPH20 rapidly and sustainably depleted HA, inducing the re-expansion of PDA blood vessels and increasing the intratumoral delivery of two chemotherapeutic agents, doxorubicin and gemcitabine. Moreover, PEGPH20 triggered fenestrations and interendothelial junctional gaps in PDA tumour endothelia and promoted a tumour-specific increase in macromolecular permeability. Finally, combination therapy with PEGPH20 and gemcitabine led to inhibition of PDA tumour growth and prolonged survival over gemcitabine monotherapy, suggesting immediate clinical utility. CONCLUSIONS: The authors demonstrate that HA impedes the intratumoral vasculature in PDA and propose that its enzymatic depletion be explored as a means to improve drug delivery and response in patients with pancreatic cancer.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Biomarcadores de Tumor/fisiología , Carcinoma Ductal Pancreático/tratamiento farmacológico , Sistemas de Liberación de Medicamentos , Resistencia a Antineoplásicos/fisiología , Ácido Hialurónico/fisiología , Neoplasias Pancreáticas/tratamiento farmacológico , Animales , Antineoplásicos/administración & dosificación , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Carcinoma Ductal Pancreático/irrigación sanguínea , Carcinoma Ductal Pancreático/mortalidad , Carcinoma Ductal Pancreático/fisiopatología , Moléculas de Adhesión Celular/administración & dosificación , Moléculas de Adhesión Celular/farmacología , Desoxicitidina/administración & dosificación , Desoxicitidina/análogos & derivados , Doxorrubicina/administración & dosificación , Resistencia a Antineoplásicos/efectos de los fármacos , Hialuronoglucosaminidasa/administración & dosificación , Hialuronoglucosaminidasa/farmacología , Inmunohistoquímica , Estimación de Kaplan-Meier , Ratones , Ratones Transgénicos , Neoplasias Pancreáticas/irrigación sanguínea , Neoplasias Pancreáticas/mortalidad , Neoplasias Pancreáticas/fisiopatología , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/farmacología , Análisis de Matrices Tisulares , Resultado del Tratamiento , Gemcitabina
7.
Neurobiol Dis ; 59: 86-99, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23851308

RESUMEN

Traumatic brain injury (TBI) causes multiple long-term defects including a loss of working memory that is frequently incapacitating. Administrations of mesenchymal stem/stromal cells (MSCs) previously produced beneficial effects in models of TBI as well as other disease models. In several models, the beneficial effects were explained by the MSCs being activated to express TSG-6, a multifunctional protein that modulates inflammation. In a mouse model of TBI, we found the initial mild phase of the inflammatory response persisted for at least 24h and was followed by secondary severe response that peaked at 3days. Intravenous human MSCs or TSG-6 during initial mild phase decreased neutrophil extravasation, expression of matrix metalloproteinase 9 by endothelial cells and neutrophils, and the subsequent blood brain barrier leakage in secondary phase. Administration of TSG-6 also decreased the lesion size at 2weeks. Importantly, the acute administration of TSG-6 within 24h of TBI was followed 6 to 10weeks later by improvements in memory, depressive-like behavior and the number of newly born-neurons. The data suggested that acute administration of TSG-6 may be an effective therapy for decreasing some of the long-term consequences of TBI.


Asunto(s)
Lesiones Encefálicas/complicaciones , Moléculas de Adhesión Celular/administración & dosificación , Trastornos de la Memoria/tratamiento farmacológico , Trastornos de la Memoria/etiología , Animales , Lesiones Encefálicas/terapia , Células Cultivadas , Modelos Animales de Enfermedad , Proteínas de Dominio Doblecortina , Proteína Doblecortina , Conducta Exploratoria/efectos de los fármacos , Conducta Alimentaria/efectos de los fármacos , Conducta Alimentaria/fisiología , Humanos , Masculino , Metaloproteinasa 9 de la Matriz/metabolismo , Aprendizaje por Laberinto/efectos de los fármacos , Aprendizaje por Laberinto/fisiología , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/fisiología , Ratones , Ratones Endogámicos C57BL , Proteínas Asociadas a Microtúbulos/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Neuropéptidos/metabolismo , Natación/psicología , Factor de von Willebrand/metabolismo
8.
Acta Pharmacol Sin ; 34(6): 784-92, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23474707

RESUMEN

AIM: To investigate the effects of mesenchymal stem cells (MSCs) transplantation on rat global cerebral ischemia and the underlying mechanisms. METHODS: Adult male SD rats underwent asphxial cardiac arrest to induce global cerebral ischemia, then received intravenous injection of 5×10(6) cultured MSCs of SD rats at 2 h after resuscitation. In another group of cardiac arrest rats, tumor necrosis factor-α-induced protein 6 (TSG-6, 6 µg) was injected into the right lateral ventricle. Functional outcome was assessed at 1, 3, and 7 d after resuscitation. Donor MSCs in the brains were detected at 3 d after resuscitation. The level of serum S-100B and proinflammatory cytokines in cerebral cortex were assayed using ELISA. The expression of TSG-6 and proinflammatory cytokines in cerebral cortex was assayed using RT-PCR. Western blot was performed to determine the levels of TSG-6 and neutrophil elastase in cerebral cortex. RESULTS: MSCs transplantation significantly reduced serum S-100B level, and improved neurological function after global cerebral ischemia compared to the PBS-treated group. The MSCs injected migrated into the ischemic brains, and were observed mainly in the cerebral cortex. Furthermore, MSCs transplantation significantly increased the expression of TSG-6, and reduced the expression of neutrophil elastase and proinflammatory cytokines in the cerebral cortex. Intracerebroventricular injection of TSG-6 reproduced the beneficial effects of MSCs transplantation in rats with global cerebral ischemia. CONCLUSION: MSCs transplantation improves functional recovery and reduces inflammatory responses in rats with global cerebral ischemia, maybe via upregulation of TSG-6 expression.


Asunto(s)
Isquemia Encefálica/terapia , Moléculas de Adhesión Celular/genética , Inflamación/terapia , Trasplante de Células Madre Mesenquimatosas/métodos , Animales , Western Blotting , Isquemia Encefálica/fisiopatología , Moléculas de Adhesión Celular/administración & dosificación , Moléculas de Adhesión Celular/metabolismo , Corteza Cerebral/metabolismo , Corteza Cerebral/patología , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática , Inflamación/patología , Mediadores de Inflamación/metabolismo , Inyecciones Intravenosas , Elastasa de Leucocito/genética , Elastasa de Leucocito/metabolismo , Masculino , Ratas , Ratas Sprague-Dawley , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Subunidad beta de la Proteína de Unión al Calcio S100/sangre , Factores de Tiempo , Resultado del Tratamiento , Regulación hacia Arriba
9.
Mol Ther ; 20(1): 14-20, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22008910

RESUMEN

Recent observations have demonstrated that one of the functions of mesenchymal stem/stromal cells (MSCs) is to serve as guardians against excessive inflammatory responses. One mode of action of the cells is that they are activated to express the interleukin (IL)-1 receptor antagonist. A second mode of action is to create a negative feedback loop in which tumor necrosis factor-α (TNF-α) and other proinflammatory cytokines from resident macrophages activate MSCs to secrete the multifunctional anti-inflammatory protein TNF-α stimulated gene/protein 6 (TSG-6). The TSG-6 then reduces nuclear factor-κB (NF-κB) signaling in the resident macrophages and thereby modulates the cascade of proinflammatory cytokines. A third mode of action is to create a second negative feedback loop whereby lipopolysaccharide, TNF-α, nitric oxide, and perhaps other damage-associated molecular patterns (DAMPs) from injured tissues and macrophages activate MSCs to secrete prostaglandin E(2) (PGE(2)). The PGE(2) converts macrophages to the phenotype that secretes IL-10. There are also suggestions that MSCs may produce anti-inflammatory effects through additional modes of action including activation to express the antireactive oxygen species protein stanniocalcin-1.


Asunto(s)
Inflamación/metabolismo , Células Madre Mesenquimatosas/metabolismo , Animales , Antiinflamatorios/metabolismo , Moléculas de Adhesión Celular/administración & dosificación , Moléculas de Adhesión Celular/genética , Moléculas de Adhesión Celular/metabolismo , Modelos Animales de Enfermedad , Humanos , Inflamación/inmunología , Inflamación/terapia , Activación de Macrófagos/inmunología , Macrófagos/metabolismo , Trasplante de Células Madre Mesenquimatosas , Ratones , Investigación con Células Madre
10.
Mol Ther ; 20(11): 2143-52, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22929658

RESUMEN

Mesenchymal stem/progenitor cells (MSCs) were reported to enhance the survival of cellular and organ transplants. However, their mode of action was not established. We here used a mouse model of corneal allotransplantation and demonstrated that peri-transplant intravenous (i.v.) infusion of human MSCs (hMSCs) decreased the early surgically induced inflammation and reduced the activation of antigen-presenting cells (APCs) in the cornea and draining lymph nodes (DLNs). Subsequently, immune rejection was decreased, and allograft survival was prolonged. Quantitative assays for human GAPDH revealed that <10 hMSCs out of 1 × 10(6) injected cells were recovered in the cornea 10 hours to 28 days after i.v. infusion. Most of hMSCs were trapped in lungs where they were activated to increase expression of the gene for a multifunctional anti-inflammatory protein tumor necrosis factor-α stimulated gene/protein 6 (TSG-6). i.v. hMSCs with a knockdown of TSG-6 did not suppress the early inflammation and failed to prolong the allograft survival. Also, i.v. infusion of recombinant TSG-6 reproduced the effects of hMSCs. Results suggest that hMSCs improve the survival of corneal allografts without engraftment and primarily by secreting TSG-6 that acts by aborting early inflammatory responses. The same mechanism may explain previous reports that MSCs decrease rejection of other organ transplants.


Asunto(s)
Trasplante de Córnea/métodos , Rechazo de Injerto/prevención & control , Trasplante de Células Madre Mesenquimatosas , Administración Intravenosa , Animales , Antiinflamatorios/administración & dosificación , Células Presentadoras de Antígenos/inmunología , Células Presentadoras de Antígenos/fisiología , Moléculas de Adhesión Celular/administración & dosificación , Moléculas de Adhesión Celular/genética , Moléculas de Adhesión Celular/metabolismo , Células Cultivadas , Córnea/inmunología , Córnea/metabolismo , Córnea/patología , Femenino , Técnicas de Silenciamiento del Gen , Rechazo de Injerto/inmunología , Antígenos de Histocompatibilidad Clase II/metabolismo , Humanos , Mediadores de Inflamación/metabolismo , Pulmón/inmunología , Pulmón/patología , Ganglios Linfáticos/inmunología , Ganglios Linfáticos/patología , Macrófagos/inmunología , Macrófagos/metabolismo , Células Madre Mesenquimatosas/metabolismo , Ratones , Ratones Endogámicos C57BL , FN-kappa B/metabolismo , ARN Interferente Pequeño/genética , Transducción de Señal , Transcriptoma , Trasplante Homólogo
11.
Ann Oncol ; 23(9): 2306-2313, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22357251

RESUMEN

BACKGROUND: Targeted therapy options in HER2-negative breast cancer are limited. This open-label, multicenter phase IB dose-escalation trial was conducted to determine safety, tolerability, and antitumor activity of a combination of docetaxel (Taxotere) and increasing doses of adecatumumab, a human IgG1 antibody targeting epithelial cell adhesion molecule (EpCAM), in EpCAM-positive relapsed or primary refractory advanced-stage breast cancer. PATIENTS AND METHODS: Patients pretreated with up to four prior chemotherapy regimens received increasing adecatumumab doses either every 3 weeks (q3w) or weekly (qw) combined with docetaxel (100 mg/m(2) q3w). Primary end points were safety and tolerability. Antitumor activity was evaluated according to RECIST. Clinical benefit was defined as complete or partial response or stable disease for ≥24 weeks. RESULTS: Thirty-one evaluable patients were treated. Most adverse events were mild to moderate in severity. Neutropenia, leukocytopenia, lymphopenia, and diarrhea (dose-limiting) were the most frequent toxic effects. Maximum tolerated doses of adecatumumab given in combination with docetaxel were 550 mg/m(2) q3w and 360 mg/m(2) qw. Clinical benefit was observed in 44% of patients treated with q3w adecatumumab and docetaxel, increasing to 63% in patients with high EpCAM-expressing tumors. CONCLUSION: Combination therapy of adecatumumab and docetaxel is safe, feasible, and potentially active in heavily pretreated advanced-stage breast cancer.


Asunto(s)
Antígenos de Neoplasias/metabolismo , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Neoplasias de la Mama/tratamiento farmacológico , Moléculas de Adhesión Celular/metabolismo , Resistencia a Antineoplásicos , Neoplasias Hepáticas/tratamiento farmacológico , Recurrencia Local de Neoplasia/tratamiento farmacológico , Adulto , Anciano , Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Monoclonales Humanizados , Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Protocolos de Quimioterapia Combinada Antineoplásica/farmacocinética , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Moléculas de Adhesión Celular/administración & dosificación , Docetaxel , Esquema de Medicación , Molécula de Adhesión Celular Epitelial , Femenino , Humanos , Trastornos Leucocíticos/inducido químicamente , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/secundario , Persona de Mediana Edad , Recurrencia Local de Neoplasia/metabolismo , Recurrencia Local de Neoplasia/patología , Taxoides/administración & dosificación , Resultado del Tratamiento
12.
J Neuroinflammation ; 9: 59, 2012 Mar 27.
Artículo en Inglés | MEDLINE | ID: mdl-22452753

RESUMEN

BACKGROUND: Inflammatory responses are detected in the retina of patients with age-related macular degeneration and Ccl2-/-/Cx3cr1-/- mice on rd8 background,(Ccl2-/-/Cx3cr1-/- mice) a model that develops progressive age-related macular degeneration-like retinal lesions including focal photoreceptor degeneration, abnormal retinal pigment epithelium and A2E accumulation. Tumor necrosis factor-inducible gene 6 protein is an anti-inflammatory protein and has been shown to improve myocardial infarction outcome and chemically injured cornea in mice by suppressing inflammation. In this study, we evaluated the effect of an intravitreous injection of recombinant TSG-6 on the retinal lesions of Ccl2-/-/Cx3cr1-/- mice. METHODS: Recombinant TSG-6 (400 ng) was administered by intravitreous injection into the right eye of six-week-old Ccl2-/-/Cx3cr1-/- mice. Their left eye was injected with phosphate-buffered saline as a control. Funduscopic pictures were taken before injection and sequentially once a month after injection. The mice were killed two months after injection and the ocular histology examined. Retinal A2E, a major component of lipofuscin, was measured by high performance liquid chromatography. The microarray of ocular mRNA of 92 immunological genes was performed. The genes showing differentiated expression in microarray were further compared between the injected right eye and the contralateral (control) eye by [real-time quantitative reverse transcription polymerase chain reaction] qRT-PCR. RESULTS: The continuous monitoring of the fundus for two months showed a slower progression or alleviation of retinal lesions in the treated right eyes as compared with the untreated left eyes. Among 23 pairs of eyes, the lesion levels improved in 78.3%, stayed the same in 8.7% and progressed in 13.0%. Histology confirmed the clinical observation. Even though there was no difference in the level of A2E between the treated and the untreated eyes, microarray analysis of 92 immune genes showed that IL-17a was substantially decreased after the treatment. Expression of TNF-α showed a similar pattern to IL-17a. The results were consistent in duplicated arrays and confirmed by qRT-PCR. CONCLUSIONS: We concluded that intravitreous administration of recombinant TSG-6 might stabilize retinal lesions in Ccl2-/-/Cx3cr1-/- mice on rd8 background. Modulation of ocular immunological gene expressions, especially IL-17a, could be one of the mechanisms.


Asunto(s)
Antiinflamatorios/administración & dosificación , Moléculas de Adhesión Celular/administración & dosificación , Degeneración Retiniana/tratamiento farmacológico , Animales , Receptor 1 de Quimiocinas CX3C , Quimiocina CCL2/deficiencia , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/genética , Interleucina-17/metabolismo , Inyecciones Intravítreas , Lipofuscina/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microscopía Electrónica de Transmisión , Microscopía Inmunoelectrónica , Oftalmoscopios , Receptores de Quimiocina/deficiencia , Retina/metabolismo , Retina/patología , Retina/ultraestructura , Degeneración Retiniana/patología , Factor de Necrosis Tumoral alfa/metabolismo
13.
Tissue Eng Regen Med ; 19(4): 839-852, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35199306

RESUMEN

BACKGROUND: In the aftermath of bone injuries, such as cranium and sternum, bone wax (BW) is used to control bleeding from the bone surfaces during surgery. Made up of artificial substances, however, it is associated with many complications such as inflammation, increased risk for infection, and bone repair delay. We, therefore, in this study set out to design and evaluate a novel BW without the above-mentioned side-effects reported for other therapies. METHODS: The pastes (new BW(s)) were prepared in the laboratory and examined by MTT, MIC, MBC, and degradability tests. Then, 60 adult male Wistar rats, divided into six equal groups including chitosan (CT), CT-octacalcium phosphate (OCP), CT-periostin (Post), CT-OCP-Post, Control (Ctrl), and BW, underwent sternotomy surgery. Once the surgeries were completed, the bone repair was assessed radiologically and thereafter clinically in vivo and in vitro using CT-scan, H&E, ELISA, and qRT-PCR. RESULTS: All pastes displayed antibacterial properties and the CT-Post group had the highest cell viability compared to the control group. In contrast to the BW, CT-Post group demonstrated weight changes in the degradability test. In the CT-Post group, more number of osteocyte cells, high trabeculae percentage, and the least fibrous connective tissue were observed compared to other groups. Additionally, in comparison to the CT and Ctrl groups, higher alkaline phosphatase activity, as well as decreased level of serum tumor necrosis factor-α, interleukin-6, and OCN in the CT-Post group was evident. Finally, Runx2, OPG, and RANKL genes' expression was significantly higher in the CT-Post group than in other groups. CONCLUSION: Our results provide insights into the desirability of pastes in terms of cellular viability, degradability, antibacterial properties, and surgical site restoration compared to the BW group. Besides, Periostin could enhance the osteogenic properties of bone tissue defect site.


Asunto(s)
Materiales Biocompatibles , Moléculas de Adhesión Celular , Quitosano , Esternón , Animales , Antibacterianos , Moléculas de Adhesión Celular/administración & dosificación , Quitosano/farmacología , Interleucina-6/sangre , Masculino , Ratas , Ratas Wistar , Esternotomía , Esternón/cirugía , Andamios del Tejido , Factor de Necrosis Tumoral alfa/sangre
14.
J Reprod Immunol ; 145: 103325, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33930667

RESUMEN

Contraceptive vaccine (CV) is a valuable, non-invasive, and alternative method for purposeful contraception. Sperm antigens are useful targets for producing CVs due to their specialized expression in sperm. In this study, a recombinant protein containing three main sperm epitopes (IZUMO1, SACA3, and PH-20) was designed and evaluated as CV to control fertility in male mice. The chimeric recombinant protein was expressed and purified in E. coli. Male mice were immunized by 100 µg purified protein and sera were collected to assess IgG antibodies. Evaluating the reproductive performance, immunized male mice mated with normal-fertile female mice and mating rate and the number of newborns was studied. Immunized mice were sacrificed and necropsy and histopathology studies were conducted. The results revealed that the designed chimeric protein stimulated the immune system of the mice effectively. The level of IgG antibody was significantly higher in vaccinated mouse rather than control mouse. Eighty percent of the vaccinated mice became infertile and in the remaining ones, the number of children decreased to 4-6 offspring instead of 10-12 in normal mice. Histopathological studies showed that no organs including heart, brain, lung, liver, kidney and intestine were damaged. However, Normal spermatogenesis has been disrupted and necrotic spermatogonia cells were reported in Seminiferous tubules. We concluded that the designed chimeric protein containing IZUMO1, SACA3, and PH-20 epitopes can stimulate the immune system and cause male contraception without any side effects.


Asunto(s)
Anticoncepción Inmunológica/métodos , Infertilidad Masculina/inmunología , Proteínas Recombinantes de Fusión/inmunología , Vacunas Anticonceptivas/inmunología , Animales , Moléculas de Adhesión Celular/administración & dosificación , Moléculas de Adhesión Celular/genética , Moléculas de Adhesión Celular/inmunología , Modelos Animales de Enfermedad , Epítopos/administración & dosificación , Epítopos/genética , Epítopos/inmunología , Humanos , Hialuronoglucosaminidasa/administración & dosificación , Hialuronoglucosaminidasa/genética , Hialuronoglucosaminidasa/inmunología , Inmunoglobulinas/administración & dosificación , Inmunoglobulinas/genética , Inmunoglobulinas/inmunología , Infertilidad Masculina/patología , Isoantígenos/administración & dosificación , Isoantígenos/genética , Isoantígenos/inmunología , Masculino , Proteínas de la Membrana/administración & dosificación , Proteínas de la Membrana/genética , Proteínas de la Membrana/inmunología , Ratones , Proteínas Recombinantes de Fusión/administración & dosificación , Proteínas Recombinantes de Fusión/genética , Proteínas de Plasma Seminal/administración & dosificación , Proteínas de Plasma Seminal/genética , Proteínas de Plasma Seminal/inmunología , Túbulos Seminíferos/citología , Túbulos Seminíferos/inmunología , Túbulos Seminíferos/patología , Espermatogonias/inmunología , Espermatogonias/patología , Vacunas Anticonceptivas/administración & dosificación , Vacunas Anticonceptivas/genética
15.
Ann Oncol ; 21(2): 275-282, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19633042

RESUMEN

BACKGROUND: High-level expression of epithelial cell adhesion molecule (EpCAM) is associated with unfavorable prognosis in breast cancer. This study was designed to investigate two doses of the fully human IgG1 anti-EpCAM antibody adecatumumab (MT201) in patients with metastatic breast cancer (MBC). METHODS: A total of 109 patients were stratified into high- and low-level EpCAM expression by immunohistochemical staining of primary tumors and subsequently randomly assigned to receive monotherapy with either high- (6 mg/kg every two weeks (q2w)) or low-dose adecatumumab (2 mg/kg/ q2w) until disease progression. RESULTS: No complete or partial tumor responses could be confirmed by central RECIST assessment. The probability for tumor progression was significantly lower in patients receiving high-dose adecatumumab and expressing high levels of EpCAM (hazard ratio 0.43; P = 0.0057 versus low dose and low EpCAM). Three of 18 patients with highest EpCAM expression treated with adecatumumab developed new metastases up to week 6, compared with 14 of 29 patients with low EpCAM. Most frequent treatment-related adverse events (high dose/low dose) were chills (59%/20%), nausea (55%/18%), fatigue (39%/23%) and diarrhea (43%/7%). CONCLUSIONS: Single-agent adecatumumab shows dose- and target-dependent clinical activity in EpCAM-positive MBC, albeit no objective tumor regression. Further investigation of adecatumumab in patients with EpCAM-overexpressing tumors and lower tumor burden is warranted.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Antígenos de Neoplasias/inmunología , Neoplasias de la Mama/tratamiento farmacológico , Moléculas de Adhesión Celular/inmunología , Moléculas de Adhesión Celular/uso terapéutico , Adulto , Anciano , Anciano de 80 o más Años , Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Monoclonales/efectos adversos , Anticuerpos Monoclonales Humanizados , Antineoplásicos/administración & dosificación , Antineoplásicos/efectos adversos , Antineoplásicos/uso terapéutico , Neoplasias de la Mama/patología , Moléculas de Adhesión Celular/administración & dosificación , Moléculas de Adhesión Celular/efectos adversos , Progresión de la Enfermedad , Supervivencia sin Enfermedad , Relación Dosis-Respuesta a Droga , Molécula de Adhesión Celular Epitelial , Femenino , Humanos , Persona de Mediana Edad , Metástasis de la Neoplasia , Resultado del Tratamiento
16.
Theranostics ; 10(1): 36-49, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-31903104

RESUMEN

Mesenchymal stem cells (MSCs) transplantation is a promising antifibrotic strategy but facing clinical controversies. Inspired by advances in nanomedicine, we aimed to bypass these clinical barriers of MSCs by identifying the key antifibrotic molecule of MSCs and developing a specific liver-targeting nanocarrier. Methods: Cytokines secreted by MSCs were examined with serum stimulation of cirrhotic patients. Immunohistochemistry, microarray, immunoblotting, and quantitative real-time PCR (qRT-PCR) were applied to identify the critical antifibrotic cytokine and to discover its role in modulating antifibrotic effects. Biomineralization method was used to prepare calcium phosphate nanoparticles (NPs). The targeting and therapeutic efficiency of NPs were evaluated by in vivo imaging and biochemical studies on fibrotic mice induced by CCl4. Results: The stimulated MSCs exhibited high-level expression of Tumor necrosis factor (TNF)-stimulated gene 6 (TSG-6). On animal study, exogenous administration of TSG-6 alone can ameliorate liver fibrosis while TSG-6 knocked MSCs (Lv-TSG-6 MSCs) lost antifibrotic effects. Further studies verified the importance of TSG-6 and identified its antifibrotic mechanism by modulating M2 macrophages and increasing matrix metalloproteinase 12 (MMP12) expression. Additionally, we found a feedback loop between TSG-6, MMP12 and pro-inflammatory cytokines (TNF-α, IL-6, and IL-1ß), which may improve our understanding of the aggravating process of cirrhosis and antifibrotic mechanisms of TSG-6 and MSCs. Based on these findings, we developed calcium phosphate nanoparticles (CaP@BSA NPs) by biomineralization method using bovine serum albumin (BSA) as the biotemplate. Imaging tracking and drug loading studies showed specific liver targeting and high TSG-6 loading efficacy of as-prepared CaP@BSA NPs. In vivo therapeutic study further demonstrated the improved therapeutic effects of TSG-6 loaded CaP@BSA. Conclusions: TSG-6 was a major antifibrotic cytokine of MSCs, TSG-6 loaded CaP@BSA NPs showed specific liver accumulation and improved therapeutic effects, which indicated translational potentials of CaP@BSA as a promising drug carrier for the liver disease management.


Asunto(s)
Moléculas de Adhesión Celular , Portadores de Fármacos/química , Cirrosis Hepática/tratamiento farmacológico , Nanopartículas/química , Animales , Fosfatos de Calcio/química , Moléculas de Adhesión Celular/administración & dosificación , Moléculas de Adhesión Celular/farmacología , Citocinas/metabolismo , Humanos , Hígado/efectos de los fármacos , Hígado/patología , Macrófagos/metabolismo , Masculino , Metaloproteinasa 12 de la Matriz/metabolismo , Ratones , Ratones Endogámicos C57BL , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/farmacología , Albúmina Sérica Bovina/química
17.
Inflammation ; 43(2): 455-465, 2020 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-31840212

RESUMEN

Although the up-regulation of periostin in osteoarthritic (OA) is found, its function on OA condyle caused by disc displacement is not clear. Our objective was to explore whether periostin has any effect on condylar resorption. We initially identified periostin-positive cells in temporomandibular joint osteoarthritic (TMJ-OA) cartilage. Furthermore, the vitro analysis confirmed that the expression of periostin in chondrocytes treated with a static pressure of 150 kpa and 200 kpa for 3 h by an in-house-designed pressure chamber. To explore the underlying mechanism, we found that periostin can induce IκBα phosphorylation and its subsequent degradation, leading to consequent p65 nuclear translocation and subsequent induction of ADAMTS5 expression, which is known to be detrimental to cartilage extracellular matrix production. Importantly, inhibiting NF-κB signaling, by BAY 11-7082 treatment, rescued periostin-induced ADAMTS5 up-regulation. This study elucidated the direct role of periostin in condylar resorption, which was found to occur via NF-κB-ADAMTS5 signaling. Inhibition of this pathway might provide a new strategy for TMJ-OA treatment.


Asunto(s)
Proteína ADAMTS5/biosíntesis , Resorción Ósea/inducido químicamente , Resorción Ósea/metabolismo , Moléculas de Adhesión Celular/administración & dosificación , Cóndilo Mandibular/metabolismo , FN-kappa B/biosíntesis , Resorción Ósea/diagnóstico por imagen , Cartílago Articular/efectos de los fármacos , Cartílago Articular/metabolismo , Moléculas de Adhesión Celular/toxicidad , Células Cultivadas , Condrocitos/efectos de los fármacos , Condrocitos/metabolismo , Relación Dosis-Respuesta a Droga , Humanos , Cóndilo Mandibular/diagnóstico por imagen , Cóndilo Mandibular/efectos de los fármacos , FN-kappa B/antagonistas & inhibidores , Nitrilos/farmacología , Presión , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Sulfonas/farmacología , Articulación Temporomandibular/diagnóstico por imagen , Articulación Temporomandibular/efectos de los fármacos , Articulación Temporomandibular/metabolismo
18.
Curr Eye Res ; 44(8): 873-881, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-30935217

RESUMEN

Purpose: To investigate the therapeutic efficacy of tumor necrosis factor (TNF)-α stimulated gene/protein 6 (TSG-6) in a rat model of corneal alkali injury. Methods: Corneal alkali injury was produced by placing an NaOH-soaked filter paper disk on the central cornea of the right eye of an anesthetized male Lewis (LEW/Crl) rat. Recombinant human TSG-6, or an equal volume of phosphate-buffered saline (PBS), was administered intravenously (IV), by anterior chamber (AC) injection, or as a topical drop. The affected eyes were photographed daily using a dissecting microscope and documented for clinical time course analysis of corneal opacification. Corneal tissue was excised at pre-determined therapeutic endpoints, with subsequent qRT-PCR or histological analyses. Results: The continuous monitoring of corneal alkali injury progression revealed TSG-6 treatments do not show sufficient effectiveness in vivo regardless of IV injection, AC injection, or topical application. Corneal opacification and neovascularization were not diminished, and gene expression was not impacted by these treatments. However, both IV and AC administration of TSG-6 significantly suppressed pro-inflammatory cytokines compared to PBS-treated eyes. Conclusion: We conclude that the therapeutic potential of TSG-6 is insufficient in a rat corneal alkali injury model.


Asunto(s)
Quemaduras Químicas/tratamiento farmacológico , Moléculas de Adhesión Celular/uso terapéutico , Enfermedades de la Córnea/tratamiento farmacológico , Modelos Animales de Enfermedad , Quemaduras Oculares/inducido químicamente , Administración Oftálmica , Animales , Cámara Anterior/efectos de los fármacos , Quemaduras Químicas/etiología , Quemaduras Químicas/patología , Moléculas de Adhesión Celular/administración & dosificación , Enfermedades de la Córnea/inducido químicamente , Enfermedades de la Córnea/patología , Infusiones Intravenosas , Inyecciones Intraoculares , Masculino , Ratas , Ratas Endogámicas Lew , Reacción en Cadena en Tiempo Real de la Polimerasa , Hidróxido de Sodio
19.
Drug Deliv ; 26(1): 98-106, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-30744432

RESUMEN

ENHANZE® drug delivery technology is based on the proprietary recombinant human hyaluronidase PH20 enzyme (rHuPH20; Halozyme Therapeutics, Inc.) that facilitates the subcutaneous (SC) delivery of co-administered therapeutics. rHuPH20 works by degrading the glycosaminoglycan hyaluronan (HA), which plays a role in resistance to bulk fluid flow in the SC space, limiting large volume SC drug delivery, dispersion, and absorption. Co-administration of rHuPH20 with partner therapies can overcome administration time and volume barriers associated with existing SC therapeutic formulations, and has been shown to reduce the burden on patients and healthcare providers compared with intravenous formulations. rHuPH20 (as HYLENEX® recombinant) is currently FDA-approved for subcutaneous fluid administration for achieving hydration, to increase the dispersion and absorption of other injected drugs, and in subcutaneous urography for improving resorption of radiopaque agents. rHuPH20 is also co-formulated with two anticancer therapies, trastuzumab (i.e. Herceptin® SC) and rituximab (i.e. RITUXAN HYCELA®/RITUXAN® SC/MabThera® SC) and dosed sequentially with human immunoglobin to treat primary immunodeficiency (i.e. HyQvia®/HYQVIA®). This article reviews pharmaceutical properties of rHuPH20, its current applications with approved therapeutics, and the potential for future developments.


Asunto(s)
Antineoplásicos Inmunológicos/administración & dosificación , Moléculas de Adhesión Celular/administración & dosificación , Sistemas de Liberación de Medicamentos/métodos , Hialuronoglucosaminidasa/administración & dosificación , Inmunoglobulinas/administración & dosificación , Animales , Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Monoclonales/metabolismo , Antígenos de Superficie/administración & dosificación , Antígenos de Superficie/metabolismo , Antineoplásicos Inmunológicos/metabolismo , Moléculas de Adhesión Celular/metabolismo , Sistemas de Liberación de Medicamentos/tendencias , Quimioterapia Combinada , Humanos , Hialuronoglucosaminidasa/metabolismo , Inmunoglobulinas/metabolismo , Inyecciones Subcutáneas , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA