Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 133
Filtrar
1.
Int J Mol Sci ; 24(12)2023 Jun 18.
Artículo en Inglés | MEDLINE | ID: mdl-37373457

RESUMEN

Epithelial ovarian cancer (EOC) is the most lethal gynecological malignancy owing to relapse caused by resistance to chemotherapy. We previously reported that cluster of differentiation 109 (CD109) expression is positively correlated with poor prognosis and chemoresistance in patients with EOC. To further explore the role of CD109 in EOC, we explored the signaling mechanism of CD109-induced drug resistance. We found that CD109 expression was upregulated in doxorubicin-resistant EOC cells (A2780-R) compared with that in their parental cells. In EOC cells (A2780 and A2780-R), the expression level of CD109 was positively correlated with the expression level of ATP-binding cassette (ABC) transporters, such as ABCB1 and ABCG2, and paclitaxel (PTX) resistance. Using a xenograft mouse model, it was confirmed that PTX administration in xenografts of CD109-silenced A2780-R cells significantly attenuated in vivo tumor growth. The treatment of CD109-overexpressed A2780 cells with cryptotanshinone (CPT), a signal transducer and activator of transcription 3 (STAT3) inhibitor, inhibited the CD109 overexpression-induced activation of STAT3 and neurogenic locus notch homolog protein 1 (NOTCH1), suggesting a STAT3-NOTCH1 signaling axis. The combined treatment of CD109-overexpressed A2780 cells with CPT and N-[N-(3,5-difluorophenacetyl)-l-alanyl]-S-phenylglycine t-butyl ester (DAPT), a NOTCH inhibitor, markedly abrogated PTX resistance. These results suggest that CD109 plays a key role in the acquisition of drug resistance by activating the STAT3-NOTCH1 signaling axis in patients with EOC.


Asunto(s)
Neoplasias Ováricas , Humanos , Femenino , Animales , Ratones , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Ováricas/genética , Neoplasias Ováricas/metabolismo , Factor de Transcripción STAT3/metabolismo , Línea Celular Tumoral , Recurrencia Local de Neoplasia , Paclitaxel/farmacología , Carcinoma Epitelial de Ovario/tratamiento farmacológico , Resistencia a Antineoplásicos , Transportadoras de Casetes de Unión a ATP , Proteínas de Neoplasias/metabolismo , Antígenos CD/uso terapéutico , Proteínas Ligadas a GPI/metabolismo , Receptor Notch1/genética , Receptor Notch1/metabolismo
2.
Expert Rev Mol Med ; 25: e16, 2023 05 03.
Artículo en Inglés | MEDLINE | ID: mdl-37132370

RESUMEN

This review discusses current research on acute paediatric leukaemia, the leukaemic bone marrow (BM) microenvironment and recently discovered therapeutic opportunities to target leukaemia-niche interactions. The tumour microenvironment plays an integral role in conferring treatment resistance to leukaemia cells, this poses as a key clinical challenge that hinders management of this disease. Here we focus on the role of the cell adhesion molecule N-cadherin (CDH2) within the malignant BM microenvironment and associated signalling pathways that may bear promise as therapeutic targets. Additionally, we discuss microenvironment-driven treatment resistance and relapse, and elaborate the role of CDH2-mediated cancer cell protection from chemotherapy. Finally, we review emerging therapeutic approaches that directly target CDH2-mediated adhesive interactions between the BM cells and leukaemia cells.


Asunto(s)
Médula Ósea , Leucemia Mieloide Aguda , Niño , Humanos , Médula Ósea/metabolismo , Médula Ósea/patología , Cadherinas/genética , Cadherinas/metabolismo , Cadherinas/uso terapéutico , Recurrencia Local de Neoplasia/metabolismo , Recurrencia Local de Neoplasia/patología , Leucemia Mieloide Aguda/terapia , Leucemia Mieloide Aguda/tratamiento farmacológico , Adhesión Celular , Microambiente Tumoral , Antígenos CD/metabolismo , Antígenos CD/uso terapéutico
3.
ACS Nano ; 17(3): 2101-2113, 2023 02 14.
Artículo en Inglés | MEDLINE | ID: mdl-36479877

RESUMEN

Intracellular delivery and genetic modification have brought a significant revolutionary to tumor immunotherapy, yet existing methods are still limited by low delivery efficiency, poor throughput, excessive cell damage, or unsuitability for suspension immune cells, specifically the natural killer cell, which is highly resistant to transfection. Here, we proposed a vibration-assisted nanoneedle/microfluidic composite system that uses large-area nanoneedles to rapidly puncture and detach the fast-moving suspension cells in the microchannel under vibration to achieve continuous high-throughput intracellular delivery. The nanoneedle arrays fabricated based on the large-area self-assembly technique and microchannels can maximize the delivery efficiency. Cas9 ribonucleoprotein complexes (Cas9/RNPs) can be delivered directly into cells due to the sufficient cellular membrane nanoperforation size; for difficult-to-transfect immune cells, the delivery efficiency can be up to 98%, while the cell viability remains at about 80%. Moreover, the throughput is demonstrated to maintain a mL/min level, which is significantly higher than that of conventional delivery techniques. Further, we prepared CD96 knockout NK-92 cells via this platform, and the gene-edited NK-92 cells possessed higher immunity by reversing exhaustion. The high-throughput, high-efficiency, and low-damage performance of our intracellular delivery strategy has great potential for cellular immunotherapy in clinical applications.


Asunto(s)
Edición Génica , Microfluídica , Supervivencia Celular , Edición Génica/métodos , Transfección , Vibración , Inmunoterapia/métodos , Humanos , Antígenos CD/genética , Antígenos CD/uso terapéutico , Ribonucleoproteínas/genética , Ribonucleoproteínas/uso terapéutico , Tratamiento Basado en Trasplante de Células y Tejidos/métodos
4.
Int J Mol Sci ; 22(2)2021 Jan 18.
Artículo en Inglés | MEDLINE | ID: mdl-33477692

RESUMEN

HIV-1 infection rapidly leads to a loss of the proliferative response of memory CD4+ T lymphocytes, when cultured with recall antigens. We report here that CD73 expression defines a subset of resting memory CD4+ T cells in peripheral blood, which highly express the α-chain of the IL-7 receptor (CD127), but not CD38 or Ki-67, yet are highly proliferative in response to mitogen and recall antigens, and to IL-7, in vitro. These cells also preferentially express CCR5 and produce IL-2. We reasoned that CD73+ memory CD4+ T cells decrease very early in HIV-1 infection. Indeed, CD73+ memory CD4+ T cells comprised a median of 7.5% (interquartile range: 4.5-10.4%) of CD4+ T cells in peripheral blood from healthy adults, but were decreased in primary HIV-1 infection to a median of 3.7% (IQR: 2.6-6.4%; p = 0.002); and in chronic HIV-1 infection to 1.9% (IQR: 1.1-3%; p < 0.0001), and were not restored by antiretroviral therapy. Moreover, we found that a significant proportion of CD73+ memory CD4+ T cells were skewed to a gut-homing phenotype, expressing integrins α4 and ß7, CXCR3, CCR6, CD161 and CD26. Accordingly, 20% of CD4+ T cells present in gut biopsies were CD73+. In HIV+ subjects, purified CD73+ resting memory CD4+ T cells in PBMC were infected with HIV-1 DNA, determined by real-time PCR, to the same level as for purified CD73-negative CD4+ T cells, both in untreated and treated subjects. Therefore, the proliferative CD73+ subset of memory CD4+ T cells is disproportionately reduced in HIV-1 infection, but, unexpectedly, their IL-7 dependent long-term resting phenotype suggests that residual infected cells in this subset may contribute significantly to the very long-lived HIV proviral DNA reservoir in treated subjects.


Asunto(s)
Antígenos CD/inmunología , Proliferación Celular/genética , Infecciones por VIH/genética , Terapia Molecular Dirigida , 5'-Nucleotidasa/genética , 5'-Nucleotidasa/inmunología , Antígenos CD/genética , Antígenos CD/uso terapéutico , Linaje de la Célula/genética , Linaje de la Célula/inmunología , Infecciones por VIH/inmunología , Infecciones por VIH/patología , Infecciones por VIH/virología , VIH-1/inmunología , VIH-1/patogenicidad , Humanos , Subunidad alfa del Receptor de Interleucina-7/genética , Subunidad alfa del Receptor de Interleucina-7/inmunología , Memoria a Largo Plazo/fisiología
5.
J Immunother Cancer ; 8(2)2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-33219094

RESUMEN

BACKGROUND: To evaluate the safety, tolerability, pharmacokinetics and pharmacodynamics of eftilagimod alpha (efti), a soluble lymphocyte activation gene-3 protein, in combination with the programmed cell death-1 (PD-1) antagonist pembrolizumab. METHODS: The study was divided into two parts; parts A and B, where part A was the dose escalation part and part B was an extension part of the study. Patients with metastatic melanoma were treated with efti plus the standard dose of pembrolizumab. Blood samples were assayed to determine plasma pharmacokinetic parameters, detect efti antibody formation and determine long-lived CD8 T cell responses and associated pharmacodynamic parameters. RESULTS: Twenty-four patients with melanoma received pembrolizumab and bi-weekly subcutaneous (s.c.) injections of efti at doses 1 mg, 6 mg or 30 mg/injection for up to 6 months (part A) or 30 mg/injection for up 12 months (part B). No dose-limiting toxicities were reported and the main adverse event for efti was injection site reactions. Sustained systemic exposure to the product was obtained in all patients following s.c. injections of 30 mg dose. Treatment induced an increase in activated CD8 and CD4 T cell counts, and in some of the soluble biomarkers, particularly interferon (IFN)-γ, a Th1 signature cytokine. An overall response rate (ORR) of 33% was observed in patients partly with pembrolizumab-refractory of part A and ORR of 50% was observed in patients with PD-1 naïve of part B. CONCLUSIONS: Efti was well tolerated in combination with pembrolizumab with encouraging antitumor activity. This warrants further clinical studies of this new combination therapy combining an antigen-presenting cell activator with an immune checkpoint inhibitor.


Asunto(s)
Anticuerpos Monoclonales Humanizados/uso terapéutico , Antígenos CD/uso terapéutico , Antineoplásicos Inmunológicos/uso terapéutico , Activación de Linfocitos/efectos de los fármacos , Melanoma/tratamiento farmacológico , Anticuerpos Monoclonales Humanizados/farmacología , Antígenos CD/farmacología , Antineoplásicos Inmunológicos/farmacología , Femenino , Humanos , Masculino , Melanoma/patología , Proteína del Gen 3 de Activación de Linfocitos
6.
Int Immunopharmacol ; 86: 106695, 2020 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-32570035

RESUMEN

BACKGROUND: Sialic acid-binding immunoglobulin-like lectins (Siglecs) are a superfamily of immunoreceptors recognizing sialic acid. Siglec-9 has been shown to mediate inhibitory immune responses. The aim of this study was to evaluate the effect of a soluble form of Siglec-9 (sSiglec-9) on inflamed intestinal epithelial cells (IECs), murine macrophages, and experimental murine colitis models. METHODS: COLO 205 human IECs and RAW 264.7 murine macrophages were pretreated with sSiglec-9 and then stimulated with TNF-α or lipopolysaccharides, respectively. The expression of proinflammatory cytokines such as IL-8 and TNF-α was measured using real-time RT-PCR and ELISA. To demonstrate the inhibitory effects of sSiglec-9 on the NF-κB pathway, IκBα phosphorylation/degradation was determined using western blotting and the DNA binding activity of NF-κB was evaluated using an electrophoretic mobility shift assay. Further, mouse models with dextran sulfate sodium-induced acute colitis and piroxicam-induced IL-10-/- chronic colitis were generated. Intraperitoneal injections of sSiglec-9 were performed, and body weight, colon length, and histopathologic findings were examined. RESULTS: sSiglec-9 suppressed IL-8 and TNF-α gene expression in stimulated COLO 205 and RAW 264.7 cells. sSiglec-9 inhibited IκBα phosphorylation/degradation and the DNA binding activity of NF-κB. sSiglec-9 injections significantly ameliorated weight loss, colon shortening, and the severity of intestinal inflammation in acute and chronic colitis mouse models. CONCLUSION: sSiglec-9 may inhibit NF-κB activation in IECs and macrophages and alleviate experimental colitis in mice, suggesting that sSiglec-9 is a potential therapeutic agent for the treatment of inflammatory bowel disease.


Asunto(s)
Antígenos CD/farmacología , Inflamación/tratamiento farmacológico , Intestinos/efectos de los fármacos , FN-kappa B/antagonistas & inhibidores , Lectinas Similares a la Inmunoglobulina de Unión a Ácido Siálico/farmacología , Animales , Antígenos CD/uso terapéutico , Línea Celular , Colitis/inducido químicamente , Colitis/tratamiento farmacológico , Sulfato de Dextran/toxicidad , Modelos Animales de Enfermedad , Células Epiteliales/efectos de los fármacos , Humanos , Inflamación/inducido químicamente , Inflamación/metabolismo , Interleucina-8/metabolismo , Intestinos/patología , Lipopolisacáridos/toxicidad , Macrófagos/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Inhibidor NF-kappaB alfa/metabolismo , Piroxicam/toxicidad , Proteínas Recombinantes/farmacología , Proteínas Recombinantes/uso terapéutico , Lectinas Similares a la Inmunoglobulina de Unión a Ácido Siálico/uso terapéutico , Transducción de Señal/efectos de los fármacos , Factor de Necrosis Tumoral alfa/metabolismo , Factor de Necrosis Tumoral alfa/toxicidad
7.
J Transl Med ; 17(1): 263, 2019 08 13.
Artículo en Inglés | MEDLINE | ID: mdl-31409424

RESUMEN

BACKGROUND: Mesenchymal stromal cells (MSC) demonstrate innate and regulatory immunologic functions and have been widely explored for cell therapy applications. Mechanisms by which MSCs achieve therapeutic effects are theorized, though appropriate dosing and duration of these mechanisms in vivo warrant deeper investigation. One, rapid immunosuppressive function of MSCs is through ectoenzyme expression of CD73 and CD39 which cooperatively hydrolyze inflammatory, extracellular adenosine triphosphate (ATP) to anti-inflammatory adenosine. Extracellular ATP has a key role in autoimmune and inflammatory diseases, which administered MSCs have the potential to modulate in a timescale that is befitting of shorter acting therapeutic function. METHODS: In vitro experiments were performed to determine the hydrolysis rates of ATP by MSCs. Through kinetic modeling from experimental results, the rate at which a single cell can metabolize ATP was determined. Based on these rates, the ability of MSCs to downregulate inflammatory signaling pathways was prospectively validated using model system parameters with respect to two different mechanisms: extracellular ATP stimulates lymphocytes to suppress proliferation and induce apoptosis and with co-stimulation, it stimulates monocytes to release pro-inflammatory IL-1ß. MSCs were co-cultured with immune cells using transwell inserts and compared to immune cell only groups. RESULTS: Hydrolysis of ATP was efficiently modeled by first-order enzyme kinetics. For in vitro culture, the rate at which a single cell can hydrolyize ATP is 8.9 nmol/min. In the presence of extracellular ATP, cocultures of MSCs reduced cytotoxicity and allows for proliferation of lymphocytes while limiting IL-1ß secretion from monocytes. CONCLUSIONS: Such use of these models may allow for better dosing predictions for MSCs to be used therapeutically for chronic inflammatory diseases such as rheumatoid arthritis, diabetes, pancreatitis, and other systemic inflammatory response syndromes. For the first time, the effect of MSCs on ATP hydrolysis in immune cell response is quantitatively analyzed on a cell-molecule basis by modeling the hydrolysis as an enzyme-substrate reaction. The results also give insight into MSCs' dynamic response mechanisms to ameliorate effects of extracellular ATP whether it be through positive or negative regulation.


Asunto(s)
5'-Nucleotidasa/uso terapéutico , Antígenos CD/uso terapéutico , Apirasa/uso terapéutico , Inmunomodulación , Células Madre Mesenquimatosas/citología , Adenosina/metabolismo , Adenosina Trifosfato/metabolismo , Muerte Celular , Proliferación Celular , Humanos , Hidrólisis , Interleucina-1beta/metabolismo , Cinética , Linfocitos/citología , Trasplante de Células Madre Mesenquimatosas , Especificidad por Sustrato
8.
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
9.
Curr Opin Allergy Clin Immunol ; 18(5): 425-431, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30015639

RESUMEN

PURPOSE OF REVIEW: Symptomatic management of chronic spontaneous urticaria (CSU) basically depends on second-generation H1 antihistamines and omalizumab. Omalizumab is a game changer in the management, but still there is a need for new targets and new biologics targeting new pathways in the treatment which will provide long-lasting remission, which will be given orally and which will be cheaper. This review will focus on new biologics that are underway of production or are already under use for different disorders but could be beneficial for the treatment of Chronic urticaria. RECENT FINDINGS: In this review, the treatment targets are classified according to the cells which are involved in the pathogenesis of CSU. Those are mast cells/basophils, B cells, T cells and eosinophils. The treatments that are under clinical trials for CSU are anti-IgE treatments such as ligelizumab, molecules targeting intracellular signaling pathways such as spleen tyrosine kinase inhibitors, surface inhibitory molecules such as siglec-8, anti-IL-1s such as canakinumab, Bruton kinase (BTK) inhibitors such as GDC-0853 and anti-IL-5s such as benralizumab and mepolizumab. SUMMARY: The ongoing clinical trials on new targets of treatment hold new hopes not only for a better care of the disease but also a better understanding of the pathomechanisms lying underneath.


Asunto(s)
Antialérgicos/uso terapéutico , Inhibidores de Proteínas Quinasas/uso terapéutico , Urticaria/tratamiento farmacológico , Agammaglobulinemia Tirosina Quinasa/antagonistas & inhibidores , Anticuerpos Antiidiotipos/uso terapéutico , Anticuerpos Monoclonales/uso terapéutico , Anticuerpos Monoclonales Humanizados/uso terapéutico , Antígenos CD/uso terapéutico , Antígenos de Diferenciación de Linfocitos B/uso terapéutico , Linfocitos B/inmunología , Basófilos/inmunología , Productos Biológicos/uso terapéutico , Enfermedad Crónica , Eosinófilos/inmunología , Humanos , Inmunoglobulina E/inmunología , Interleucina-1/antagonistas & inhibidores , Interleucina-5/antagonistas & inhibidores , Lectinas/uso terapéutico , Mastocitos/inmunología , Terapia Molecular Dirigida , Omalizumab/uso terapéutico , Piperazinas/uso terapéutico , Piridonas/uso terapéutico , Quinasa Syk/antagonistas & inhibidores , Linfocitos T/inmunología , Urticaria/inmunología
10.
Epilepsia ; 59(6): 1257-1268, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29799628

RESUMEN

OBJECTIVE: We previously discovered a role for the extracellular domain of the transmembrane protein semaphorin 4D (Sema4D) as a fast-acting, selective, and positive regulator of functional γ-aminobutyric acid (GABA)ergic synapse formation in hippocampal neuronal culture. We also demonstrated that Sema4D treatment increases inhibitory tone and suppresses hyperexcitability in an organotypic hippocampal slice culture model of epilepsy. Here, we investigate the ability of Sema4D to promote GABAergic synapse formation and suppress seizure activity in vivo in adult mice. METHODS: We performed a 3-hour, intrahippocampal infusion of Sema4D or control protein into the CA1 region of adult mice. To quantify GABAergic presynaptic bouton density, we performed immunohistochemistry on hippocampal tissue sections isolated from these animals using an antibody that specifically recognizes the glutamic acid decarboxylase isoform 65 protein (GAD65), which is localized to presynaptic GABAergic boutons. To assess seizure activity, we employed 2 in vivo mouse models of epilepsy, intravenous (iv) pentylenetetrazol (PTZ) and hippocampal electrical kindling, in the presence or absence of Sema4D treatment. We monitored seizure activity by behavioral observation or electroencephalography (EEG). To assay the persistence of the Sema4D effect, we monitored seizure activity and measured the density of GAD65-positive presynaptic boutons 3 or 48 hours after Sema4D infusion. RESULTS: Sema4D-treated mice displayed an elevated density of GABAergic presynaptic boutons juxtaposed to hippocampal pyramidal neuron cell bodies, consistent with the hypothesis that Sema4D promotes the formation of new inhibitory synapses in vivo. In addition, Sema4D acutely suppressed seizures in both the PTZ and electrical kindling models. When we introduced a 48-hour gap between Sema4D treatment and the seizure stimulus, seizure activity was indistinguishable from controls. Moreover, immunohistochemistry on brain sections or hippocampal slices isolated 3 hours, but not 48 hours, after Sema4D treatment displayed an increase in GABAergic bouton density, demonstrating temporal correlation between the effects of Sema4D on seizures and GABAergic synaptic components. SIGNIFICANCE: Our findings suggest a novel approach to treating acute seizures: harnessing synaptogenic molecules to enhance connectivity in the inhibitory network.


Asunto(s)
Anticonvulsivantes/uso terapéutico , Antígenos CD/uso terapéutico , Terminales Presinápticos/efectos de los fármacos , Convulsiones/tratamiento farmacológico , Semaforinas/uso terapéutico , Animales , Animales Recién Nacidos , Células Cultivadas , Convulsivantes/efectos adversos , Modelos Animales de Enfermedad , Estimulación Eléctrica/efectos adversos , Femenino , Neuronas GABAérgicas/efectos de los fármacos , Neuronas GABAérgicas/metabolismo , Neuronas GABAérgicas/patología , Glutamato Descarboxilasa/metabolismo , Hipocampo/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Neuronas/efectos de los fármacos , Técnicas de Cultivo de Órganos , Pentilenotetrazol/toxicidad , Convulsiones/patología
11.
N Engl J Med ; 378(17): 1604-1610, 2018 04 26.
Artículo en Inglés | MEDLINE | ID: mdl-29694819

RESUMEN

Genetic deficiency of ectodysplasin A (EDA) causes X-linked hypohidrotic ectodermal dysplasia (XLHED), in which the development of sweat glands is irreversibly impaired, an condition that can lead to life-threatening hyperthermia. We observed normal development of mouse fetuses with Eda mutations after they had been exposed in utero to a recombinant protein that includes the receptor-binding domain of EDA. We administered this protein intraamniotically to two affected human twins at gestational weeks 26 and 31 and to a single affected human fetus at gestational week 26; the infants, born in week 33 (twins) and week 39 (singleton), were able to sweat normally, and XLHED-related illness had not developed by 14 to 22 months of age. (Funded by Edimer Pharmaceuticals and others.).


Asunto(s)
Antígenos CD/uso terapéutico , Displasia Ectodermal Anhidrótica Tipo 1/terapia , Ectodisplasinas/genética , Ectodisplasinas/uso terapéutico , Terapias Fetales/métodos , Terapia Genética/métodos , Fragmentos Fc de Inmunoglobulinas/uso terapéutico , Diagnóstico Prenatal , Receptores Fc/uso terapéutico , Proteínas Recombinantes de Fusión/uso terapéutico , Adulto , Líquido Amniótico , Displasia Ectodermal Anhidrótica Tipo 1/diagnóstico por imagen , Displasia Ectodermal Anhidrótica Tipo 1/genética , Ectodisplasinas/deficiencia , Femenino , Humanos , Inyecciones , Masculino , Mutación , Embarazo , Radiografía , Proteínas Recombinantes/uso terapéutico , Glándulas Sudoríparas/anomalías , Glándulas Sudoríparas/diagnóstico por imagen , Germen Dentario/diagnóstico por imagen
12.
J Mol Neurosci ; 64(2): 200-210, 2018 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-29280053

RESUMEN

To explore the hypothesis that CD200Fc, a CD200R1 agonist with anti-inflammatory properties, will inhibit retinal glial cells hyperactivation and retinal ganglion cells (RGCs) apoptosis after optic nerve injury. CD200Fc was immediately administered after optic nerve crush (ONC) once by intravitreal injection. Rats were euthanized at 5 days after ONC. The density of RGCs was counted by immunostaining of retina flat mounts for Brn3a. TUNEL assay, immunoblotting analysis of ionized calcium-binding adapter molecule 1(iba1) (microglia marker) and glial fibrillary acidic protein (GFAP) (astrocytes and Müller cells marker), RT-PCR analysis of cyclooxygenase-2 (COX-2), inducible nitric oxide synthase (iNOS), monocyte chemotactic protein 1 (MCP-1), tumor necrosis factor-α (TNF-α), interleukin (IL)-8 and IL-10, ELISA measure protein levels of inflammatory cytokines and western blot analysis of CD200 and CD200R1 were evaluated. CD200Fc treatment suppressed ONC-induced RGCs loss through inhibition of RGCs apoptosis. Additionally, expression of glial cells activation markers GFAP and iba1 and production of pro-inflammatory cytokines (COX-2, iNOS, MCP-1, TNF-α, IL-8) were decreased in CD200Fc treated animals after ONC. Meanwhile, anti-inflammatory cytokine IL-10 was increased by CD200Fc treatment in ONC-induced rat retina. Finally, we found that CD200Fc significantly inhibited ONC-induced increased in expression of CD200 and raised the already high basal CD200R1 expression in the rat retina after ONC. Our results demonstrated that the anti-inflammatory effects of CD200Fc in ONC rats model through inhibited the activation of retinal glial cells via the interaction between CD200 and CD200R1, and the neuroprotective effects of CD200Fc on RGCs thought inhibited its apoptosis.


Asunto(s)
Antígenos CD/uso terapéutico , Apoptosis , Neuroglía/efectos de los fármacos , Traumatismos del Nervio Óptico/tratamiento farmacológico , Receptores Inmunológicos/metabolismo , Animales , Antígenos CD/administración & dosificación , Antígenos CD/farmacología , Quimiocina CCL2/metabolismo , Ciclooxigenasa 2/metabolismo , Interleucina-8/metabolismo , Inyecciones Intravítreas , Masculino , Neuroglía/metabolismo , Óxido Nítrico Sintasa de Tipo II/metabolismo , Unión Proteica , Proteínas RGS/metabolismo , Ratas , Ratas Sprague-Dawley , Receptores Inmunológicos/agonistas , Retina/citología , Retina/efectos de los fármacos , Retina/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
13.
Proc Natl Acad Sci U S A ; 114(10): 2687-2692, 2017 03 07.
Artículo en Inglés | MEDLINE | ID: mdl-28209777

RESUMEN

CD6 was established as a marker of T cells more than three decades ago, and recent studies have identified CD6 as a risk gene for multiple sclerosis (MS), a disease in which autoreactive T cells are integrally involved. Nevertheless, the precise role of CD6 in regulating T-cell responses is controversial and its significance in the pathogenesis of various diseases remains elusive, partly due to the lack of animals engineered to alter expression of the CD6 gene. In this report, we found that CD6 KO mice showed decreased pathogenic T-cell responses, reduced spinal cord T-cell infiltration, and attenuated disease severity in experimental autoimmune encephalomyelitis (EAE), an animal model of MS. CD6-deficient T cells exhibited augmented activation, but also significantly reduced survival and proliferation after activation, leading to overall decreased Th1 and Th17 polarization. Activated CD6-deficient T cells also showed impaired infiltration through brain microvascular endothelial cell monolayers. Furthermore, by developing CD6 humanized mice, we identified a mouse anti-human CD6 monoclonal antibody that is highly effective in treating established EAE without depleting T cells. These results suggest that (i) CD6 is a negative regulator of T-cell activation, (ii) at the same time, CD6 is a positive regulator of activated T-cell survival/proliferation and infiltration; and (iii) CD6 is a potential new target for treating MS and potentially other T-cell-driven autoimmune conditions.


Asunto(s)
Anticuerpos Monoclonales Humanizados/inmunología , Antígenos CD/inmunología , Antígenos de Diferenciación de Linfocitos T/inmunología , Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Esclerosis Múltiple/tratamiento farmacológico , Animales , Anticuerpos Monoclonales Humanizados/uso terapéutico , Antígenos CD/uso terapéutico , Antígenos de Diferenciación de Linfocitos T/uso terapéutico , Linfocitos T CD4-Positivos/inmunología , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/inmunología , Proliferación Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Encefalomielitis Autoinmune Experimental/inmunología , Encefalomielitis Autoinmune Experimental/patología , Humanos , Activación de Linfocitos/efectos de los fármacos , Activación de Linfocitos/inmunología , Ratones , Ratones Noqueados , Esclerosis Múltiple/inmunología , Esclerosis Múltiple/patología , Médula Espinal/efectos de los fármacos , Médula Espinal/inmunología , Médula Espinal/patología , Células TH1/inmunología , Células Th17/inmunología , Células Th17/patología
14.
Curr Drug Targets ; 17(6): 640-50, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-25777272

RESUMEN

CD6, one of the first antigens to be identified on T cells, is a membrane glycoprotein that physically associates with the antigen receptor complex. Because of this, its main function seems to involve the modulation of TCR-mediated signaling pathways. However, growing evidence indicates that this ancient and conserved scavenger-like receptor may also play a role as pattern recognition receptor (PRR), similar to other members of the scavenger receptor cysteine rich superfamily (SRCR-SF). Here, we discuss the functional interactions of CD6 with microbe- and damage-associated signals and the potential use of soluble forms of CD6 in the therapeutic treatment of bacterial infections, in particular multi-drug resistant bacterial strains. Importantly, microbe recognition by CD6 may also have functional consequences on T cell activation and differentiation, which remain to be explored.


Asunto(s)
Antígenos CD/metabolismo , Antígenos de Diferenciación de Linfocitos T/metabolismo , Receptores de Antígenos de Linfocitos T/metabolismo , Receptores Depuradores/metabolismo , Linfocitos T/citología , Animales , Antígenos CD/farmacología , Antígenos CD/uso terapéutico , Antígenos de Diferenciación de Linfocitos T/farmacología , Antígenos de Diferenciación de Linfocitos T/uso terapéutico , Bacterias/inmunología , Infecciones Bacterianas/tratamiento farmacológico , Diferenciación Celular , Farmacorresistencia Bacteriana Múltiple/efectos de los fármacos , Humanos , Activación de Linfocitos , Transducción de Señal , Linfocitos T/inmunología
15.
Acta Oncol ; 54(10): 1706-13, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26361073

RESUMEN

BACKGROUND: Immunotherapy with immune checkpoint inhibitors has emerged as promising treatment modality for cancer based on the success of anti-CTLA-4 and -PD-1/PD-L1 antibodies. LAG-3 and TIM-3 are two new immune checkpoints. The aim of this work is to review the role and application of LAG-3 and TIM-3 for cancer immunotherapy. MATERIAL AND METHODS: Literatures were searched and collected in Medline/PubMed. RESULTS: LAG-3 is presented as a CD4 homolog type I transmembrane protein which binds MHC class II molecules. LAG-3 negatively regulates T cell proliferation, homeostasis and function. IMP321 is formed of an extracellular portion of human LAG-3 fused to the Fc fraction of human IgG1 and has shown increased T cell responses and tolerability in phase I studies on advanced renal cell cancer. When combined with paclitaxel, IMP321 has exerted immune enhancement and tumor inhibition with no significant IMP321-related adverse events. TIM-3 belongs to the TIM family and mainly negatively regulates Th1 immunity. The TIM-3/galectin-9 pathway contributes to the suppressive tumor microenvironment. TIM-3 overexpression is associated with poor prognosis in a variety of cancers. Both LAG-3 and TIM-3 are coexpressed with other immune checkpoints. The application of LAG-3 or TIM-3 does play an important role in anti-tumor responses, and maybe better when combing with anti-CTLA-4 and anti-PD-1/L1 antibodies. CONCLUSIONS: These two immune checkpoints play crucial roles in cancer development and may be used in future clinical practice of cancer therapy.


Asunto(s)
Antígenos CD/inmunología , Antígenos CD/uso terapéutico , Inmunoterapia/métodos , Proteínas de la Membrana/antagonistas & inhibidores , Proteínas de la Membrana/inmunología , Neoplasias/terapia , Antígenos CD/metabolismo , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Receptor 2 Celular del Virus de la Hepatitis A , Humanos , Proteínas de la Membrana/metabolismo , Células TH1/inmunología , Microambiente Tumoral/inmunología , Proteína del Gen 3 de Activación de Linfocitos
16.
Cancer Res ; 75(11): 2166-76, 2015 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-25840983

RESUMEN

Cancer stem cells (CSC) drive tumorigenesis and contribute to genotoxic therapy resistance, diffuse infiltrative invasion, and immunosuppression, which are key factors for the incurability of glioblastoma multiforme (GBM). The AC133 epitope of CD133 is an important CSC marker for GBM and other tumor entities. Here, we report the development and preclinical evaluation of a recombinant AC133×CD3 bispecific antibody (bsAb) that redirects human polyclonal T cells to AC133(+) GBM stem cells (GBM-SC), inducing their strong targeted lysis. This novel bsAb prevented the outgrowth of AC133-positive subcutaneous GBM xenografts. Moreover, upon intracerebral infusion along with the local application of human CD8(+) T cells, it exhibited potent activity in prophylactic and treatment models of orthotopic GBM-SC-derived invasive brain tumors. In contrast, normal hematopoietic stem cells, some of which are AC133-positive, were virtually unaffected at bsAb concentrations effective against GBM-SCs and retained their colony-forming abilities. In conclusion, our data demonstrate the high activity of this new bsAb against patient-derived AC133-positive GBM-SCs in models of local therapy of highly invasive GBM.


Asunto(s)
Anticuerpos Biespecíficos/uso terapéutico , Antígenos CD/inmunología , Glioblastoma/terapia , Glicoproteínas/inmunología , Células Madre Neoplásicas/inmunología , Péptidos/inmunología , Antígeno AC133 , Anticuerpos Biespecíficos/inmunología , Antígenos CD/uso terapéutico , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/patología , Carcinogénesis/inmunología , Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Epítopos/inmunología , Glioblastoma/inmunología , Glioblastoma/patología , Glicoproteínas/uso terapéutico , Humanos , Inmunoterapia/métodos , Células Madre Neoplásicas/patología , Péptidos/uso terapéutico
17.
Mediators Inflamm ; 2015: 405629, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25767333

RESUMEN

Glucocorticoids are commonly used for treating asthma and its exacerbations but have well-recognised adverse effects and are not always effective. Few alternative treatments exist. Using a murine model of an acute exacerbation of asthma, we assessed the ability of ISU201, a novel protein drug, to suppress the inflammatory response when administered after induction of an exacerbation. Sensitised mice were chronically challenged with a low mass concentration of aerosolised ovalbumin, and then received a single moderate-level challenge to simulate an allergen-induced exacerbation. ISU201 was administered to mice 2 and 8 hours later, while pulmonary inflammation and expression of mRNA for chemokines and proinflammatory cytokines were assessed after 4, 12, and 24 hours. Relative to vehicle-treated controls, ISU201 suppressed accumulation of pulmonary neutrophils and eosinophils, while accelerating the decline in CXCL1, TNF-α, and IL-6 in lavage fluid and lung tissue. ISU201 significantly reduced peak expression of mRNA for the chemokines Cxcl9 and Cxcl10, the adhesion molecules Icam1 and Vcam1, and the proinflammatory cytokines Il1b, Il12p40, and Csf1. The ability of ISU201 to promote resolution of inflammation suggests that it may have potential as an alternative to glucocorticoids in the management of asthma, including when administered after the onset of an acute exacerbation.


Asunto(s)
Antígenos CD/uso terapéutico , Asma/tratamiento farmacológico , Asma/inmunología , Inflamación/tratamiento farmacológico , Inflamación/inmunología , Pulmón/inmunología , Pulmón/metabolismo , Fragmentos de Péptidos/uso terapéutico , Animales , Asma/metabolismo , Quimiocina CXCL10/genética , Quimiocina CXCL9/genética , Modelos Animales de Enfermedad , Femenino , Técnicas para Inmunoenzimas , Inflamación/metabolismo , Molécula 1 de Adhesión Intercelular/genética , Interleucina-1beta/genética , Pulmón/patología , Ratones , Ratones Endogámicos BALB C , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética
18.
Am J Respir Cell Mol Biol ; 53(2): 276-84, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25569356

RESUMEN

In allergic asthma, homeostatic pathways are dysregulated, which leads to an immune response toward normally innocuous antigens. The CD200-CD200 receptor pathway is a central regulator of inflammation, and CD200 expression was recently found to be down-regulated in circulating leukocytes of patients with asthma. Given the antiinflammatory properties of CD200, we investigated whether local delivery of recombinant CD200 (rCD200) could reinstate lung homeostasis in an experimental model of asthma. Brown Norway rats were sensitized with ovalbumin (OVA) and alum. rCD200 was intratracheally administered 24 hours before OVA challenge, and airway responsiveness to methacholine was measured 24 hours after the allergen challenge. Inflammation was also assessed by measuring cell recruitment and cytokine levels in bronchoalveolar lavages, as well as lung and draining lymph node accumulation of dendritic cells (DCs) and T cells. In sensitized rats, rCD200 abolished airway hyperresponsiveness, whereas the sham treatment had no effect. In addition, rCD200 strongly reduced OVA-induced lung accumulation of myeloid DCs, CD4(+) T cells, and T helper type 2 cells. This was associated with a strong reduction of OVA-induced IL-13 level and with an increase of IL-10 in supernatants of bronchoalveolar lavages. Lung eosinophilia and draining lymph node accumulation of myeloid DCs and T cells were not affected by rCD200. Overall, these data reveal that rCD200 can inhibit airway hyperresponsiveness in a model of asthma by a multistep mechanism associated with local alterations of the T cell response and the cytokine milieu.


Asunto(s)
Antígenos CD/uso terapéutico , Asma/metabolismo , Receptores Inmunológicos/fisiología , Animales , Antígenos CD/farmacología , Asma/tratamiento farmacológico , Asma/inmunología , Citocinas/metabolismo , Evaluación Preclínica de Medicamentos , Masculino , Contracción Muscular , Músculo Liso/fisiopatología , Ratas , Células Th2/inmunología , Tráquea/fisiopatología
19.
J Infect Dis ; 209(7): 1077-86, 2014 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-24265437

RESUMEN

Gram-positive bacteria cause a broad spectrum of infection-related diseases in both immunocompetent and immunocompromised hosts, ranging from localized infections to severe systemic conditions such as septic and toxic shock syndromes. This situation has been aggravated by the recent emergence of multidrug-resistant strains, thus stressing the need for alternative therapeutic approaches. One such possibility would be modulating the host's immune response. Herein, the potential use of a soluble form of the scavenger-like human lymphocyte receptor CD6 (shCD6) belonging to an ancient family of innate immune receptors has been evaluated. shCD6 can bind to a broad spectrum of gram-positive bacteria thanks to the recognition of highly conserved cell wall components (lipoteichoic acid [LTA] and peptidoglycan [PGN]), which are essential for their viability and pathogenicity and are not amenable to antibiotic resistance. shCD6 has in vitro inhibitory effects on both bacterial growth and Toll-like receptor-mediated inflammatory response induced by LTA plus PGN. In vivo infusion of shCD6 improves survival on mouse models of septic shock by Staphylococcus aureus (either multidrug-resistant or -sensitive) or their endotoxins (LTA + PGN) or exotoxins (TSST-1). These results support the use of shCD6 and/or other scavenger-like immune receptors in the treatment of severe gram-positive-induced infectious conditions.


Asunto(s)
Antígenos CD/inmunología , Antígenos de Diferenciación de Linfocitos T/inmunología , Productos Biológicos/inmunología , Peptidoglicano/inmunología , Staphylococcus aureus/inmunología , Ácidos Teicoicos/inmunología , Factores de Virulencia/inmunología , Animales , Antígenos CD/uso terapéutico , Antígenos de Diferenciación de Linfocitos T/uso terapéutico , Productos Biológicos/uso terapéutico , Modelos Animales de Enfermedad , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Peptidoglicano/metabolismo , Unión Proteica , Choque Séptico/tratamiento farmacológico , Infecciones Estafilocócicas/tratamiento farmacológico , Ácidos Teicoicos/metabolismo , Factores de Virulencia/metabolismo
20.
Curr Opin Oncol ; 25(6): 701-6, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24097105

RESUMEN

PURPOSE OF REVIEW: To describe the current new targeted therapy with monoclonal and bispecific antibodies in adult acute lymphoblastic leukemia (ALL), to improve response rates and outcome. RECENT FINDINGS: Blast cells in ALL express a variety of specific antigens, such as CD19, CD20, CD22, CD33 and CD52, and recently monoclonal antibodies (MoAbs) became available to target these antigens. The anti-CD20 MoAb rituximab has substantially improved the outcome in Burkitt lymphoma/leukemia, and is currently applied in de-novo B-precursor ALL. The MoAbs directed against CD22, linked to cytotoxic agents, either to calicheamicin (inotuzomab ozogamicin) or to plant or bacterial toxins (epratuzumab) are explored in refractory/relapsed childhood and adult ALL. Targeting CD19 is of great interest, as it is expressed in all B-lineage cells, including early precursors. The new bispecific antibody blinatumomab combines single chain antibodies to CD19 and CD3, and thereby T cells lyse the CD19 bearing B cells and is effective in patients with positive minimal residual disease (MRD) or refractory/relapsed ALL. SUMMARY: Antibody therapy in ALL is very promising, with high rate of complete remission and MRD-negativity in advanced ALL. It is currently explored in de-novo ALL to establish the best setting in combination with chemotherapy or even as a monotherapy.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Antígenos CD/uso terapéutico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Terapia Molecular Dirigida , Recurrencia Local de Neoplasia/prevención & control , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamiento farmacológico , Adulto , Anciano , Anticuerpos Biespecíficos/administración & dosificación , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales de Origen Murino/administración & dosificación , Antígenos CD/inmunología , Terapia Combinada , Femenino , Humanos , Masculino , Persona de Mediana Edad , Terapia Molecular Dirigida/métodos , Terapia Molecular Dirigida/tendencias , Leucemia-Linfoma Linfoblástico de Células Precursoras/inmunología , Leucemia-Linfoma Linfoblástico de Células Precursoras/patología , Rituximab , Resultado del Tratamiento
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA