Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 112
Filtrar
1.
Nat Commun ; 15(1): 5330, 2024 Jun 22.
Artículo en Inglés | MEDLINE | ID: mdl-38909062

RESUMEN

Porcine deltacoronavirus (PDCoV) is an emerging enteric pathogen that has recently been detected in humans. Despite this zoonotic concern, the antigenic structure of PDCoV remains unknown. The virus relies on its spike (S) protein for cell entry, making it a prime target for neutralizing antibodies. Here, we generate and characterize a set of neutralizing antibodies targeting the S protein, shedding light on PDCoV S interdomain crosstalk and its vulnerable sites. Among the four identified antibodies, one targets the S1A domain, causing local and long-range conformational changes, resulting in partial exposure of the S1B domain. The other antibodies bind the S1B domain, disrupting binding to aminopeptidase N (APN), the entry receptor for PDCoV. Notably, the epitopes of these S1B-targeting antibodies are concealed in the prefusion S trimer conformation, highlighting the necessity for conformational changes for effective antibody binding. The binding footprint of one S1B binder entirely overlaps with APN-interacting residues and thus targets a highly conserved epitope. These findings provide structural insights into the humoral immune response against the PDCoV S protein, potentially guiding vaccine and therapeutic development for this zoonotic pathogen.


Asunto(s)
Anticuerpos Neutralizantes , Anticuerpos Antivirales , Deltacoronavirus , Epítopos , Glicoproteína de la Espiga del Coronavirus , Glicoproteína de la Espiga del Coronavirus/inmunología , Glicoproteína de la Espiga del Coronavirus/metabolismo , Glicoproteína de la Espiga del Coronavirus/química , Animales , Anticuerpos Neutralizantes/inmunología , Porcinos , Anticuerpos Antivirales/inmunología , Epítopos/inmunología , Humanos , Deltacoronavirus/inmunología , Deltacoronavirus/metabolismo , Antígenos CD13/metabolismo , Antígenos CD13/inmunología , Infecciones por Coronavirus/inmunología , Infecciones por Coronavirus/virología , Dominios Proteicos , Unión Proteica , Enfermedades de los Porcinos/virología , Enfermedades de los Porcinos/inmunología , Células HEK293
2.
Eur J Med Chem ; 260: 115752, 2023 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-37647727

RESUMEN

Aminopeptidase N (APN/CD13) plays a role in tumors progression, but its inhibitor lacks cytotoxicity and is used as an adjuvant drug in cancer treatment. Histone deacetylases (HDACs) are a type of epigenetic targets, and HDAC inhibitors are cytotoxic and exhibit synergistic effects with other anticancer agents. Herein, a novel series of HDAC/CD13 dual inhibitors were rationally designed and synthesized to combine the anti-metastasis and anti-invasion of CD13 inhibitor with the cytotoxic of HDAC inhibitor. The representative compound 12 exhibited more potent inhibitory activity against human CD13, HDAC1-3, and antiproliferative activity than positive controls bestatin and SAHA. Compound 12 effectively induced apoptosis in MV4-11 cells, while arresting A549 cells in G2/M phase. Moreover, 12 exhibited significantly better anti-metastasis and anti-invasion effects than mono-inhibitors 32 and 38, indicating that it is a promising anti-cancer agent for further investigation.


Asunto(s)
Inhibidores de Histona Desacetilasas , Neoplasias , Humanos , Células A549 , Apoptosis , División Celular , Epigenómica , Inhibidores de Histona Desacetilasas/farmacología , Neoplasias/tratamiento farmacológico , Antígenos CD13/química , Antígenos CD13/inmunología
3.
Front Immunol ; 12: 742292, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34887854

RESUMEN

For a long time, proteins with enzymatic activity have not been usually considered to carry out other functions different from catalyzing chemical reactions within or outside the cell. Nevertheless, in the last few years several reports have uncovered the participation of numerous enzymes in other processes, placing them in the category of moonlighting proteins. Some moonlighting enzymes have been shown to participate in complex processes such as cell adhesion. Cell adhesion plays a physiological role in multiple processes: it enables cells to establish close contact with one another, allowing communication; it is a key step during cell migration; it is also involved in tightly binding neighboring cells in tissues, etc. Importantly, cell adhesion is also of great importance in pathophysiological scenarios like migration and metastasis establishment of cancer cells. Cell adhesion is strictly regulated through numerous switches: proteins, glycoproteins and other components of the cell membrane. Recently, several cell membrane enzymes have been reported to participate in distinct steps of the cell adhesion process. Here, we review a variety of examples of membrane bound enzymes participating in adhesion of immune cells.


Asunto(s)
Adhesión Celular/fisiología , Leucocitos/enzimología , 5'-Nucleotidasa/inmunología , 5'-Nucleotidasa/fisiología , Proteínas ADAM/inmunología , Proteínas ADAM/fisiología , ADP-Ribosil Ciclasa/inmunología , ADP-Ribosil Ciclasa/fisiología , ADP-Ribosil Ciclasa 1/inmunología , ADP-Ribosil Ciclasa 1/fisiología , Antígenos CD/inmunología , Antígenos CD/fisiología , Antígenos CD13/inmunología , Antígenos CD13/fisiología , Adhesión Celular/inmunología , Membrana Celular/enzimología , Membrana Celular/inmunología , Dipeptidil Peptidasa 4/inmunología , Dipeptidil Peptidasa 4/fisiología , Proteínas Ligadas a GPI/inmunología , Proteínas Ligadas a GPI/fisiología , Humanos , Leucocitos/inmunología , Leucocitos/fisiología , Glicoproteínas de Membrana/inmunología , Glicoproteínas de Membrana/fisiología , Proteínas de la Membrana/inmunología , Proteínas de la Membrana/fisiología , Modelos Biológicos
4.
Front Immunol ; 12: 753371, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34721427

RESUMEN

Many pathogens enter the host via the gut, causing disease in animals and humans. A robust intestinal immune response is necessary to protect the host from these gut pathogens. Despite being best suited for eliciting intestinal immunity, oral vaccination remains a challenge due to the gastrointestinal environment, a poor uptake of vaccine antigens by the intestinal epithelium and the tolerogenic environment pervading the gut. To improve uptake, efforts have focused on targeting antigens towards the gut mucosa. An interesting target is aminopeptidase N (APN), a conserved membrane protein present on small intestinal epithelial cells shown to mediate epithelial transcytosis. Here, we aimed to further optimize this oral vaccination strategy in a large animal model. Porcine APN-specific monoclonal antibodies were generated and the most promising candidate in terms of epithelial transcytosis was selected to generate antibody fusion constructs, comprising a murine IgG1 or porcine IgA backbone and a low immunogenic antigen: the F18-fimbriated E. coli tip adhesin FedF. Upon oral delivery of these recombinant antibodies in piglets, both mucosal and systemic immune responses were elicited. The presence of the FedF antigen however appeared to reduce these immune responses. Further analysis showed that F18 fimbriae were able to disrupt the antigen presenting capacity of intestinal antigen presenting cells, implying potential tolerogenic effects of FedF. Altogether, these findings show that targeted delivery of molecules to epithelial aminopeptidase N results in their transcytosis and delivery to the gut immune systems. The results provide a solid foundation for the development of oral subunit vaccines to protect against gut pathogens.


Asunto(s)
Adhesinas Bacterianas/inmunología , Anticuerpos Monoclonales/inmunología , Antígenos Bacterianos/inmunología , Antígenos CD13/inmunología , Proteínas de Escherichia coli/inmunología , Inmunoconjugados/inmunología , Inmunoglobulina A/biosíntesis , Mucosa Intestinal/inmunología , Intestino Delgado/inmunología , Porcinos/inmunología , Transcitosis , Vacunas Sintéticas/inmunología , Adhesinas Bacterianas/administración & dosificación , Administración Oral , Animales , Anticuerpos Antibacterianos/biosíntesis , Anticuerpos Antibacterianos/inmunología , Anticuerpos Monoclonales/administración & dosificación , Afinidad de Anticuerpos , Células Presentadoras de Antígenos/inmunología , Antígenos Bacterianos/administración & dosificación , Antígenos CD13/fisiología , Escherichia coli Enterotoxigénica/inmunología , Células Epiteliales/metabolismo , Proteínas de Escherichia coli/administración & dosificación , Femenino , Fimbrias Bacterianas/inmunología , Inmunoconjugados/administración & dosificación , Inmunoglobulina A/administración & dosificación , Inmunoglobulina A/inmunología , Inmunoglobulina G/inmunología , Intestino Delgado/enzimología , Ratones , Proteínas Recombinantes de Fusión/administración & dosificación , Proteínas Recombinantes de Fusión/inmunología , Transcitosis/fisiología , Vacunación/veterinaria
5.
Front Immunol ; 12: 729086, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34512663

RESUMEN

A successful malaria transmission blocking vaccine (TBV) requires the induction of a high antibody titer that leads to abrogation of parasite traversal of the mosquito midgut following ingestion of an infectious bloodmeal, thereby blocking the cascade of secondary human infections. Previously, we developed an optimized construct UF6b that elicits an antigen-specific antibody response to a neutralizing epitope of Anopheline alanyl aminopeptidase N (AnAPN1), an evolutionarily conserved pan-malaria mosquito midgut-based TBV target, as well as established a size-controlled lymph node targeting biodegradable nanoparticle delivery system that leads to efficient and durable antigen-specific antibody responses using the model antigen ovalbumin. Herein, we demonstrate that co-delivery of UF6b with the adjuvant CpG oligodeoxynucleotide immunostimulatory sequence (ODN ISS) 1018 using this biodegradable nanoparticle vaccine delivery system generates an AnAPN1-specific immune response that blocks parasite transmission in a standard membrane feeding assay. Importantly, this platform allows for antigen dose-sparing, wherein lower antigen payloads elicit higher-quality antibodies, therefore less antigen-specific IgG is needed for potent transmission-reducing activity. By targeting lymph nodes directly, the resulting immunopotentiation of AnAPN1 suggests that the de facto assumption that high antibody titers are needed for a TBV to be successful needs to be re-examined. This nanovaccine formulation is stable at -20°C storage for at least 3 months, an important consideration for vaccine transport and distribution in regions with poor healthcare infrastructure. Together, these data support further development of this nanovaccine platform for malaria TBVs.


Asunto(s)
Adyuvantes Inmunológicos/farmacología , Anopheles/inmunología , Ganglios Linfáticos/efectos de los fármacos , Vacunas contra la Malaria/farmacología , Malaria/prevención & control , Nanopartículas , Oligodesoxirribonucleótidos/farmacología , Plasmodium/inmunología , Desarrollo de Vacunas , Animales , Anopheles/parasitología , Anticuerpos Neutralizantes/sangre , Anticuerpos Antiprotozoarios/sangre , Antígenos CD13/antagonistas & inhibidores , Antígenos CD13/inmunología , Antígenos CD13/metabolismo , Composición de Medicamentos , Epítopos , Femenino , Interacciones Huésped-Parásitos , Inmunoglobulina G/sangre , Ganglios Linfáticos/inmunología , Ganglios Linfáticos/parasitología , Malaria/inmunología , Malaria/parasitología , Malaria/transmisión , Vacunas contra la Malaria/inmunología , Ratones , Nanomedicina , Plasmodium/patogenicidad , Vacunación
7.
Elife ; 92020 09 02.
Artículo en Inglés | MEDLINE | ID: mdl-32876563

RESUMEN

Porcine reproductive and respiratory syndrome virus (PRRSV) and transmissible gastroenteritis virus (TGEV) are two highly infectious and lethal viruses causing major economic losses to pig production. Here, we report generation of double-gene-knockout (DKO) pigs harboring edited knockout alleles for known receptor proteins CD163 and pAPN and show that DKO pigs are completely resistant to genotype 2 PRRSV and TGEV. We found no differences in meat-production or reproductive-performance traits between wild-type and DKO pigs, but detected increased iron in DKO muscle. Additional infection challenge experiments showed that DKO pigs exhibited decreased susceptibility to porcine deltacoronavirus (PDCoV), thus offering unprecedented in vivo evidence of pAPN as one of PDCoV receptors. Beyond showing that multiple gene edits can be combined in a livestock animal to achieve simultaneous resistance to two major viruses, our study introduces a valuable model for investigating infection mechanisms of porcine pathogenic viruses that exploit pAPN or CD163 for entry.


Pig epidemics are the biggest threat to the pork industry. In 2019 alone, hundreds of billions of dollars worldwide were lost due to various pig diseases, many of them caused by viruses. The porcine reproductive and respiratory virus (PRRS virus for short), for instance, leads to reproductive disorders such as stillbirths and premature labor. Two coronaviruses ­ the transmissible gastroenteritis virus (or TGEV) and the porcine delta coronavirus ­ cause deadly diarrhea and could potentially cross over into humans. Unfortunately, there are still no safe and effective methods to prevent or control these pig illnesses, but growing disease-resistant pigs could reduce both financial and animal losses. Traditionally, breeding pigs to have a particular trait is a slow process that can take many years. But with gene editing technology, it is possible to change or remove specific genes in a single generation of animals. When viruses infect a host, they use certain proteins on the surface of the host's cells to find their inside: the PRRS virus relies a protein called CD163, and TGEV uses pAPN. Xu, Zhou, Mu et al. used gene editing technology to delete the genes that encode the CD163 and pAPN proteins in pigs. When the animals were infected with PRRS virus or TGEV, the non-edited pigs got sick but the gene-edited animals remained healthy. Unexpectedly, pigs without CD163 and pAPN also coped better with porcine delta coronavirus infections, suggesting that CD163 and pAPN may also help this coronavirus infect cells. Finally, the gene-edited pigs reproduced and produced meat as well as the control pigs. These experiments show that gene editing can be a powerful technology for producing animals with desirable traits. The gene-edited pigs also provide new knowledge about how porcine viruses infect pigs, and may offer a starting point to breed disease-resistant animals on a larger scale.


Asunto(s)
Antígenos CD13/deficiencia , Infecciones por Coronavirus/prevención & control , Coronavirus/patogenicidad , Gastroenteritis Porcina Transmisible/prevención & control , Síndrome Respiratorio y de la Reproducción Porcina/prevención & control , Virus del Síndrome Respiratorio y Reproductivo Porcino/patogenicidad , Receptores de Superficie Celular/deficiencia , Virus de la Gastroenteritis Transmisible/patogenicidad , Animales , Animales Modificados Genéticamente , Antígenos CD/genética , Antígenos CD/inmunología , Antígenos de Diferenciación Mielomonocítica/genética , Antígenos de Diferenciación Mielomonocítica/inmunología , Composición Corporal , Antígenos CD13/genética , Antígenos CD13/inmunología , Coronavirus/inmunología , Infecciones por Coronavirus/genética , Infecciones por Coronavirus/inmunología , Infecciones por Coronavirus/virología , Susceptibilidad a Enfermedades , Gastroenteritis Porcina Transmisible/genética , Gastroenteritis Porcina Transmisible/inmunología , Gastroenteritis Porcina Transmisible/virología , Técnicas de Silenciamiento del Gen , Interacciones Microbiota-Huesped , Industria para Empaquetado de Carne , Fenotipo , Síndrome Respiratorio y de la Reproducción Porcina/genética , Síndrome Respiratorio y de la Reproducción Porcina/inmunología , Síndrome Respiratorio y de la Reproducción Porcina/virología , Virus del Síndrome Respiratorio y Reproductivo Porcino/inmunología , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/inmunología , Sus scrofa/genética , Porcinos , Virus de la Gastroenteritis Transmisible/inmunología , Aumento de Peso
8.
J Hematol Oncol ; 13(1): 32, 2020 04 07.
Artículo en Inglés | MEDLINE | ID: mdl-32264921

RESUMEN

BACKGROUND: In the search for novel antibody-drug conjugates (ADCs) with therapeutic potential, it is imperative to identify novel targets to direct the antibody moiety. CD13 seems an attractive ADC target as it shows a differential pattern of expression in a variety of tumors and cell lines and it is internalized upon engagement with a suitable monoclonal antibody. PM050489 is a marine cytotoxic compound tightly binding tubulin and impairing microtubule dynamics which is currently undergoing clinical trials for solid tumors. METHODS: Anti-CD13 monoclonal antibody (mAb) TEA1/8 has been used to prepare a novel ADC, MI130110, by conjugation to the marine compound PM050489. In vitro and in vivo experiments have been carried out to demonstrate the activity and specificity of MI130110. RESULTS: CD13 is readily internalized upon TEA1/8 mAb binding, and the conjugation with PM050489 did not have any effect on the binding or the internalization of the antibody. MI130110 showed remarkable activity and selectivity in vitro on CD13-expressing tumor cells causing the same effects than those described for PM050489, including cell cycle arrest at G2, mitosis with disarrayed and often multipolar spindles consistent with an arrest at metaphase, and induction of cell death. In contrast, none of these toxic effects were observed in CD13-null cell lines incubated with MI130110. Furthermore, in vivo studies showed that MI130110 exhibited excellent antitumor activity in a CD13-positive fibrosarcoma xenograft murine model, with total remissions in a significant number of the treated animals. Mitotic catastrophes, typical of the payload mechanism of action, were also observed in the tumor cells isolated from mice treated with MI130110. In contrast, MI130110 failed to show any activity in a xenograft mouse model of myeloma cells not expressing CD13, thereby corroborating the selectivity of the ADC to its target and its stability in circulation. CONCLUSION: Our results show that MI130110 ADC combines the antitumor potential of the PM050489 payload with the selectivity of the TEA1/8 monoclonal anti-CD13 antibody and confirm the correct intracellular processing of the ADC. These results demonstrate the suitability of CD13 as a novel ADC target and the effectiveness of MI130110 as a promising antitumor therapeutic agent.


Asunto(s)
Antineoplásicos Inmunológicos/farmacología , Antígenos CD13/inmunología , Inmunoconjugados/farmacología , Neoplasias/tratamiento farmacológico , Policétidos/farmacología , Pironas/farmacología , Animales , Antineoplásicos Inmunológicos/química , Antineoplásicos Inmunológicos/uso terapéutico , Línea Celular Tumoral , Femenino , Humanos , Inmunoconjugados/química , Inmunoconjugados/uso terapéutico , Ratones , Ratones Desnudos , Neoplasias/inmunología , Policétidos/química , Policétidos/uso terapéutico , Pironas/química , Pironas/uso terapéutico
9.
Sci Transl Med ; 11(497)2019 06 19.
Artículo en Inglés | MEDLINE | ID: mdl-31217340

RESUMEN

Advanced ovarian cancer is frequently treated with combination chemotherapy, but high recurrence rates show the need for therapies that can produce durable responses and extend overall survival. Bispecific antibodies that interact with tumor antigens on cancer cells and activating receptors on immune cells offer an innovative immunotherapy approach. Here, we describe a human bispecific antibody (REGN4018) that binds both Mucin 16 (MUC16), a glycoprotein that is highly expressed on ovarian cancer cells, and CD3, thus bridging MUC16-expressing cells with CD3+ T cells. REGN4018 induced T cell activation and killing of MUC16-expressing tumor cells in vitro. Binding and cytotoxicity of REGN4018 in vitro were minimally affected by high concentrations of CA-125, the shed form of MUC16, which is present in patients. In preclinical studies with human ovarian cancer cells and human T cells in immunodeficient mice, REGN4018 potently inhibited growth of intraperitoneal ovarian tumors. Moreover, in a genetically engineered immunocompetent mouse expressing human CD3 and human MUC16 [humanized target (HuT) mice], REGN4018 inhibited growth of murine tumors expressing human MUC16, and combination with an anti-PD-1 antibody enhanced this efficacy. Immuno-PET imaging demonstrated localization of REGN4018 in MUC16-expressing tumors and in T cell-rich organs such as the spleen and lymph nodes. Toxicology studies in cynomolgus monkeys showed minimal and transient increases in serum cytokines and C-reactive protein after REGN4018 administration, with no overt toxicity. Collectively, these data demonstrate potent antitumor activity and good tolerability of REGN4018, supporting clinical evaluation of REGN4018 in patients with MUC16-expressing advanced ovarian cancer.


Asunto(s)
Anticuerpos Biespecíficos/inmunología , Anticuerpos Biespecíficos/uso terapéutico , Antígeno Ca-125/inmunología , Antígeno Ca-125/metabolismo , Proteínas de la Membrana/inmunología , Proteínas de la Membrana/metabolismo , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Ováricas/inmunología , Linfocitos T/metabolismo , Animales , Antígenos CD13/inmunología , Antígenos CD13/metabolismo , Femenino , Citometría de Flujo , Humanos , Inmunoglobulina G/inmunología , Inmunoglobulina G/metabolismo , Células Jurkat , Macaca fascicularis , Ratones , Neoplasias Ováricas/metabolismo , Linfocitos T/inmunología
10.
Nanoscale ; 11(4): 1737-1744, 2019 Jan 23.
Artículo en Inglés | MEDLINE | ID: mdl-30623954

RESUMEN

Quantitative profiling of membrane proteins on the cell surface is of great interest in tumor targeted therapy and single cell biology. However, the existing technologies are either of insufficient resolution, or unable to provide precise information on the localization of individual proteins. Here, we report a new method that combines the use of quantum dot labeling, super-resolution microscopy (structured illumination microscopy, SIM) and software modeling. In this proof-of-principle study, we assessed the biological effects of Bestatin on individual cells from different AML cell lines expressing CD13 proteins, a potential target for tumor targeted therapy. Using the proposed method, we found that the different AML cell lines exhibit different CD13 expression densities, ranging from 0.1 to 1.3 molecules per µm2 cell surface, respectively. Importantly, Bestatin treatment assays shows that its effects on cell growth inhibition, apoptosis and cell cycle change are directly proportional to the density of CD13 on the cell surface of these cell lines. The results suggest that the proposed method advances the quantitative analysis of single cell surface proteins, and that the quantitative profiling information of the target protein on single cells has potential value in targeted drug susceptibility assessment.


Asunto(s)
Antígenos CD13/metabolismo , Leucemia Mieloide Aguda/diagnóstico por imagen , Anticuerpos/química , Anticuerpos/inmunología , Apoptosis/efectos de los fármacos , Antígenos CD13/inmunología , Puntos de Control del Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Humanos , Leucina/análogos & derivados , Leucina/farmacología , Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/patología , Microscopía Confocal , Puntos Cuánticos/química
11.
Sci Rep ; 7(1): 14373, 2017 10 30.
Artículo en Inglés | MEDLINE | ID: mdl-29085061

RESUMEN

A subset of monoclonal anti-DNA autoantibodies enters a variety of living cells. Here, we aimed to identify the endocytic receptors recognized by an internalizing anti-nucleic acid autoantibody, the 3D8 single-chain variable fragment (scFv). We found that cell surface binding and internalization of 3D8 scFv were inhibited markedly in soluble heparan sulfate (HS)/chondroitin sulfate (CS)-deficient or -removed cells and in the presence of soluble HS and CS. 3D8 scFv colocalized intracellularly with either HS proteoglycans (HSPGs) or CSPGs in HeLa cells. 3D8 scFv was co-endocytosed and co-precipitated with representative individual HSPG and CSPG molecules: syndecan-2 (a transmembrane HSPG), glypican-3 (a glycosylphosphatidylinositol (GPI)-anchored HSPG); CD44 (a transmembrane CSPG); and brevican (a GPI-anchored CSPG). Collected data indicate that 3D8 scFv binds to the negatively charged sugar chains of both HSPGs and CSPGs and is then internalized along with these molecules, irrespective of how these proteoglycans are associated with the cell membrane. This is the first study to show that anti-DNA antibodies enter cells via both HSPGs and CSPGs simultaneously. The data may aid understanding of endocytic receptors that bind anti-DNA autoantibodies. The study also provides insight into potential cell membrane targets for macromolecular delivery.


Asunto(s)
Proteoglicanos Tipo Condroitín Sulfato/fisiología , Proteoglicanos de Heparán Sulfato/fisiología , Animales , Anticuerpos Antinucleares/fisiología , Antígenos CD13/inmunología , Membrana Celular/metabolismo , Proteoglicanos Tipo Condroitín Sulfato/metabolismo , Sulfatos de Condroitina/metabolismo , Citoplasma/metabolismo , Endocitosis/fisiología , Glicosaminoglicanos/metabolismo , Glipicanos/inmunología , Células HeLa , Proteoglicanos de Heparán Sulfato/metabolismo , Heparitina Sulfato/metabolismo , Humanos , Receptores de Hialuranos/inmunología , Ácidos Nucleicos/metabolismo , Vesículas Transportadoras
12.
J Immunol ; 199(5): 1672-1681, 2017 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-28739875

RESUMEN

Human monocytic myeloid-derived suppressor cells (MO-MDSCs) within the hepatic compartment suppress inflammation and impair immune surveillance in liver cancer. It is currently not known whether recruitment of MO-MDSCs from blood via hepatic sinusoidal endothelium (HSEC) contributes to their enrichment within the hepatic compartment. We compared the transmigratory potential of MO-MDSCs and monocytes after adhesion to hepatic endothelial monolayers in flow-based assays that mimic in vivo shear stress in the sinusoids. Despite comparable binding to HSEC monolayers, proportionally fewer MO-MDSCs underwent transendothelial migration, indicating that the final steps of extravasation, where actin polymerization plays an important role, are impaired in MO-MDSCs. In this article, we found reduced levels of CD13 on MO-MDSCs, which has recently been reported to control cell motility in monocytes, alongside reduced VLA-4 expression, an integrin predominantly involved in adherence to the apical side of the endothelium. CD13 and VLA-4 blocking and activating Abs were used in flow-based adhesion assays, live-cell imaging of motility, and actin polymerization studies to confirm a role for CD13 in impaired MO-MDSC transmigration. These findings indicate that CD13 significantly contributes to tissue infiltration by MO-MDSCs and monocytes, thereby contributing to the pathogenesis of hepatic inflammation.


Asunto(s)
Antígenos CD13/metabolismo , Endotelio Corneal/fisiología , Hemocromatosis/inmunología , Hepatitis/inmunología , Hígado/inmunología , Células Supresoras de Origen Mieloide/inmunología , Migración Transendotelial y Transepitelial , Actinas/metabolismo , Anticuerpos Bloqueadores/farmacología , Antígenos CD13/genética , Antígenos CD13/inmunología , Adhesión Celular , Movimiento Celular , Células Cultivadas , Regulación hacia Abajo , Humanos , Integrina alfa4beta1/genética , Integrina alfa4beta1/inmunología , Integrina alfa4beta1/metabolismo
13.
J Cancer Res Clin Oncol ; 143(11): 2159-2170, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-28669053

RESUMEN

PURPOSE: Treatment of cancer using standard chemotherapy still offers a poor prognosis combined with severe side effects. Novel antibody-based therapies have been shown to overcome low efficiency and lack of selectivity by targeting cancer-associated antigens, such as aminopeptidase CD13. METHODS: We isolated a high-affinity CD13-specific single-chain fragment variable (scFv13) from a phage display library of V-genes from mice immunized with soluble antigen. An immunotoxin comprising the scFv13 and a truncated version of the exotoxin A of Pseudomonas aeruginosa (ETA', scFv13-ETA') and a bispecific scFv targeting CD13 and CD16 simultaneously (bsscFv[13xds16]) was generated and investigated for their therapeutic potential. RESULTS: Both fusion proteins bound specifically to target cells with high affinity. Furthermore, scFv13-ETA' inhibited the proliferation of human cancer cell lines efficiently at low concentrations (IC50 values of 408 pM-7 nM) and induced apoptosis (40-85% of target cells). The bsscFv triggered dose-dependent antibody-dependent cell-mediated cytotoxicity, resulting in the lysis of up to 23.9% A2058 cells, 18.0% MDA-MB-468 cells and 19.1% HL-60 cells. CONCLUSION: The provided data demonstrate potent therapeutic activity of the scFv13-ETA' and the bsscFv[13xds16]. The CD13-specific scFv is therefore suitable for the direct and specific delivery of both cytotoxic agents and effector cells to cancer-derived cells, making it ideal for further therapeutic evaluation.


Asunto(s)
ADP Ribosa Transferasas/inmunología , Anticuerpos Biespecíficos/farmacología , Apoptosis/efectos de los fármacos , Toxinas Bacterianas/inmunología , Antígenos CD13/antagonistas & inhibidores , Proliferación Celular/efectos de los fármacos , Exotoxinas/inmunología , Inmunotoxinas/farmacología , Neoplasias/tratamiento farmacológico , Anticuerpos de Cadena Única/inmunología , Factores de Virulencia/inmunología , Antígenos CD13/inmunología , Humanos , Neoplasias/inmunología , Neoplasias/patología , Proteínas Recombinantes/farmacología , Células Tumorales Cultivadas , Exotoxina A de Pseudomonas aeruginosa
14.
Biosci Rep ; 37(2)2017 04 28.
Artículo en Inglés | MEDLINE | ID: mdl-28270576

RESUMEN

Intestinal epithelial cells are the targets for transmissible gastroenteritis (TGE) virus (TGEV) infection. It is urgent to develop a novel candidate against TGEV entry. Bacillus subtilis is a probiotic with excellent anti-microorganism properties and one of its secretions, surfactin, has been regarded as a versatile weapon for most plant pathogens, especially for the enveloped virus. We demonstrate for the first time that B. subtilis OKB105 and its surfactin can effectively inhibit one animal coronavirus, TGEV, entering the intestinal porcine epithelial cell line (IPEC-J2). Then, several different experiments were performed to seek the might mechanisms. The plaque assays showed that surfactant could reduce the plaque generation of TGEV in a dose-dependent manner. Meanwhile, after incubation with TGEV for 1.5 h, B. subtilis could attach TGEV particles to their surface so that the number of virus to bind to the host cells was declined. Furthermore, our data showed that the inhibition of B. subtilis was closely related to the competition with TGEV for the viral entry receptors, including epidermal growth factor receptor (EGFR) and aminopeptidase N (APN) protein. In addition, Western blotting and apoptosis analysis indicated that B. subtilis could enhance the resistance of IPEC-J2 cells by up-regulating the expression of toll-like receptor (TLR)-6 and reducing the percentage of apoptotic cells. Taken together, our results suggest that B. subtilis OKB105 and its surfactin can antagonize TGEV entry in vitro and may serve as promising new candidates for TGEV prevention.


Asunto(s)
Antivirales/inmunología , Bacillus subtilis/inmunología , Células Epiteliales/inmunología , Lipopéptidos/inmunología , Péptidos Cíclicos/inmunología , Virus de la Gastroenteritis Transmisible/inmunología , Animales , Antivirales/metabolismo , Bacillus subtilis/metabolismo , Bacillus subtilis/fisiología , Western Blotting , Antígenos CD13/inmunología , Antígenos CD13/metabolismo , Línea Celular , Células Epiteliales/microbiología , Células Epiteliales/virología , Receptores ErbB/inmunología , Receptores ErbB/metabolismo , Interacciones Huésped-Patógeno/inmunología , Mucosa Intestinal/citología , Lipopéptidos/metabolismo , Péptidos Cíclicos/metabolismo , Porcinos , Receptor Toll-Like 6/inmunología , Receptor Toll-Like 6/metabolismo , Virus de la Gastroenteritis Transmisible/fisiología , Internalización del Virus
15.
Vet Parasitol ; 229: 15-21, 2016 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-27809971

RESUMEN

Forty goats, aged from 2 to 5 months were subjected to two different immunization protocols with a vaccine containing Haemonchus contortus gut membrane proteins H11/H-gal-GP to evaluate protection against H. contortus on pre-contaminated pastures. Goats were allocated to four groups of ten, three of them received their first vaccination before turnout. One group (V4) was then vaccinated at 4-week-intervals whereas another two groups (V6 and V6SEP) were vaccinated at 6-week-intervals. A control group (CTRL) remained unvaccinated. In May, after the second vaccination, all goats were turned out on pastures which had been previously contaminated with H. contortus eggs by seeder sheep for a period of six weeks. Goats of groups V4, V6 and CTRL were grazed together, whereas V6SEP was kept separately at an identical stocking rate. Clinical (PCV, FAMACHA, body weight), parasitological (faecal egg count, FEC) and serological (antibody titres) parameters were measured fortnightly. All goats were stabled in October, drenched with levamisole and two weeks later infected with 5000 L3 of H. contortus and slaughtered four weeks later for determination of abomasal worm burdens. Group mean FEC peaked 42-56days after turnout. Significantly lower FEC were observed in V6SEP vs CTRL between D 28 and 70 (p<0.01). Mean egg output of all groups decreased substantially and fluctuated at low levels until the end of the grazing period (D 154). Goats responded to vaccination with increasing antibody titres peaking after every booster. Mean worm burdens deriving from experimental infections were reduced by 89, 65 and 47% in groups V4, V6 and V6SEP, respectively, compared with the controls. The difference was significant for V4 (p<0.01). Antibody titres measured 14days before slaughter did not correlate statistically with the worm burdens. It was concluded that the vaccination protocol did not result in sufficient protection on pasture, as antibody titres were still low at the time the goats were exposed to larval contamination on pasture after turnout.


Asunto(s)
Enfermedades de las Cabras/prevención & control , Hemoncosis/veterinaria , Proteínas del Helminto/inmunología , Vacunación/veterinaria , Vacunas/administración & dosificación , Animales , Antígenos CD13/inmunología , Endopeptidasas/inmunología , Heces/parasitología , Femenino , Enfermedades de las Cabras/parasitología , Cabras , Hemoncosis/prevención & control , Masculino , Glicoproteínas de Membrana/inmunología , Proteínas de la Membrana/inmunología , Recuento de Huevos de Parásitos/veterinaria
16.
PLoS One ; 11(7): e0160108, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27467268

RESUMEN

Aminopeptidase N (CD13) is a widely expressed cell surface metallopeptidase involved in the migration of cancer and endothelial cells. Apart from our demonstration that CD13 modulates the efficacy of tumor necrosis factor-α-induced apoptosis in neutrophils, no other function for CD13 has been ascribed in this cell. We hypothesized that CD13 may be involved in neutrophil migration and/or homotypic aggregation. Using purified human blood neutrophils we confirmed the expression of CD13 on neutrophils and its up-regulation by pro-inflammatory agonists. However, using the anti-CD13 monoclonal antibody WM-15 and the aminopeptidase enzymatic inhibitor bestatin we were unable to demonstrate any direct involvement of CD13 in neutrophil polarisation or chemotaxis. In contrast, IL-8-mediated neutrophil migration in type I collagen gels was significantly impaired by the anti-CD13 monoclonal antibodies WM-15 and MY7. Notably, these antibodies also induced significant homotypic aggregation of neutrophils, which was dependent on CD13 cross-linking and was attenuated by phosphoinositide 3-kinase and extracellular signal-related kinase 1/2 inhibition. Live imaging demonstrated that in WM-15-treated neutrophils, where homotypic aggregation was evident, the number of cells entering IL-8 impregnated collagen I gels was significantly reduced. These data reveal a novel role for CD13 in inducing homotypic aggregation in neutrophils, which results in a transmigration deficiency; this mechanism may be relevant to neutrophil micro-aggregation in vivo.


Asunto(s)
Antígenos CD13/metabolismo , Colágeno Tipo I/metabolismo , Neutrófilos/citología , Adulto , Anticuerpos Monoclonales/inmunología , Antígenos CD13/inmunología , Polaridad Celular , Células Cultivadas , Quimiotaxis de Leucocito , Humanos , N-Formilmetionina Leucil-Fenilalanina/farmacología , Neutrófilos/efectos de los fármacos , Neutrófilos/inmunología
17.
Clin Res Hepatol Gastroenterol ; 40(4): 494-503, 2016 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26774363

RESUMEN

BACKGROUND: Scirrhous gastric cancer is associated with peritoneal dissemination and advanced lymph node metastasis from an early stage, and the prognosis is still poor. In this study, we aimed to analyze candidate molecules for targeted therapy of scirrhous gastric cancer. We searched for molecules/metabolic activity that might be predominantly expressed in a subpopulation of scirrhous gastric cancer cells and might function as cancer stem cell markers. RESULTS: For this purpose, we investigated the expression of various cell surface markers and of aldehyde dehydrogenase (ALDH) activity. These analyses showed that the scirrhous gastric cancer cell lines HSC-58 and HSC-44PE heterogeneously expressed CD13, while CD44, CDCP1, EpCAM and ABCG2 were expressed uniformly. Moreover, 10% of the total HSC-58 cell population expressed ALDH enzyme activity. A subpopulation of cells strongly positive for ALDH also expressed high levels of CD13, both of which are known as cancer stem cell markers. HSC-58 cells expressing high levels of CD13 showed lower sensitivity to a cancer drug cisplatin than cells with low levels of CD13. In contrast, CD13(-high) subpopulation of HSC-58 was more sensitive to an aminopeptidase N inhibitor bestatin. In terms of antibody-drug therapy, anti-CD13-immunotoxin was highly cytotoxic towards HSC-58 cells and was more cytotoxic than anti-EpCAM-immunotoxin. CONCLUSION: These data suggest that CD13 is a suitable cell surface candidate for targeted antibody-drug therapy of scirrhous gastric cancer.


Asunto(s)
Antígenos CD13/metabolismo , Neoplasias Gástricas/metabolismo , Aldehído Deshidrogenasa/metabolismo , Antineoplásicos/farmacología , Antígenos CD13/inmunología , Línea Celular Tumoral , Cisplatino/farmacología , Humanos , Inmunotoxinas/farmacología , Leucina/análogos & derivados , Leucina/farmacología , Neoplasias Gástricas/tratamiento farmacológico , Neoplasias Gástricas/patología
18.
J Control Release ; 220(Pt A): 149-159, 2015 Dec 28.
Artículo en Inglés | MEDLINE | ID: mdl-26485047

RESUMEN

Enteric diseases still have a devastating impact on global health. Oral vaccination is crucial to prevent intestinal infections, since only vaccines delivered to the intestinal tract elicit potent immune responses at the site of pathogen entry. However, oral vaccines encounter multiple barriers, including poor uptake and tolerance mechanisms, preventing the immune system to react to innocuous environmental antigens. Antigen delivery systems combined with selective targeting seem a promising strategy to overcome these obstacles. The current study evaluates the capacity of aminopeptidase N (APN)-targeted ß-glucan microparticles (GPs) as antigen delivery system. Antibodies against APN, an intestinal epithelial receptor, are efficiently oriented conjugated to GPs via the biolinker protein G. The resultant microparticles were analyzed for their antigen load, adjuvanticity and interaction with enterocytes and dendritic cells (DCs). Functionalization of GPs with antibodies neither impedes antigen load nor adjuvanticity. In addition, targeting to APN increases the uptake of microparticles by enterocytes and DCs, leading to an enhanced maturation of the latter as evidenced by an upregulation of maturation markers and a strong pro-inflammatory cytokine response. Finally, oral administration of APN-targeted antigen-loaded particles to piglets elicits higher serum antigen-specific antibody responses as compared to control particles. Taken together, these data support the use of APN-targeted GPs for oral delivery of antigens.


Asunto(s)
Antígenos/administración & dosificación , Antígenos CD13/inmunología , beta-Glucanos/química , Animales , Anticuerpos Monoclonales/química , Línea Celular , Células Cultivadas , Sistemas de Liberación de Medicamentos , Inmunización , Mucosa Intestinal/metabolismo , Albúmina Sérica Bovina/administración & dosificación , Porcinos , beta-Glucanos/inmunología
19.
J Virol ; 89(22): 11203-12, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26311872

RESUMEN

UNLABELLED: Insect-borne plant viruses cause significant agricultural losses and jeopardize sustainable global food production. Although blocking plant virus transmission would allow for crop protection, virus receptors in insect vectors are unknown. Here we identify membrane alanyl aminopeptidase N (APN) as a receptor for pea enation mosaic virus (PEMV) coat protein (CP) in the gut of the pea aphid, Acyrthosiphon pisum, using a far-Western blot method. Pulldown and immunofluorescence binding assays and surface plasmon resonance were used to confirm and characterize CP-APN interaction. PEMV virions and a peptide comprised of PEMV CP fused to a proline-rich hinge (-P-) and green fluorescent protein (CP-P-GFP) specifically bound to APN. Recombinant APN expressed in Sf9 cells resulted in internalization of CP-P-GFP, which was visualized by confocal microscopy; such internalization is an expected hallmark of a functional gut receptor. Finally, in assays with aphid gut-derived brush border membrane vesicles, binding of CP-P-GFP competed with binding of GBP3.1, a peptide previously demonstrated to bind to APN in the aphid gut and to impede PEMV uptake into the hemocoel; this finding supports the hypothesis that GBP3.1 and PEMV bind to and compete for the same APN receptor. These in vitro data combined with previously published in vivo experiments (S. Liu, S. Sivakumar, W. O. Sparks, W. A. Miller, and B. C. Bonning, Virology 401:107-116, 2010, http://dx.doi.org/10.1016/j.virol.2010.02.009) support the identification of APN as the first receptor in a plant virus vector. Knowledge of this receptor will provide for technologies based on PEMV-APN interaction designed to block plant virus transmission and to suppress aphid populations. IMPORTANCE: A significant proportion of global food production is lost to insect pests. Aphids, in addition to weakening plants by feeding on their sap, are responsible for transmitting about half of the plant viruses vectored by insects. Growers rely heavily on the application of chemical insecticides to manage both aphids and aphid-vectored plant viral disease. To increase our understanding of plant virus-aphid vector interaction, we provide in vitro evidence supporting earlier in vivo work for identification of a receptor protein in the aphid gut called aminopeptidase N, which is responsible for entry of the plant virus pea enation mosaic virus into the pea aphid vector. Enrichment of proteins found on the surface of the aphid gut epithelium resulted in identification of this first aphid gut receptor for a plant virus. This discovery is particularly important since the disruption of plant virus binding to such a receptor may enable the development of a nonchemical strategy for controlling aphid-vectored plant viruses to maximize food production.


Asunto(s)
Áfidos/virología , Antígenos CD13/metabolismo , Proteínas de la Cápside/metabolismo , Virus de Plantas/genética , Receptores Virales/metabolismo , Animales , Anticuerpos/inmunología , Antígenos CD13/inmunología , Línea Celular , Escherichia coli/genética , Escherichia coli/metabolismo , Proteínas Fluorescentes Verdes/metabolismo , Insectos Vectores/virología , Luteovirus/metabolismo , Microvellosidades/virología , Virus del Mosaico/genética , Enfermedades de las Plantas/virología , Unión Proteica/fisiología , Células Sf9 , Spodoptera , Vicia faba
20.
Am J Clin Pathol ; 144(2): 305-14, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26185316

RESUMEN

OBJECTIVES: Abnormalities of the RUNX1 gene in childhood B-acute lymphoblastic leukemia (B-ALL) are manifested by ETV6-RUNX1 or RUNX1 amplification. A detailed comparison between the two regarding clinicopathologic features with genetic analysis has not been performed previously. This parallel study assessed how different RUNX1 abnormalities affect the clinicopathology of B-ALL. METHODS: We compared clinicopathologic factors, including age, sex, WBC count, cerebrospinal fluid (CSF) involvement, immunophenotype, and blast proliferation rate between B-ALL with RUNX1 amplification (10 cases) and B-ALL with ETV6-RUNX1 translocation (67 cases) in childhood B-ALL. RESULTS: CD7 was often expressed in RUNX1 amplification but not in ETV6-RUNX1 (44% vs 0%, P = .0001) and appeared to correlate with CSF involvement in the former group (3/4 [75%]). CD13 was often detected in ETV6-RUNX1 with additional RUNX1 gain (38%) with an even higher frequency in double ETV6-RUNX1 translocation (77%), but was not detected in RUNX1 amplification (0%, P < .05). Children with RUNX1 amplification were older and more often CSF positive, while those with ETV6-RUNX1 were younger, more frequently had hyperleukocytosis, and had higher blast proliferation rates. CONCLUSIONS: RUNX1 copy numbers seem to be proportional to the age of B-ALL onset and the frequency of CSF involvement, while RUNX1 amplification vs translocation causes aberrant expression of CD7 and CD13, respectively.


Asunto(s)
Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/patología , Adolescente , Edad de Inicio , Animales , Antígenos CD7/biosíntesis , Antígenos CD7/inmunología , Antígenos CD13/biosíntesis , Antígenos CD13/inmunología , Preescolar , Femenino , Citometría de Flujo , Amplificación de Genes , Humanos , Inmunofenotipificación , Hibridación Fluorescente in Situ , Masculino , Leucemia-Linfoma Linfoblástico de Células Precursoras/líquido cefalorraquídeo , Conejos , Translocación Genética , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...