Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
1.
PeerJ ; 8: e10176, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33335804

RESUMEN

BACKGROUND: Antimicrobial peptides have a broad spectrum of antimicrobial activities and are attracting attention as promising next-generation antibiotics against multidrug-resistant (MDR) bacteria. The all-d-enantiomer [D(KLAKLAK)2] has been reported to have antimicrobial activity against Escherichia coli and Pseudomonas aeruginosa, and to be resistant to protein degradation in bacteria because it is composed of D-enantiomer compounds. In this study, we demonstrated that modification of [D(KLAKLAK)2] by the addition of an L-cysteine residue to its N- or C- terminus markedly enhanced its antimicrobial activities against Gram-negative bacteria such as MDR Acinetobacter baumannii, E. coli, and P. aeruginosa. METHODS: The peptides [D(KLAKLAK)2] (DP), DP to which L-cysteine was added at the N-terminus C-DP, and DP to which L-cysteine was added at the C-terminus DP-C, were synthesized at >95% purity. The minimum inhibitory concentrations of peptides and antibiotics were determined by the broth microdilution method. The synergistic effects of the peptides and the antibiotics against MDR P. aeruginosa were evaluated using the checkerboard dilution method. In order to assess how these peptides affect the survival of human cells, cell viability was determined using a Cell Counting Kit-8. RESULTS: C-DP and DP-C enhanced the antimicrobial activities of the peptide against MDR Gram-negative bacteria, including A. baumannii, E. coli, and P. aeruginosa. The antimicrobial activity of DP-C was greater than that of C-DP, with these peptides also having antimicrobial activity against drug-susceptible P. aeruginosa and drug-resistant P. aeruginosa overexpressing the efflux pump components. C-DP and DP-C also showed antimicrobial activity against colistin-resistant E. coli harboring mcr-1, which encodes a lipid A modifying enzyme. DP-C showed synergistic antimicrobial activity against MDR P. aeruginosa when combined with colistin. The LD50 of DP-C against a human cell line HepG2 was six times higher than the MIC of DP-C against MDR P. aeruginosa. The LD50 of DP-C was not altered by incubation with low-dose colistin. CONCLUSION: Attachment of an L-cysteine residue to the N- or C-terminus of [D(KLAKLAK)2] enhanced its antimicrobial activity against A. baumannii, E. coli, and P. aeruginosa. The combination of C-DP or DP-C and colistin had synergistic effects against MDR P. aeruginosa. In addition, DP-C and C-DP showed much stronger antimicrobial activity against MDR A. baumannii and E. coli than against P. aeruginosa.

2.
Eur J Pharmacol ; 887: 173596, 2020 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-32979353

RESUMEN

Bifidobacterium is a nonpathogenic strain of anaerobic bacteria that selectively localizes and proliferates in tumors. It has emerged as a specific carrier of anticancer proteins against malignant tumors. Claudins are tetraspanin transmembrane proteins that form tight junctions. Claudin-4 is overexpressed in certain epithelial malignant cancers. The C-terminal fragment of the Clostridium perfringens enterotoxin (C-CPE), an exotoxin without the cytotoxic domain, strongly binds to claudin-4. The C-CPE fusion toxin (C-CPE-PE23), which targets claudin-4, strongly suppresses tumor growth; however, C-CPE fusion toxins exhibit hepatic toxicity. In this study, we successfully generated a strain of Bifidobacterium longum that secreted C-CPE-PE23 (B. longum-C-CPE-PE23) and was specific to and cross reactive with human and mouse claudin-4. We evaluated the therapeutic potential of this strain against triple-negative breast cancer using a mouse model. C-CPE-PE23 decreased cell viability in a dose-dependent manner in human and mouse breast cancer cell lines. After intravenous injection, Bifidobacterium was specifically distributed in the tumors of mice bearing breast cancer tumors. Moreover, B. longum-C-CPE-PE23 significantly suppressed tumor growth in mice with breast cancer without serious side effects, such as weight loss or hepatic and renal damage. We suggest that B. longum-C-CPE-PE23 is a good candidate for breast cancer treatment. Bifidobacterium could also be used as a drug delivery system for hepatotoxic agents.


Asunto(s)
Antineoplásicos/administración & dosificación , Antineoplásicos/uso terapéutico , Bifidobacterium/metabolismo , Claudinas/metabolismo , Sistemas de Liberación de Medicamentos , Neoplasias de la Mama Triple Negativas/terapia , Animales , Línea Celular Tumoral , Claudina-4/metabolismo , ADN Recombinante , Relación Dosis-Respuesta a Droga , Enterotoxinas/administración & dosificación , Enterotoxinas/uso terapéutico , Femenino , Humanos , Ratones , Ratones Endogámicos BALB C , Plásmidos/genética
3.
Yakugaku Zasshi ; 138(7): 923-930, 2018.
Artículo en Japonés | MEDLINE | ID: mdl-29962470

RESUMEN

 Intravenously administered obligate anaerobic bacteria, such as bifidobacteria, grow specifically in tumor tissues. This specificity is attributed to the following: (1) Vascular walls in tumor tissues have nanometer- to micrometer-wide cracks, which allow the bacteria to pass through; (2) the intratumoral environment is hypoxic, due to poor vascularization, and therefore bifidobacteria can survive and proliferate in this anaerobic environment; (3) bifidobacteria cannot survive in well-oxygenated normal tissues. Moreover, unlike gram-negative bacteria, the gram-positive bifidobacteria do not produce endotoxins; therefore, there is no risk of endotoxin shock associated with their intravenous administration. Recently, the utility of bifidobacteria for specific drug delivery to tumor tissues has been highlighted. We have established a novel anti-cancer drug-delivery system using Bifidobacterium longum for the specific release of anti-tumor antibodies (e.g., antibody-drug complexes or single-chain antibodies) to targeted tumor tissues. Here, we introduce the results of our investigation.


Asunto(s)
Antineoplásicos/administración & dosificación , Bifidobacterium longum , Sistemas de Liberación de Medicamentos , Neoplasias/tratamiento farmacológico , Neoplasias/microbiología , Anaerobiosis , Animales , Humanos , Inmunotoxinas , Ratones , Neoplasias/irrigación sanguínea , Neoplasias/patología , Recombinación Genética
4.
J Infect Dis ; 203(11): 1574-81, 2011 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-21592986

RESUMEN

BACKGROUND: There is still no effective method to prevent or treat severe acute respiratory syndrome (SARS), which is caused by SARS coronavirus (CoV). In the present study, we evaluated the efficacy of a fully human monoclonal antibody capable of neutralizing SARS-CoV in vitro in a Rhesus macaque model of SARS. METHODS: The antibody 5H10 was obtained by vaccination of KM mice bearing human immunoglobulin genes with Escherichia coli-producing recombinant peptide containing the dominant epitope of the viral spike protein found in convalescent serum samples from patients with SARS. RESULTS: 5H10, which recognized the same epitope that is also a cleavage site critical for the entry of SARS-CoV into host cells, inhibited propagation of the virus and pathological changes found in Rhesus macaques infected with the virus through the nasal route. In addition, we analyzed the mode of action of 5H10, and the results suggested that 5H10 inhibited fusion between the virus envelope and host cell membrane. 5H10 has potential for use in prevention and treatment of SARS if it reemerges. CONCLUSIONS: This study represents a platform to produce fully human antibodies against emerging infectious diseases in a timely and safe manner.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/farmacología , Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/inmunología , Glicoproteínas de Membrana/inmunología , Síndrome Respiratorio Agudo Grave/terapia , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo/inmunología , Proteínas del Envoltorio Viral/inmunología , Enzima Convertidora de Angiotensina 2 , Animales , Animales Modificados Genéticamente , Anticuerpos Monoclonales/metabolismo , Anticuerpos Neutralizantes/metabolismo , Anticuerpos Antivirales/metabolismo , Western Blotting , Dominio Catalítico , Fusión Celular , Modelos Animales de Enfermedad , Células Gigantes/efectos de los fármacos , Humanos , Inmunohistoquímica , Pulmón/patología , Pulmón/virología , Macaca mulatta , Glicoproteínas de Membrana/genética , Ratones , Peptidil-Dipeptidasa A , Proteínas Recombinantes/genética , Proteínas Recombinantes/inmunología , Proteínas Recombinantes/farmacología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Síndrome Respiratorio Agudo Grave/inmunología , Síndrome Respiratorio Agudo Grave/virología , Glicoproteína de la Espiga del Coronavirus , Proteínas del Envoltorio Viral/genética
5.
Stem Cells Dev ; 20(1): 159-68, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20497033

RESUMEN

The use of induced pluripotent stem cells (iPSCs) is an exciting frontier in the study and treatment of human diseases through the generation of specific cell types. Here we show the derivation of iPSCs from human nonmobilized peripheral blood (PB) and bone marrow (BM) mononuclear cells (MNCs) by retroviral transduction of OCT3/4, SOX2, KLF4, and c-MYC. The PB- and BM-derived iPSCs were quite similar to human embryonic stem cells with regard to morphology, expression of surface antigens and pluripotency-associated transcription factors, global gene expression profiles, and differentiation potential in vitro and in vivo. Infected PB and BM MNCs gave rise to iPSCs in the presence of several cytokines, although transduction efficiencies were not high. We found that 5 × 10(5) PB MNCs, which corresponds to less than 1 mL of PB, was enough for the generation of several iPSC colonies. Generation of iPSCs from MNCs of nonmobilized PB, with its relative efficiency and ease of harvesting, could enable the therapeutic use of patient-specific pluripotent stem cells.


Asunto(s)
Células Sanguíneas/citología , Técnicas de Cultivo de Célula/métodos , Células Madre Pluripotentes Inducidas/citología , Adulto , Biomarcadores/metabolismo , Células de la Médula Ósea/citología , Diferenciación Celular/genética , Células Clonales , Metilación de ADN/genética , Reordenamiento Génico/genética , Genoma Humano/genética , Células Germinativas/citología , Células Germinativas/metabolismo , Humanos , Cadenas Pesadas de Inmunoglobulina/genética , Células Madre Pluripotentes Inducidas/metabolismo , Factor 4 Similar a Kruppel , Leucocitos Mononucleares/citología , Leucocitos Mononucleares/metabolismo , Masculino , Regiones Promotoras Genéticas/genética , Receptores de Antígenos de Linfocitos T alfa-beta/genética , Donantes de Tejidos
7.
Biochem Biophys Res Commun ; 395(2): 251-7, 2010 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-20363213

RESUMEN

Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) and its receptors (TRAIL-R1 and TRAIL-R2) are promising targets for tumor therapy. However, their clinical use is limited because some tumors show resistance to TRAIL-treatment. Here, we analyzed epitopes of nine TRAIL-R1-specific human monoclonal antibodies and demonstrated at least five tentative epitopes on human TRAIL-R1. We found that some of the five were post-translationally modified on some tumor cell lines. Interestingly, one of them, an epitope of TR1-272 antibody (TR1-272-epitope) disappeared on the tumor cells that are more susceptible to TRAIL-induced apoptosis compared to TR1-272-epitope positive cells. Treatment of TR1-272-epitope negative cells with TRAIL induced large cluster formation of TRAIL-R1, while treatment of TR1-272-epiope positive cells with TRAIL did not. These results suggest that TR1-272-antibody might distinguish the TRAIL-R1 conformation that could deliver stronger death signals. Further analysis of epitope-appearance and sensitivity to TRAIL should clarify the mechanisms of TRAIL-induced apoptosis of tumor cells and would provide useful information about tumor therapy using TRAIL and TRAIL-R signaling.


Asunto(s)
Apoptosis , Neoplasias/metabolismo , Procesamiento Proteico-Postraduccional , Receptores del Factor de Necrosis Tumoral/metabolismo , Anticuerpos Monoclonales/inmunología , Línea Celular Tumoral , Epítopos/inmunología , Epítopos/metabolismo , Humanos , Neoplasias/patología , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF , Receptores del Factor de Necrosis Tumoral/inmunología , Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo
8.
Stem Cells Dev ; 19(2): 229-38, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19558219

RESUMEN

Reprogramming of somatic cells provides potential for the generation of specific cell types, which could be a key step in the study and treatment of human diseases. In vitro reprogramming of somatic cells into a pluripotent embryonic stem (ES) cell-like state has been reported by retroviral transduction of murine fibroblasts using four embryonic transcription factors or through cell fusion of somatic and pluripotent stem cells. Here we show that mouse adult bone marrow mononuclear cells (BM MNCs) are competent as donor cells and can be reprogrammed into pluripotent ES cell-like cells. We isolated BM MNCs and mouse embryonic fibroblasts (MEFs) from Oct4-GFP transgenic mice, fused them with ES cells, or infected them with retroviruses expressing Oct4, Sox2, Klf4, and c-Myc. Fused BM MNCs formed more ES-like colonies than did MEFs. Infected BM MNCs gave rise to induced pluripotent stem (iPS) cells, although transduction efficiencies were not high. It was more efficient to pick up iPS colonies as compared with MEFs. BM-derived iPS (BM iPS) cells expressed ES cell markers, formed teratomas, and contributed to chimera mice with germ line development. Clonal analysis revealed that BM iPS clones had diversity, although some clones were found to be genetically identical with different phenotypes. Our findings imply that BM MNCs have potential advantages to generate iPS cells for the clinical application.


Asunto(s)
Células de la Médula Ósea/citología , Células Madre Embrionarias/citología , Células Madre Pluripotentes Inducidas/citología , Leucocitos Mononucleares/citología , Animales , Células de la Médula Ósea/metabolismo , Fusión Celular , Trasplante de Células/métodos , Células Cultivadas , Embrión de Mamíferos/citología , Células Madre Embrionarias/metabolismo , Femenino , Fibroblastos/citología , Fibroblastos/metabolismo , Perfilación de la Expresión Génica , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Inmunohistoquímica , Células Madre Pluripotentes Inducidas/metabolismo , Células Madre Pluripotentes Inducidas/trasplante , Factor 4 Similar a Kruppel , Leucocitos Mononucleares/metabolismo , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Ratones Transgénicos , Factor 3 de Transcripción de Unión a Octámeros/genética , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Retroviridae/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Teratoma/genética , Teratoma/metabolismo , Teratoma/patología , Transducción Genética
9.
Int J Cancer ; 2009 Dec 21.
Artículo en Inglés | MEDLINE | ID: mdl-20027630

RESUMEN

We observed previously that two carbohydrate epitopes, extended type 1 chain Le(a)-Le(a) and Le(b)-Le(a), are expressed strongly in human gastric or colorectal cancer and cell lines derived therefrom, but their expression in human normal colorectal cells is highly limited. A monoclonal antibody, termed GNX-8, was established through immunization of "KM mice" with colonic cancer cell line Colo205, and with purified Le(b)-Le(a) glycosphingolipid, followed by screening human IgG directed to this antigen. KM mice possess human chromosome fragments and are capable of producing human immunoglobulin. GNX-8 reacted specifically with extended type 1 chain epitope Le(b)-Le(a), bound to all five colonic cancer cell lines so far tested, and displayed strong complement-dependent cytotoxicity (CDC) and antibody-dependent cellular cytotoxicity (ADCC). The antigens defined by GNX-8, expressed in Colo205 cells, were: (i) glycosphingolipids with epitope Le(b)-Le(a), whose reactivity was abolished upon defucosylation; (ii) glycoproteins with molecular mass range from 32 to >175 kDa, which were depleted in cells cultured in the presence of benzyl-alpha-GalNAc, indicating that these epitopes are O-linked glycans.Immunohistological reactivity of GNX-8 at 1 mug/ml, applied on tissue sections from colorectal and various other types of cancer, was much stronger than that with various normal cells and tissues. GNX-8 reactivity with normal cells required a much higher concentration (150 mug/ml), and this reactivity was based on cross-reaction with non-extended, normal blood group Le(b) antigen. Growth of subcutaneous xenograft of human colonic cancer cells, Colo205 or DLD-1, in nude mice or SCID mice, was strongly inhibited by administration of GNX-8. These observations, taken together, indicate that antibody GNX-8, directed specifically to Le(b)-Le(a) antigen, provides a novel direction of immunotherapy for human colorectal cancer. (c) 2009 UICC.

11.
Int J Oncol ; 34(3): 619-27, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19212666

RESUMEN

Tumor development is a complex and dynamic process that involves malignant, vascular, and stromal cells. Endosialin is a tumor endothelial marker (TEM) present in the microvasculature and stroma of human tumors. Cancer-associated fibroblasts (CAF) have been implicated in promoting tumor development and have been associated with mesenchymal stem cells (MSC). Since stem/progenitor cells recruited either from bone marrow or residing in nearby tissues can contribute to pathological processes we investigated endosialin in MSC using a novel monoclonal antibody. Endosialin is highly expressed by CAF and human bone marrow-derived MSC. MSC can form networks in a tube formation assay that is inhibited by an anti-endosialin antibody. Immunohistochemistry for human endosialin in xenograft tumors following co-injection of MSC and cancer cells identified MSC in tumor stroma. MSC are a potential target for anticancer therapeutic intervention and endosialin expression offers a new tool for the identification of MSC. Endosialin expression by both CAF and MSC further implies the potential contribution of MSC to tumor stroma via differentiation into tumor stromal fibroblasts.


Asunto(s)
Antígenos CD/biosíntesis , Antígenos de Neoplasias/biosíntesis , Biomarcadores de Tumor/biosíntesis , Células de la Médula Ósea/metabolismo , Células de la Médula Ósea/patología , Endotelio Vascular/metabolismo , Neoplasias/metabolismo , Animales , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Diferenciación Celular/fisiología , Endotelio Vascular/citología , Endotelio Vascular/patología , Fibroblastos/metabolismo , Fibroblastos/patología , Humanos , Masculino , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Células Madre Mesenquimatosas/patología , Ratones , Ratones SCID , Neoplasias/irrigación sanguínea , Neoplasias/genética , Neoplasias/patología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células del Estroma/citología , Células del Estroma/metabolismo , Células del Estroma/patología
12.
Clin Cancer Res ; 14(22): 7223-36, 2008 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-19010839

RESUMEN

PURPOSE: Endosialin/CD248/tumor endothelial marker 1 is expressed in stromal cells, endothelial cells, and pericytes in various tumors; however, few studies have focused on expression in malignant cells. EXPERIMENTAL DESIGN: We studied expression of endosialin in clinical specimens, cell culture, and animal models and designed an anti-endosialin therapeutic prototype. RESULTS: Fifty human tumor cell lines and 6 normal cell types in culture were assayed by reverse transcription-PCR and/or flow cytometry for endosialin. Cell surface protein was found on 7 sarcoma lines, 1 neuroblastoma, and 4 normal cell types in culture. A fully human anti-endosialin antibody bound to human A-673 Ewing's sarcoma cells and SK-N-AS neuroblastoma cells but not HT-1080 cells. Exposure of cells to an anti-human IgG conjugated to saporin resulted in growth inhibition only of endosialin-expressing cells. Endosialin expression was assessed by immunohistochemistry in 250 clinical specimens of human cancer including 20 cancer subtypes. Endosialin is frequently found in human cancers. Endosialin expression is mainly a perivascular feature in carcinomas, with some expression in stromal cells. In sarcomas, endosialin is expressed by malignant cells, perivascular cells, and stromal cells. Development and characterization of experimental models for studying endosialin biology in sarcomas and evaluating anti-endosialin therapies is presented. CONCLUSIONS: Findings suggest that an anti-endosialin immunotoxin might be a promising therapeutic approach for endosialin-positive neoplasia, especially synovial sarcoma, fibrosarcoma, malignant fibrous histiocytoma, liposarcoma, and osteosarcoma. Thus, a diagnostic/therapeutic targeted therapeutic approach to treatment of endosialin-expressing tumors may be possible.


Asunto(s)
Antígenos CD/biosíntesis , Antígenos de Neoplasias/biosíntesis , Carcinoma/metabolismo , Inmunotoxinas/farmacología , Neoplasias/metabolismo , Sarcoma/metabolismo , Animales , Antígenos CD/genética , Antígenos de Neoplasias/genética , Carcinoma/genética , Línea Celular Tumoral , Citometría de Flujo , Humanos , Inmunoglobulina G/farmacología , Inmunohistoquímica , Neoplasias/genética , ARN Mensajero/análisis , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteínas Inactivadoras de Ribosomas Tipo 1/toxicidad , Saporinas , Sarcoma/genética
13.
Microvasc Res ; 76(3): 180-8, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18761022

RESUMEN

The formation of functional, mature blood vessels depends on the interaction between endothelial cells and pericytes. Commonality exists in the processes involved in vasculature development between tissues whether healthy or diseased. Endosialin/TEM 1 is a cell membrane protein that is expressed in blood vessels during embryogenesis and tumorigenesis but not in normal mature vessels. Antibodies developed to human endosialin were used to investigate endosialin expression and function in human prenatal brain pericytes and pericytes residing in tumors. Anti-endosialin was capable of preventing pericyte tube formation in culture and inhibited migration. Brain pericytes in culture had higher levels of endosialin/TEM 1 than TEMs-2, -3, -4, -5, -7, and -8. Immunocytochemistry revealed that endosialin was present in the cytoplasmic body and in the elongated extensions essential to pericyte function. Transgenic mice engineered to express human endosialin bred on an immunocompromised background allowed the growth of human tumor xenografts. In human colon carcinoma Colo205 and HT29 xenografts grown in human endosialin-transgenic mice, endosialin expression was largely confined to NG2-expressing perivascular cells and not CD31-positive endothelial cells. Similar methods applied to human ovarian and colon tumors confirmed endosialin expression by pericytes. The data indicate that endosialin is strongly expressed by pericytes during periods of active angiogenesis during embryonic and tumor development. Anti-endosialin antibodies may have value in identifying vasculature in malignant tissues. With the appropriate agent, targeting endosialin may interfere with blood vessel growth during tumor development.


Asunto(s)
Antígenos CD/metabolismo , Antígenos de Neoplasias/metabolismo , Neoplasias Experimentales/irrigación sanguínea , Neoplasias Experimentales/metabolismo , Neovascularización Patológica , Neovascularización Fisiológica , Pericitos/citología , Pericitos/metabolismo , Animales , Antígenos CD/genética , Antígenos de Neoplasias/genética , Secuencia de Bases , Línea Celular Tumoral , Células Cultivadas , Cartilla de ADN/genética , Desarrollo Embrionario , Femenino , Expresión Génica , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones SCID , Ratones Transgénicos , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Trasplante Heterólogo
14.
Mol Cancer Ther ; 7(8): 2536-46, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18723498

RESUMEN

Angiogenesis occurs during normal physiologic processes as well as under pathologic conditions such as tumor growth. Serial analysis of gene expression profiling revealed genes [tumor endothelial markers (TEM)] that are overexpressed in tumor endothelial cells compared with normal adult endothelial cells. Because blood vessel development of malignant tumors under certain conditions may include endothelial precursor cells (EPC) recruited from bone marrow, we investigated TEM expression in EPC. The expression of TEM1 or endosialin (CD248) and other TEM has been discovered in a population of vascular endothelial growth factor receptor 2+/CD31+/CD45-/VE-cadherin+ EPC derived from human CD133+/CD34+ cells. EPC share some properties with fully differentiated endothelial cells from normal tissue, yet reverse transcription-PCR and flow cytometry reveal that EPC express higher levels of endosialin at the molecular and protein levels. The elevated expression of endosialin in EPC versus mature endothelial cells suggests that endosialin is involved in the earlier stages of tumor angiogenesis. Anti-endosialin antibodies inhibited EPC migration and tube formation in vitro. In vivo, immunohistochemistry indicated that human EPC continued to express endosialin protein in a Matrigel plug angiogenesis assay established in nude mice. Anti-endosialin antibodies delivered systemically at 25 mg/kg were also able to inhibit circulating murine EPC in nude mice bearing s.c. SKNAS tumors. EPC and bone marrow-derived cells have been shown previously to incorporate into malignant blood vessels in some instances, yet they remain controversial in the field. The data presented here on endothelial genes that are up-regulated in tumor vasculature and in EPC support the hypothesis that the angiogenesis process in cancer can involve EPC.


Asunto(s)
Antígenos CD/metabolismo , Antígenos de Neoplasias/metabolismo , Endotelio Vascular/metabolismo , Antígenos CD/inmunología , Antígenos de Neoplasias/inmunología , Secuencia de Bases , Células Cultivadas , Cartilla de ADN , Endotelio Vascular/citología , Citometría de Flujo , Humanos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
15.
J Immunol ; 180(4): 2294-8, 2008 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-18250438

RESUMEN

Infusion reactions are a major side effect of the administration of therapeutic Abs and are the result of a complex immune reaction. In this study, we report that substitutions of Fc amino acids in the anti-HLA-DR Ab HD8 reduce its ability to induce infusion reactions in rats and monkeys. We first showed that i.v. administration of IgG1- and IgG2-subclass HD8 Abs induces severe infusion reactions in monkeys. These Abs express strong complement-dependent cytotoxicity (CDC), and in vivo depletion of complement in rats by pretreatment with cobra venom factor abrogated the lethal infusion reactions generated by HD8-IgG1. Thus, the infusion reactions appear to be largely driven by the complement system. To reduce the CDC function of HD8-IgG1, its Fc region was modified by two amino acid substitutions at Pro(331)Ser and Lys(322)Ala. The modified Ab was incapable of expressing CDC in vitro and did not induce severe infusion reactions in rats and monkeys, even at extremely high doses. The modified Ab retained its Ab-dependent cellular cytotoxicity function as well as its antitumor activity in a tumor-bearing mouse model. In summary, complement appears to drive infusion reactions, and modifications that eliminate the CDC activity of an Ab also reduce its ability to induce infusion reactions.


Asunto(s)
Citotoxicidad Celular Dependiente de Anticuerpos/inmunología , Activación de Complemento/inmunología , Isoanticuerpos/toxicidad , Animales , Animales Congénicos , Antineoplásicos/administración & dosificación , Antineoplásicos/uso terapéutico , Células CHO , Línea Celular , Línea Celular Tumoral , Proteínas del Sistema Complemento/toxicidad , Cricetinae , Cricetulus , Antígenos HLA-DR/inmunología , Antígenos HLA-DR/metabolismo , Humanos , Fragmentos Fc de Inmunoglobulinas/administración & dosificación , Fragmentos Fc de Inmunoglobulinas/genética , Fragmentos Fc de Inmunoglobulinas/metabolismo , Infusiones Intravenosas/efectos adversos , Isoanticuerpos/administración & dosificación , Isoanticuerpos/uso terapéutico , Linfoma/inmunología , Linfoma/mortalidad , Linfoma/terapia , Macaca fascicularis , Masculino , Ratones , Ratones SCID , Ratas , Trasplante Heterólogo
16.
Clin Exp Nephrol ; 12(1): 20-7, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18175057

RESUMEN

BACKGROUND: The KM mouse lacks endogenous genes for immunoglobulins and carries the entire human IgH locus and the IgLk transgene. Therefore, human IgA1 does not provoke a hetero-immune response. We had observed mesangial IgA deposits in KM mice given desialo-degalacto (DeS/DeGal) IgA1. METHODS: In this study, the mice were immunized with synthetic IgA1 hinge (glyco-)peptide before administration of DeS/DeGal IgA1, and the effects of the pre-immunization were evaluated. Mice were divided into sHP, 5GalNAc-sHP and non-immunization groups. In two pre-immunization groups, KLH-conjugated sHP or KLH-5GalNAc-sHP, which has five GalNAc residues, was subcutaneously given three times every 2 weeks. Two weeks after the final pre-immunization, DeS/DeGal IgA1 was administered daily for 5 weeks. Serial serum levels of anti-sHP and anti-IgA1 antibodies were evaluated by ELISA. On the day of the last administration of IgA1, renal biopsy was performed. RESULTS: Mesangial IgA deposits were observed in all non-immunized mice. In pre-immunized mice, IgA deposition was not detected in 6 of 13 sHP mice and 1 of 4 5GalNAc-sHP mice. The intensities of IgA deposits were significantly different between sHP groups and non-immunized (P = 0.003) groups. There was a significant inverse correlation between the intensities of IgA deposits and the anti-sHP antibody titers (P = 0.016). CONCLUSIONS: These results suggest that the anti-IgA1 hinge peptide antibody plays a role in the inhibition of glomerular IgA deposition.


Asunto(s)
Anticuerpos/uso terapéutico , Asialoglicoproteínas/metabolismo , Glomerulonefritis por IGA/inmunología , Inmunoglobulina A/inmunología , Inmunoglobulina A/metabolismo , Glomérulos Renales/metabolismo , Animales , Asialoglicoproteínas/inmunología , Humanos , Glomérulos Renales/ultraestructura , Ratones , Microscopía Electrónica
17.
Toxicon ; 51(3): 345-52, 2008 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-18061641

RESUMEN

Human monoclonal antibodies (HuMAbs) against HR1a from Protobothrops (formerly Trimeresurus) flavoviridis venom were obtained by the fusion of SP2/0-Ag14 myeloma cells and spleen cells from KM mice immunized with purified HR1a. The ability of HuMAbs to neutralize the HR1a was determined by in vitro neutralization assay and by neutralization of the hemorrhagic activity. The initial screening of over 300 hybridoma fusion wells resulted in the establishment of 80 HR1a-reactive hybridomas. Of the reactive clones, HuMAb HR1a-7 and HR1a-18 neutralized both proteolytic and hemorrhagic activity of HR1a. Mapping of epitope recognized by the reactive clones was performed by using an ELISA that measured antibody binding to overlapping peptides (15 amino acid peptide offset frameshifted by three residues) covering the metalloproteinase domain sequence of HR1a. HuMAbs HR1a-7 and HR1a-18 neutralized HR1a by reacting with peptides of EQQRYLNNFRFIELV and IVNTLNETYRYL. The three-dimensional structure of HR1a based on a homology modeling predicted that these two epitopes are surface exposed.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Metaloproteasas/antagonistas & inhibidores , Venenos de Víboras/enzimología , Viperidae , Secuencia de Aminoácidos , Animales , Anticuerpos Monoclonales/inmunología , Mapeo Epitopo , Humanos , Hibridomas , Masculino , Metaloproteasas/química , Metaloproteasas/inmunología , Metaloproteasas/metabolismo , Ratones , Modelos Moleculares , Datos de Secuencia Molecular , Pruebas de Neutralización , Conformación Proteica , Venenos de Víboras/antagonistas & inhibidores , Venenos de Víboras/inmunología
18.
J Periodontol ; 78(5): 933-9, 2007 May.
Artículo en Inglés | MEDLINE | ID: mdl-17470029

RESUMEN

BACKGROUND: Porphyromonas gingivalis has been implicated as an important pathogen in the development of adult periodontitis, and its colonization of subgingival sites is critical in the pathogenic process. We recently reported the construction and characterization of human immunoglobulin G isotype clones, which were specifically reactive with recombinant (r) 40-kDa outer membrane protein (OMP) of P. gingivalis. The aim of this study was to investigate the efficacy of human monoclonal antibody (hMAb) against r40-kDa OMP of P. gingivalis to the protection alveolar bone loss by P. gingivalis in rats. METHODS: The role of 40-kDa OMP in the adherence of P. gingivalis to human gingival epithelial cells (HGECs) was examined by preincubating with r40-kDa OMP hMAb before adding the HGECs. Moreover, we used a rat model to examine the effect of the anti-r40-kDa OMP hMAb in alveolar bone loss by oral infection. Forty-six days after the last infection, the periodontal bone level was assessed morphometrically on defleshed rat jaws. RESULTS: The adherence to HGECs was reduced by 84% compared to adherence levels without the antibody. P. gingivalis could not be detected from rats in a P. gingivalis-non-infected group and a group that was administered the anti-r40-kDa OMP hMAb. The bone loss in P. gingivalis-infected animals that were administered the anti-r40-kDa OMP hMAb was significantly lower than that of P. gingivalis-infected rats. CONCLUSIONS: Our results suggest that transchromosomic mouse-derived hMAb against r40-kDa OMP of P. gingivalis protects against periodontal bone loss. This newly constructed anti-r40-kDa OMP hMAb was used to protect against periodontal diseases caused by P. gingivalis infection.


Asunto(s)
Pérdida de Hueso Alveolar/prevención & control , Anticuerpos Antibacterianos/inmunología , Anticuerpos Monoclonales/inmunología , Proteínas de la Membrana Bacteriana Externa/inmunología , Infecciones por Bacteroidaceae/complicaciones , Porphyromonas gingivalis/inmunología , Pérdida de Hueso Alveolar/inmunología , Pérdida de Hueso Alveolar/microbiología , Animales , Anticuerpos Antibacterianos/uso terapéutico , Anticuerpos Monoclonales/uso terapéutico , Células Cultivadas , Modelos Animales de Enfermedad , Células Epiteliales/inmunología , Células Epiteliales/microbiología , Encía/citología , Humanos , Inmunoglobulina G/inmunología , Inmunoglobulina G/uso terapéutico , Isotipos de Inmunoglobulinas , Masculino , Periodontitis/inmunología , Periodontitis/microbiología , Porphyromonas gingivalis/patogenicidad , Ratas , Ratas Wistar , Proteínas Recombinantes , Estadísticas no Paramétricas
19.
Cancer Sci ; 98(6): 921-8, 2007 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17428256

RESUMEN

HD8, a fully human monoclonal antibody specific for human leukocyte antigen-DR (HLA-DR), was generated by using the transchromosome mouse that bears the human immunoglobulin genes. HD8 could bind to all 13 tested HLA-DR-positive cell lines and 35 B-cells from healthy donors. Epitope mapping revealed that while the antibody recognizes the most polymorphic region of the HLA-DRB chain, its critical epitope residues are conserved in the major alleles. Indeed, HD8 could recognize 99.2% of HLA-DRB alleles. Since its essential epitope residues are also largely conserved in HLA-DP and HLA-DQ, HD8 could recognize 100% and 66% of the HLA-DP and HLA-DQ alleles tested, respectively. HD8 exerted strong antibody-dependent cellular cytotoxicity and complement-dependent cytotoxicity in vitro, and significantly extended the life span of immunocompromised mice inoculated with non-Hodgkin lymphoma cell lines. The HD8 antibody may be highly useful in HLA-DR-targeted immunotherapy as it is likely to evoke similarly strong responses in individuals carrying different HLA-DR alleles.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Antígenos HLA-DR/inmunología , Linfoma/terapia , Adenosina Trifosfatasas , Animales , Anticuerpos Monoclonales/uso terapéutico , Linfocitos B/inmunología , Línea Celular , Citotoxicidad Inmunológica , Mapeo Epitopo , Proteínas de Escherichia coli , Humanos , Linfoma/genética , Ratones , Ratones Transgénicos
20.
Anticancer Res ; 25(6A): 3733-9, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-16302733

RESUMEN

For antibody-based therapy of cancer, monoclonal antibodies (mAbs) of human origin are superior to mouse, mouse/human chimeric or humanized mAbs, because of their minimum immunogenicity to humans and their efficient collaboration with human effector cells. In the present study, human mAbs were prepared against a pancarcinoma antigen, MK-1 (Ep-CAM), using a genetically-engineered mouse (KM mouse) that contains the human immunoglobulin genes. Spleen cells from KM mice, immunized with recombinant MK-1, were fused with P3-U1 mouse myeloma cells. Of 44 anti-MK-1 clones analyzed, two were of IgG4 and the others of IgM clones. Although the two IgG4 clones were suggested to recognize the same antigenic determinant or two closely located determinants, their VK regions were encoded by different light-chain genes while their VH sequences were identical. The two IgG4 and one of the IgM clones tested revealed antibody-dependent cell-mediated cytotoxicity and complement-dependent cytotoxicity, respectively, against MK-1-expressing cells in vitro, suggesting that these fully human mAbs produced against MK-1 and their V-region genes, which are applicable for the preparation of engineered antibody fragments that may be useful for antibody-based therapy of cancer.


Asunto(s)
Anticuerpos Monoclonales/genética , Anticuerpos Monoclonales/inmunología , Antígenos de Neoplasias/inmunología , Moléculas de Adhesión Celular/inmunología , Inmunoglobulina G/inmunología , Inmunoglobulina M/inmunología , Secuencia de Aminoácidos , Animales , Anticuerpos Monoclonales/biosíntesis , Anticuerpos Monoclonales/farmacología , Citotoxicidad Celular Dependiente de Anticuerpos , Células CHO , Línea Celular Tumoral , Clonación Molecular , Cricetinae , Molécula de Adhesión Celular Epitelial , Técnicas de Transferencia de Gen , Humanos , Inmunoglobulina G/genética , Inmunoglobulina M/genética , Región Variable de Inmunoglobulina/genética , Región Variable de Inmunoglobulina/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Datos de Secuencia Molecular , Proteínas Recombinantes de Fusión/biosíntesis , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/inmunología , Neoplasias Gástricas/inmunología , Neoplasias Gástricas/terapia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA