Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
Más filtros

Intervalo de año de publicación
1.
J Virol ; 95(17): e0186820, 2021 08 10.
Artículo en Inglés | MEDLINE | ID: mdl-34132574

RESUMEN

Pathogenic clade B New World mammarenaviruses (NWM) can cause Argentine, Venezuelan, Brazilian, and Bolivian hemorrhagic fevers. Sequence variability among NWM glycoproteins (GP) poses a challenge to the development of broadly neutralizing therapeutics against the entire clade of viruses. However, blockade of their shared binding site on the apical domain of human transferrin receptor 1 (hTfR1/CD71) presents an opportunity for the development of effective and broadly neutralizing therapeutics. Here, we demonstrate that the murine monoclonal antibody OKT9, which targets the apical domain of hTfR1, can sterically block cellular entry by viral particles presenting clade B NWM glycoproteins (GP1-GP2). OKT9 blockade is also effective against viral particles pseudotyped with glycoproteins of a recently identified pathogenic Sabia-like virus. With nanomolar affinity for hTfR1, the OKT9 antigen binding fragment (OKT9-Fab) sterically blocks clade B NWM-GP1s and reduces infectivity of an attenuated strain of Junin virus. Binding of OKT9 to the hTfR1 ectodomain in its soluble, dimeric state produces stable assemblies that are observable by negative-stain electron microscopy. A model of the OKT9-sTfR1 complex, informed by the known crystallographic structure of sTfR1 and a newly determined structure of the OKT9 antigen binding fragment (Fab), suggests that OKT9 and the Machupo virus GP1 share a binding site on the hTfR1 apical domain. The structural basis for this interaction presents a framework for the design and development of high-affinity, broadly acting agents targeting clade B NWMs. IMPORTANCE Pathogenic clade B NWMs cause grave infectious diseases, the South American hemorrhagic fevers. Their etiological agents are Junin (JUNV), Guanarito (GTOV), Sabiá (SABV), Machupo (MACV), Chapare (CHAV), and a new Sabiá-like (SABV-L) virus recently identified in Brazil. These are priority A pathogens due to their high infectivity and mortality, their potential for person-to-person transmission, and the limited availability of effective therapeutics and vaccines to curb their effects. While low homology between surface glycoproteins of NWMs foils efforts to develop broadly neutralizing therapies targeting NWMs, this work provides structural evidence that OKT9, a monoclonal antibody targeting a single NWM glycoprotein binding site on hTfR1, can efficiently prevent their entry into cells.


Asunto(s)
Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Neutralizantes/administración & dosificación , Anticuerpos Antivirales/administración & dosificación , Arenavirus del Nuevo Mundo/fisiología , Glicoproteínas/inmunología , Fiebre Hemorrágica Americana/prevención & control , Receptores de Transferrina/inmunología , Células A549 , Secuencia de Aminoácidos , Anticuerpos Monoclonales/inmunología , Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/inmunología , Fiebre Hemorrágica Americana/inmunología , Fiebre Hemorrágica Americana/virología , Humanos , Estructura Terciaria de Proteína , Receptores de Transferrina/química , Receptores de Transferrina/genética
2.
Cytokine ; 120: 220-226, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31121497

RESUMEN

An antibody-cytokine fusion protein, composed of the murine single-chain cytokine interleukin-12 (IL-12) genetically fused to a human IgG3 specific for the human tumor-associated antigen HER2/neu maintains antigen binding, cytokine bioactivity, and IL-12 heparin-binding activity. This latter property is responsible for the binding of the cytokine to glycosaminoglycans (GAGs) on the cell surface and the extracellular matrix and has been implicated in modulating IL-12 bioactivity. Previous studies indicate that the p40 subunit of human and murine IL-12 is responsible for the heparin-binding activity of this heterodimeric cytokine. In the present study we used bioinformatic analysis and site-directed mutagenesis to develop a version of the antibody-(IL-12) fusion protein without heparin-binding activity. This was accomplished by replacing the basic arginine (R) and lysine (K) residues in the cluster of amino acids 254-260 (RKKEKMK) of the murine IL-12 p40 subunit by the neutral non-polar amino acid alanine (A), generating an AAAEAMA mutant fusion protein. ELISA and flow cytometry demonstrated that the antibody fusion protein lacks heparin-binding activity but retains antigen binding. A T-cell proliferation assay showed IL-12 bioactivity in this construct. However, the IL-12 bioactivity is decreased compared to its non-mutated counterpart, which is consistent with an ancillary role of the heparin-binding site of IL-12 in modulating its activity. Thus, we have defined a cluster of amino acid residues with a crucial role in the heparin-binding activity of murine IL-12 in the context of an antibody-cytokine fusion protein.


Asunto(s)
Aminoácidos/metabolismo , Anticuerpos/metabolismo , Heparina/metabolismo , Interleucina-12/química , Interleucina-12/metabolismo , Proteínas Recombinantes de Fusión/metabolismo , Animales , Antígenos de Neoplasias/metabolismo , Sitios de Unión , Línea Celular Tumoral , Proliferación Celular , Humanos , Ratones , Unión Proteica , Receptor ErbB-2/metabolismo , Linfocitos T/citología
4.
Biochim Biophys Acta ; 1820(3): 291-317, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21851850

RESUMEN

BACKGROUND: Traditional cancer therapy can be successful in destroying tumors, but can also cause dangerous side effects. Therefore, many targeted therapies are in development. The transferrin receptor (TfR) functions in cellular iron uptake through its interaction with transferrin. This receptor is an attractive molecule for the targeted therapy of cancer since it is upregulated on the surface of many cancer types and is efficiently internalized. This receptor can be targeted in two ways: 1) for the delivery of therapeutic molecules into malignant cells or 2) to block the natural function of the receptor leading directly to cancer cell death. SCOPE OF REVIEW: In the present article we discuss the strategies used to target the TfR for the delivery of therapeutic agents into cancer cells. We provide a summary of the vast types of anti-cancer drugs that have been delivered into cancer cells employing a variety of receptor binding molecules including Tf, anti-TfR antibodies, or TfR-binding peptides alone or in combination with carrier molecules including nanoparticles and viruses. MAJOR CONCLUSIONS: Targeting the TfR has been shown to be effective in delivering many different therapeutic agents and causing cytotoxic effects in cancer cells in vitro and in vivo. GENERAL SIGNIFICANCE: The extensive use of TfR for targeted therapy attests to the versatility of targeting this receptor for therapeutic purposes against malignant cells. More advances in this area are expected to further improve the therapeutic potential of targeting the TfR for cancer therapy leading to an increase in the number of clinical trials of molecules targeting this receptor. This article is part of a Special Issue entitled Transferrins: molecular mechanisms of iron transport and disorders.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Antineoplásicos/administración & dosificación , Terapia Molecular Dirigida/métodos , Neoplasias/tratamiento farmacológico , Receptores de Transferrina/metabolismo , Antineoplásicos/metabolismo , Antineoplásicos/uso terapéutico , Apoptosis/efectos de los fármacos , Proteínas Portadoras/administración & dosificación , Proteínas Portadoras/metabolismo , Sistemas de Liberación de Medicamentos , Vectores Genéticos , Humanos , Hierro/metabolismo
5.
J Virol ; 86(7): 4024-8, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22278244

RESUMEN

Five New World (NW) arenaviruses cause human hemorrhagic fevers. Four of these arenaviruses are known to enter cells by binding human transferrin receptor 1 (hTfR1). Here we show that the fifth arenavirus, Chapare virus, similarly uses hTfR1. We also identify an anti-hTfR1 antibody, ch128.1, which efficiently inhibits entry mediated by the glycoproteins of all five viruses, as well as replication of infectious Junín virus. Our data indicate that all NW hemorrhagic fever arenaviruses utilize a common hTfR1 apical-domain epitope and suggest that therapeutic agents targeting this epitope, including ch128.1 itself, can be broadly effective in treating South American hemorrhagic fevers.


Asunto(s)
Anticuerpos/inmunología , Antígenos CD/química , Antígenos CD/inmunología , Arenavirus del Nuevo Mundo/fisiología , Regulación hacia Abajo , Fiebres Hemorrágicas Virales/virología , Receptores de Transferrina/química , Receptores de Transferrina/inmunología , Internalización del Virus , Secuencia de Aminoácidos , Animales , Antígenos CD/genética , Línea Celular , Fiebres Hemorrágicas Virales/genética , Fiebres Hemorrágicas Virales/inmunología , Humanos , Datos de Secuencia Molecular , Estructura Terciaria de Proteína , Receptores de Transferrina/genética , Receptores Virales/química , Receptores Virales/genética , Receptores Virales/inmunología , Alineación de Secuencia
6.
BMC Cancer ; 13: 195, 2013 Apr 17.
Artículo en Inglés | MEDLINE | ID: mdl-23594731

RESUMEN

BACKGROUND: Prostate cancer (PCa) is the second leading cause of cancer deaths in men in the United States. The prostate-specific antigen (PSA), often found at high levels in the serum of PCa patients, has been used as a marker for PCa detection and as a target of immunotherapy. The murine IgG1 monoclonal antibody AR47.47, specific for human PSA, has been shown to enhance antigen presentation by human dendritic cells and induce both CD4 and CD8 T-cell activation when complexed with PSA. In this study, we explored the properties of a novel mouse/human chimeric anti-PSA IgE containing the variable regions of AR47.47 as a potential therapy for PCa. Our goal was to take advantage of the unique properties of IgE in order to trigger immune activation against PCa. METHODS: Binding characteristics of the antibody were determined by ELISA and flow cytometry. In vitro degranulation was determined by the release of ß-hexosaminidase from effector cells. In vivo degranulation was monitored in human FcεRIα transgenic mice using the passive cutaneous anaphylaxis assay. These mice were also used for a vaccination study to determine the in vivo anti-cancer effects of this antibody. Significant differences in survival were determined using the Log Rank test. In vitro T-cell activation was studied using human dendritic cells and autologous T cells. RESULTS: The anti-PSA IgE, expressed in murine myeloma cells, is properly assembled and secreted, and binds the antigen and FcεRI. In addition, this antibody is capable of triggering effector cell degranulation in vitro and in vivo when artificially cross-linked, but not in the presence of the natural soluble antigen, suggesting that such an interaction will not trigger systemic anaphylaxis. Importantly, the anti-PSA IgE combined with PSA also triggers immune activation in vitro and in vivo and significantly prolongs the survival of human FcεRIα transgenic mice challenged with PSA-expressing tumors in a prophylactic vaccination setting. CONCLUSIONS: The anti-PSA IgE exhibits the expected biological properties and is capable of triggering immune activation and anti-tumor protection. Further studies on this antibody as a potential PCa therapy are warranted.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Degranulación de la Célula/efectos de los fármacos , Inmunoglobulina E/uso terapéutico , Factores Inmunológicos/uso terapéutico , Antígeno Prostático Específico/inmunología , Neoplasias de la Próstata/terapia , Receptores de IgE/inmunología , Animales , Degranulación de la Célula/inmunología , Células Dendríticas , Humanos , Inmunoglobulina E/farmacología , Estimación de Kaplan-Meier , Activación de Linfocitos/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Transgénicos , Linfocitos T , Vacunación
7.
Cancer Immunol Immunother ; 61(7): 991-1003, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22127364

RESUMEN

Breast and ovarian cancer are two of the leading causes of cancer deaths among women in the United States. Overexpression of the HER2/neu oncoprotein has been reported in patients affected with breast and ovarian cancers, and is associated with poor prognosis. To develop a novel targeted therapy for HER2/neu expressing tumors, we have constructed a fully human IgE with the variable regions of the scFv C6MH3-B1 specific for HER2/neu. This antibody was expressed in murine myeloma cells and was properly assembled and secreted. The Fc region of this antibody triggers in vitro degranulation of rat basophilic cells expressing human FcεRI (RBL SX-38) in the presence of murine mammary carcinoma cells that express human HER2/neu (D2F2/E2), but not the shed (soluble) antigen (ECD(HER2)) alone. This IgE is also capable of inducing passive cutaneous anaphylaxis in a human FcεRIα transgenic mouse model, in the presence of a cross-linking antibody, but not in the presence of soluble ECD(HER2). Additionally, IgE enhances antigen presentation in human dendritic cells and facilitates cross-priming, suggesting that the antibody is able to stimulate a secondary T-cell anti-tumor response. Furthermore, we show that this IgE significantly prolongs survival of human FcεRIα transgenic mice bearing D2F2/E2 tumors. We also report that the anti-HER2/neu IgE is well tolerated in a preliminary study conducted in Macaca fascicularis (cynomolgus) monkeys. In summary, our results suggest that this IgE should be further explored as a potential therapeutic against HER2/neu overexpressing tumors, such as breast and ovarian cancers.


Asunto(s)
Neoplasias de la Mama/inmunología , Neoplasias de la Mama/terapia , Inmunización Pasiva/métodos , Inmunoglobulina E/farmacología , Receptor ErbB-2/inmunología , Animales , Especificidad de Anticuerpos , Neoplasias de la Mama/enzimología , Células CHO , Línea Celular Tumoral , Cricetinae , Femenino , Humanos , Inmunoglobulina E/inmunología , Macaca fascicularis , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Transgénicos , Ratas , Ensayos Antitumor por Modelo de Xenoinjerto
8.
Proc Natl Acad Sci U S A ; 106(46): 19322-7, 2009 Nov 17.
Artículo en Inglés | MEDLINE | ID: mdl-19880745

RESUMEN

Trypanosoma brucei, a parasitic protist with a single flagellum, is the causative agent of African sleeping sickness. Propulsion of T. brucei was long believed to be by a drill-like, helical motion. Using millisecond differential interference-contrast microscopy and analyzing image sequences of cultured procyclic-form and bloodstream-form parasites, as well as bloodstream-form cells in infected mouse blood, we find that, instead, motility of T. brucei is by the propagation of kinks, separating left-handed and right-handed helical waves. Kink-driven motility, previously encountered in prokaryotes, permits T. brucei a helical propagation mechanism while avoiding the large viscous drag associated with a net rotation of the broad end of its tapering body. Our study demonstrates that millisecond differential interference-contrast microscopy can be a useful tool for uncovering important short-time features of microorganism locomotion.


Asunto(s)
Movimiento Celular , Flagelos/fisiología , Trypanosoma brucei brucei/fisiología , Tripanosomiasis Africana/microbiología , Animales , Células Cultivadas , Ratones , Ratones Endogámicos BALB C , Microscopía/métodos , Trypanosoma brucei brucei/citología
9.
Biosensors (Basel) ; 12(7)2022 Jul 14.
Artículo en Inglés | MEDLINE | ID: mdl-35884329

RESUMEN

To produce innovative biopharmaceuticals, highly flexible, adaptable, robust, and affordable bioprocess platforms for bioreactors are essential. In this article, we describe the development of a large-area microfluidic bioreactor (LM bioreactor) for mammalian cell culture that works at laminar flow and perfusion conditions. The 184 cm2 32 cisterns LM bioreactor is the largest polydimethylsiloxane (PDMS) microfluidic device fabricated by photopolymer flexographic master mold methodology, reaching a final volume of 2.8 mL. The LM bioreactor was connected to a syringe pump system for culture media perfusion, and the cells' culture was monitored by photomicrograph imaging. CHO-ahIFN-α2b adherent cell line expressing the anti-hIFN-a2b recombinant scFv-Fc monoclonal antibody (mAb) for the treatment of systemic lupus erythematosus were cultured on the LM bioreactor. Cell culture and mAb production in the LM bioreactor could be sustained for 18 days. Moreover, the anti-hIFN-a2b produced in the LM bioreactor showed higher affinity and neutralizing antiproliferative activity compared to those mAbs produced in the control condition. We demonstrate for the first-time, a large area microfluidic bioreactor for mammalian cell culture that enables a controlled microenvironment suitable for the development of high-quality biologics with potential for therapeutic use.


Asunto(s)
Reactores Biológicos , Microfluídica , Animales , Anticuerpos Monoclonales , Células CHO , Técnicas de Cultivo de Célula/métodos , Cricetinae , Cricetulus , Proteínas Recombinantes
10.
Nat Commun ; 13(1): 558, 2022 01 28.
Artículo en Inglés | MEDLINE | ID: mdl-35091550

RESUMEN

Five New World mammarenaviruses (NWMs) cause life-threatening hemorrhagic fever (HF). Cellular entry by these viruses is mediated by human transferrin receptor 1 (hTfR1). Here, we demonstrate that an antibody (ch128.1/IgG1) which binds the apical domain of hTfR1, potently inhibits infection of attenuated and pathogenic NWMs in vitro. Computational docking of the antibody Fab crystal structure onto the known structure of hTfR1 shows an overlapping receptor-binding region shared by the Fab and the viral envelope glycoprotein GP1 subunit that binds hTfR1, and we demonstrate competitive inhibition of NWM GP1 binding by ch128.1/IgG1 as the principal mechanism of action. Importantly, ch128.1/IgG1 protects hTfR1-expressing transgenic mice against lethal NWM challenge. Additionally, the antibody is well-tolerated and only partially reduces ferritin uptake. Our findings provide the basis for the development of a novel, host receptor-targeted antibody therapeutic broadly applicable to the treatment of HF of NWM etiology.


Asunto(s)
Antígenos CD/metabolismo , Arenaviridae/metabolismo , Fiebre Hemorrágica Americana/metabolismo , Receptores de Transferrina/metabolismo , Proteínas del Envoltorio Viral/metabolismo , Células A549 , Animales , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/metabolismo , Anticuerpos Monoclonales/farmacología , Antígenos CD/inmunología , Arenaviridae/efectos de los fármacos , Arenaviridae/fisiología , Chlorocebus aethiops , Fiebre Hemorrágica Americana/prevención & control , Fiebre Hemorrágica Americana/virología , Interacciones Huésped-Patógeno/efectos de los fármacos , Humanos , Virus Junin/efectos de los fármacos , Virus Junin/fisiología , Ratones Endogámicos C57BL , Ratones Transgénicos , Simulación del Acoplamiento Molecular , Unión Proteica/efectos de los fármacos , Receptores de Transferrina/antagonistas & inhibidores , Receptores de Transferrina/inmunología , Células Vero
11.
Physiol Genomics ; 36(2): 89-97, 2009 Jan 08.
Artículo en Inglés | MEDLINE | ID: mdl-19001510

RESUMEN

The process of parturition involves the complex interplay of factors that change the excitability and contractile activity of the uterus. We have compared the relative gene expression profile of myometrium from rats before parturition (21 days pregnant) and during delivery, using high-density DNA microarray. Of 8,740 sequences available in the array, a total of 3,782 were detected as present. From the sequences that were significantly altered, 59 genes were upregulated and 82 genes were downregulated. We were able to detect changes in genes described to have altered expression level at term, including connexin 43 and 26, cyclooxygenase 2, and oxytocin receptor, as well as novel genes that have been not previously associated with parturition. Quantitative real-time PCR on selected genes further confirmed the microarray data. Here we report for the first time that aquaporin5 (AQP5), a member of the aquaporin water channel family, was dramatically downregulated during parturition (approximately 100-fold by microarray and approximately 50-fold by real-time PCR). The emerging profile highlights biochemical cascades occurring in a period of approximately 36 h that trigger parturition and the initiation of myometrium reverse remodeling postpartum. The microarray analysis uncovered genes that were previously suspected to play a role in parturition. This regulation involves genes from immune/inflammatory response, steroid/lipid metabolism, calcium homeostasis, cell volume regulation, cell signaling, cell division, and tissue remodeling, suggesting the presence of multiple and redundant mechanisms altered in the process of birth.


Asunto(s)
Expresión Génica , Miometrio/metabolismo , Parto/genética , Preñez/genética , Animales , Acuaporina 5/genética , Acuaporina 5/metabolismo , Conexina 43/genética , Conexina 43/metabolismo , Ciclooxigenasa 2/genética , Ciclooxigenasa 2/metabolismo , Femenino , Perfilación de la Expresión Génica , Análisis de Secuencia por Matrices de Oligonucleótidos , Parto/metabolismo , Embarazo , Ratas , Receptores de Oxitocina/genética , Receptores de Oxitocina/metabolismo
12.
J Gene Med ; 11(8): 655-63, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19455593

RESUMEN

BACKGROUND: Targeted gene transduction in vivo is the ultimate preferred method for gene delivery. We previously developed targeting lentiviral vectors that specifically recognize cell surface molecules with conjugated antibodies and mediate targeted gene transduction both in vitro and in vivo. Although effective in some experimental settings, the conjugation of virus with antibodies is mediated by the interaction between protein A and the Fc region of antibodies, which is not as stable as covalent conjugation. We have now developed a more stable conjugation strategy utilizing the interaction between avidin and biotin. METHODS: We inserted the biotin-adaptor-peptide, which was biotinylated by secretory biotin ligase at specific sites, into our targeting envelope proteins, enabling conjugation of the pseudotyped virus with avidin, streptavidin or neutravidin. RESULTS: When conjugated with avidin-antibody fusion proteins or the complex of avidin and biotinylated targeting molecules, the vectors could mediate specific transduction to targeted cells recognized by the targeting molecules. When conjugated with streptavidin-coated magnetic beads, transduction by the vectors was targeted to the locations of magnets. CONCLUSIONS: This targeting vector system can be used for broad applications of targeted gene transduction using biotinylated targeting molecules or targeting molecules fused with avidin.


Asunto(s)
Biotina/metabolismo , Técnicas de Transferencia de Gen , Vectores Genéticos/genética , Lentivirus/genética , Péptidos/metabolismo , Avidina/metabolismo , Biotinilación , Western Blotting , Línea Celular , Electroforesis en Gel de Poliacrilamida , Humanos , Magnetismo , Mutagénesis Insercional , Receptores de Transferrina/metabolismo , Proteínas Recombinantes de Fusión/metabolismo , Virus Sindbis/genética , Transducción Genética , Proteínas del Envoltorio Viral/metabolismo , Virión/metabolismo
13.
Mol Cancer Ther ; 7(3): 721-9, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18347157

RESUMEN

We have documented previously that a multiple chaperone protein vaccine termed chaperone-rich cell lysate (CRCL) promotes tumor-specific T-cell responses leading to cancer regression in several mouse tumor models. We report here that CRCL vaccine generated from a mouse breast cancer (TUBO, HER2/neu positive) is also capable of eliciting humoral immunity. Administration of TUBO CRCL triggered anti-HER2/neu antibody production and delayed the progression of established tumors. This antitumor activity can be transferred through the serum isolated from TUBO CRCL-immunized animals and involved both B cells and CD4(+) T lymphocytes. Further evaluation of the mechanisms underlying TUBO CRCL-mediated humoral immunity highlighted the role of antibody-dependent cell-mediated cytotoxicity. These results suggest that tumor-derived CRCL vaccine has a wider applicability as a cancer vaccine because it can target both T-cell- and B-cell-specific responses and may represent a promising approach for the immunotherapy of cancer.


Asunto(s)
Anticuerpos Antineoplásicos/biosíntesis , Vacunas contra el Cáncer/inmunología , Modelos Animales de Enfermedad , Neoplasias Mamarias Experimentales/inmunología , Chaperonas Moleculares/metabolismo , Animales , Citotoxicidad Celular Dependiente de Anticuerpos , Línea Celular , Ensayo de Inmunoadsorción Enzimática , Femenino , Citometría de Flujo , Sueros Inmunes , Neoplasias Mamarias Experimentales/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Transgénicos
14.
Mol Cancer Ther ; 6(11): 2995-3008, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18025284

RESUMEN

We have previously developed an antibody fusion protein composed of a mouse/human chimeric IgG3 specific for the human transferrin receptor genetically fused to avidin (anti-hTfR IgG3-Av) as a universal delivery system for cancer therapy. This fusion protein efficiently delivers biotinylated FITC into cancer cells via TfR-mediated endocytosis. In addition, anti-hTfR IgG3-Av alone exhibits intrinsic cytotoxic activity and interferes with hTfR recycling, leading to the rapid degradation of the TfR and lethal iron deprivation in certain malignant B-cell lines. We now report on the cytotoxic effects of a conjugate composed of anti-hTfR IgG3-Av and biotinylated saporin 6 (b-SO6), a toxin derived from the plant Saponaria officinalis that inhibits protein synthesis. Conjugation of anti-hTfR IgG3-Av with b-SO6 enhances the cytotoxic effect of the fusion protein in sensitive cells and also overcomes the resistance of malignant cells that show low sensitivity to the fusion protein alone. Our results show for the first time that loading anti-hTfR IgG3-Av with a biotinylated toxin enhances the cytotoxicity of the fusion protein alone. These results suggest that anti-hTfR IgG3-Av has great potential as a therapeutic agent for a wide range of applications due to its intrinsic cytotoxic activity plus its ability to deliver biotinylated molecules into cancer cells.


Asunto(s)
Avidina/metabolismo , Biotinilación , Neoplasias Hematológicas/patología , Inmunoglobulina G/farmacología , Proteínas de Plantas/metabolismo , Receptores de Transferrina/antagonistas & inhibidores , Proteínas Recombinantes de Fusión/metabolismo , Proteínas Inactivadoras de Ribosomas Tipo 1/metabolismo , Caspasas/metabolismo , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Ensayos de Selección de Medicamentos Antitumorales , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos , Activación Enzimática/efectos de los fármacos , Neoplasias Hematológicas/enzimología , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/efectos de los fármacos , Humanos , Deficiencias de Hierro , Biosíntesis de Proteínas/efectos de los fármacos , Receptores de Transferrina/metabolismo , Saporinas
15.
Integr Biol (Camb) ; 10(3): 136-144, 2018 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-29488523

RESUMEN

Herein, a microfluidic device with cistern design for cultivation of adherent eukaryotic cells for the production of recombinant proteins is presented. The geometric configuration of the microchannels in the device provided laminar flow with reduced velocity profiles in the cisterns, resulting in an adequate microenvironment for long-term adherent cell growth with passive pumping flow cycles of 24 hours. CHO-ahIFNα2b and HEK-ahIFNα2b adherent cell lines expressing a novel anti-hIFN-α2b recombinant monoclonal antibody (MAb) for the treatment of systemic lupus erythematosus were cultured on the surface of PDMS/glass microchannels coated with poly-d-lysine. A 24 day culture of CHO-ahIFNα2b cells resulted in MAb concentrations up to 166.4 µg mL-1 per day. The productivity of CHO-ahIFNα2b and HEK-ahIFNα2b cell lines was higher in the microdevice compared to that obtained using the adherent cell culture method (T-flask), with a 5.89- and 7.31-fold increase, respectively. Moreover, biological analysis of the MAbs produced in the microdevice showed no significant differences in the neutralizing antiproliferative activity of the hIFN-α2b or the cytokine cell signaling compared to the MAbs produced with cell adherent methods. These results suggest that this microfluidic device is suitable for long-term culture of mammalian cells and can improve the productivity of cells expressing recombinant MAbs with potential for therapeutic use without affecting the quality attributes of the product.


Asunto(s)
Anticuerpos Monoclonales/química , Técnicas de Cultivo de Célula/métodos , Dispositivos Laboratorio en un Chip , Animales , Células CHO , Adhesión Celular , Proliferación Celular , Cricetinae , Cricetulus , Medios de Cultivo , Dimetilpolisiloxanos/química , Diseño de Equipo , Vidrio , Células HEK293 , Humanos , Lupus Eritematoso Sistémico/terapia , Polilisina/química , Proteínas Recombinantes/química
16.
PLoS One ; 13(3): e0193605, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29494694

RESUMEN

Over the past few years, image analysis has emerged as a powerful tool for analyzing various cell biology parameters in an unprecedented and highly specific manner. The amount of data that is generated requires automated methods for the processing and analysis of all the resulting information. The software available so far are suitable for the processing of fluorescence and phase contrast images, but often do not provide good results from transmission light microscopy images, due to the intrinsic variation of the acquisition of images technique itself (adjustment of brightness / contrast, for instance) and the variability between image acquisition introduced by operators / equipment. In this contribution, it has been presented an image processing software, Python based image analysis for cell growth (PIACG), that is able to calculate the total area of the well occupied by cells with fusiform and rounded morphology in response to different concentrations of fetal bovine serum in microfluidic chips, from microscopy images in transmission light, in a highly efficient way.


Asunto(s)
Técnicas de Cultivo de Célula/instrumentación , Procesamiento de Imagen Asistido por Computador/métodos , Técnicas Analíticas Microfluídicas/instrumentación , Células HEK293 , Humanos , Microscopía de Contraste de Fase , Programas Informáticos
17.
Nat Struct Mol Biol ; 25(2): 131-134, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29335561

RESUMEN

The atomic structure of the infectious, protease-resistant, ß-sheet-rich and fibrillar mammalian prion remains unknown. Through the cryo-EM method MicroED, we reveal the sub-ångström-resolution structure of a protofibril formed by a wild-type segment from the ß2-α2 loop of the bank vole prion protein. The structure of this protofibril reveals a stabilizing network of hydrogen bonds that link polar zippers within a sheet, producing motifs we have named 'polar clasps'.


Asunto(s)
Amiloide/química , Microscopía por Crioelectrón , Enlace de Hidrógeno , Priones/química , Proteínas Amiloidogénicas/química , Animales , Carbamazepina/química , Bovinos , Cricetinae , Ciervos , Electrones , Humanos , Ratones , Péptidos/química , Filogenia , Estructura Secundaria de Proteína , Proteoma , Ovinos , Propiedades de Superficie , Difracción de Rayos X
18.
Mol Cancer Ther ; 5(4): 1029-40, 2006 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-16648575

RESUMEN

We have previously generated antihuman HER2/neu-humanized IgG3 fused to interleukin-2 (IL-2), IL-12, or granulocyte macrophage colony-stimulating factor (GM-CSF) [monofunctional fusion proteins (mono-AbFP)] or fused to IL-2 and IL-12 or IL-12 and GM-CSF [bifunctional fusion proteins (bi-AbFP)]. These AbFPs retained cytokine and antigen-binding activities. We have now further characterized the AbFPs and determined the heparin-binding activity of the fused cytokines, their ability to trigger IFN-gamma secretion and natural killer (NK) activation, and their direct antitumor efficacy. Flow cytometry revealed heparin-binding activity in the AbFPs containing IL-12 and IL-2, although this activity seems to be decreased in the bi-AbFPs. However, both bi-AbFPs retained the capacity to stimulate IL-12-dependent IFN-gamma secretion in the NK cell line KY-1, and IL-12/IL-2 bi-AbFP induced NK activity in splenocytes. The antitumor effectiveness of bi-AbFPs and mono-AbFP combinations was studied in mice challenged i.p. with three different human HER2/neu murine syngeneic models (D2F2/E2, CT26-HER2/neu, and MC38-HER2/neu). Although a significant variability in the profile of antitumor response was observed in the different tumor models, the combination of IL-12 and GM-CSF mono-AbFPs protected 100% of D2F2/E2-challenged and 75% of CT26-HER2/neu-challenged mice. In contrast, bi-AbFPs protected less than the combination of mono-AbFPs and, in some models, even less than mono-AbFPs alone. However, in all cases, most of long-term survivors showed protection after s.c. rechallenge with the tumors and later with the parental tumors not expressing HER2/neu. These results show that, although the pattern of protection is tumor model dependent, treatments with AbFPs can effectively generate high levels of protection against peritoneal tumors expressing HER2/neu, which may be relevant in patients with primary or metastatic peritoneal carcinomatosis that may be observed in ovarian, colon, stomach, bladder, lung, and breast cancers.


Asunto(s)
Anticuerpos/uso terapéutico , Neoplasias del Colon/inmunología , Citocinas/inmunología , Citocinas/uso terapéutico , Genes erbB-2 , Mieloma Múltiple/inmunología , Animales , Línea Celular Tumoral , Neoplasias del Colon/tratamiento farmacológico , Genes erbB-2/efectos de los fármacos , Factor Estimulante de Colonias de Granulocitos y Macrófagos/uso terapéutico , Interleucina-12/uso terapéutico , Interleucina-2/uso terapéutico , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Mieloma Múltiple/tratamiento farmacológico
20.
Bol Med Hosp Infant Mex ; 73(6): 372-379, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-29421281

RESUMEN

Iron is essential for cell growth and is imported into cells in part through the action of transferrin (Tf), a protein that binds its receptor (TfR1 or CD71) on the surface of a cell, and then releases iron into endosomes. TfR1 is a single pass type-II transmembrane protein expressed at basal levels in most tissues. High expression of TfR1 is typically associated with rapidly proliferating cells, including various types of cancer. TfR1 is targeted by experimental therapeutics for several reasons: its cell surface accessibility, constitutive endocytosis into cells, essential role in cell growth and proliferation, and its overexpression by cancer cells. Among the therapeutic agents used to target TfR1, antibodies stand out due to their remarkable specificity and affinity. Clinical trials are being conducted to evaluate the safety and efficacy of agents targeting TfR1 in cancer patients with promising results. These observations suggest that therapies targeting TfR1 as direct therapeutics or delivery conduits remain an attractive alternative for the treatment of cancers that overexpress the receptor.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA