Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 58
Filtrar
Más filtros

Bases de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Mol Cell Biochem ; 479(3): 579-590, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-37129769

RESUMEN

Antibody drug conjugates (ADCs) with twelve FDA approved drugs, known as a novel category of anti-neoplastic treatment created to merge the monoclonal antibody specificity with cytotoxicity effect of chemotherapy. However, despite many undeniable advantages, ADCs face certain problems, including insufficient internalization after binding, complex structures and large size of full antibodies especially in targeting of solid tumors. Camelid single domain antibody fragments (Nanobody®) offer solutions to this challenge by providing nanoscale size, high solubility and excellent stability, recombinant expression in bacteria, in vivo enhanced tissue penetration, and conjugation advantages. Here, an anti-human CD22 Nanobody was expressed in E.coli cells and conjugated to Mertansine (DM1) as a cytotoxic payload. The anti-CD22 Nanobody was expressed and purified by Ni-NTA resin. DM1 conjugated anti-CD22 Nanobody was generated by conjugation of SMCC-DM1 to Nanobody lysine groups. The conjugates were characterized using SDS-PAGE and Capillary electrophoresis (CE-SDS), RP-HPLC, and MALDI-TOF mass spectrometry. Additionally, flow cytometry analysis and a competition ELISA were carried out for binding evaluation. Finally, cytotoxicity of conjugates on Raji and Jurkat cell lines was assessed. The drug-to-antibody ratio (DAR) of conjugates was calculated 2.04 using UV spectrometry. SDS-PAGE, CE-SDS, HPLC, and mass spectrometry confirmed conjugation of DM1 to the Nanobody. The obtained results showed the anti-CD22 Nanobody cytotoxicity was enhanced almost 80% by conjugation with DM1. The binding of conjugates was similar to the non-conjugated anti-CD22 Nanobody in flow cytometry experiments. Concludingly, this study successfully suggest that the DM1 conjugated anti-CD22 Nanobody can be used as a novel tumor specific drug delivery system.


Asunto(s)
Inmunoconjugados , Maitansina , Neoplasias , Anticuerpos de Dominio Único , Anticuerpos Monoclonales/farmacología , Antineoplásicos/inmunología , Línea Celular Tumoral , Inmunoconjugados/química , Inmunoconjugados/uso terapéutico , Maitansina/química , Neoplasias/tratamiento farmacológico , Lectina 2 Similar a Ig de Unión al Ácido Siálico/inmunología , Camelidae/inmunología
2.
Prep Biochem Biotechnol ; 54(3): 307-316, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-37452673

RESUMEN

Inhibition of FGFR2 signaling is promising in targeted therapy of FGFR2-related tumors. In this study, anti-FGFR2 nanobodies (Nbs) were isolated through screening of an immune camelid phage display library. Four rounds of biopanning were carried out with commercial human FGFR2 antigen and enrichment was assessed by ELISA and phage titration. The gene of Nb was sub-cloned into the expression vector, and the recombinant vector was transformed into Escherichia coli WK6 cells. The recombinant protein was purified using Ni-NTA affinity chromatography. The anti-FGFR2 Nb (C13) was characterized by SDS-PAGE, western blotting, competitive inhibition ELISA, flow cytometry, MTT, and migration assay. C13 Nb recognized FGFR2 with high specificity and no cross-reactivity was observed with other tested antigens. The affinity of C13 Nb was calculated to be 1.5 × 10-9 M. Results of cytotoxicity showed that C13 Nb (10 µg/ml) inhibited 85% of the proliferation of T-47D cells (p < 0.001). In addition, C13 inhibited the migration of 68% of T-47D toward the source of the growth factor (p < 0.01). The flow cytometry showed that C13 Nb bound to the surface of FGFR2+ cells, T-47D cell line (96%). Results indicate the potential of anti-FGFR2 Nb for targeted therapy of FGFR2-overexpressing tumors after complementary investigations.


Asunto(s)
Bacteriófagos , Neoplasias , Humanos , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/genética , Bioprospección , Western Blotting , Escherichia coli/genética
3.
J Cell Mol Med ; 26(23): 5929-5942, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36412036

RESUMEN

Different growth factors can regulate stem cell differentiation. We used keratinocyte growth factor (KGF) to direct adipose-derived stem cells (ASCs) differentiation into keratinocytes. To enhance KGF bioavailability, we targeted KGF for collagen by fusing it to collagen-binding domain from Vibrio mimicus metalloprotease (vibrioCBD-KGF). KGF and vibrioCBD-KGF were expressed in Escherichia coli and purified to homogeneity. Both proteins displayed comparable activities in stimulating proliferation of HEK-293 and MCF-7 cells. vibrioCBD-KGF demonstrated enhanced collagen-binding affinity in immunofluorescence and ELISA. KGF and vibrioCBD-KGF at different concentrations (2, 10, and 20 ng/ml) were applied for 21 days on ASCs cultured on collagen-coated plates. Keratinocyte differentiation was assessed based on morphological changes, the expression of keratinocyte markers (Keratin-10 and Involucrin), and stem cell markers (Collagen-I and Vimentin) by real-time PCR or immunofluorescence. Our results indicated that the expression of keratinocyte markers was substantially increased at all concentrations of vibrioCBD-KGF, while it was observed for KGF only at 20 ng/ml. Immunofluorescence staining approved this finding. Moreover, down-regulation of Collagen-I, an indicator of differentiation commitment, was more significant in samples treated with vibrioCBD-KGF. The present study showed that vibrioCBD-KGF is more potent in inducing the ASCs differentiation into keratinocytes compared to KGF. Our results have important implications for effective skin regeneration using collagen-based biomaterials.


Asunto(s)
Diferenciación Celular , Factor 7 de Crecimiento de Fibroblastos , Queratinocitos , Células Madre , Humanos , Colágeno , Colágeno Tipo I/genética , Factor 7 de Crecimiento de Fibroblastos/farmacología , Células HEK293 , Queratinocitos/citología , Queratinocitos/efectos de los fármacos , Células Madre/citología , Células Madre/efectos de los fármacos
4.
Anal Biochem ; 652: 114745, 2022 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-35609685

RESUMEN

Expression of proteins in bacterial host cells, particularly E.coli, has gained much attention in recent years. Low expression outcome is the main technical drawback associated with this procedure, further restricting its largescale application in industry. Therefore, application of new amendments or reformations are required before further proceedings. Extremely low frequency magnetic fields (ELF-MFs) have shown to significantly affect biological processes, including gene expression, in E.coli. In current study, we investigated whether application of ELF-MF could result in overexpression of proteins in E.coli or not. Cluster of differentiation-22 (CD22), as a model protein, was expressed in E.Coli Rosetta (DE3) under continuous exposure to ELF-MF after applying various concentrations of Isopropyl ß-d-1-thiogalactopyranoside (IPTG) (0.25-1.25 mM) as inducer. The strength and frequency of electromagnetic fields (EMFs) ranged between 15 and 100 mT and 2.5-20 Hz respectively. Interestingly, application of 55 mT EMFs with frequencies ranging from 2.5 to 2.8 Hz significantly enhanced the yield of expression at all studied IPTG concentrations. Contrarily, EMFs with intensities other than 55 mT meaningfully declined protein expression at IPTG concentrations equal to 1 and 1.25 mM. In conclusion, application of specific range of ELF-MFs may be exploited as a new modification for enhancing heterologous expression of proteins in E.coli.


Asunto(s)
Campos Electromagnéticos , Campos Magnéticos , Isopropil Tiogalactósido , Proteínas Recombinantes/genética
5.
Biochem Biophys Res Commun ; 565: 14-20, 2021 08 06.
Artículo en Inglés | MEDLINE | ID: mdl-34087508

RESUMEN

Regardless of their target and mechanism, anticancer drugs directly influence biological behavior of cancer cells by activating chemical signaling pathways. Due to the complex interaction between diverse signaling pathways, these drugs may profoundly impact the physical characteristics of cancer cells and regulate their mechanical properties. In this study, the effects of two Aromatase Inhibitor (Letrozole and Exemestane), and one mTOR Inhibitor (Everolimus) on cell mechanical properties, actin content/distribution, and nuclear areas of two invasive and non-invasive breast cancer cell line after 24 h treatment with concentrations previously reported were investigated. While metabolic activity of cell lines was highly affected by drug treatment, significant alterations in Young's modulus of cell bodies, nuclear areas, and actin content and distribution were reported with higher impact on invasive cells. It was concluded that regulation of mechanical behavior of cells by all three drugs emphasizes the cross talk between chemical and physical signaling cascades, and describes a correlation between biological and physical behaviors of cancer cells which might give an insight to a better understanding of mechanisms by which anti-cancer drugs function to enhance their performances.


Asunto(s)
Androstadienos/farmacología , Antineoplásicos/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Citoesqueleto/efectos de los fármacos , Everolimus/farmacología , Letrozol/farmacología , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Supervivencia Celular/efectos de los fármacos , Citoesqueleto/metabolismo , Femenino , Humanos , Células Tumorales Cultivadas
6.
J Cell Physiol ; 234(3): 2693-2704, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30246298

RESUMEN

Targeted therapy using specific monoclonal antibodies (mAbs) conjugated to chemotherapeutic agents or toxins has become one of the top priorities in cancer therapy. Antibody-drug conjugates (ADCs) are emerging as a promising strategy for cancer-targeted therapy. In this study, trastuzumab, a humanized monoclonal anti-HER2 antibody, was reduced by dithiothreitol and conjugated to the microtubule-disrupting agent monomethyl auristatin E (MMAE) through a valine-citrulline peptide linker (trastuzumab-MC-Val-Cit-PABC-MMAE [trastuzumab-vcMMAE]). After conjugation, ADCs were characterized by using UV-vis, sodium dodecyl sulfate polyacrylamide gel electrophoresis (SDS-PAGE), and flow cytometry. The antitumor activity of the ADC was evaluated in breast cancer cells in vitro. In addition, ADCs were further characterized using purification by the protein A chromatography, followed by assessment using apoptosis and MTT (3-(4,5-dimethylthiazol-2-yl)-2, 5-diphenyltetrazolium bromide) assays. Hydrophobic interaction chromatography was used to determine drug-to-antibody ratio species of ADCs produced. Our finding showed that approximately 5.12 drug molecules were conjugated to each mAb. H2L2, H2L, HL, H2, H, and L forms of ADCs were detected in nonreducing SDS-PAGE. The binding of trastuzumab-vcMMAE to HER2-positive cells was comparable with that of the parental mAb. The MTT assay showed that our ADCs induced significant cell death in HER2-positive cells, but not in HER2-negative cells. The ADCs produced was a mixture of species, unconjugated trastuzumab (14.147%), as well as trastuzumab conjugated with two (44.868%), four (16.886%), six (13.238%), and eight (10.861%) molecules of MMAE. These results indicated that MMAE-conjugated trastuzumab significantly increases the cytotoxic activity of trastuzumab, demonstrating high affinity, specificity, and antitumor activity in vitro. Trastuzumab-vcMMAE is an effective and selective agent for the treatment of HER2-positive breast tumors.


Asunto(s)
Neoplasias de la Mama/tratamiento farmacológico , Oligopéptidos/farmacología , Receptor ErbB-2/metabolismo , Trastuzumab/farmacología , Anticuerpos Monoclonales/farmacología , Antineoplásicos/farmacología , Antineoplásicos Inmunológicos/farmacología , Línea Celular Tumoral , Humanos , Inmunoconjugados/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto/métodos
7.
Microb Pathog ; 122: 122-129, 2018 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-29908308

RESUMEN

BACKGROUND: Despite the fact that pneumococcal conjugate vaccines (PCVs) have significantly reduced the rate of invasive pneumococcal diseases through the use of vaccine serotypes, infection with Streptococcus pneumoniae remains a major public health hazard. Serotype-independent vaccines that are economically viable species of common protein antigens such as whole-cell vaccines (WCVs) are needed. Considering the ongoing debate about the effectiveness of WCVs, a systematic literature review and meta-analysis was carried out to determine the efficacy of WCVs against colonization in mice. MATERIAL AND METHODS: A systematic review was undertaken of published studies on the protection (colonized/uncolonized) of whole cell pneumococcal vaccine in mice. The search terms used were "whole cell vaccine" and "Streptococcus pneumoniae" in PubMed, Google Scholar, Embase, Web of Science and Scopus engines. Data was extracted from original publications and a meta-analysis was performed on studies divided into sub-groups by the number of inoculations, type of sample, type of adjuvant, time of sampling, design of study and quality of study. RESULTS: Ten eligible articles published from 2000 to 2016 were included in this review. The meta-analysis was performed on eight out of 10 studies and demonstrated that the estimated pooled risk ratios (RRs) for comparison of colonization between the vaccinated and unvaccinated mice for outcomes 1 and 2 were 0.18 and 0.24, respectively. Lower RRs were observed in sub-groups that were inoculated with vaccines three times, those using cholera toxin (CT) adjuvants and those obtained as tracheal specimens from the mice. CONCLUSIONS: The best protocol for use of a WCV is its application with CT adjuvant administered intranasally in three inoculations at doses of 108 CFU. Further studies performed under similar conditions to obtain accurate results on the effectiveness of this vaccine are recommended.


Asunto(s)
Infecciones Neumocócicas/prevención & control , Vacunas Neumococicas/inmunología , Vacunas de Productos Inactivados/inmunología , Adyuvantes Inmunológicos/administración & dosificación , Administración Intranasal , Animales , Modelos Animales de Enfermedad , Ratones , Vacunas Neumococicas/administración & dosificación , Vacunas de Productos Inactivados/administración & dosificación
8.
Anal Biochem ; 544: 34-41, 2018 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-29246751

RESUMEN

Antibody-based targeting of angiogenesis is a key approach for cancer treatment. Delta-like ligand 4 (DLL4) plays a pivotal role in tumor neovascular development and angiogenesis during tumor progression. It forecasts the prognosis of human malignancies and blocking its signaling can help to inhibit neovascularization and tumor metastasis. Nanobodies are the smallest antigen-binding domains of heavy chain antibodies in camelidae. The aim of this study was to develop a Nanobody against DLL4 and apply binding and functional approaches to target it. In this work, a Nanobody library against human recombinant DLL4 was developed. After panning, the periplasmic-extract (PE) of individual colonies were screened through ELISA. The interactions between Nanobody and DLL4 were assessed using immunohistochemistry and FACS. The functional assessment was carried out via tube formation assay. We selected a Nanobody (3Nb3) with a high binding signal to DLL4, associated with a binding affinity of 3.6 nM. It was demonstrated that 3Nb3 binds to native DLL4 on the surface of MKN cells and gastric carcinoma tissue, and also inhibits the maturation of capillary-like structures in HUVECs. The results were indicative of the potential of Nanobody for DLL4 identification and can broaden the scope for development of cancer diagnosis and treatment techniques.


Asunto(s)
Péptidos y Proteínas de Señalización Intercelular/inmunología , Neovascularización Patológica/inmunología , Anticuerpos de Dominio Único/inmunología , Neoplasias Gástricas/inmunología , Proteínas Adaptadoras Transductoras de Señales , Animales , Proteínas de Unión al Calcio , Línea Celular , Ensayo de Inmunoadsorción Enzimática , Humanos , Neovascularización Patológica/diagnóstico , Neoplasias Gástricas/diagnóstico
9.
Biotechnol Appl Biochem ; 65(5): 718-725, 2018 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-29543347

RESUMEN

CD22 is a B-cell-specific trans-membrane glycoprotein, which is found on the surface of the most B cells and modulates their function, survival, and apoptosis. Recently, targeting this cell surface biomarker in B-cell malignancies and disorders has attracted a lot of attention. The variable domain of camelid single-chain antibodies (VHH, nanobody) is a form of antibodies with novel properties including small size (15-17 kDa), thermal and chemical stability, high affinity and homology to human antibody sequences. In this study, a novel anti-CD22-specific VHH (Nb) has been developed and characterized by the screening of an immunized phage display library and its binding to CD22+ B cells is evaluated. Produced anti-CD22 VHH had a single protein band about 17 kDa of molecular size in Western blotting and its binding affinity was approximately 9 × 10-9  M. Also, this product had high specificity and it was able to recognize the natural CD22 antigen in CD22+ cell lysate as well as on the cell surface (93%). This anti-CD22 VHH with both high affinity and specificity recognizes CD22 antigen well and can be used in diagnosis and treatment of B cell disorders and malignancies.


Asunto(s)
Lectina 2 Similar a Ig de Unión al Ácido Siálico/inmunología , Anticuerpos de Dominio Único/inmunología , Animales , Afinidad de Anticuerpos , Formación de Anticuerpos , Especificidad de Anticuerpos , Biomarcadores/metabolismo , Western Blotting , Camelus , Electroforesis en Gel de Poliacrilamida , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Humanos , Masculino
10.
Immunopharmacol Immunotoxicol ; 40(5): 368-374, 2018 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-30183441

RESUMEN

OBJECTIVES: Angiogenesis targeting is an attractive approach for cancer treatment. Delta-like ligand 4 (DLL4) plays a pivotal role in neovascular development and its inhibitors have recently entered clinical trials for solid tumors. The aim of this study was to evaluate the possibilities of using anti-DLL4 antibody fragment as an angiogenesis maturation inhibitor. MATERIALS AND METHODS: In this study, a DLL4-specific Nanobody, named 3Nb3, was selected and assessed by western blotting and internalization assays. Functional assessments included MTT, apoptosis, and chicken chorioallantoic membrane (CAM) assays. RESULTS: Based on the results, 3Nb3 specifically binds to DLL4 and internalizes into MKN cell. Furthermore, 3Nb3 significantly inhibited the proliferation of cells and also neovascularization in the CAM. CONCLUSIONS: These data demonstrated the potential of Nanobody for application in targeting DLL4. Our findings may provide a basis for the development of novel therapeutic techniques to inhibit growth and neovascularization of tumors.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Anticuerpos Monoclonales/farmacología , Proliferación Celular/efectos de los fármacos , Fragmentos de Inmunoglobulinas/farmacología , Péptidos y Proteínas de Señalización Intracelular/inmunología , Proteínas de la Membrana/inmunología , Neovascularización Patológica/tratamiento farmacológico , Animales , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Embrión de Pollo , Membrana Corioalantoides/efectos de los fármacos , Células HEK293 , Humanos
11.
Anal Biochem ; 511: 27-30, 2016 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-27156812

RESUMEN

Antibody engineering involves a range of custom modifications of immunoglobulins to improve their affinity, valency, and pharmacokinetics, ensuring a better target therapy achievement. A number of therapeutic antibodies have been used for cell surface receptor blockage, interfering with the ligand binding and inhibiting receptor-driven activation of cells. Here we describe the construction and characterization of a recombinant bivalent single-chain Fv (biscFv) that targets CD123. On conversion of anti-CD123 scFv to biscFv format, the recognition of the cognate ligand is not altered. Moreover, the increased overall efficacy of the anti-CD123 biscFv in binding and inhibition of CD123/IL-3 (interleukin-3) interactions in TF-1 cells is demonstrated.


Asunto(s)
Anticuerpos Biespecíficos , Subunidad alfa del Receptor de Interleucina-3/antagonistas & inhibidores , Anticuerpos de Cadena Única , Animales , Anticuerpos Biespecíficos/biosíntesis , Anticuerpos Biespecíficos/genética , Anticuerpos Biespecíficos/inmunología , Anticuerpos Biespecíficos/farmacología , Línea Celular , Humanos , Interleucina-3/inmunología , Subunidad alfa del Receptor de Interleucina-3/inmunología , Ratones , Anticuerpos de Cadena Única/biosíntesis , Anticuerpos de Cadena Única/genética , Anticuerpos de Cadena Única/inmunología , Anticuerpos de Cadena Única/farmacología
12.
Protein Expr Purif ; 123: 19-25, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-26996993

RESUMEN

Angiogenesis is the formation of new blood vessels which is involved in migration, growth and differentiation of endothelial cells. This process regularly occurs during growth and development in children however, in adults is usually part of a disease process such as cancer. The vascular endothelial growth factor (VEGF) is a vital player in the vascular development and angiogenesis in physiological and pathological processes. Camelid's immune system has unique antibodies which are composed of only a heavy chain homodimer and the variable domain (VHH, Nanobody). Nanobodies are small, around 15 kDa and stable. In this study, we engineered and constructed a new Nanobody-Fc fusion protein (fusionbody) composed of an anti-VEGFR2 Nanobody and an Fc fragment of human IgG1 antibody. The recombinant vector was transfected into NS0 host cells. Stable producer clones were developed and the recombinant fusionbody was expressed and purified. Functional assay showed the anti-VEGFR2 fusionbody could bind to VEGFR2 on cell surface via VHH part and could mediate killing the targeted cells through direct cell death and complement-dependent cytotoxicity (CDC).


Asunto(s)
Fragmentos Fc de Inmunoglobulinas/inmunología , Anticuerpos de Dominio Único/inmunología , Receptor 2 de Factores de Crecimiento Endotelial Vascular/inmunología , Animales , Muerte Celular , Línea Celular , Clonación Molecular , Proteínas del Sistema Complemento/inmunología , Citotoxicidad Inmunológica , Humanos , Fragmentos Fc de Inmunoglobulinas/genética , Ratones , Ingeniería de Proteínas , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/inmunología , Anticuerpos de Dominio Único/genética
13.
Mol Cell Biochem ; 423(1-2): 129-139, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27696309

RESUMEN

Loss of cell-cell adhesion function is a common characteristic of many human epithelial carcinomas that is frequently due to loss of E-cadherin expression. In cancer progression, loss of E-cadherin is associated with invasion and metastasis potential, hence restoration of its function may contribute to the metastasis inhibition. This study examined effect of Epidermal Growth Factor Receptor (EGFR/Her1) blockade on the E-cadherin expression, cellular adherence, and cell elasticity in two human epithelial cancer cell lines, MCF7 and A431. EGFR blocking agents as antibodies or small molecules target EGFR directly. Furthermore, due to intracellular signaling pathways they influence cell behavior and activities. The idea here is to investigate the effect of reduced activity of this signaling pathway using anti-EGFR Antibody (Cetuximab) and tyrosine kinase inhibitor (Lapatinib) on cell-cell adhesion and cell mechanical properties. Real-Time PCR analysis demonstrated that treatment of cells with considered drugs increased the expression of E-cadherin gene among samples. The atomic force microscopy-based single cell force spectroscopy technique was used to measure adhesive force of cancerous cells. Results indicated that inhibition of EGFR activity elevated cell-cell adhesion force, accompanied by stiffening of the cell bodies. In summary, Cetuximab and Lapatinib have been found to mediate cell-cell adhesion by restoration of E-cadherin expression and function. Our data suggest possible therapeutic potential for inhibition of metastasis via the blockade of EGFR signaling.


Asunto(s)
Neoplasias de la Mama , Cetuximab/farmacología , Receptores ErbB/metabolismo , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Proteínas de Neoplasias/metabolismo , Quinazolinas/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/ultraestructura , Adhesión Celular/efectos de los fármacos , Femenino , Humanos , Lapatinib , Células MCF-7 , Microscopía de Fuerza Atómica
14.
FASEB J ; 28(9): 4004-14, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24891523

RESUMEN

Hemiscorpius lepturus scorpionism poses one of the most dangerous health problems in many parts of the world. The common therapy consists of using antivenom antibody fragments derived from a polyclonal immune response raised in horses. However, this immunotherapy creates serious side effects, including anaphylactic shock sometimes even leading to death. Thus, many efforts have been made to introduce new replacement therapeutics that cause less adverse reactions. One of the most attractive approaches to replacing the available therapy is offered by single-domain antibody fragments, or nanobodies (Nbs). We immunized dromedaries with H. lepturus toxin and identified a functional recombinant Nb (referred to as F7Nb) against heminecrolysin (HNc), the major known hemolytic and dermonecrotic fraction of H. lepturus venom. This Nb was retrieved from the immune library by phage display selection. The in vitro neutralization tests indicated that 17.5 nmol of the F7Nb can inhibit 45% of the hemolytic activity of 1 EC100 (7.5 µg/ml) of HNc. The in vivo neutralization tests demonstrated that F7Nb had good antihemolytic and antidermonecrotic effects against HNc in all tested mice. Surprisingly, F7Nb (8.75 nmol) neutralized 1 LD100 of HNc (10 µg) via an intracerebroventricular route or 1 LD100 (80 µg) via a subcutaneous route. All of the control mice died. Hence, this Nb is a potential leading novel candidate for treating H. lepturus scorpionism in the near future.


Asunto(s)
Anticuerpos Neutralizantes/inmunología , Antivenenos/uso terapéutico , Camelus/inmunología , Picaduras de Escorpión/terapia , Venenos de Escorpión/química , Escorpiones/metabolismo , Anticuerpos de Dominio Único/uso terapéutico , Secuencia de Aminoácidos , Animales , Western Blotting , Ensayo de Inmunoadsorción Enzimática , Eritrocitos/efectos de los fármacos , Femenino , Hemólisis/efectos de los fármacos , Inmunización , Ratones , Ratones Endogámicos BALB C , Datos de Secuencia Molecular , Necrosis , Picaduras de Escorpión/inmunología , Picaduras de Escorpión/parasitología , Enfermedades de la Piel/patología , Enfermedades de la Piel/prevención & control
15.
Protein Expr Purif ; 116: 139-43, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26297626

RESUMEN

Cluster of Differentiation 90 (CD90, Thy-1) has been proposed as one of the most important biomarkers in several cancer cells including cancer stem cells (CSCs). CD90 is considered as a potential normal stem cell and CSCs biomarker and also has been identified in lung cancer stem cells, hepatocellular carcinoma cells and high-grade gliomas. Using eukaryotic host systems involves complex procedures and frequently results in low protein yields. The expression of recombinant proteins in Escherichia coli is comparatively easier than eukaryotic host cells. The potential of large scale production of recombinant protein has made this system an economic production platform. In this study we expressed the extra-membrane domain of human CD90 (exCD90) antigen (Gln15-Cys130) in E. coli expression host cells. The epitope integrity of purified recombinant antigen was confirmed by antibody-antigen interaction using 5E10 anti-CD90 monoclonal antibody and binding study through ELISA and florescent staining of CD90(+) cells in a flow cytometry experiment.


Asunto(s)
Escherichia coli/genética , Antígenos Thy-1/genética , Antígenos Thy-1/inmunología , Animales , Anticuerpos/inmunología , Reacciones Antígeno-Anticuerpo , Secuencia de Bases , Clonación Molecular , Ensayo de Inmunoadsorción Enzimática , Femenino , Humanos , Datos de Secuencia Molecular , Estructura Terciaria de Proteína , Conejos , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/inmunología , Antígenos Thy-1/química
16.
Cell J ; 26(1): 51-61, 2024 Jan 31.
Artículo en Inglés | MEDLINE | ID: mdl-38351729

RESUMEN

OBJECTIVE: Type 1 diabetes (T1Ds) is an autoimmune disease in which the immune system invades and destroys insulin-producing cells. Nevertheless, at the time of diagnosis, about 30-40% of pancreatic beta cells are healthy and capable of producing insulin. Bi-specific antibodies, chimeric antigen receptor regulatory T cells (CAR-Treg cells), and labeled antibodies could be a new emerging option for the treatment or diagnosis of type I diabetic patients. The aim of the study is to choose appropriate cell surface antigens in the pancreas tissue for generating an antibody for type I diabetic patients. MATERIALS AND METHODS: In this bioinformatics study, we extracted pancreas-specific proteins from two large databases; the Human Protein Atlas (HPA) and Genotype-Tissue Expression (GTEx) Portal. Pancreatic-enriched genes were chosen and narrowed down by Protter software for the investigation of accessible extracellular domains. The immunohistochemistry (IHC) data of the protein atlas database were used to evaluate the protein expression of selected antigens. We explored the function of candidate antigens by using the GeneCards database to evaluate the potential dysfunction or activation/hyperactivation of antigens after antibody binding. RESULTS: The results showed 429 genes are highly expressed in the pancreas tissue. Also, eighteen genes encoded plasma membrane proteins that have high expression in the microarray (GEO) dataset. Our results introduced four structural proteins, including NPHS1, KIRREL2, GP2, and CUZD1, among all seventeen candidate proteins. CONCLUSION: The presented antigens can potentially be used to produce specific pancreatic antibodies that guide CARTreg, bi-specific, or labeling molecules to the pancreas for treatment, detection, or other molecular targeted therapy scopes for type I diabetes.

17.
Sci Rep ; 14(1): 3689, 2024 02 14.
Artículo en Inglés | MEDLINE | ID: mdl-38355744

RESUMEN

In type 1 diabetes, the immune system destroys pancreatic beta cells in an autoimmune condition. To overcome this disease, a specific monoclonal antibody that binds to pancreatic beta cells could be used for targeted immunotherapy. Protein tyrosine phosphatase receptor N (PTPRN) is one of the important surface antigen candidates. Due to its high sequence homology among mammals, so far, no single-chain monoclonal antibody has been produced against this receptor. In this study, we developed a novel single-chain variable fragment (scFv) against the PTPRN extracellular domain. To this aim, ostrich species was used as a host is far phylogenetically birds from mammals to construct a phage display library for the first time. An ostrich-derived scfv phage display library was prepared and biopanning steps were done to enrich and screen for isolating the best anti-PTPRN binders. An scFv with appropriate affinity and specificity to the PTPRN extracellular domain was selected and characterized by ELISA, western blotting, and flow cytometry. The anti-PTPRN scFv developed in this study could be introduced as an effective tool that can pave the way for the creation of antibody-based targeting systems in cooperation with the detection and therapy of type I diabetes.


Asunto(s)
Anticuerpos de Cadena Única , Struthioniformes , Animales , Struthioniformes/metabolismo , Biblioteca de Péptidos , Monoéster Fosfórico Hidrolasas/metabolismo , Anticuerpos Monoclonales , Ensayo de Inmunoadsorción Enzimática , Mamíferos/metabolismo
18.
Mol Biotechnol ; 65(12): 1968-1978, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-36906729

RESUMEN

As a member of the tumor necrosis factor (TNF) superfamily, the B-cell activating factor (BAFF) plays a crucial role in B-cell survival and differentiation. Overexpression of this protein has been closely linked to autoimmune disorders and some B-cell malignancies. Using monoclonal antibodies (mAbs) against the BAFF soluble domain appears to be a complementary treatment for some of these diseases. This study aimed to produce and develop a specific Nanobody (Nb), a variable camelid antibody domain, against the soluble domain of BAFF protein. After camel immunization with recombinant protein and preparing cDNA from total RNAs separated from camel lymphocytes, an Nb library was developed. Individual colonies capable of binding selectively to rBAFF were obtained by periplasmic-ELISA, sequenced, and expressed in a bacterial expression system. The specificity and affinity of selected Nb were determined and its target identification and functionality were evaluated using flow cytometry.


Asunto(s)
Factor Activador de Células B , Camelus , Animales , Factor Activador de Células B/genética , Interleucina-4 , Linfocitos B , Anticuerpos Monoclonales/genética
19.
J Biomed Mater Res A ; 111(8): 1216-1227, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-36752269

RESUMEN

The human amniotic membrane dressing has been shown to accelerate the wound healing process in the clinic. In this study, heparin was conjugated to a human Acellular Amniotic Membrane (hAAM) to provide affinity binding sites for immobilizing growth factors. To study the acceleration of the wound healing process, we bound epidermal growth factor and fibroblast growth factor 1 to heparinized hAAMs (GF-Hep-hAAMs). The heparinized hAAMs (Hep-hAAMs) were characterized by toluidine blue staining and infrared spectroscopy. The quality control of hAAM was performed by hematoxylin staining, swelling capacity test and biomechanical evaluation. The cytotoxicity, adhesion, and migration in vitro assays of GF-Hep-hAAMs on L-929 fibroblast cells were also studied by MTT assay, scanning electron microscopy, and scratch assay, respectively. Finally, in vivo skin wound healing study was performed to investigate the wound closure rate, re-epithelization, collagen deposition, and formation of new blood vessels. The results showed that GF-Hep-hAAMs enhance the rate of wound closure and epidermal regeneration in BALB/c mice. In conclusion, GF-Hep-hAAMs could accelerate the wound healing process, significantly in the first week.


Asunto(s)
Apósitos Biológicos , Cicatrización de Heridas , Ratones , Animales , Humanos , Colágeno/metabolismo , Factor de Crecimiento Epidérmico/farmacología , Amnios , Piel
20.
Mol Biotechnol ; 65(5): 766-773, 2023 May.
Artículo en Inglés | MEDLINE | ID: mdl-36203034

RESUMEN

Angiogenesis, the formation of new vessels, is a critical step in the malignancy progression of solid tumors. Many investigations have demonstrated the usefulness of immunotoxins to halt angiogenesis in solid tumors. Pharmaceutically, Vascular Endothelial Growth Factor (VEGF) can deliver coupled toxins to the tumor vessels through VEGF Receptors. In the current study, we designed, expressed, and assessed the in vitro and in vivo toxicities of a novel immunotoxin consisting of mouse VEGF and heminecrolysin toxin (mVEGF-HNc). The fusion protein was expressed in E. coli and purified via Ni+2 affinity chromatography. The biological activity of immunotoxin was evaluated on NIH/3T3 cells and TC1-tumorized mouse model. The mVEGF-NHc showed significant cytotoxicity on the cells as VEGFR-expressing cells. Moreover, the size of the tumor in the mVEGF-HNc-treated group started to reduce after six injections, while it continued to grow in the PBS-received mice. Efficacious targeting of solid tumor cells via mVEGF-HNc suggests its prospective therapeutic potential for cancer therapy.


Asunto(s)
Inmunotoxinas , Neoplasias , Ratones , Animales , Factor A de Crecimiento Endotelial Vascular , Escherichia coli/metabolismo , Factores de Crecimiento Endotelial Vascular , Neoplasias/tratamiento farmacológico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA