Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
1.
Front Immunol ; 12: 669496, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34040611

RESUMEN

Natural killer cell engagers gained enormous interest in recent years due to their potent anti-tumor activity and favorable safety profile. Simultaneously, chicken-derived antibodies entered clinical studies paving the way for avian-derived therapeutics. In this study, we describe the affinity maturation of a common light chain (cLC)-based, chicken-derived antibody targeting EGFR, followed by utilization of the same light chain for the isolation of CD16a- and PD-L1-specific monoclonal antibodies. The resulting binders target their respective antigen with single-digit nanomolar affinity while blocking the ligand binding of all three respective receptors. Following library-based humanization, bispecific and trispecific variants in a standard 1 + 1 or a 2 + 1 common light chain format were generated, simultaneously targeting EGFR, CD16a, and PD-L1. The trispecific antibody mediated an elevated antibody-dependent cellular cytotoxicity (ADCC) in comparison to the EGFR×CD16a bispecific variant by effectively bridging EGFR/PD-L1 double-positive cancer cells with CD16a-positive effector cells. These findings represent, to our knowledge, the first detailed report on the generation of a trispecific 2 + 1 antibodies exhibiting a common light chain and illustrate synergistic effects of trispecific antigen binding. Overall, this generic procedure paves the way for the engineering of tri- and oligospecific therapeutic antibodies derived from avian immunizations.


Asunto(s)
Anticuerpos Biespecíficos/farmacología , Anticuerpos Monoclonales Humanizados/farmacología , Diseño de Fármacos , Epítopos , Inhibidores de Puntos de Control Inmunológico/farmacología , Cadenas Ligeras de Inmunoglobulina/farmacología , Células Asesinas Naturales/efectos de los fármacos , Neoplasias Cutáneas/tratamiento farmacológico , Animales , Anticuerpos Biespecíficos/inmunología , Anticuerpos Monoclonales Humanizados/inmunología , Especificidad de Anticuerpos , Antígeno B7-H1/antagonistas & inhibidores , Antígeno B7-H1/inmunología , Antígeno B7-H1/metabolismo , Línea Celular Tumoral , Pollos , Citotoxicidad Inmunológica/efectos de los fármacos , Receptores ErbB/antagonistas & inhibidores , Receptores ErbB/inmunología , Receptores ErbB/metabolismo , Inhibidores de Puntos de Control Inmunológico/inmunología , Inmunización , Cadenas Ligeras de Inmunoglobulina/inmunología , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/metabolismo , Receptores de IgG/antagonistas & inhibidores , Receptores de IgG/inmunología , Receptores de IgG/metabolismo , Neoplasias Cutáneas/inmunología , Neoplasias Cutáneas/metabolismo , Neoplasias Cutáneas/patología
2.
JCI Insight ; 5(14)2020 07 23.
Artículo en Inglés | MEDLINE | ID: mdl-32544092

RESUMEN

Free light chains (FLCs) induce inflammatory pathways in proximal tubule cells (PTCs). The role of TLRs in these responses is unknown. Here we present findings on the role of TLRs in FLC-induced PTC injury. We exposed human kidney PTC cultures to κ and λ FLCs and used cell supernatants and pellets for ELISA and gene expression studies. We also analyzed tissues from Stat1-/- and littermate control mice treated with daily i.p. injections of a κ FLC for 10 days. FLCs increased the expression of TLR2, TLR4, and TLR6 via HMGB1, a damage-associated molecular pattern. Countering TLR2, TLR4, and TLR6 through GIT-27 or specific TLR siRNAs reduced downstream cytokine responses. Blocking HMGB1 through siRNA or pharmacologic inhibition, or via STAT1 inhibition, reduced FLC-induced TLR2, TLR4, and TLR6 expression. Blocking endocytosis of FLCs through silencing of megalin/cubilin, with bafilomycin A1 or hypertonic sucrose, attenuated FLC-induced cytokine responses in PTCs. IHC showed decreased TLR4 and TLR6 expression in kidney sections from Stat1-/- mice compared with their littermate controls. PTCs exposed to FLCs released HMGB1, which induced expression of TLR2, TLR4, and TLR6 and downstream inflammation. Blocking FLCs' endocytosis, Stat1 knockdown, HMGB1 inhibition, and TLR knockdown each rescued PTCs from FLC-induced injury.


Asunto(s)
Proteína HMGB1/genética , Inflamación/genética , Túbulos Renales Proximales/metabolismo , Factor de Transcripción STAT1/genética , Animales , Endocitosis/efectos de los fármacos , Endocitosis/inmunología , Humanos , Cadenas Ligeras de Inmunoglobulina/metabolismo , Cadenas Ligeras de Inmunoglobulina/farmacología , Inflamación/inmunología , Inflamación/patología , Riñón/inmunología , Riñón/metabolismo , Riñón/patología , Túbulos Renales Proximales/efectos de los fármacos , Túbulos Renales Proximales/inmunología , Ratones , Ratones Noqueados , Receptores de Superficie Celular/genética , Receptor Toll-Like 2/genética , Receptor Toll-Like 4/genética , Receptor Toll-Like 6/genética
3.
Protein Expr Purif ; 170: 105596, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32036001

RESUMEN

Antibodies that block interaction of immune checkpoint receptors with its ligands have revolutionized the treatment of several cancers. Despite the success of this approach, the high cost has been restricted the use of this class of drugs. In this context, the development of biosimilar can be an important strategy for reducing prices and expanding access after patent has been dropped. Here, we evaluated the use of HEK293 cells for transient expression of an immune checkpoint-blocking antibody as a first step for biosimilar development. Antibody light and heavy chain genes were cloned into pCI-neo vector and transiently expressed in HEK293 cells. The culture supernatant was then subjected to protein A affinity chromatography, which allowed to obtain the antibody with high homogeneity. For physicochemical comparability, biosimilar antibody and reference drug were analyzed by SDS-PAGE, isoelectric focusing, circular dichroism and fluorescence spectroscopy. The results indicated that the both antibodies have a high degree of structural similarity. Lastly, the biosimilar antibody binding capacity to target receptor was shown to be similar to reference product in ELISA and flow cytometry assays. These data demonstrate that the HEK293 system can be used as an important tool for candidate selection and early development of biosimilar antibodies.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Biosimilares Farmacéuticos/farmacología , Inhibidores de Puntos de Control Inmunológico/farmacología , Proteínas de Punto de Control Inmunitario/genética , Cadenas Pesadas de Inmunoglobulina/farmacología , Cadenas Ligeras de Inmunoglobulina/farmacología , Anticuerpos Monoclonales/biosíntesis , Afinidad de Anticuerpos , Especificidad de Anticuerpos , Biosimilares Farmacéuticos/metabolismo , Cromatografía de Afinidad , Vectores Genéticos/química , Vectores Genéticos/metabolismo , Células HEK293 , Humanos , Inhibidores de Puntos de Control Inmunológico/inmunología , Proteínas de Punto de Control Inmunitario/inmunología , Cadenas Pesadas de Inmunoglobulina/biosíntesis , Cadenas Ligeras de Inmunoglobulina/biosíntesis , Focalización Isoeléctrica
4.
Front Immunol ; 11: 606878, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33424853

RESUMEN

Bispecific (BsAb) and biparatopic (BpAb) antibodies emerged as promising formats for therapeutic biologics exhibiting tailor-made functional properties. Over recent years, chicken-derived antibodies have gained traction for diagnostic and therapeutic applications due to their broad epitope coverage and convenience of library generation. Here we report the first generation of a biparatopic common light chain (cLC) chicken-derived antibody by an epitope binning-based screening approach using yeast surface display. The resulting monospecific antibodies target conformational epitopes on domain II or III of the epidermal growth factor receptor (EGFR) with lower double- or single-digit nanomolar affinities, respectively. Furthermore, the domain III targeting variant was shown to interfere with epidermal growth factor (EGF) binding. Utilizing the Knob-into-Hole technology (KiH), a biparatopic antibody with subnanomolar affinity was generated that facilitates clustering of soluble and cell-bound EGFR and displayed enhanced antibody-dependent cell-mediated cytotoxicity (ADCC) compared to the parental antibodies. This strategy for generating cLC-based biparatopic antibodies from immunized chickens may pave the way for their further development in therapeutic settings.


Asunto(s)
Anticuerpos Biespecíficos/metabolismo , Antineoplásicos Inmunológicos/metabolismo , Técnicas de Visualización de Superficie Celular , Pollos/inmunología , Inmunización , Cadenas Ligeras de Inmunoglobulina/metabolismo , Saccharomyces cerevisiae/metabolismo , Animales , Anticuerpos Biespecíficos/inmunología , Anticuerpos Biespecíficos/farmacología , Especificidad de Anticuerpos , Citotoxicidad Celular Dependiente de Anticuerpos , Antineoplásicos Inmunológicos/inmunología , Antineoplásicos Inmunológicos/farmacología , Bioprospección , Carcinoma de Células Escamosas/tratamiento farmacológico , Carcinoma de Células Escamosas/inmunología , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patología , Línea Celular Tumoral , Mapeo Epitopo , Epítopos , Receptores ErbB/administración & dosificación , Receptores ErbB/inmunología , Humanos , Cadenas Ligeras de Inmunoglobulina/inmunología , Cadenas Ligeras de Inmunoglobulina/farmacología , Saccharomyces cerevisiae/genética
5.
AIDS ; 30(11): 1691-701, 2016 07 17.
Artículo en Inglés | MEDLINE | ID: mdl-27058352

RESUMEN

OBJECTIVE: To develop a novel and potent fusion inhibitor of HIV infection based on a rational strategy for synthetic antibody library construction. DESIGN: The reduced molecular weight of single-domain antibodies (sdAbs) allows targeting of cryptic epitopes, the most conserved and critical ones in the context of HIV entry. Heavy-chain sdAbs from camelids are particularly suited for this type of epitope recognition because of the presence of long and flexible antigen-binding regions [complementary-determining regions (CDRs)]. METHODS: We translated camelid CDR features to a rabbit light-chain variable domain (VL) and constructed a library of minimal antibody fragments with elongated CDRs. Additionally to elongation, CDRs' variability was restricted to binding favorable amino acids to potentiate the selection of high-affinity sdAbs. The synthetic library was screened against a conserved, hidden, and crucial-to-fusion sequence on the heptad-repeat 1 (HR1) region of the HIV-1 envelope glycoprotein. RESULTS: Two anti-HR1 VLs, named F63 and D104, strongly inhibited laboratory-adapted HIV-1 infectivity. F63 also inhibited infectivity of HIV-1 and HIV-2 primary isolates similarly to the Food and Drug Administration-approved fusion inhibitor T-20 and HIV-1 strains resistant to T-20. Moreover, epitope mapping of F63 revealed a novel target sequence within the highly conserved hydrophobic pocket of HR1. F63 was also capable of interacting with viral and cell lipid membrane models, a property previously associated with T-20's inhibitory mechanism. CONCLUSION: In summary, to our best knowledge, we developed the first potent and broad VL sdAb fusion inhibitor of HIV infection. Our study also gives insights into engineering strategies that could be explored to enhance the development of antiviral drugs.


Asunto(s)
Productos Biológicos/farmacología , Anticuerpos Anti-VIH/farmacología , Inhibidores de Fusión de VIH/farmacología , VIH-1/efectos de los fármacos , Cadenas Ligeras de Inmunoglobulina/farmacología , Anticuerpos de Dominio Único/farmacología , Productos del Gen env del Virus de la Inmunodeficiencia Humana/metabolismo , Animales , Anticuerpos Anti-VIH/genética , VIH-2/efectos de los fármacos , Células HeLa , Humanos , Cadenas Ligeras de Inmunoglobulina/genética , Conejos , Anticuerpos de Dominio Único/genética
6.
Kidney Int ; 86(4): 669-71, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25265948

RESUMEN

AL amyloidosis is a severe complication of plasma-cell disorders, secondary to monoclonal immunoglobulin light chain (LC) deposition in the kidney and other organs. Though the physicochemical properties of amyloid-forming monoclonal LCs have been demonstrated to be involved in their propensity to aggregate, it remains unclear where, when, and finally why amyloid fibrils are formed in vivo. Teng et al. shed light on this long-standing issue thanks to a new animal model.


Asunto(s)
Amiloide/biosíntesis , Amiloidosis/metabolismo , Modelos Animales de Enfermedad , Mesangio Glomerular/metabolismo , Cadenas Ligeras de Inmunoglobulina/farmacología , Enfermedades Renales/metabolismo , Lisosomas/fisiología , Animales , Masculino
7.
Kidney Int ; 86(4): 738-46, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24786709

RESUMEN

In vitro and ex vivo studies have elucidated the step-by-step process whereby some physicochemically abnormal light chains are processed by mesangial cells to form amyloid fibrils. Although crucial steps in the cascade of events have been determined, these findings have not been reproduced in vivo. This has led to some doubts as to the significance and clinical application of the information that has been deciphered. Here, we developed an animal model which uses mice injected with amyloidogenic light chains purified from the urine of patients with biopsy-proven, light-chain-associated glomerular amyloidosis which validated in vitro/ex vivo findings. This animal model showed internalization of the light chains utilizing caveolae followed by trafficking to the mature lysosomal compartment where fibrils were formed. This model permits evaluation of mesangial amyloidogenesis for prolonged periods of time, is potentially useful to test maneuvers to modulate events that take place, and can be used to design novel therapeutic interventions.


Asunto(s)
Amiloide/biosíntesis , Amiloidosis/metabolismo , Modelos Animales de Enfermedad , Mesangio Glomerular/metabolismo , Cadenas Ligeras de Inmunoglobulina/farmacología , Enfermedades Renales/metabolismo , Lisosomas/fisiología , Amiloide/metabolismo , Amiloide/ultraestructura , Animales , Antígenos CD/análisis , Antígenos de Diferenciación Mielomonocítica/análisis , Arteriolas/metabolismo , Espacio Extracelular , Mesangio Glomerular/química , Mesangio Glomerular/ultraestructura , Inmunohistoquímica , Masculino , Ratones , Ratones Noqueados , Microscopía Electrónica de Rastreo , Fenotipo
8.
J Neuroimmunol ; 264(1-2): 14-23, 2013 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-24051200

RESUMEN

Plasma B cells secrete immunoglobulinfree light chains (IgLC) which by binding to mast cells can mediate hypersensitivity responses and are involved in several immunological disorders. To investigate the effects of antigen-specific IgLC activation, intracellular recordings were made from cultured murine dorsal root ganglion (DRG) neurons, which can specifically bind IgLC. The neurons were sensitized with IgLC for 90min and subsequently activated by application of the corresponding antigen (DNP-HSA). Antigen application induced a decrease in the rate of rise of the action potentials of non-nociceptive neurons (MANOVA, p=2.10(-6)), without affecting the resting membrane potential or firing threshold. The action potentials of the nociceptive neurons (p=0.57) and the electrical excitability of both types of neurons (p>0.35) were not affected. We conclude that IgLC can mediate antigen-specific responses by reducing the rate of rise of action potentials in non-nociceptive murine DRG neurons. We suggest that antigen-specific activation of IgLC-sensitized non-nociceptive DRG neurons may contribute to immunological hypersensitivity responses and neuroinflammation.


Asunto(s)
Potenciales de Acción/efectos de los fármacos , Ganglios Espinales/citología , Cadenas Ligeras de Inmunoglobulina/farmacología , Neuronas/efectos de los fármacos , Análisis de Varianza , Animales , Antígenos/metabolismo , Fenómenos Biofísicos/efectos de los fármacos , Biofisica , Células Cultivadas , Estimulación Eléctrica , Masculino , Ratones , Ratones Endogámicos BALB C , Neuronas/clasificación , Neuronas/fisiología , Factores de Tiempo
9.
J Biol Chem ; 288(27): 19558-68, 2013 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-23677996

RESUMEN

Human antibody light chains belonging to subgroup II of germ line genes were amplified by a seminested PCR technique using B-lymphocytes taken from a human adult infected with influenza virus. Each gene of the human light chains was transferred into the Escherichia coli system. The recovered light chain was highly purified using a two-step purification system. Light chain 22F6 showed interesting catalytic features. The light chain cleaved a peptide bond of synthetic peptidyl-4-methyl-coumaryl-7-amide (MCA) substrates, such as QAR-MCA and EAR-MCA, indicating amidase activity. It also hydrolyzed a phosphodiester bond of both DNA and RNA. From the analysis of amino acid sequences and molecular modeling, the 22F6 light chain possesses two kinds of active sites as amidase and nuclease in close distances. The 22F6 catalytic light chain could suppress the infection of influenza virus type A (H1N1) of Madin-Darby canine kidney cells in an in vitro assay. In addition, the catalytic light chain clearly inhibited the infection of the influenza virus of BALB/c mice via nasal administration in an in vivo assay. In the experiment, the titer in the serum of the mice coinfected with the 22F6 light chain and H1N1 virus became considerably lowered compared with that of 22F6-non-coinfected mice. Note that the catalytic light chain was prepared from human peripheral lymphocyte and plays an important role in preventing infection by influenza virus. Considering the fact that the human light chain did not show any acute toxicity for mice, our procedure developed in this study must be unique and noteworthy for developing new drugs.


Asunto(s)
Anticuerpos Monoclonales , Anticuerpos Antivirales , Linfocitos B/inmunología , Cadenas Ligeras de Inmunoglobulina , Subtipo H1N1 del Virus de la Influenza A/inmunología , Infecciones por Orthomyxoviridae/tratamiento farmacológico , Adulto , Animales , Anticuerpos Monoclonales/genética , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/farmacología , Anticuerpos Antivirales/genética , Anticuerpos Antivirales/inmunología , Anticuerpos Antivirales/farmacología , Secuencia de Bases , Perros , Femenino , Humanos , Cadenas Ligeras de Inmunoglobulina/genética , Cadenas Ligeras de Inmunoglobulina/inmunología , Cadenas Ligeras de Inmunoglobulina/farmacología , Células de Riñón Canino Madin Darby , Masculino , Ratones , Datos de Secuencia Molecular , Infecciones por Orthomyxoviridae/genética , Infecciones por Orthomyxoviridae/inmunología
10.
Atherosclerosis ; 225(1): 220-3, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22981431

RESUMEN

UNLABELLED: Misfolded immunoglobulin light chain proteins (LC) in light chain amyloidosis (AL) are toxic to vascular tissues. We tested the hypothesis that chaperone protein clusterin preserves endothelial function and cell survival during LC exposure. METHODS: LC (20 µg/mL) were given to human aortic endothelial cells (EC) for 24-h and clusterin protein/gene expression and secretion were measured. DNA fragmentation was measured with/without recombinant clusterin (Clu, 300 ng/mL). Adipose arterioles (non-AL subjects) were tested for dilator responses to acetylcholine/papaverine at baseline and after 1-h of LC ± Clu. RESULTS: LC reduced EC clusterin secretion, protein and gene expression while increasing DNA fragmentation. Clu attenuated LC-induced DNA fragmentation and restored dilator response to acetylcholine (logEC50: control -7.05 ± 0.2, LC + Clu -6.53 ± 0.4, LC -4.28 ± 0.7, p < 0.05 versus control, LC + Clu). CONCLUSIONS: LC induced endothelial cell death and dysfunction while reducing clusterin protein/gene expression and secretion. Exogenous clusterin attenuated LC toxicity. This represents a new pathobiologic mechanism and therapeutic target for AL amyloidosis.


Asunto(s)
Amiloidosis/fisiopatología , Clusterina/fisiología , Células Endoteliales/fisiología , Cadenas Ligeras de Inmunoglobulina , Arteriolas/fisiopatología , Muerte Celular/efectos de los fármacos , Clusterina/biosíntesis , Fragmentación del ADN/efectos de los fármacos , Células Endoteliales/efectos de los fármacos , Femenino , Humanos , Cadenas Ligeras de Inmunoglobulina/metabolismo , Cadenas Ligeras de Inmunoglobulina/farmacología , Masculino , Persona de Mediana Edad , Vasodilatación/efectos de los fármacos
11.
Blood ; 117(4): 1301-7, 2011 Jan 27.
Artículo en Inglés | MEDLINE | ID: mdl-21098396

RESUMEN

One of the major attendant complications of multiple myeloma is renal injury, which contributes significantly to morbidity and mortality in this disease. Monoclonal immunoglobulin free light chains (FLCs) are usually directly involved, and tubulointerstitial renal injury and fibrosis are prominent histologic features observed in myeloma. The present study examined the role of monoclonal FLCs in altering the nuclear factor κ light chain enhancer of activated B cells (NF-κB) activity of renal epithelial cells. Human proximal tubule epithelial cells exposed to 3 different human monoclonal FLCs demonstrated Src kinase-dependent activation of the NF-κB pathway, which increased production of monocyte chemoattractant protein-1 (MCP-1). Tyrosine phosphorylation of inhibitor of κB kinases (IKKs) IKKα and IKKß and a concomitant increase in inhibitor of κB (IκB) kinase activity in cell lysates were observed. Time-dependent, Src kinase-dependent increases in serine and tyrosine phosphorylation of IκBα and NF-κB activity were also demonstrated. Proteasome inhibition partially blocked FLC-induced MCP-1 production. These findings fit into a paradigm characterized by FLC-induced redox-signaling events that activated the canonical and atypical (IKK-independent) NF-κB pathways to promote a proinflammatory, profibrotic renal environment.


Asunto(s)
Células Epiteliales/efectos de los fármacos , Cadenas Ligeras de Inmunoglobulina/farmacología , Riñón/efectos de los fármacos , FN-kappa B/metabolismo , Familia-src Quinasas/fisiología , Antineoplásicos/farmacología , Ácidos Borónicos/farmacología , Bortezomib , Células Cultivadas , Quimiocina CCL2/metabolismo , Relación Dosis-Respuesta a Droga , Evaluación Preclínica de Medicamentos , Células Epiteliales/metabolismo , Humanos , Quinasa I-kappa B/metabolismo , Cadenas Ligeras de Inmunoglobulina/fisiología , Riñón/metabolismo , Fosforilación/efectos de los fármacos , Pirazinas/farmacología , Transducción de Señal/efectos de los fármacos , Tirosina/metabolismo , Familia-src Quinasas/metabolismo
12.
Circ Res ; 106(9): 1533-40, 2010 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-20360247

RESUMEN

RATIONALE: In recent work, we have demonstrated a crucial role of mast cells in the development of viral myocarditis. Viral infection could lead to increased synthesis of free immunoglobulin light chains (FLC) and our earlier work showed that FLC can trigger mast cell activation. OBJECTIVE: We studied the possible involvement of FLC in the pathogenesis of viral myocarditis, and therapeutic effects of FLC using an animal model of viral myocarditis. METHODS AND RESULTS: DBA/2 mice were inoculated intraperitoneally with encephalomyocarditis (EMC) virus. Serum levels and concentrations in the heart of kappa FLC on day 14 in mice inoculated with EMC virus were significantly increased compared with controls. Myocardial viral concentration was significantly inhibited, the area of myocardial lesions was smaller in mice treated with kappa or lambda FLC, and survival of mice given FLC significantly improved. In contrast, an FLC antagonist deteriorated myocarditis. kappa and lambda FLC chains inhibited EMC viral replication in human amnion cells in vitro. lambda FLC significantly increased the gene expression of interleukin-10 in the heart which was previously shown to improve viral myocarditis when given exogenously. FLC also tended to increase the gene expressions of interferon-alpha and -gamma in the heart mice. CONCLUSIONS: FLC have antiviral and antiinflammatory effects and improved viral myocarditis in mice. FLC may be promising agents for the treatment of viral myocarditis.


Asunto(s)
Antiinflamatorios/farmacología , Antivirales/farmacología , Infecciones por Cardiovirus/tratamiento farmacológico , Cadenas Ligeras de Inmunoglobulina/farmacología , Miocarditis/tratamiento farmacológico , Miocarditis/virología , Animales , Antiinflamatorios/sangre , Antiinflamatorios/uso terapéutico , Antivirales/sangre , Antivirales/uso terapéutico , Infecciones por Cardiovirus/patología , Infecciones por Cardiovirus/virología , Modelos Animales de Enfermedad , Virus de la Encefalomiocarditis/efectos de los fármacos , Virus de la Encefalomiocarditis/fisiología , Humanos , Cadenas Ligeras de Inmunoglobulina/sangre , Cadenas Ligeras de Inmunoglobulina/uso terapéutico , Masculino , Ratones , Ratones Endogámicos DBA , Pruebas de Sensibilidad Microbiana , Miocarditis/patología , Tasa de Supervivencia , Replicación Viral/efectos de los fármacos
13.
Biotechnol Bioeng ; 106(1): 161-6, 2010 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-20047190

RESUMEN

The tumor necrosis factor-alpha (TNFalpha) plays an important role in a number of chronic inflammatory disorders. Monoclonal camelidae variable heavy chain domain-only antibodies (V(H)H) have been developed to antagonize the action of human TNFalpha (anti-TNF-V(H)H). Here we describe a strategy to obtain functional dimeric anti-TNF-V(H)H molecules, based on the C-terminal fusion of a kappa light chain domain to the anti-TNF-V(H)H. The resulting fusion protein was transiently expressed by use of viral vectors in Nicotiana benthamiana((Nb)) leaves and purified. Competitive ELISA and cell cytotoxicity assays revealed that the dimerized anti-(Nb)TNF-V(H)H(Ckappa) proteins blocked TNFalpha-activity more effectively than either the monomeric Escherichia coli((Ec)) produced anti-(Ec)TNF-V(H)H or the monomeric anti-(Nb)TNF-V(H)H(Ckappa). We suggest that enhanced inhibition shown by dimeric anti-(Nb)TNF-V(H)H(Ckappa) proteins is achieved by an increase in avidity.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Anticuerpos Neutralizantes/farmacología , Cadenas Ligeras de Inmunoglobulina/farmacología , Factores Inmunológicos/farmacología , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Anticuerpos Monoclonales/genética , Anticuerpos Neutralizantes/genética , Dimerización , Expresión Génica , Vectores Genéticos , Humanos , Cadenas Ligeras de Inmunoglobulina/genética , Factores Inmunológicos/genética , Virus de Plantas/genética , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/farmacología , Nicotiana/genética
14.
J Immunol Methods ; 346(1-2): 26-37, 2009 Jul 31.
Artículo en Inglés | MEDLINE | ID: mdl-19427867

RESUMEN

Natural IgA antibodies are abundantly produced in vivo to protect serosal surfaces from invading infectious organisms. However, the immunotherapeutic potential of IgA has hardly been explored, although there is evidence that recombinant IgA antibodies may broaden the armentarium to combat certain infectious or malignant diseases. One of the limitations for exploring IgA's therapeutic activity has been the difficulty to obtain enough recombinant material with desired specificity for in vivo studies. Here, we describe the production and purification of monomeric recombinant IgA1 and IgA2 antibodies under serum-free conditions. For antibody production, suspension adapted CHO-K1 cells and a glutamine synthetase selection vector were used, which resulted in specific production rates of up to 2.2 pg/cell/day. Purities of >95% of monomeric antibodies were obtained by a combination of affinity chromatography-using an anti-kappa-light chain matrix-and size exclusion chromatography. Purified antibodies displayed the expected biochemical characteristics and were functionally fully active. Importantly, all required reagents and methods are commercially available and not dependent on the specificity of the desired antibody. In addition, all employed technologies and methodologies are similar to those used for the production of therapeutic IgG antibodies - thus allowing further up-scaling and streamlining according to existing antibody production technologies. In conclusion, the described methodology may assist in the development of recombinant IgA antibodies for therapeutic applications.


Asunto(s)
Inmunoglobulina A/biosíntesis , Inmunoglobulina A/aislamiento & purificación , Cadenas Pesadas de Inmunoglobulina/biosíntesis , Cadenas Pesadas de Inmunoglobulina/aislamiento & purificación , Cadenas Ligeras de Inmunoglobulina/biosíntesis , Cadenas Ligeras de Inmunoglobulina/aislamiento & purificación , Animales , Especificidad de Anticuerpos , Citotoxicidad Celular Dependiente de Anticuerpos , Sitios de Unión de Anticuerpos , Células CHO , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Cromatografía de Afinidad , Cromatografía en Gel , Cricetinae , Cricetulus , Receptores ErbB/antagonistas & inhibidores , Receptores ErbB/inmunología , Humanos , Hibridomas , Inmunoglobulina A/genética , Inmunoglobulina A/metabolismo , Inmunoglobulina A/farmacología , Cadenas Pesadas de Inmunoglobulina/genética , Cadenas Pesadas de Inmunoglobulina/metabolismo , Cadenas Pesadas de Inmunoglobulina/farmacología , Cadenas Ligeras de Inmunoglobulina/genética , Cadenas Ligeras de Inmunoglobulina/metabolismo , Cadenas Ligeras de Inmunoglobulina/farmacología , Leucocitos Mononucleares/efectos de los fármacos , Leucocitos Mononucleares/inmunología , Proteínas Recombinantes de Fusión/biosíntesis , Proteínas Recombinantes de Fusión/aislamiento & purificación , Transfección
15.
Kidney Int ; 75(6): 634-45, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18923384

RESUMEN

Nodular glomerulosclerosis results from increased deposition of extracellular matrix proteins and monotypic light chains. The inability of mesangial cells to degrade abnormal levels of tenascin-C--along with the increased expression of some growth factors such as platelet-derived growth factor (PDGF) and transforming growth factor-beta (TGF-beta)--is crucial to the pathogenesis of light chain deposition disease (LCDD). In order to study the molecular processes contributing to LCDD, we grew mesangial cells in three-dimensional matrices and incubated the cells with free light chains purified from the urine of patients with biopsy-proven LCDD, immunoglobulin-associated amyloid deposits, or myeloma cast nephropathy. Light chains of the latter two cohorts served as controls. Mesangial cells incubated with light chains from patients with LCDD show a significant increase in tenascin-C expression, centrally located within newly formed nodules, along with increased expression of PDGF and TGF-betas, compared to mesangial cells incubated with control light chains. There was less extracellular MMP-7 even though its intracellular expression is markedly increased compared to the control. Addition of active MMP-7 degraded this excess tenascin-C in vitro, a process that could be prevented by an exogenous MMP inhibitor. Our in vitro model recapitulates in vivo findings in patients with LCDD, thus allowing definition of the sequential pathologic processes associated with glomerulopathic light chain interactions with mesangial cells.


Asunto(s)
Nefropatías Diabéticas/patología , Cadenas Ligeras de Inmunoglobulina/farmacología , Estudios de Casos y Controles , Células Cultivadas , Nefropatías Diabéticas/etiología , Humanos , Metaloproteinasa 7 de la Matriz/análisis , Células Mesangiales/patología , Factor de Crecimiento Derivado de Plaquetas/análisis , Tenascina/análisis , Factor de Crecimiento Transformador beta/análisis
16.
Nephrol Dial Transplant ; 23(3): 860-70, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17933841

RESUMEN

BACKGROUND: To determine the role of epithelial-mesenchymal transition (EMT) as a potential mechanism contributing to the characteristic tubulointerstitial renal fibrosis in multiple myeloma, we examined whether myeloma light chains (LCs) directly induce EMT in human renal proximal tubule epithelial cells (PTECs). METHODS: As positive controls we used TGF-beta1 and cyclosporine A (CsA), two agents known to induce EMT in PTECs. Human LCs were isolated and purified from the urine of myeloma patients with modest renal insufficiency without evidence of glomerular involvement. HK-2 cells were exposed to kappa LC (25 microM) for periods up to 72 h. RESULTS: LCs induced marked cellular morphological alterations in PTECs, accompanied with increased expression levels of profibrotic TGF-beta1, FSP-1 and extracellular matrix components. Using semiquantitative immunoblotting and RT-PCR, we observed that the expression of E-cadherin decreased after 24 h, while the expression of alpha-SMA increased in PTEC after continuous exposure to kappa-LCs. Human serum albumin (HSA; 160 microM) had less potent effect on the expression of EMT-related molecules. Neutralizing TGF-beta1 antibody blocked CsA-induced EMT but had no effect on LC-exposed cells. LC-induced EMT and the secretions of IL-6 and MCP-1 were, however, markedly attenuated by p38 MAPK interference. The use of bone morphogenetic protein-7 or pituitary adenylate cyclase-activating polypeptide (PACAP) induced the formation of cell aggregates, and the reacquisition of E-cadherin expression and renal proximal tubule epithelial morphology within the confluent cell monolayer during and after LC exposure. CONCLUSIONS: These findings demonstrate that LC is a direct stimulus for EMT in PTECs. LC-induced EMT involved multiple cytokines, is modulated by p38 MAPK, but appeared independent of the action of TGF-beta1. LC-induced EMT may be an important mechanism of kidney injury associated with myeloma and may be reversible upon the administration of exogenous PACAP.


Asunto(s)
Transdiferenciación Celular/efectos de los fármacos , Células Epiteliales/patología , Cadenas Ligeras de Inmunoglobulina/metabolismo , Cadenas Ligeras de Inmunoglobulina/farmacología , Túbulos Renales Proximales/patología , Mesodermo/patología , Mieloma Múltiple/metabolismo , Proteína Morfogenética Ósea 7 , Proteínas Morfogenéticas Óseas/farmacología , Cadherinas/metabolismo , Proteínas de Unión al Calcio/metabolismo , Agregación Celular/efectos de los fármacos , Células Cultivadas , Quimiocina CCL2/metabolismo , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Proteínas de la Matriz Extracelular/metabolismo , Fibrosis/metabolismo , Fibrosis/patología , Humanos , Interleucina-6/metabolismo , Túbulos Renales Proximales/efectos de los fármacos , Túbulos Renales Proximales/metabolismo , Mesodermo/efectos de los fármacos , Mesodermo/metabolismo , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/farmacología , Proteína de Unión al Calcio S100A4 , Factor de Crecimiento Transformador beta/farmacología , Factor de Crecimiento Transformador beta1/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
17.
J Am Soc Nephrol ; 18(4): 1239-45, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17360948

RESUMEN

As low molecular weight proteins, restriction from glomerular filtration is minimized, permitting significant amounts of Ig light chains to be endocytosed into the proximal tubule epithelium, particularly in plasma cell dyscrasias. Recent studies have shown that this effect of concentrating light chains within the proximal tubule alters cell function. This study demonstrated that light chains belonged to a class of proteins that are capable of catalyzing the formation of hydrogen peroxide. Sufficient amounts of hydrogen peroxide were produced in HK-2 cells to stimulate the production of monocyte chemoattractant protein-1 (MCP-1), a key chemokine involved in monocyte/macrophage migration and activation of the proximal tubule, and to increase lactate dehydrogenase release into the medium. The light chain-mediated effect on MCP-1 production was inhibited by co-incubation with 1,3-dimethyl-2-thiourea, which also inhibited lactate dehydrogenase release, and by pyrrolidine dithiocarbamate, an inhibitor of NF-kappaB. The amount of light chain that stimulated an intracellular redox-signaling pathway in the proximal tubule cells was well within levels that are seen in patients who have plasma cell dyscrasias. The conclusion is that light chains possess a unique property that permits the development of intracellular oxidative stress that in turn promotes activation of the proximal tubule and elaboration of MCP-1.


Asunto(s)
Peróxido de Hidrógeno/metabolismo , Cadenas Ligeras de Inmunoglobulina/farmacología , Estrés Oxidativo , Catálisis , Células Cultivadas , Quimiocina CCL2/biosíntesis , Humanos , Riñón/metabolismo , L-Lactato Deshidrogenasa/metabolismo , Tiourea/análogos & derivados , Tiourea/farmacología
18.
Am J Pathol ; 166(1): 197-208, 2005 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-15632012

RESUMEN

Amyloidoses are a group of disorders characterized by abnormal folding of proteins that impair organ function. We investigated the cellular response of primary cardiac fibroblasts to amyloidogenic light chains and determined the corresponding change in proteoglycan expression and localization. The cellular response to 11 urinary immunoglobulin light chains of kappa1, lambda6, and lambda 3 subtypes was evaluated. The localization of the light chains was monitored by conjugating them to Oregon Green 488 and performing live cell confocal microscopy. Sulfation of the proteoglycans was determined after elution over Q1-columns with a single-step salt gradient (1.5 mol/L NaCl) via dimethylmethylene blue. Light chains were detected inside cells within 4 hours and demonstrated perinuclear localization. Over 80% of the cells showed intracellular localization of the amyloid light chains. The light chains induced sulfation of the secreted glycosaminoglycans, but the cell fraction possessed only minimal sulfation. Furthermore, the light chains caused a translocation of heparan sulfate proteoglycan to the nucleus. The conformation and thermal stability of light chains was altered when they were incubated in the presence of heparan sulfate and destabilization of the amyloid light chains was detected. These studies indicate that internalization of the light chains mediates the expression and localization of heparan sulfate proteoglycans.


Asunto(s)
Amiloidosis/patología , Fibroblastos/patología , Miocardio/patología , Amiloidosis/inmunología , Amiloidosis/orina , Fibroblastos/efectos de los fármacos , Fibroblastos/inmunología , Glicosaminoglicanos/análisis , Humanos , Cadenas Ligeras de Inmunoglobulina/aislamiento & purificación , Cadenas Ligeras de Inmunoglobulina/farmacología , Cadenas Ligeras de Inmunoglobulina/orina , Microscopía Confocal , Miocardio/citología , Procesamiento Proteico-Postraduccional
19.
Lab Invest ; 84(10): 1322-38, 2004 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-15286662

RESUMEN

Human mesangial cells (HMCs) are injured by either excessive amounts or abnormal light chains (LCs), or a combination of both in patients with plasma cell dyscrasias. Consequently, these HMCs undergo phenotypic transformations. HMCs were incubated with eight different light-chains (LCs) for 96 h. These cells, in addition to 51 patient samples from patients with AL-amyloidosis (AL-Am), light-chain deposition disease (LCDD), myeloma cast nephropathy (MCN) and controls were analyzed by immunohistochemistry for CD68, muscle-specific actin (MSA), smooth muscle actin (SMA), CD14, and Ham56 protein expressions. All samples were also studied using electron microscopy. Greater staining (four- and three-fold) expressions of CD68 and Ham56, respectively, were observed in the HMCs incubated with AL-Am-LCs compared to those with LCDD-LCs and control. SMA expression levels were five-fold higher in LCDD-LC-treated cells compared to the other categories of LC-treated and control cells. Similar results were obtained in the renal specimens, however, CD68 levels were 12-fold higher in the AL-Am cases compared to the LCDD cases, respectively. Conversely, MSA and SMA levels were three fold higher in the LCDD cases than in the AL-Am ones. No CD14 expression was noted in any of the samples and CD-34 staining of HMCs treated with the various LCs only showed rare positive cells. Dynamic real-time studies to visualize the rough endoplasmic reticulum (RER) and lysosomal compartments in HMCs incubated with LCDD and AL-Am-LCs showed striking expansion of each of the above-mentioned compartments, respectively. This indicates the presence of more RER in the LCDD-LC-treated HMCs and a striking increase in lysosomes noticeable in the AL-Am-LC-treated cells. Data obtained in this study highlighted that HMCs incubated with LCDD-LCs undergo a myofibroblastic phenotypic transformation, while AL-Am-LCs induce a macrophage-like phenotype in these cells.


Asunto(s)
Amiloide/inmunología , Amiloidosis/inmunología , Mesangio Glomerular/efectos de los fármacos , Cadenas Ligeras de Inmunoglobulina/farmacología , Enfermedades Renales/inmunología , Actinas/metabolismo , Amiloide/metabolismo , Amiloidosis/metabolismo , Amiloidosis/patología , Antígenos CD/metabolismo , Antígenos de Diferenciación Mielomonocítica/metabolismo , Biomarcadores/análisis , Células Cultivadas , Proteínas de la Matriz Extracelular/metabolismo , Mesangio Glomerular/metabolismo , Mesangio Glomerular/ultraestructura , Humanos , Cadenas Ligeras de Inmunoglobulina/metabolismo , Inmunohistoquímica , Enfermedades Renales/metabolismo , Enfermedades Renales/patología , Mieloma Múltiple/inmunología , Mieloma Múltiple/metabolismo , Mieloma Múltiple/patología , Fenotipo
20.
Biochemistry ; 43(31): 9999-10007, 2004 Aug 10.
Artículo en Inglés | MEDLINE | ID: mdl-15287727

RESUMEN

Beta-amyloid (Abeta), a peptide generated by proteolytic cleavage of the amyloid precursor protein (APP), is a major constituent of the neuritic plaques associated with Alzheimer's disease (AD). Up-regulation of alpha-secretase, which can hydrolyze Abeta between Lys16 and Leu17, has been proposed as a potential therapeutic strategy in the treatment of AD. Previously, we identified two light-chain antibody fragments that had proteolytic activity against Abeta, one with alpha-secretase-like activity and one with carboxypeptidase-like activity. Here we show that cleavage of Abeta40 by hk14, the light-chain antibody having carboxypeptidase-like activity, alters aggregation of Abeta and neutralizes any cytotoxic effects of the peptide. Cleavage of Abeta40 with c23.5, the light chain having alpha-secretase-like cleavage, substantially increases the aggregation rate of Abeta; however, it does not show any corresponding increase in cytotoxicity. The increase in aggregation resulting from hydrolysis by c23.5 can be attributed to the decreased solubility of the hydrolyzed products relative to the parent Abeta40, primarily the Abeta17-40 fragment. These results demonstrate that antibody fragment mediated proteolytic degradation of Abeta peptide can be a potential therapeutic route to control Abeta aggregation and toxicity in vivo. Our results also suggest that increasing alpha-secretase activity as a therapeutic route must be approached with some caution because this can lead to a substantial increase in aggregation.


Asunto(s)
Péptidos beta-Amiloides/antagonistas & inhibidores , Péptidos beta-Amiloides/metabolismo , Cadenas Ligeras de Inmunoglobulina/metabolismo , Cadenas Ligeras de Inmunoglobulina/farmacología , Fragmentos de Péptidos/antagonistas & inhibidores , Fragmentos de Péptidos/metabolismo , Secretasas de la Proteína Precursora del Amiloide , Péptidos beta-Amiloides/toxicidad , Péptidos beta-Amiloides/ultraestructura , Ácido Aspártico Endopeptidasas , Sitios de Unión de Anticuerpos , Carboxipeptidasas/metabolismo , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Endopeptidasas/metabolismo , Humanos , Hidrólisis , Fragmentos de Inmunoglobulinas/metabolismo , Fragmentos de Inmunoglobulinas/farmacología , Fragmentos de Inmunoglobulinas/ultraestructura , Cadenas Ligeras de Inmunoglobulina/ultraestructura , Microscopía de Fuerza Atómica , Fragmentos de Péptidos/toxicidad , Fragmentos de Péptidos/ultraestructura
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...