Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
Proc Natl Acad Sci U S A ; 113(28): 7864-9, 2016 07 12.
Artículo en Inglés | MEDLINE | ID: mdl-27325759

RESUMEN

Oligoclonal Ig bands (OCBs) of the cerebrospinal fluid are a hallmark of multiple sclerosis (MS), a disabling inflammatory disease of the central nervous system (CNS). OCBs are locally produced by clonally expanded antigen-experienced B cells and therefore are believed to hold an important clue to the pathogenesis. However, their target antigens have remained unknown, mainly because it was thus far not possible to isolate distinct OCBs against a background of polyclonal antibodies. To overcome this obstacle, we copurified disulfide-linked Ig heavy and light chains from distinct OCBs for concurrent analysis by mass spectrometry and aligned patient-specific peptides to corresponding transcriptome databases. This method revealed the full-length sequences of matching chains from distinct OCBs, allowing for antigen searches using recombinant OCB antibodies. As validation, we demonstrate that an OCB antibody from a patient with an infectious CNS disorder, neuroborreliosis, recognized a Borrelia protein. Next, we produced six recombinant antibodies from four MS patients and identified three different autoantigens. All of them are conformational epitopes of ubiquitous intracellular proteins not specific to brain tissue. Our findings indicate that the B-cell response in MS is heterogeneous and partly directed against intracellular autoantigens released during tissue destruction. In addition to helping elucidate the role of B cells in MS, our approach allows the identification of target antigens of OCB antibodies in other neuroinflammatory diseases and the production of therapeutic antibodies in infectious CNS diseases.


Asunto(s)
Autoantígenos/inmunología , Esclerosis Múltiple/inmunología , Bandas Oligoclonales/inmunología , Borrelia/inmunología , Células HEK293 , Humanos , Neuroborreliosis de Lyme/inmunología
2.
Brain ; 137(Pt 10): 2703-14, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25060097

RESUMEN

Intrathecal oligoclonal bands of the cerebrospinal fluid are considered the most important immunological biomarkers of multiple sclerosis. They typically consist of clonally expanded IgG antibodies that underwent affinity maturation during sustained stimulation by largely unknown antigens. In addition, ∼40% of patients with multiple sclerosis have oligoclonal bands that consist of expanded IgM antibodies. We investigated the molecular composition of IgM- and IgG-chains from cerebrospinal fluid of 12 patients with multiple sclerosis, seven patients with other neurological diseases, and eight healthy control subjects by high-throughput deep-sequencing and single-cell PCR. Further, we studied the expression of activation-induced cytidine deaminase, the key enzyme for affinity maturation of antibodies, in cerebrospinal fluid samples of 16 patients. From the cerebrospinal fluid of two multiple sclerosis patients we isolated single B cells and investigated the co-expression of antibody chains with activation-induced cytidine deaminase. In striking contrast to IgM-chains from peripheral blood, IgM-chains from cerebrospinal fluid of patients with multiple sclerosis or neuroborreliosis showed a high degree of somatic hypermutation. We found a high content of mutations that caused amino acid exchanges as compared to silent mutations. In addition, more mutations were found in the complementarity determining regions of the IgM-chains, which interact with yet unknown antigens, as compared to framework regions. Both observations provide evidence for antigen-driven affinity maturation. Furthermore, single B cells from the cerebrospinal fluid of patients with multiple sclerosis co-expressed somatically hypermutated IgM-chains and activation-induced cytidine deaminase, an enzyme that is crucial for somatic hypermutation and class switch recombination of antibodies and is normally expressed during activation of B cells in germinal centres. Clonal tracking of particular IgM(+) B cells allowed us to relate unmutated ancestor clones in blood to hypermutated offspring clones in CSF. Unexpectedly, however, we found no evidence for intrathecal isotype switching from IgM to IgG. Our data suggest that the intrathecal milieu sustains a germinal centre-like reaction with clonal expansion and extensive accumulation of somatic hypermutation in IgM-producing B cells.


Asunto(s)
Inmunoglobulina M/líquido cefalorraquídeo , Inmunoglobulina M/genética , Inflamación/genética , Inflamación/inmunología , Esclerosis Múltiple/genética , Esclerosis Múltiple/inmunología , Hipermutación Somática de Inmunoglobulina/genética , Médula Espinal/inmunología , Médula Espinal/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Secuencia de Aminoácidos , Anticuerpos Monoclonales Humanizados/uso terapéutico , Linfocitos B/inmunología , Secuencia de Bases , Proliferación Celular , Femenino , Humanos , Inmunoglobulina G/líquido cefalorraquídeo , Inmunoglobulina G/genética , Masculino , Persona de Mediana Edad , Datos de Secuencia Molecular , Natalizumab , Análisis de la Célula Individual , Adulto Joven
3.
J Biol Chem ; 287(25): 20986-95, 2012 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-22549773

RESUMEN

In polymyositis and inclusion body myositis, muscle fibers are surrounded and invaded by CD8-positive cytotoxic T cells expressing the αß-T cell receptor (αß-TCR) for antigen. In a rare variant of myositis, muscle fibers are similarly attacked by CD8-negative T cells expressing the γδ-TCR (γδ-T cell-mediated myositis). We investigated the antigen specificity of a human γδ-TCR previously identified in an autoimmune tissue lesion of γδ-T cell-mediated myositis. We show that this Vγ1.3Vδ2-TCR, termed M88, recognizes various proteins from different species. Several of these proteins belong to the translational apparatus, including some bacterial and human aminoacyl-tRNA synthetases (AA-RS). Specifically, M88 recognizes histidyl-tRNA synthetase, an antigen known to be also targeted by autoantibodies called anti-Jo-1. The M88 target epitope is strictly conformational, independent of post-translational modification, and exposed on the surface of the respective antigenic protein. Extensive mutagenesis of the translation initiation factor-1 from Escherichia coli (EcIF1), which served as a paradigm antigen with known structure, showed that a short α-helical loop around amino acids 39 to 42 of EcIF1 is a major part of the M88 epitope. Mutagenesis of M88 showed that the complementarity determining regions 3 of both γδ-TCR chains contribute to antigen recognition. M88 is the only known example of a molecularly characterized γδ-TCR expressed by autoaggressive T cells in tissue. The observation that AA-RS are targeted by a γδ-T cell and by autoantibodies reveals an unexpected link between T cell and antibody responses in autoimmune myositis.


Asunto(s)
Enfermedades Autoinmunes/inmunología , Epítopos de Linfocito T/inmunología , Proteínas Musculares/inmunología , Polimiositis/inmunología , Receptores de Antígenos de Linfocitos T gamma-delta/inmunología , Linfocitos T/inmunología , Autoanticuerpos/genética , Autoanticuerpos/inmunología , Enfermedades Autoinmunes/genética , Epítopos de Linfocito T/genética , Escherichia coli/genética , Escherichia coli/inmunología , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/inmunología , Histidina-ARNt Ligasa/genética , Histidina-ARNt Ligasa/inmunología , Humanos , Proteínas Musculares/genética , Mutagénesis , Polimiositis/genética , Estructura Secundaria de Proteína , Receptores de Antígenos de Linfocitos T gamma-delta/genética
4.
Methods Mol Biol ; 2492: 333-352, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35733055

RESUMEN

The blood-brain barrier (BBB) is a highly specialized structure, constituted by endothelial cells that together with astrocytes and pericytes provide a functional interface between the central nervous system and the periphery. Several pathological conditions may affect its functions, and lately BBB involvement in the pathogenesis of Alzheimer's disease has been demonstrated. Both endothelial cells and astrocytes can be differentially affected during the course of the disease. In vitro BBB models present a powerful tool in evaluating the effects that ß-amyloid (Aß), or other pathogenic stimuli, play on the BBB at cellular level. In vitro BBB models derived from human cell sources are rare and not easily implemented. We generated two conditionally immortalized human cell lines, brain microvascular endothelial cells (TY10), and astrocytes (hAST), that, when co-cultured under appropriate conditions, exhibit BBB-like characteristics. This model allowed us to evaluate the transmigration of peripheral blood mononuclear cells (PBMCs) through the in vitro barrier exposed to Aß and the role played by astrocytes in the modulation of this phenomenon. We describe here the methodology used in our lab to set up our in vitro model of the BBB and to carry out a PBMC transmigration assay.


Asunto(s)
Enfermedad de Alzheimer , Barrera Hematoencefálica , Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Astrocitos/metabolismo , Barrera Hematoencefálica/metabolismo , Células Endoteliales/metabolismo , Humanos , Leucocitos Mononucleares/metabolismo
5.
Front Bioeng Biotechnol ; 8: 573775, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33117784

RESUMEN

We describe here the design and implementation of an in vitro microvascular open model system using human brain microvascular endothelial cells. The design has several advantages over other traditional closed microfluidic platforms: (1) it enables controlled unidirectional flow of media at physiological rates to support vascular function, (2) it allows for very small volumes which makes the device ideal for studies involving biotherapeutics, (3) it is amenable for multiple high resolution imaging modalities such as transmission electron microscopy (TEM), 3D live fluorescence imaging using traditional spinning disk confocal microscopy, and advanced lattice light sheet microscopy (LLSM). Importantly, we miniaturized the design, so it can fit within the physical constraints of LLSM, with the objective to study physiology in live cells at subcellular level. We validated barrier function of our brain microvessel-on-a-chip by measuring permeability of fluorescent dextran and a human monoclonal antibody. One potential application is to investigate mechanisms of transcytosis across the brain microvessel-like barrier of fluorescently-tagged biologics, viruses or nanoparticles.

6.
Fluids Barriers CNS ; 15(1): 23, 2018 Aug 31.
Artículo en Inglés | MEDLINE | ID: mdl-30165870

RESUMEN

BACKGROUND: Receptor-mediated transcytosis is one of the major routes for drug delivery of large molecules into the brain. The aim of this study was to develop a novel model of the human blood-brain barrier (BBB) in a high-throughput microfluidic device. This model can be used to assess passage of large biopharmaceuticals, such as therapeutic antibodies, across the BBB. METHODS: The model comprises human cell lines of brain endothelial cells, astrocytes, and pericytes in a two-lane or three-lane microfluidic platform that harbors 96 or 40 chips, respectively, in a 384-well plate format. In each chip, a perfused vessel of brain endothelial cells was grown against an extracellular matrix gel, which was patterned by means of surface tension techniques. Astrocytes and pericytes were added on the other side of the gel to complete the BBB on-a-chip model. Barrier function of the model was studied using fluorescent barrier integrity assays. To test antibody transcytosis, the lumen of the model's endothelial vessel was perfused with an anti-transferrin receptor antibody or with a control antibody. The levels of antibody that penetrated to the basal compartment were quantified using a mesoscale discovery assay. RESULTS: The perfused BBB on-a-chip model shows presence of adherens and tight junctions and severely limits the passage of a 20 kDa FITC-dextran dye. Penetration of the antibody targeting the human transferrin receptor (MEM-189) was markedly higher than penetration of the control antibody (apparent permeability of 2.9 × 10-5 versus 1.6 × 10-5 cm/min, respectively). CONCLUSIONS: We demonstrate successful integration of a human BBB microfluidic model in a high-throughput plate-based format that can be used for drug screening purposes. This in vitro model shows sufficient barrier function to study the passage of large molecules and is sensitive to differences in antibody penetration, which could support discovery and engineering of BBB-shuttle technologies.


Asunto(s)
Anticuerpos/metabolismo , Barrera Hematoencefálica/metabolismo , Dispositivos Laboratorio en un Chip , Astrocitos/metabolismo , Permeabilidad Capilar , Línea Celular , Evaluación Preclínica de Medicamentos/métodos , Células Endoteliales/metabolismo , Humanos , Técnicas Analíticas Microfluídicas , Microvasos/metabolismo , Modelos Neurológicos , Pericitos/metabolismo
7.
Neurol Neuroimmunol Neuroinflamm ; 4(1): e311, 2017 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-28018943

RESUMEN

OBJECTIVE: To address the hypothesis that physiologic interactions between astrocytes and endothelial cells (EC) at the blood-brain barrier (BBB) are afflicted by pathogenic inflammatory signaling when astrocytes are exposed to aquaporin-4 (AQP4) antibodies present in the immunoglobulin G (IgG) fraction of serum from patients with neuromyelitis optica (NMO), referred to as NMO-IgG. METHODS: We established static and flow-based in vitro BBB models incorporating co-cultures of conditionally immortalized human brain microvascular endothelial cells and human astrocyte cell lines with or without AQP4 expression. RESULTS: In astrocyte-EC co-cultures, exposure of astrocytes to NMO-IgG decreased barrier function, induced CCL2 and CXCL8 expression by EC, and promoted leukocyte migration under flow, contingent on astrocyte expression of AQP4. NMO-IgG selectively induced interleukin (IL)-6 production by AQP4-positive astrocytes. When EC were exposed to IL-6, we observed decreased barrier function, increased CCL2 and CXCL8 expression, and enhanced leukocyte transmigration under flow. These effects were reversed after application of IL-6 neutralizing antibody. CONCLUSIONS: Our results indicate that NMO-IgG induces IL-6 production by AQP4-positive astrocytes and that IL-6 signaling to EC decreases barrier function, increases chemokine production, and enhances leukocyte transmigration under flow.

8.
Sci Transl Med ; 9(397)2017 07 05.
Artículo en Inglés | MEDLINE | ID: mdl-28679661

RESUMEN

Neuromyelitis optica (NMO) is an inflammatory disorder mediated by antibodies to aquaporin-4 (AQP4) with prominent blood-brain barrier (BBB) breakdown in the acute phase of the disease. Anti-AQP4 antibodies are produced mainly in the periphery, yet they target the astrocyte perivascular end feet behind the BBB. We reasoned that an endothelial cell-targeted autoantibody might promote BBB transit of AQP4 antibodies and facilitate NMO attacks. Using monoclonal recombinant antibodies (rAbs) from patients with NMO, we identified two that strongly bound to the brain microvascular endothelial cells (BMECs). Exposure of BMECs to these rAbs resulted in nuclear translocation of nuclear factor κB p65, decreased claudin-5 protein expression, and enhanced transit of macromolecules. Unbiased membrane proteomics identified glucose-regulated protein 78 (GRP78) as the rAb target. Using immobilized GRP78 to deplete GRP78 antibodies from pooled total immunoglobulin G (IgG) of 50 NMO patients (NMO-IgG) reduced the biological effect of NMO-IgG on BMECs. GRP78 was expressed on the surface of murine BMECs in vivo, and repeated administration of a GRP78-specific rAb caused extravasation of serum albumin, IgG, and fibrinogen into mouse brains. Our results identify GRP78 antibodies as a potential component of NMO pathogenesis and GRP78 as a candidate target for promoting central nervous system transit of therapeutic antibodies.


Asunto(s)
Autoanticuerpos/metabolismo , Barrera Hematoencefálica/inmunología , Barrera Hematoencefálica/patología , Proteínas de Choque Térmico/inmunología , Neuromielitis Óptica/inmunología , Neuromielitis Óptica/patología , Adulto , Albúminas/metabolismo , Animales , Acuaporina 4/metabolismo , Membrana Celular/metabolismo , Chaperón BiP del Retículo Endoplásmico , Células Endoteliales/patología , Fibrinógeno/metabolismo , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Inmunoglobulina G/metabolismo , Ratones Endogámicos C57BL , Microvasos/patología , Neuromielitis Óptica/líquido cefalorraquídeo , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/aislamiento & purificación , Proteínas Recombinantes/farmacología
9.
Handb Clin Neurol ; 133: 39-59, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27112670

RESUMEN

In autoimmune neurologic disorders, the blood-brain barrier (BBB) plays a central role in immunopathogenesis, since this vascular interface is an entry path for cells and effector molecules of the peripheral immune system to reach the target organ, the central nervous system (CNS). The BBB's unique anatomic structure and the tightly regulated interplay of its cellular and acellular components allow for maintenance of brain homeostasis, regulation of influx and efflux, and protection from harm; these ensure an optimal environment for the neuronal network to function properly. In both health and disease, the BBB acts as mediator between the periphery and the CNS. For example, immune cell trafficking through the cerebral vasculature is essential to clear microbes or cell debris from neural tissues, while poorly regulated cellular transmigration can underlie or worsen CNS pathology. In this chapter, we focus on the specialized multicellular structure and function of the BBB/neurovascular unit and discuss how BBB breakdown can precede or be a consequence of neuroinflammation. We introduce the blood-cerebrospinal fluid barrier and include a brief aside about evolutionary aspects of barrier formation and refinements. Lastly, since restoration of barrier function is considered key to ameliorate neurologic disease, we speculate about new therapeutic avenues to repair a damaged BBB.


Asunto(s)
Barrera Hematoencefálica/fisiología , Neovascularización Fisiológica/fisiología , Animales , Transporte Biológico/fisiología , Barrera Hematoencefálica/anatomía & histología , Líquido Cefalorraquídeo/fisiología , Células Endoteliales/fisiología , Humanos , Neuroglía/fisiología , Neuronas/fisiología
10.
PLoS One ; 10(7): e0133392, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26197437

RESUMEN

The ability of the Blood Brain Barrier (BBB) to maintain proper barrier functions, keeping an optimal environment for central nervous system (CNS) activity and regulating leukocytes' access, can be affected in CNS diseases. Endothelial cells and astrocytes are the principal BBB cellular constituents and their interaction is essential to maintain its function. Both endothelial cells and astrocytes express the receptors for the bioactive sphingolipid S1P. Fingolimod, an immune modulatory drug whose structure is similar to S1P, has been approved for treatment in multiple sclerosis (MS): fingolimod reduces the rate of MS relapses by preventing leukocyte egress from the lymph nodes. Here, we examined the ability of S1P and fingolimod to act on the BBB, using an in vitro co-culture model that allowed us to investigate the effects of S1P on endothelial cells, astrocytes, and interactions between the two. Acting selectively on endothelial cells, S1P receptor signaling reduced cell death induced by inflammatory cytokines. When acting on astrocytes, fingolimod treatment induced the release of a factor, granulocyte macrophage colony-stimulating factor (GM-CSF) that reduced the effects of cytokines on endothelium. In an in vitro BBB model incorporating shear stress, S1P receptor modulation reduced leukocyte migration across the endothelial barrier, indicating a novel mechanism that might contribute to fingolimod efficacy in MS treatment.


Asunto(s)
Astrocitos/citología , Barrera Hematoencefálica/metabolismo , Células Endoteliales/citología , Lisofosfolípidos/metabolismo , Esclerosis Múltiple/inmunología , Receptores de Lisoesfingolípidos/metabolismo , Esfingosina/análogos & derivados , Adulto , Astrocitos/metabolismo , Movimiento Celular , Supervivencia Celular , Citocinas/metabolismo , Clorhidrato de Fingolimod/química , Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Voluntarios Sanos , Humanos , Inflamación , Leucocitos/citología , Microcirculación , Persona de Mediana Edad , Transducción de Señal , Esfingolípidos/química , Esfingosina/metabolismo , Estrés Mecánico , Adulto Joven
11.
J Neurosci Methods ; 232: 165-72, 2014 Jul 30.
Artículo en Inglés | MEDLINE | ID: mdl-24858797

RESUMEN

BACKGROUND: In vitro blood-brain barrier (BBB) models can be useful for understanding leukocyte-endothelial interactions at this unique vascular-tissue interface. Desirable features of such a model include shear stress, non-transformed cells and co-cultures of brain microvascular endothelial cells with astrocytes. Recovery of transmigrated leukocytes for further analysis is also appealing. NEW METHODS: We report an in vitro BBB model for leukocyte transmigration incorporating shear stress with co-culture of conditionally immortalized human endothelial cell line (hBMVEC) and human astrocyte cell line (hAST). Transmigrated leukocytes can be recovered for comparison with input and non-transmigrated cells. RESULT: hBMVEC and hAST exhibited physiological and morphological BBB properties when cocultured back-to-back on membranes. In particular, astrocyte processes protruded through 3 µm membrane pores, terminating in close proximity to the hBMVEC with a morphology reminiscent of end-feet. Co-culture with hAST also decreased the permeability of hBMVEC. In our model, astrocytes promoted transendothelial leukocyte transmigration. COMPARISON WITH EXISTING METHODS: This model offers the opportunity to evaluate whether BBB properties and leukocyte transmigration across cytokine-activated hBMVEC are influenced by human astrocytes. CONCLUSIONS: We present a BBB model for leukocyte transmigration incorporating shear stress with co-culture of hBMVEC and hAST. We demonstrate that hAST promoted leukocyte transmigration and also increased certain barrier functions of hBMVEC. This model provides reproducible assays for leukocyte transmigration with robust results, which will enable further defining the relationships among leukocytes and the cellular elements of the BBB.


Asunto(s)
Astrocitos/fisiología , Barrera Hematoencefálica/fisiología , Células Endoteliales/fisiología , Estrés Mecánico , Adulto , Transporte Biológico , Barrera Hematoencefálica/ultraestructura , Células Cultivadas , Claudina-5/metabolismo , Técnicas de Cocultivo , Células Endoteliales/ultraestructura , Femenino , Regulación de la Expresión Génica/fisiología , Proteína Ácida Fibrilar de la Glía/metabolismo , Humanos , Masculino , Microvasos/citología , Persona de Mediana Edad , Modelos Biológicos , Ocludina/metabolismo , Temperatura , Migración Transendotelial y Transepitelial , Adulto Joven , Proteína de la Zonula Occludens-1/metabolismo , Factor de von Willebrand/metabolismo
12.
Nat Med ; 19(12): 1584-96, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24309662

RESUMEN

The interface between the blood circulation and the neural tissue features unique characteristics that are encompassed by the term 'blood-brain barrier' (BBB). The main functions of this barrier, namely maintenance of brain homeostasis, regulation of influx and efflux transport, and protection from harm, are determined by its specialized multicellular structure. Every constituent cell type makes an indispensable contribution to the BBB's integrity. But if one member of the BBB fails, and as a result the barrier breaks down, there can be dramatic consequences and neuroinflammation and neurodegeneration can occur. In this Review, we highlight recently gained mechanistic insights into the development and maintenance of the BBB. We then discuss how BBB disruption can cause or contribute to neurological disease. Finally, we examine how this knowledge can be used to explore new possibilities for BBB repair.


Asunto(s)
Barrera Hematoencefálica/embriología , Barrera Hematoencefálica/lesiones , Barrera Hematoencefálica/fisiología , Enfermedades del Sistema Nervioso/etiología , Animales , Vasos Sanguíneos/embriología , Encéfalo/irrigación sanguínea , Encéfalo/embriología , Humanos , Neovascularización Fisiológica/fisiología , Enfermedades del Sistema Nervioso/terapia , Factor A de Crecimiento Endotelial Vascular/fisiología , Vía de Señalización Wnt/fisiología
13.
J Neuroimmunol ; 233(1-2): 245-8, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21353315

RESUMEN

We investigated the overlap shared between the immunoglobulin (Ig) proteome of the cerebrospinal fluid (CSF) and the B cell Ig-transcriptome of CSF and the central nervous system (CNS) tissue of three patients with multiple sclerosis. We determined the IgG-proteomes of CSF by mass spectrometry, and compared them to the IgG-transcriptomes from CSF and brain lesions, which were analyzed by cDNA cloning. Characteristic peptides that were identified in the CSF-proteome could also be detected in the transcriptomes of both, brain lesions and CSF, providing evidence for a strong overlap of the IgG repertoires in brain lesions and in the CSF.


Asunto(s)
Subgrupos de Linfocitos B/inmunología , Subgrupos de Linfocitos B/metabolismo , Inmunoglobulinas/biosíntesis , Inmunoglobulinas/líquido cefalorraquídeo , Esclerosis Múltiple/líquido cefalorraquídeo , Esclerosis Múltiple/inmunología , Subgrupos de Linfocitos B/patología , Células Clonales/inmunología , Células Clonales/metabolismo , Células Clonales/patología , Humanos , Esclerosis Múltiple/genética
14.
Nat Med ; 14(6): 688-93, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18488038

RESUMEN

We describe a method for correlating the immunoglobulin (Ig) proteomes with the B cell transcriptomes in human fluid and tissue samples, using multiple sclerosis as a paradigm. Oligoclonal Ig bands and elevated numbers of clonally expanded B cells in the cerebrospinal fluid (CSF) are diagnostic hallmarks of multiple sclerosis. Here we compared the Ig transcriptomes of B cells with the corresponding Ig proteomes in CSF samples from four subjects with multiple sclerosis. We created individual Ig transcriptome databases that contained the subject-specific mutations introduced by V(D)J recombination and somatic hypermutation and then searched the CSF for corresponding characteristic peptides by mass spectrometry. In each sample, the Ig transcriptomes and proteomes strongly overlapped, showing that CSF B cells indeed produce the oligoclonal Ig bands. This approach can be applied to other organ-specific diagnostic fluid or tissue samples to compare the Ig transcripts of local B cells with the corresponding antibody proteomes of individual subjects.


Asunto(s)
Proteínas del Líquido Cefalorraquídeo/inmunología , Esclerosis Múltiple/líquido cefalorraquídeo , Bandas Oligoclonales/genética , Proteoma/análisis , Transcripción Genética , Secuencia de Aminoácidos , Linfocitos B/inmunología , Proteínas del Líquido Cefalorraquídeo/genética , Bases de Datos Genéticas , Geles , Humanos , Cadenas Pesadas de Inmunoglobulina/genética , Cadenas Ligeras de Inmunoglobulina/genética , Región Variable de Inmunoglobulina/genética , Focalización Isoeléctrica , Espectrometría de Masas , Datos de Secuencia Molecular , Esclerosis Múltiple/inmunología , Bandas Oligoclonales/inmunología , Homología de Secuencia de Aminoácido
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA